2 December 2016

Dendritic Cells in the Immune System—History, Lineages, Tissues, Tolerance, and Immunity

ABSTRACT

The aim of this review is to provide a coherent framework for understanding dendritic cells (DCs). It has seven sections. The introduction provides an overview of the immune system and essential concepts, particularly for the nonspecialist reader. Next, the “History” section outlines the early evolution of ideas about DCs and highlights some sources of confusion that still exist today. The “Lineages” section then focuses on five different populations of DCs: two subsets of “classical” DCs, plasmacytoid DCs, monocyte-derived DCs, and Langerhans cells. It highlights some cellular and molecular specializations of each, and also notes other DC subsets that have been proposed. The following “Tissues” section discusses the distribution and behavior of different DC subsets within nonlymphoid and secondary lymphoid tissues that are connected by DC migration pathways between them. In the “Tolerance” section, the role of DCs in central and peripheral tolerance is considered, including their ability to drive the differentiation of different populations of regulatory T cells. In contrast, the “Immunity” section considers the roles of DCs in sensing of infection and tissue damage, the initiation of primary responses, the T-cell effector phase, and the induction of immunological memory. The concluding section provides some speculative ideas about the evolution of DCs. It also revisits earlier concepts of generation of diversity and clonal selection in terms of DCs driving the evolution of T-cell responses. Throughout, this review highlights certain areas of uncertainty and suggests some avenues for future investigation.
Ralph Steinman was posthumously awarded a Nobel Prize in 2011 “for his discovery of the dendritic cell and its role in adaptive immunity.” He first coined the term “dendritic cell” in 1973 while working with Zanvil Cohn. His findings laid the foundations for a new field of immunology. This has enormous therapeutic potential for new vaccination strategies against cancer and infectious diseases. It also holds promise for new therapies for autoimmune disease, allergy, and transplantation reactions.
This review seeks to provide a conceptual framework for understanding dendritic cells (DCs). It focuses mainly on DCs of mice, and to a lesser extent humans, although a menagerie of other creatures will be mentioned. It deals mainly with DCs and “conventional” T cells in the adaptive immune system, but other, specialized “innate-like” T cells will be noted. A number of recent reviews (cited later) contain much additional information, including many molecular details not covered here.
The introduction to this review provides essential background information on the immune system and mechanisms of tolerance and immunity, particularly for nonspecialist readers. Further general information can be found in standard texts (e.g., references 14). With this background, the remaining sections may be read selectively or consecutively. To aid cross-referencing, the five central sections are cited in brief as “History,” “Lineages,” “Tissues,” “Tolerance,” and “Immunity.” The review ends with a discussion and conclusion.

INTRODUCTION

A crucial function of the immune system is to provide defense against infection, or immunity. It is conventionally divided into a phylogenetically ancient innate immune system, which exists in all animals, and an adaptive immune system, which evolved later in jawed vertebrates (gnathostomes), including mice and humans. The immune system has been described as “the most efficient killing machine ever created.” It is therefore essential that it does not attack its host and maintains a state of unresponsiveness, or “tolerance,” to its own tissues. In addition, it must establish a state of mutualism with the beneficial populations of commensal organisms that populate the skin and tracts of mucosal tissues, and which have helped shape the evolution of the immunity system. DCs appear to have evolved around the time that adaptive immunity “suddenly” appeared. They play a fundamental role in linking the innate and adaptive immune systems together so they can work together synergistically. DCs are crucially important in tolerance and immunity.

Infection and Immunity

All living things, both plants and animals, must defend themselves against infection. The types of infectious agent that would infect and inhabit larger eukaryotic animals are the viruses; the prokaryotic bacteria; and the eukaryotic groups of fungi, single-celled protozoa, and multicellular helminths. To infect, they must cross the defensive barriers provided by host epithelia, including the external surfaces of the skin and the internal (but topologically external) mucosal surfaces. These preexisting barriers to infection have their own forms of constitutive defense, provided, for example, by antiviral and antimicrobial proteins such as defensins and cathelicidins. Having gained access to the internal milieu of the host, in order to survive and replicate, some infectious agents invade intracellular niches (typically viruses, intracellular bacteria, and protozoa), while others adopt an extracellular habit (commonly pyogenic bacteria and other protozoa). Other organisms survive and replicate either within or outside of the epithelial barriers, as exemplified by fungi colonizing the skin or helminths cohabiting within the mucosal tracts of the intestine. Hence, to survive, any multicellular animal must be able to eliminate or at least control pathogens from any class of infectious agent. In the case of humans, these range in size over 8 or more orders of magnitude from the smallest viruses to longest tapeworms, can be located both within and outside of host cells, and may have enormously complex and transformative life cycles (the malarial parasite, for example).

Innate Immunity

All plants can defend themselves against infection, although they do not possess an immune system as such. The innate immune system emerged with the dawning of the animal kingdom and particularly with the evolution of the first multicellular organisms. Innate immunity provides the earliest molecular and cellular, and often system-wide, mechanisms of defense that are rapidly triggered by infection. For example, phagocytes such as macrophages can capture and internalize many bacteria, which are then degraded. The soluble complement system can also rapidly be activated to coat microbes and help macrophages do this. Fundamentally, innate responses have several crucial functions. First, they may rapidly and efficiently eliminate infectious agents. In general, immunological mechanisms that effect, or bring about, the elimination or control of infection are termed effector mechanisms. Second, early innate responses typically cause local inflammation. This enables yet more molecular and cellular effectors to be recruited from the blood into local sites of infection, further amplifying their efficacy. Third, innate responses can lead to later system-wide modulations of organ function. These result, for example, in fever and “sickness behavior” through actions on the brain, and the so-called acute-phase response produced largely by the liver. In the latter case, extraordinarily high quantities of new molecules are pumped into the bloodstream. Many of these seem to be opsonins, which coat infectious agents, bind to complementary receptors on cells such as macrophages, and promote uptake and clearance. Finally, innate responses are absolutely essential for the subsequent initiation and regulation of adaptive immune responses (see below).
So-called pattern recognition receptors (PRRs) are essential for innate immunity. These cellular receptors enable different types of infectious agents to be sensed with a high degree of accuracy. This is because PRRs can recognize highly conserved molecular motifs on which infectious agents depend for their integrity, replication, and survival. These components, which cannot be synthesized by the host, are generally known as pathogen-associated molecular patterns (PAMPs). The PRRs also facilitate precise discrimination between classes of infectious agents, for example, by recognition of lipopolysaccharide as a component of Gram-negative bacterial cell walls, absent in Gram-positive species. Similar molecules also exist in soluble form and are sometimes termed pattern recognition molecules. They include molecules such as C1q and mannose-binding lectin of the complement system. These can bind directly to bacterial surfaces or to opsonic molecules attached to them, and trigger the so-called classical and lectin pathways, which both result in complement activation. Arguably, PRRs enable the innate immune system to discriminate perfectly between infectious nonself and healthy self.
Some PRRs, such as scavenger receptors and members of the C-type lectin receptor family, can promote the uptake and internalization of infectious agents through receptor-mediated endocytosis or phagocytosis, by macrophages, for example. Other PRRs, however, trigger rapid secretion of proinflammatory cytokines and/or type I interferons (IFN-I), which contribute to rapid initiation of inflammatory and antiviral responses, respectively. These PRRs include the Toll-like receptors (TLRs), associated with the plasma membrane and endocytic compartments of cells such as macrophages, and the RIG-I (retinoic acid-inducible gene I)-like and NOD (nucleotide oligomerization domain)-like receptors in the cytosol. PRRs are also known to sense cell or tissue damage and death, whether or not it results from infection. This is through their capacity to recognize so-called damage-associated molecular patterns (DAMPs). One example is extracellular ATP, which in fact acts as a DAMP in animals as well as plants. Hence, through such mechanisms, innate immunity can rapidly sense and discriminate between different types of “danger” that may be presented to the host and trigger the responses needed for defense and restoration of homeostasis.
The rapid initiation of innate responses critically depends on the different types of cells that are present at sites of infection, all of which express PRRs. These include epithelial cells of tissues such as skin and the mucosae (the gut, lung and airways, and urogenital tract). Under physiological conditions, most tissues also contain resident populations of macrophages and mast cells and, in many, recently described innate lymphoid cells (ILCs). Collectively, it may be convenient to think of these as “alarm cells” that sense danger and rapidly sound “alarm signals” that are provided by molecular alarmins. The latter include the proinflammatory cytokines and lipid mediators that induce local inflammation. Subsequently, large numbers of reinforcements are recruited from the blood to help fight the danger and repair the damage. These include the granulocytes—neutrophils, or basophils and eosinophils—and later monocytes, as well as complement components and “natural” antibodies.
The abundant cellular expression of multiple types of different PRRs that are “hard-wired” to induce rapid cellular responses, as well as soluble forms that can be quickly activated, ensures that innate responses to infection are extremely fast, generally large, and often (though not always) highly effective. DCs are also present in tissues. They too can detect infection, tissue damage, and cell death through their own expression of PRRs and by sensing of alarmins, and also contribute to innate responses. However, they also have pivotal functions in the adaptive immune system, in the relatively few species that have one.

Adaptive Immunity

The vast majority of animal species on the planet survive and continue to evolve with the innate immune system as their only means of defense against infection. And yet, around the time of the Cambrian explosion some 540 million years ago, the adaptive immune system evolved within a remarkably short period of geological time (5). This provides immunity that is based on two main types of lymphocytes, T cells and antibody-producing B cells. Each has its own specialized type of receptor that enables it to recognize components of infectious agents or antigens: T-cell receptors (TCRs), and B-cell receptors (BCRs) which can be secreted as antibodies. This new type of immunity, as recognized in humans, probably first evolved in jawed fish. Through the millennia, with the emergence of all other vertebrates, adaptive immunity has coopted the mechanisms of innate immunity, with which it coexists, for its own purposes. Recent evidence indicates that an alternative form of adaptive immunity originated even earlier in jawless fish (agnathans), such as lampreys and hagfish, but is apparently an evolutionary dead end. In this review, the term “adaptive immunity” will be used only for the type that exists in jawed vertebrates (gnathostomes).
Precisely why the adaptive immune system “suddenly” appeared remains an almost complete mystery. However, according to the 2R hypothesis, it may have been facilitated by two entire genome replications before or around the time of the Cambrian explosion; even a third round may have occurred in some bony fish. If so, it would have enabled the different classes of antigen receptors (BCRs and TCRs) to evolve on paralogous chromosomes from an ancestral gene locus. This form of immunity probably also depended on the incorporation into the genome of a transposon, perhaps from a virus, that encodes the recombinase-activating gene (RAG). (There are in fact two RAG genes in gnathostomes, but only one in agnathans.) RAG enables a remarkable genetic mechanism known as somatic recombination to operate in developing T cells and B cells (but in no other cell type). This generates the extremely diverse sets of TCRs and BCRs. These lymphocyte antigen receptors are clonally distributed such that, at any one time, all those expressed by any T cell or B cell are essentially the same. They are in fact encoded by germ line gene segments that can be joined by somatic recombination to form functional genes. Together with additional mechanisms of diversification, a quite astonishing total number of receptors could be generated from extremely small portions of the genome that would otherwise require billions or trillions more DNA.
T cells and B cells, and their respective receptors, have very different but complementary functions in adaptive immunity. BCRs recognize small portions (epitopes) of extracellular molecules that typically retain their native, three-dimensional conformations. Hence, antibodies can bind directly to antigens such as viruses and bacteria before they infect cells. In contrast, so-called conventional T cells express αβ TCRs. In general, αβ TCRs can recognize intracellular antigens such as viruses and bacteria that have infected or been internalized by host cells. In actual fact, they recognize peptides derived from antigens inside cells that have been bound to classical major histocompatibility complex (MHC) molecules, which enable their transport from intracellular locations up to the cell surface. Thus, αβ TCRs recognize peptide-MHC complexes but not free peptides alone, and are often said to be MHC restricted. Classical MHC molecules are expressed by host cells so that they can be continuously surveyed by T cells and recognized if they have become infected by infectious agents or otherwise contain them. Conventionally, host cells are said to present their antigens to T cells and are hence termed antigen-presenting cells (APCs); the process as a whole is known as antigen presentation. In this review, the terms “conventional” and “classical” (see above) will normally be omitted, but they are used here since, for example, some “unconventional” T cells express γδ TCRs instead and some of these can recognize other, “nonclassical” MHC molecules.
Overall, antigen recognition by lymphocytes is anticipatory in nature, since vast repertoires of antigen receptors are generated in any individual even before any infection has occurred. It is also highly discriminatory, since even the slightest molecular change in an epitope may abolish recognition. It is, however, highly promiscuous, in that any given receptor has the potential to cross-react with perhaps millions of different epitopes from unrelated antigens that just happen to have the same shape (6). (The same is actually also true of MHC molecules binding peptides.) Functionally, what this means is that there is an extremely high chance that the adaptive immune system will be able to recognize an antigen from any given infectious agent it may encounter in the future, but the frequency of lymphocytes that can recognize any given epitope is extremely low. Hence, in any adaptive immune response, only these extremely rare clones of lymphocytes that recognize antigen can participate. Originating from a hypothesis put forward decades ago, this is known as clonal selection (by antigen). These few clones first need to be activated, since naive (antigen-inexperienced) lymphocytes are resting and impotent. They also need to be greatly expanded in numbers through multiple rounds of proliferation (or clonal expansion) before they can reach a critical mass to function effectively. In addition, lymphocytes are in fact highly plastic cells that can generate different types of effector responses (see below) but need to be “instructed” as to which type they should produce, as well as where they should go. All this takes time. For these reasons, the first, primary adaptive response produced against any infectious agent appears to be extremely slow. As measured, for example, by the frequency of antigen-specific T cells and antibodies, it may take days to reach a peak, in contrast to innate responses measured in minutes or hours. These responses are, however, much faster if the same infection occurs again, because the adaptive immune system can generate an immunological memory of what it has previously encountered (see below). DCs have essential roles in all stages of adaptive immune responses, and we now know they also enable these responses to be initiated much more quickly than it might seem.

Evolution of lymphoid tissues

The evolution of adaptive immunity necessitated the coevolution of specialized lymphoid organs and tissues for generating lymphocytes and regulating their responses (5). These are conventionally divided into primary lymphoid organs, where lymphocytes develop, and secondary lymphoid organs (SLOs; this abbreviation also encompasses secondary lymphoid tissues), where the fully developed lymphocytes can be activated and regulated during the adaptive immune response. The former include the thymus and sometimes the bone marrow. The latter include the spleen and lymph nodes, together with specialized mucosa-associated lymphoid tissue (MALT) such as Peyer’s patches in the small intestine. All jawed vertebrates have a thymus, and a related structure known as the thymoid appears to exist even in earlier agnathans. In some species, there are additional specialized primary organs where B cells are generated. In humans and mice, B cells undergo most of their development in bone marrow, but in chickens they develop in the bursa of Fabricius, which is where the term “B” cell originated. All gnathostomes also have a spleen, which has been retained throughout evolution from jawed fish to humans. Lymph nodes evolved much later and perhaps first originated in some (but not all) species of birds, though related aggregates of lymphoid tissue are present in reptiles and amphibians. However, Peyer’s patches are absent in birds and appear to have evolved even more recently. Hence, while adaptive immunity arose within a very short period of geological time, the different lymphoid tissues have continued to evolve in both structure and most probably function. Notably, with time, lymphoid tissues also became increasingly organized. Typically, T cells became segregated in distinct T-cell zones separate from the B cells in follicles (and, in birds and mammals, within germinal centers, which can develop from them). DCs have been identified in primary lymphoid organs and SLOs of all species of jawed vertebrates that have been closely studied. This suggests that they have played important roles in adaptive immunity throughout its evolution.

Immunological tolerance

In innate immunity, the expression of PRRs that can recognize PAMPs and DAMPs enables precise discrimination between infectious nonself and normal self components. This situation is, however, completely different in adaptive immunity. Because the somatic recombination of antigen receptor gene segments during BCR and TCR generation is an inherently random process, any newly formed receptor might potentially be able to recognize a self-antigen. Therefore, during lymphocyte development, cells with newly created autoreactive antigen receptors that recognize self-antigens with a high affinity are typically deleted from the repertoire. Because this process occurs within the primary, or central, lymphoid organs (i.e., the thymus and bone marrow in mice and humans), it contributes to what is known as central tolerance. It is, however, not perfect. Hence, there are additional mechanisms that can suppress the function of lymphocytes with autoreactive receptors if they escape from the thymus and enter peripheral SLOs and nonlymphoid tissues (NLTs). This can involve deletion of the autoreactive cells or the imposition of a state of unresponsiveness, or anergy, prior to their removal. Importantly, however, specialized populations of T cells, known as regulatory T cells (Tregs), can be generated that actively suppress potentially damaging autoreactive responses in the periphery. Some are generated within the thymus itself and are known as natural or thymically induced Tregs (tTregs). Others are generated within peripheral tissues and may also be produced during adaptive responses. These are collectively known as induced or peripherally induced Tregs (pTregs). DCs may play crucial roles in both central tolerance and the induction and maintenance of peripheral tolerance.

T-cell responses

Innate immune responses are absolutely essential for the subsequent induction of adaptive immune responses that are mediated by populations of αβ T cells and conventional B cells. This is because antigen-specific lymphocytes that recognize their cognate epitopes need to be instructed as to its “context,” in other words, whether the source of that antigen is “dangerous” or not. If it is, they can be stimulated to mount a response, while if not, they may develop into Tregs, become unresponsive, or be killed. In the case of T cells, DCs are essential for helping to place the antigen in its context. Importantly, this is because if DCs sense danger themselves, they induce the expression of specialized costimulatory molecules that help to trigger T-cell activation; if not, they don’t. Hence, generally speaking, two different sets of signals need to be delivered to a T cell before it can respond: the first is delivered through the TCR, which recognizes its cognate peptide-MHC complex (derived from the antigen the DC has acquired); the second is delivered through costimulation (provided by the DC if it has sensed danger). A similar situation actually also applies to many B-cell responses. These can be greatly increased if the antigen they recognize is bound to the specialized C3d component, which can be generated by the complement system after it is activated during innate responses. The requirement for two signals for lymphocyte activation ensures that provided there is no danger, and hence no innate response, recognition of any self component in normal tissues helps to reinforce peripheral tolerance (see above).
While all conventional T cells express αβ TCRs, they can contribute to fundamentally different types of adaptive responses. There are two types of T cells, defined by expression of CD8 or CD4; two types of classical MHC molecules, class I and class II; and two main sources of peptides that can be loaded onto the latter, from intracellular and extracellular antigens, respectively. In general, CD8+ T cells recognize peptide–MHC-I complexes after peptides have been generated in the cytosol from “endogenous” antigens, such as viruses that have infected cells. These cells can develop into cytotoxic T lymphocytes (CTLs), which can kill the cells they recognize. Because essentially all cells can express MHC-I molecules, CD8+ T cells can therefore help to eliminate any host cell that has been infected. In contrast, CD4+ T cells recognize peptide–MHC-II complexes after the peptides have been generated within endosomal compartments from “exogenous” antigens, such as bacteria that were phagocytosed. These cells can develop into effector cells and acquire the capacity to secrete hormone-like cytokines and deliver other molecular signals that stimulate the effector functions of the cell that they recognize (in other cases they function as Tregs, as noted above). The constitutive expression of MHC-II is restricted to a very few specialized cell types, such as DCs and B cells, although expression can be induced in others, such as epithelial cells, during adaptive responses. In general, therefore, these two types of T-cell responses are quite segregated. An exception to this rule is cross-presentation, whereby peptides from exogenous antigens can be rerouted for loading onto MHC-I molecules. Roughly speaking, this enables specialized populations of DCs, for example, to initiate CD8+ T-cell responses even if they have not been infected. Conversely, peptides from endogenous antigens may be rerouted onto MHC-II molecules during the cellular process of autophagy, when a cell digests its own cytoplasmic organelles.
In general, so-called helper CD4+ T-cell responses are absolutely essential for subsequent induction of other types of adaptive responses. For example, they are generally required to activate conventional B cells, which may then develop into antibody-secreting plasma cells. In this way CD4+ T cells can help to stimulate robust antibody responses of different types, typically against protein antigens from infectious agents. Generally speaking, in mice and humans, these antibodies are usually IgG, IgA, or IgE for systemic, mucosal, or barrier defenses, respectively. (IgM is the first type of antibody that is secreted in all T-dependent [TD] responses, but IgD usually remains membrane bound, though it can also be secreted for defense.) In many cases, helper T cells are also essential for activation of CD8+ T cells and their subsequent development into CTLs that can kill infected cells. These types of response are therefore termed helper-dependent or TD responses. (This is to distinguish them from T-independent responses that can also be produced by other specialized populations of innate-like B cells, not considered here.)
A central paradigm in current immunology is that CD4+ T cells can adopt a relatively small number of characteristic polarized types of response in different infectious settings. The best known of these are mediated by so-called Th17, Th1, and Th2 cells, often referred to as CD4+ T-cell subsets. For example, many infections caused by extracellular bacteria and fungi can induce Th17 responses, which recruit neutrophils to the site of infection and may stimulate the production of IgA by plasma cells. Alternatively, many viruses, intracellular bacteria, and protozoa more typically induce Th1 responses, which can activate potent antimicrobial mechanisms of macrophages and induce specific types of IgG response. In contrast, helminths (and perhaps hematogenous insects and arachnids biting the skin) usually elicit Th2 responses, which can recruit basophils and eosinophils to the site of infection, elicit IgE responses, and help to maintain or restore barrier defenses. In each setting, the particular combinations of effector mechanisms that are induced can then synergize to bring about a coordinated and integrated response that is best suited to eliminate or control the infectious agent in question.

Immunological memory

During primary T-cell responses, effector cells are generated and many of these migrate from SLOs to sites of infection within NLTs. Here the effector CD4+ T cells and CD8+ CTLs can contribute to the clearance or control of infection in different ways, noted above. However, during primary responses, memory T cells can also be generated, which remain long after the antigen has been eliminated and the bulk of effector cells that were generated have been cleared. The same is true for B-cell responses, which can also generate memory B cells. These antigen-specific clones of memory cells persist in much higher numbers than were present in the naive cell repertoires, and are more easily and rapidly reactivated. Hence, they enable the adaptive immune system to respond very rapidly to subsequent (secondary, tertiary, etc.) encounters with the same antigen, so much so that an infectious agent can be rapidly eliminated without causing any disease. Memory cells can provide protection against any pathogen to which they were initially induced and are long-lived cells. Hence, they provide immunological memory that can often last for a lifetime. (Long-lived plasma cells can also secrete protective antibodies for considerable periods of time.)
Immunological memory is a unique and specialized feature of the adaptive immune system and does not apply to innate immunity. However, other types of immune memory exist in even the most ancient animals, such as sponges, which lack an adaptive immune system. More recently the term “innate memory” has also been introduced to describe the phenomenon in which innate cells, such as macrophages, for example, may respond faster or differently against the same class of PAMPs to which they were initially exposed. However, this form of memory is not clonal and arguably might be better termed “innate imprinting.” In this review, the term “immunological memory” will be used exclusively within the context of adaptive immune responses.
DCs may have the specialized capacity to trigger T-cell activation, over and above any other cell type, and hence are essential for initiating T-cell and TD responses. They also contribute directly to the effector functions of T cells by helping to polarize their responses. In addition, they may also play central roles in memory responses, not least because they may reactivate memory T cells if the antigen is encountered again. At a species level, natural selection has driven the evolution and diversification of the adaptive immune system. In turn, DCs may drive the evolution and diversification of adaptive responses required for tolerance and immunity.

DCs IN HISTORY

This section provides a brief historical introduction to the evolution of early ideas about DCs. It also highlights some sources of confusion that persist even today. To put this into some sort of context, Ralph Steinman’s first paper on DCs was published in 1973 (7). Earlier studies had indicated that an “accessory cell” was required to induce T-cell and B-cell responses, and that APCs were somehow required to “process and present” antigens in a form that could be recognized by T cells. Then, in 1974, Zinkernagel and Doherty (8) first described the phenomenon of MHC restriction, for which they later received a Nobel Prize. This phenomenon is now understood as being due to the requirement for αβ TCRs to recognize peptide-MHC complexes. However, it was not until a decade or more later that the structure of the TCR and the gene segments that encoded it became known, and precisely how peptides bound to MHC molecules was understood. All of this was completely unknown in 1973. Moreover, at that time, it seemed most likely that macrophages were essential accessory cells and APCs for T-cell responses.

Lymphoid DCs and Interdigitating Cells

The original description (7) of a Steinman-Cohn DC was of a morphologically distinct, trace cell type that was noted among adherent cells cultured from mouse SLOs, such as spleen, lymph nodes, and Peyer’s patches of the intestine. These cells, as originally defined, continually extended and contracted spiny cell extensions in culture that were termed “dendrites.” They did not phagocytose particulate tracers, in contrast to the majority population of relatively sessile macrophages, which were avid phagocytes. Subsequently, DCs were discovered to be the most potent stimulators of T cell and T-cell-mediated immune responses, compared to other cell types tested (913). They seemed to have a unique capacity to initiate T-cell activation, thus acting as accessory cells. Moreover, through elaboration of cytokines such as interleukin-12 (IL-12), it seemed likely that they could also regulate the subsequent type of immunity that was induced (13). Hence, the original definition of a DC was a distinct cell type that could be isolated from SLOs and which was nonphagocytic but highly immunostimulatory.
Soon after their description, it was suggested that isolated DCs might be counterparts of the interdigitating cells. These had previously been identified within sections of lymphoid tissues, and particularly in the T-cell-rich zones of spleen and lymph nodes (1416). It was only later realized that these cells were mostly liberated from lymphoid tissues after enzymatic digestion, for example, with collagenase. Hence, interdigitating cells appeared to be relatively fixed within the T-cell areas, in contrast to the apparently more free DCs first studied. These likely represented the nests of DCs that were later noted in more peripheral regions of tissues such as the marginal zones of spleen (17).
The development of monoclonal antibodies (MAbs) was published just 2 years after the initial description of DCs, and also rewarded with a Nobel Prize (18). This was crucial for further exploration of the phenotype and function of DCs in culture, and of their possible relationship to other cell types. It was soon appreciated that both isolated DCs and interdigitating cells expressed MHC-II molecules, and they were later found to express costimulatory molecules, thus helping to explain their capacity to initiate T-cell responses. Newly developed MAbs also showed that both cell types also expressed CD11c (part of the complement receptor type 4). However, they could be distinguished phenotypically since the majority of DCs that were readily liberated expressed 33D1 (the first MAb generated against DCs, later shown to recognize DCIR2 [DC inhibitory receptor 2]), whereas interdigitating cells expressed CD205 (a C-type lectin receptor) (17, 19).
The more general term “lymphoid DC” was often adopted for presumptive DCs isolated from or localized within SLOs. These also included cells within the medulla of the thymus, which, after isolation, were also found to be nonphagocytic but to have immunostimulatory function in culture (20, 21). However, early evidence suggested that these thymic DCs might be lymphoid in origin, deriving from the common lymphoid progenitor (CLP) rather than common myeloid progenitor (CMP) downstream of the hematopoietic stem cell. Matters were further complicated by the later finding that some DCs expressed the lymphoid markers CD4 (a subset of DCIR2+ cells) or CD8 (generally interdigitating cells). Hence, the term “lymphoid” was used rather uneasily to reflect either or both a lymphoid tissue and/or a hematopoietic lymphoid origin.

Langerhans Cells and Interstitial DCs

Attention then turned to presumptive DCs in NLTs (this abbreviation also including “nonlymphoid organs”). The first detailed studies were of Langerhans cells (LCs) isolated from the epidermis of mouse skin (22, 23). These cells have a dendritic morphology within the tissue and form a dense network of cells in the suprabasal region of the epidermis. They also contain a characteristic organelle known as the Birbeck granule. After isolation from the skin, LCs were found to be weak or inactive as stimulators of T-cell responses in vitro. They were also shown to be phagocytic (24). During culture, however, the cells acquired potent immunostimulatory activity accompanied by profound phenotypic and functional changes, including increased expression of MHC-II molecules but loss of Birbeck granules and a markedly reduced capacity for phagocytosis. They thus came to resemble Steinman-Cohn DCs.
Working around the same time, other investigators had been studying a trace cell type that expressed MHC-II molecules in NLTs such as heart and kidney (25, 26). These cells, which became known as interstitial DCs, were identified in the connective tissue of all NLTs studied, except for the central nervous system. The impetus for such studies was the suggestion, made as early as 1957, that transplant rejection was initiated by a small number of “passenger leukocytes” that were carried over in the graft, while the bulk of the transplanted material was in fact nonimmunogenic. Consistent with this idea, when interstitial cells were isolated from vascularized organs such as heart and kidney, it was found that their phenotypes and functions changed during culture similarly to what had been observed for LCs (27).
The term “myeloid DC” was introduced by some workers to discriminate between the presumptive DCs that were isolated from or identified within NLTs and the lymphoid DCs and interdigitating cells noted above (28). The use of this term probably also originated from observations that DCs in NLTs, as well as myeloid cells such as monocytes and macrophages, expressed Fc receptors and CD11b. These cells did not, however, express CD4 or CD8, and later evidence indicated that they probably originated from myeloid progenitors downstream of the CMP. Unfortunately, the focus on shared myeloid markers to characterize DC populations has bedeviled the field and caused further confusion that, to some extent, has still not been fully resolved.

Afferent Lymph Veiled Cells and Blood DCs

Because of the developments above, the concept of a DC needed to encompass both a phagocytic, nonimmunostimulatory cell in NLTs as well as the Steinman-Cohn DC isolated from lymphoid tissues. Moreover, it appeared likely that one could develop into the other. This transition was termed “maturation,” since cells in NLTs were considered to be relatively immature compared to mature cells in lymphoid tissues that had immunostimulatory activity. The concept of a DC also needed to be revised to encompass a migratory stage, as the cells apparently moved from peripheral tissues into SLOs, for example, via afferent lymph. In larger animals, it is possible to cannulate afferent lymphatics and to study migrating cells directly, but this is extremely difficult in smaller animals such as rodents. However, by surgically removing regional lymph nodes and allowing time for the lymphatics to reanastomose, one can obtain cells that would otherwise travel in afferent lymph after cannulation of the thoracic duct; this source is termed “pseudoafferent lymph” (29).
In culture, cells obtained from lymph or pseudoafferent lymph were found to have characteristic lamellipodiae, or “veils,” and became known as veiled cells (16). These cells were found to be traveling in lymph derived from tissues such as gut, liver, and skin in a continuous manner (30, 31). In the latter case they resembled LCs since they contained Birbeck granules, and similar cells were also detected in regional lymph nodes (32, 33). (An early pioneer, Brigitte Balfour, who was related to a former British prime minister, cannulated the lymphatics of her own leg in order to obtain veiled cells, and made vast numbers of cinematographic recordings of their movements in culture that now fill a back room somewhere in London [S. C. Knight, personal communication]). Veiled cells were also found to stimulate T-cell responses in culture (30, 34). The concept that veiled cells represent an intermediate stage of DC trafficking from NLTs into SLOs was reinforced by other studies. For example, studies of skin explants and transplants enabled the direct visualization of presumptive LCs leaving the epidermis and migrating through dermal lymphatics out of the skin (35, 36). Moreover, while the numbers of such cells in lymph were generally quite low, the flux of these cells increased massively after infection or inflammation in the site from which they originated. This suggested that they may have important roles in generating immune responses within SLOs.
DCs had also been identified in human blood, although it was not clear if these might be en route to or migrating from NLTs (37). In early studies, the capacity of DCs to migrate via the blood into the spleen, where they homed to areas rich in T cells, was demonstrated after adoptive transfer of labeled DCs in mice (38, 39). Moreover, DCs were observed to disappear from cardiac tissue after mouse heart transplantation, and donor cells were then detected in recipient spleen, where they were visualized often in close association with recipient T cells (40). Stimuli promoting DC maturation and migration also started to be identified. These included proinflammatory cytokines such as IL-1 and tumor necrosis factor α (TNF-α) (41, 42) and microbial products such as bacterial lipopolysaccharide, which also induced potent immunostimulatory activity in vitro (43). Hence, the general concept evolved that, in vivo, infection and inflammation in NLTs stimulated the maturation and migration of DCs into SLOs to stimulate TD immunity.

Ex Vivo-Generated Bone Marrow-Derived and Monocyte-Derived DCs

DCs were clearly shown to have a hematopoietic origin and to originate from bone marrow of adult animals. It was discovered that cells resembling DCs could also be generated in culture from mouse and rat bone marrow progenitors, particularly after supplementation of cultures with granulocyte-macrophage colony-stimulating factor (GM-CSF; CSF-2) (44, 45). There were perhaps two main drivers for these studies. First, as a trace cell type, the isolation of “primary” DCs from tissues was laborious, time-consuming, and often costly (not just expensive) in terms of the large number of animals that were needed for even a single experiment. Second, the appreciation that DCs might be central to the initiation and regulation of T-cell-mediated immunity almost immediately suggested that they may have enormous therapeutic potential. It was subsequently shown that DCs could be generated from macaque and human bone marrow CD34+ progenitors (4648). At around the same time it was also discovered that cells resembling DCs could also be generated from human monocytes in culture (49, 50), although their differentiation into veiled cells in culture had in fact been reported some years earlier (51).
The discovery that it was possible to generate monocyte-derived DCs (moDCs) suggested that, in vivo, monocytes are bipotential cells that can differentiate into either macrophages or DCs with immunostimulatory activity. It was therefore proposed that monocytes might be recruited to peripheral sites of inflammation and infection, where they differentiated into moDCs before migrating to SLOs to stimulate T-cell-mediated immunity. Later work further reinforced this view, particularly after injection of particulates into the skin or the blood (see “Lineages”). But, from a therapeutic standpoint, these findings were critical. At last it became possible to initiate human clinical trials in which cancer patients received infusions of their own “ex vivo-generated” DCs that had been pulsed with tumor antigens and matured in one form or another. Thousands of patients have now been treated in a large number of clinical trials, which continue with varying degrees of success (5254). (In some centers, perhaps a quarter of patients with end-stage [IV] melanoma who came off trial but continued to be treated with their DCs on an ad hoc basis have lived for many years afterwards, compared sadly to survival rates more in terms of months [C. Figdor and G. Schuler, personal communication].)

Plasmacytoid DCs

As early as the 1950s, pathologists had noted an unusual cell type within SLOs, particularly in disease settings, which was originally known as a plasmacytoid monocyte or plasmacytoid T cell. In 1997, it was discovered that if these cells were isolated from inflamed human tonsils and cultured with IL-3 and CD40 ligand (normally expressed by activated T cells in vivo), they developed immunostimulatory activity (55). They also appeared to be nonphagocytic and thus, functionally, came to resemble Steinman-Cohn DCs. In many other respects, however, they were very different: they appeared to circulate primarily in the blood (but not in lymph), had not been identified in many other tissues under physiological conditions, had a very different phenotype, were later shown to have the specialized capacity to secrete extremely high levels of IFN-I during viral infections, and additionally showed evidence of a possible lymphoid origin. Whether or not these cells should be considered as DCs is still debated by some investigators.

Immunostimulatory and Tolerogenic DCs

Many early studies focused particularly on the immunostimulatory properties of DCs. Parallel developments in the field, however, clearly demonstrated that at least some, if not all, of these cells might also contribute to tolerance. For example, increasing evidence showed that interdigitating DCs of the thymus were probably involved in the induction of central tolerance during T-cell development (despite their immunostimulatory function in vitro). Moreover, much later studies showed that veiled cells migrated constitutively from the gut in a relatively immature state. Some of them contained apoptotic cell contents, presumably derived from the tissue (56). It was therefore proposed that these cells might contribute to peripheral tolerance in regional lymphoid tissues. This paradigm was subsequently extended to DCs that migrated constitutively from all other NLTs in which they had been identified. Hence, DCs in different types of tissue appeared to have different cellular functions (e.g., phagocytosis versus immunostimulation); could contribute to tolerance and/or immunity depending on circumstances; and while some were clearly related (e.g., those migrating from one site to another), others (e.g., plasmacytoid DCs [pDCs]) seemed very different.

Follicular DCs Are Not “Dendritic Cells”

Another cell with dendritic morphology had been described within SLOs in 1965 (57, 58). Unlike interdigitating cells, however, these cells were identified within the germinal centers that develop from B-cell follicles during the course of adaptive immune responses. They were originally known as “antigen-retaining reticular cells.” However, they were termed “follicular dendritic cells” by Steinman in 1978 (59), since at that time he believed they might be related to the Steinman-Cohn DCs he first described. Despite this early confusion, it is now absolutely clear that the follicular DC is a completely distinct cell type, in terms of both its stromal cell origin and functions. These cells retain native antigens on their cell surface and are involved in selection of B cells during the so-called germinal center reaction (60). Hence, in this review, the term “dendritic cell” specifically excludes follicular DCs.

Multiple DC Subsets

By the turn of the century, at least eight types of DCs or dendritic-like cells had been identified: LCs and interstitial DCs within peripheral NLTs (commonly termed myeloid DCs); afferent lymph veiled cells and blood DCs in the circulation; Steinman-Cohn DCs and interdigitating cells within the lymphoid tissues (sometimes termed lymphoid DCs); moDCs in peripheral and possibly lymphoid tissues; and pDCs. Nevertheless, there appeared to be a commonality between them in that each cell type was increasingly shown to contribute to both T-cell tolerance and T-cell-mediated immune responses. Even so, into the fifth decade after Steinman’s first publication, the field of DC immunobiology appears to have become very complicated and often confused. It seems that an ever increasing number of “new DCs” are being described, particularly from phenotypic studies of cells from different tissues, some of which appear to have varying immunological functions depending on the experimental system that is tested. There is therefore understandable debate about if and how it might ever be possible to define a DC, and continuing controversy over the relationship between these and other cell types, and particularly macrophages, that seems well beyond semantics. This review focuses specifically on just a few types of DCs that seem quite distinctive, though others will be mentioned. This is to try to distinguish clearly between “the wood and the trees.” Whether or not it leads only to a clearing in a forest remains to be seen; it may be the edge of the wood.

DCs IN LINEAGES

This review is based on the premise that five distinct lineages of DCs or DC-like cells have to date been identified with some clarity. These populations comprise two subsets of “classical” DCs, pDCs, moDCs, and LCs. Evidence that these are different lineages include the findings that (i) they can develop from distinct hematopoietic or myelopoietic progenitors, (ii) they require different sets of transcription factors and cytokines for development, (iii) the fully differentiated cells adopt distinct transcriptional programs that regulate their functions, and (iv) interconversion from one type to another has not been seen (6168). In addition, (v) cells that seem to resemble these subsets can be generated from progenitors in culture (6871) (see “History”), though much care is needed in extrapolating findings from these systems to the cells in tissues (72). In this section, some of the molecular and cellular specializations of each will be highlighted. Their distributions and behavior within tissues and their functions in tolerance and immunity are discussed in the following sections.
Fate-mapping studies have been particularly helpful in elucidating the developmental origins of different populations of DCs from progenitors. The techniques used have included adoptive cell transfer, cellular barcoding, expression of reporter genes, and genetic lineage tracing in the Cre-Lox system (73). However, any such technique is potentially associated with off-target effects. For example, the use of irradiated recipients in cell transfer studies may disturb normal developmental signals within a leukopenic environment, and virus-mediated transduction during barcoding may skew cell fates. Moreover, the promoters used may not be strictly cell specific. This includes CD11c, which is often considered to be a specific marker for all five populations of DCs, but is and can be expressed on other cell types. Genetic ablation studies, using gene-targeted knockout mice, have also been very informative (74) but are likewise potentially subject to off-target effects (75). These considerations become particularly important when discoveries of “new types” of DCs are reported. Very rare individuals with DC deficiency have also recently been identified (76). However, the underlying genetic defects also affect development or homeostasis of other cell lineages, so they are rather uninformative when trying to elucidate the precise origins and functions of DCs.
It is generally accepted that the pluripotent hematopoietic stem cell generates both a CMP and a CLP (6163). Adoptive cell transfer studies have shown that both the CMP and CLP and other progenitors can at least give rise to classical DCs and pDCs in tissues under experimental conditions, and most likely during times of physiological stress. However, current evidence also indicates that, under normal steady-state and homeostatic conditions, the two subsets of classical DCs, pDCs, and moDCs derive from the CMP. In contrast, LCs are initially produced from primitive myelopoietic progenitors during embryonic development that also generate resident macrophage populations of some tissues (see below). According to one simplified, linear model, downstream progenitors of the CMP become progressively committed toward the generation of monocytes (which can subsequently develop into moDCs), pDCs, and, lastly, the two populations of classical DCs (77). In this type of model, the CMP generates a bipotential cell termed the macrophage-DC progenitor (MDP). In turn, this produces monocytes, perhaps via a common monocyte progenitor, with another bipotential cell called the common DC progenitor (CDP). The latter then generates pDCs, which complete their development in the bone marrow, together with a final bipotential cell termed the pre-DC. The pre-DC then migrates into the tissues, where it generates the two subsets of classical DCs. However, such schemes have been contested (78, 79). Apart from possible species differences that may exist, such differentiation pathways may well not conform to strictly linear models (80).

Classical DCs

The two types of classical DCs are crucially important for initiation of primary T-cell responses and make a substantial contribution to the regulation of adaptive immunity. These two subsets are as distinct from each other as are T cells and B cells (81). Classical DCs have been identified in most tissues studied closely, and all contain both types. However, these cells are absent from the parenchyma of the brain and the eye, though they do populate sites such as meninges and outer cornea. They are also absent from stratified squamous epithelia such as the epidermis of skin, which contains LCs instead. Both types of classical DCs have been identified in all warm-blooded species studied, including the mouse, rat, sheep, cow, pig, and human; the subsets in mouse and human seem extremely similar to each other at a transcriptional level (82, 83). Cells resembling classical DCs have also been identified in fish and amphibians (8486), though further study is required to determine whether or not these are classical DCs and, if so, whether there are two subsets. Nevertheless, the evolutionary conservation of two distinct subsets of classical DCs in warm-blooded species at least suggests that they play fundamentally different roles in the immune system.
In mice, pre-DCs migrate from the bone marrow via the blood into the tissues, where, locally, they differentiate into mature cells. Both the pre-DC and its presumptive upstream CDP and MDP progenitors express the CX3CR1 chemokine (fractalkine) receptor, but expression is extinguished as they differentiate into the mature cells (87). Classical DCs express the transcription factor Zbtb46 (zDC; Btbd4), which helps to discriminate between these cells and moDCs, pDCs, and LCs (64). However, this is not a stable or definitive marker, as it is downregulated on DC maturation, can be induced in monocytes, and is or can be expressed in other cell types. The homeostatic proliferation of pre-DCs and/or the development of classical DCs also depend on the Flt3L growth factor and its receptor, CD135. In general, classical DCs have a short half-life of just a few days in tissues, and this is tightly regulated: experimentally decreasing their life span has detrimental effects on the induction of immunity, while increasing it can result in autoimmune manifestations (88).
In this review, for simplicity, the two types of classical DCs will be designated as DCI and DCII subsets, and these are intended as neutral terms. The pre-DC generates two resident populations in all SLOs, and probably the thymus. These will be termed cDCI and cDCII cells, an abbreviation (cDC) that is otherwise used for classical or conventional DCs. The pre-DC also generates two populations in NLTs, which, however, are only transiently resident because they can subsequently migrate to SLOs. To distinguish these cells from the resident populations in SLOs, they will be termed migratory mDCI and mDCII cells, an abbreviation (mDC) that is otherwise used for myeloid DCs. Although detailed studies have not been performed in all cases, the respective populations of cDCI and mDCI, and cDCII and mDCII, appear to be extremely similar in many respects. However, their phenotypes do vary depending on their tissue of localization (see below), presumably reflecting tissue-specific specializations (89).
In general, classical DCs in NLTs express relatively low levels of MHC-II molecules compared to some of their counterparts in SLOs. A common view is that DCs in NLTs can endocytose or phagocytose antigens and generate intracellular peptide–MHC-II complexes. (They may also retain intracellular antigenic epitopes for considerable periods of time, even after antigen has been eliminated [90].) In response to stimuli such as TLR agonists and proinflammatory mediators, they decrease further uptake, begin to express high levels of peptide–MHC-II molecules at the cell surface, and increase their expression of costimulatory molecules such as CD86. They also express CD40, which enables cross talk between DCs and activated T cells via CD40L. Together, these regulate complex, bidirectional cascades of costimulatory (and conversely, coinhibitory) molecular interactions. The related phenotypic and functional changes are commonly referred to as DC maturation. The cells then migrate from the tissue into SLOs (91). Here, due to their new and increased expression of CCR7, they home to T-cell areas where the corresponding chemokines (CCL19 and CCL21) are produced (92, 93). Here these migratory DCs can activate antigen-specific T cells and trigger subsequent adaptive responses. An extreme view might hold that just one population of classical DCs could do the whole job itself. And yet there are two distinct subsets of migratory DCs in NLTs, and it is unlikely that the two resident subsets in SLOs are mere spectators. This strongly implies a marked division of labor between the two main types of classical DCs (DCI and DCII), and very close cooperation between all four in adaptive immune responses as a whole.

The DCI population

The DCI subset may have specialized capacities to present cell-associated antigens to CD8+ T cells, and perhaps differentially to sense PAMPs and DAMPs from these cellular sources. Differentiation of DCI cells from the pre-DCs requires transcription factors such as IFN regulatory factor 8 (IRF8), Batf3 (which, however, is also expressed by DCII cells), and Id2. Mice with targeted deletions in the corresponding genes have profound deficiencies in DCI cells. As a general rule, DCI cells in mice express molecules such as CD8 in SLOs but CD103 and/or CD207 (Langerin) in NLTs, which facilitates phenotypic discrimination from DCII cells. The function of CD8, which is expressed as an αα homodimer, in contrast to the αβ heterodimer of conventional CD8+ T cells, is unknown. (Perhaps future insights might come from consideration of other systems [94].) CD103 is the αE integrin, which can combine with β7 to form a receptor for E-cadherin, discussed later. The corresponding lineage in humans expresses CD141 (BDCA-3 [blood DC antigen 3]) or thrombomodulin (95, 96). This plays regulatory roles in both the complement and coagulation pathways, but its relevance to DC function is not clear. This aspect may be worthy of further study, especially since the complement and coagulation systems are so tightly linked.
Several examples of the specialized functions of the DCI population can be highlighted that seem to set them apart from DCII cells. First, DCI cells in mouse and human differentially express TLR3. This may enable them to sense double-stranded RNA produced during viral replication in infected cells, for example, after uptake of apoptotic or necrotic bodies. They also selectively express TLR11 and TLR5 in mouse and TLR10 in human (97, 98). Second, DCI cells differentially express receptors that can recognize DAMPs and/or capture and internalize dead and dying cells (99). For example, both mouse and human DCI cells express CD36, which is a receptor for apoptotic cells (and plasma thrombospondin) (100, 101). They also express CLEC9A (DC NK lectin group receptor-1 [DNGR-1]), which is a receptor for necrotic cells and recognizes F-actin as a DAMP (102). In mice, DCI cells also express CD205 (DEC-205), which is a receptor for uptake of both necrotic and apoptotic cells (103), as well as CD24, which can recognize high-mobility group box-1 (HMGB1) as a DAMP (104). Hence, these cells may be specialized for uptake of cell-associated antigens, and perhaps sensing of cell-associated DAMPs. Third, DCI cells express exceptionally high levels of components required for the peptide–MHC-I pathway and may constitutively cross-present exogenous peptides into the same route (105). Consequently, they seem particularly well specialized for antigen presentation to CD8+ T cells. Fourth, DCI cells can secrete high levels of cytokines such as IL-12, which is associated with polarization of CD4+ Th1 cells (13). Finally, in both mice and humans, DCI cells differentially express the chemokine receptor XCR1, which has been associated with homeostasis and tolerance (106, 107). The potential relevance of some of these specializations is discussed later (see “Tolerance” and, especially, “Immunity”).

The DCII population

In contrast to the above, the DCII subset may be specialized to present extracellular antigens to CD4+ T cells and perhaps differentially to sense extracellular PAMPs and soluble DAMPs. Differentiation of DCII cells requires a distinct set of transcription factors, which include Irf4 and RelB. Mice with targeted deletion of the corresponding genes have profound deficiencies in DCII cells. In mice and humans, these cells in probably all tissues express CD11b, which can associate with CD18 to form the complement receptor type 3 (CR3). However, expression of CD11b has proven a confounding factor in studies that have attempted to characterize DC populations in tissues since it is also expressed by monocytes, monocyte-derived cells (such as moDCs), and macrophages. Presumably the expression of CR3 (CD11b/CD18) facilitates uptake of complement-opsonized antigens by DCs and enhances antigen presentation. It seems generally presumed that this is also the case for CR4 (CD11d/CD18), which is expressed by all DC subsets. It remains possible, however, that these receptors may play other regulatory roles, and it is notable that, unusually, Fc receptors on DCs tend to be inhibitory in function (108). Mouse, rat, and human DCII cells, but not the DCI subset, also express CD172 (signal regulatory protein α [SIRPα]; see below), and this has been very helpful in distinguishing between these cell types, but again, this is not a definitive marker, as it can also be expressed by monocytes.
Several examples of the specialized functions of the DCII population can be highlighted that distinguish them from the DCI subset. First, mouse DCII cells selectively express TLR7 and TLR5 (109). These respectively may enable them to sense genomic RNA of viruses after uptake into endosomes, and perhaps the abnormal endosomal localization of host nucleic acids or chromatin as DAMPs (110), as well as bacterial flagellin at the cell surface. In contrast, DCII cells in human selectively express TLR2 and TLR4 (97), which, in addition to PAMPs, can recognize HMGB1 as a soluble DAMP (110). Second, mouse and human DCII cells selectively express CD172 (SIRPα), which is an inhibitory receptor that recognizes CD47 on host cells and prevents phagocytosis (“phagoptosis”). Interestingly, CD47 is also a receptor for thrombospondin. In addition, mouse DCII cells in SLOs express Clec4a4 (DCIR2; 33D1), which is an inhibitory receptor that has been implicated in maintenance of homeostatic tolerance (111). Third, the DCII population expresses high levels of all components required for the peptide–MHC-II pathway (105, 112), though cross-presentation may be inducible. Consequently, they may be specialized for antigen presentation to CD4+ T cells. Finally, DCII cells have been more generally associated with the selective induction of Th17 and/or Th2 responses (113). Some of these specializations are discussed later (see “Immunity”).

Plasmacytoid DCs

pDCs seem specialized to induce antiviral resistance and may regulate CD4+ T-cell responses and perhaps contribute to immunological memory of CD8+ T cells. Despite the original demonstration that pDCs can develop into immunostimulatory cells (55), subsequent studies have found that this capacity may be relatively limited compared to that of classical DCs. There seems to be little or no persuasive evidence that pDCs initiate T-cell responses in vivo. Unlike differentiation of classical DCs, the development of pDCs is dependent on the transcription factor E2-2 although it also requires Flt3L (65, 114, 115). While pDCs may normally be generated from the CMP under homeostatic conditions, they can also be generated from the CLP. Interestingly, and irrespective of myeloid or lymphoid origin, human pDCs express pre-Tα, mouse pDCs have rearranged IgH DJ segments, and both express RAG (116). These respective molecules are normally essential for formation of the pre-TCR and expression of the pre-BCR during intermediate stages of T- and B-cell differentiation, and for somatic recombination of both TCRs and BCRs. It has been proposed that this reflects the evolutionary origins of pDCs (116), a point that is further considered below.
In general, pDCs circulate in the blood and populate SLOs, which they enter via high endothelial venules (HEVs), and may also home to the thymus (see “Tolerance”). Small numbers of pDCs also populate mucosal tissues, within the lamina propria of the gut and lung and airways, for example, but appear to be excluded from other peripheral tissues under homeostatic conditions. Hence, migration of pDCs to the mucosal tissues could represent homing to lymphoid tissue within the gut wall, for example, or more generally migration in response to homeostatic inflammation (see “Tissues”). Such homing is dependent on expression of CCR9, although entry to SLOs is not. However, in pathological settings, pDCs can accumulate in large numbers in sites such as inflamed skin and within melanomas and ovarian and mammary carcinomas. Their roles in these sites are unclear.
Perhaps the best-known function of pDCs is the expression of exceptionally high levels of IFN-I in response to viral infections, and particularly of IFN-α. Both human and mouse pDCs selectively express TLR7 and TLR9, which enable them to sense genomic single-stranded RNA and DNA in endosomes. In fact, they may also sense host nucleic acids liberated during tissue damage and, in turn, facilitate wound healing through ancient (innate) mechanisms (117, 118). Nevertheless, IFN-I secretion is likely to be tightly regulated through a number of inhibitory receptors, such as Siglec-H (sialic acid-binding immunoglobulin-type lectin H) in mouse as well as CD303 (Clec4c; BDCA-2) and immunoglobulin-like transcript 7 (ILT7) in human, all of which regulate TLR responses (95, 119). Mouse pDCs also express tetherin (BST-2 [bone marrow stromal antigen 2]), which can physically bind live viruses to infected cells. In response to viral infection, the cells undergo autophagy, which may route endogenous viral antigens into the peptide–MHC-II pathway.
Through their capacity to secrete high levels of IFN-I, pDCs may play central roles in promoting antiviral resistance as well as regulating innate and adaptive responses. In the latter case, for example, at a molecular level, IFN-I can modulate functions of classical DCs by promoting extended antigen sampling of endosomal compartments for peptide–MHC-II loading (120); enhance cross-presentation and peptide–MHC-I loading; and regulate IL-12 expression, either to promote Th1 polarization or conversely to facilitate activation of CTLs (and innate-like NK cells). At the cellular level, IFN-I stimulates the differentiation of classical DCs from progenitors (121) and increases the maturation and migration of classical DCs from NLTs into SLOs. Furthermore, IFN-I decreases the generation of Tregs; increases the expression by CD8+ T cells of perforin and granzymes, which are both important for CTL killing; and promotes the survival of memory CD8+ T cells. Nevertheless, some of these proposed functions are inferences from the described roles of IFN-I in other settings, and, particularly since IFN-I is a large family of different classes, further study is required to confirm or refute a direct role of pDCs in some of them. Overall, however, pDCs may play regulatory roles in adaptive immunity either directly or by modulating the functions of other DC populations (122). This apparent synergy has, for example, been shown to be crucial for clearance of viral infections (123).

Monocyte-Derived DCs

moDCs may play particularly important roles in the effector phase of T-cell-mediated responses, and perhaps particularly those of CD4+ T cells. It now seems clear that monocytes are bipotential cells that can home to the tissues and differentiate into moDCs or macrophages, though monocytes, as such, have also been identified within some tissues (124). Other populations of probable monocyte-derived cells, described as “Tip-DCs” and “LysoDCs,” have also been reported (below, and see “Tissues”). Discrimination between moDCs, classical DCs (and particularly DCII cells), as well as monocyte-derived and other macrophages has proven highly problematic. In large part, this is because of the shared expression of molecules that can be further modulated during differentiation. For example, most or all express CD11c, and moDCs and mDCII cells both express MHC-II and CD11b. Nevertheless, a particularly useful marker of monocyte-derived macrophages in tissues is CX3CR1, since its expression increases to high levels during the differentiation of these cells from monocytes; it is also expressed by resident macrophage progenitors (125). In contrast, expression is lost during differentiation of pre-DCs into classical DCs. However, CX3CR1 is probably expressed at intermediate levels during development of moDCs in tissues, so this is not definitive. This molecule is the fractalkine receptor, which is presumed to facilitate cellular interactions with epithelial cells in tissues, though its precise functions remain unclear (126). In the mouse, many macrophage populations, including monocyte-derived populations as well as LCs, but not classical DCs, express the F4/80 molecule (127, 128). Importantly, CD64 (FcγR) also seems to be selectively expressed by moDCs but not classical DCs, thus helping to distinguish between them (108, 129).
Different monocyte subsets have been identified in the blood. In humans, those that express high levels of CD14, with or without CD16, are the likely equivalents of those that express high levels of Ly6C (Gr-1) in the mouse. Most evidence indicates that these cells can be recruited to inflammatory sites, where they have the potential to develop into moDCs or macrophages. Hence, this population has been termed inflammatory monocytes. In contrast, in physiological conditions, another human subset with high-level expression of CD16 but low CD14, which is the likely equivalent of those expressing low levels of Ly6C in mouse, are associated with the luminal surfaces of endothelial cells. Hence, they have been termed patrolling monocytes. It seems probable that both types of monocytes can be recruited to tissues under different circumstances and, depending on which and the particular microenvironment in which they find themselves, may “choose” alternative fates. Hence, they may develop into either moDCs or macrophages (130, 131). However, these tissue populations probably represent a continuum of different phenotypes and functions ranging from moDCs, LysoDCs, and Tip-DCs to macrophages (which are further subdivided by some into M1 or M2 types), and quite a lot more in between (132). Nevertheless, moDCs and macrophages, as commonly described, are probably very different in function.
There is much evidence that monocytes and/or monocyte-derived cells such as moDCs play important roles in the resolution of infections by bacteria and fungi; examples include infection with Citrobacter in the intestine and Aspergillus in the lungs (133135). They have also been associated with the polarization of Th1 responses due at least in part to their secretion of IL-12, but also with Th2 responses in other settings such as allergic asthma (136). In addition, moDCs seem able to elicit “recall” responses by activating memory T cells, though their relative importance in this respect, compared to classical DCs, is still not clear (137). In fact, both moDCs and classical DCs most likely play complementary and synergistic roles during adaptive immune responses in general (132) (see “Immunity”). In addition, populations of Tip-DCs have been identified within the spleen during infections with intracellular pathogens such as Listeria monocytogenes or Trypanosoma cruzi. These cells can secrete TNF-α and express inducible nitric oxide synthase, which are required for synthesis of reactive nitrogen intermediates, but also stimulate primary T-cell responses in culture. They are most likely monocyte-derived cells but functionally appear to represent a transitional stage between moDCs and macrophages. This fate choice appears to depend in part on which TLR agonists might be sensed (138).
A particularly potent stimulus for the differentiation of monocytes into moDCs may be phagocytosis of particles at endothelial cell surfaces. For example, in a transmigration system in vitro, CD14+ and/or CD16+ monocytes were able to traverse endothelial cell monolayers into an artificial matrix where they internalized particles, after which they crossed back and developed into moDCs (139, 140). Furthermore, injection of particulates into mouse skin was observed to recruit monocytes into the site, which subsequently migrated into the regional lymph nodes and developed into particle-laden moDCs in the T-cell-rich paracortical regions (141). Earlier work had also demonstrated that injection of particulates into the bloodstream of rats led to their uptake by cells within the marginating pool in the sinusoids of the liver, where Kupffer cells reside (142144). Subsequently, particle-laden cells were detected within the hepatic lymph, presumably en route to the regional lymph nodes. These findings raise the possibility that Kupffer cells initially phagocytosed the particles and perhaps then elaborated chemokines to recruit monocytes from the bloodstream, which, after phagocytosis, began to develop into moDCs. The cells that were subsequently detected in lymph nodes indeed had little phagocytic activity and could stimulate primary T-cell responses in culture, but additionally expressed CD103. Whether or not such expression was due to their microenvironment is not clear. However, taken together with the fact that the two main subsets of monocytes can migrate into tissues (above), it is possible that each could separately generate a distinct population of moDCs. While there seems to be no evidence to support this as yet, potential parallels with the classical DCI and DCII subsets should be obvious and worthy of closer scrutiny.
At least three different roles for moDCs in immunity may be envisaged. First, at some stages of differentiation, they may acquire effector functions, which contribute to the elimination of infectious agents in peripheral tissues, as seems likely for cells such as Tip-DCs. Second, moDCs may acquire peripheral antigens and migrate into SLOs to initiate primary T-cell responses. Evidence has been presented that, while the entry of monocytes into the tissues is CCR2 dependent, subsequent migration of moDCs into SLOs depends on expression of CCR7 and CCR8 (145). In this respect they could be viewed as a rapidly recruited source of immunostimulatory cells that assist the induction of adaptive immune responses, perhaps after classical DCs have migrated from the tissue and before repopulation from the pre-DC. Nevertheless, at present there seems to be little available evidence to support a major role of moDCs in the activation of primary T-cell responses. Finally, there is good evidence that moDCs may instead play essential roles in the effector phases of T-cell-mediated immune responses in NLTs (see “Immunity”).

Langerhans Cells

Epidermal LCs were first identified in 1868, but even today their roles in immunological responses have not been clearly elucidated. The dermis of skin, like most other tissues, contains the two subsets of classical DCs. In contrast, the epidermis contains only LCs. Related cells have been identified in all other stratified squamous epithelia, such as those of the oropharynx, vagina, ectocervix, and anus. LCs are situated in a suprabasal position deep within the epidermis, where they express MHC-II and CD11c and, in the mouse, F4/80. Epidermal LCs and dermal DCI cells also express CD207 (Langerin). This is involved in the development of Birbeck granules, which are specialized endosomal compartments of unclear function that have, however, only been identified in LCs (see “Tissues”). Unlike the development of other DC subsets, that of mouse LCs requires transcription factors such as ID2 and RUNX3 and is also dependent on transforming growth factor β (TGF-β) (which can be used to generate LC-like cells from progenitors in culture [70]). In addition, LC development is dependent on the CSF-1 receptor (M-CSFR; CD115). However, LCs persist in the skin of CD115-deficient mice, most probably because they require IL-34, which is an alternative ligand for this receptor to which it binds with a higher affinity than CSF-1.
LCs may have important functions related to monitoring the integrity of the epidermal barrier and contributing to repair. For example, in mice, skin oils apparently diffusing through the epidermis after damage to the outermost stratum corneum may be presented by LCs to a subset of specialized, innate-like NKT cells (146). These in turn may elaborate cytokines such as IL-22 to induce keratinocyte proliferation, and hence effect repair. Though their cell bodies are situated deep within the epidermis, LCs can extend cytoplasmic processes upwards between the keratinocytes and appear able to sample antigens beneath the stratum corneum (147). There seems to be little available evidence supporting the idea that LCs play any major role in initiating T-cell responses. In contrast, they may induce or maintain tolerance within the skin, even in the presence of TLR agonists (148). In part this seems due to their capacity to maintain populations of neonatal Tregs that home to the skin after birth (149) (see “Tolerance”). LCs also appear to contribute to polarized CD4+ T-cell responses of different types (Th17, Th1, and/or Th2) against antigens that may breach the uppermost layers of the skin or penetrate the epidermis. Other evidence tends to suggest that their contributions to CD8+ T-cell responses may be indirect (150).
It is, however, important to note that there are very distinct differences between the skin of different species of jawed vertebrates, which all possess epidermal LCs. For example, obviously the skin of mammals has an outer keratinized layer that is coated with skin oils and generally surrounded by air (except, of course, for aquatic mammals such as whales). In fish and amphibian tadpoles, however, the skin is a mucosal tissue surrounded by an aqueous environment, similar to that of the mouse or human embryo developing in the amniotic sac. There are even major differences between mouse and human skin. Both contain T cells in the epidermis, but those in mouse are mostly unconventional γδ T cells (dendritic epidermal T cells), whereas most in human are conventional αβ memory T cells. Possibly, therefore, the epidermal LCs of different species may have evolved some different functions, and there may be immunological differences between LCs of mouse and human.
Unlike all other DC lineages discussed here, the epidermal LCs of skin first arise very early during embryonic development. In the mouse, primitive myelopoiesis begins in the yolk sac at around embryonic day 7 (E7) and primitive myelopoietic (nonmonocytic) progenitors seed the epidermis at around E8.5. Here they generate LCs, which form a self-renewing population (151, 152); they also generate the resident populations of macrophages in the brain and liver, the microglial and Kupffer cells (153). A second wave of LCs then populates the epidermis, which develop from fetal liver-derived monocytes at E16.5 after the commencement of definitive hematopoiesis. (The same progenitors may also generate a second population of Kupffer cells in the liver, populate the lamina propria of the lung with cells that develop into alveolar macrophages, and probably give rise to resident macrophages of many other tissues [154].) The fetal-derived epidermal LCs then undergo rapid proliferation after birth and come to represent the predominant, self-renewing population in adult skin under homeostatic conditions (151). Under inflammatory conditions, at least, epidermal LCs can be further supplemented by bone marrow-derived monocytes that home to the skin epidermis and develop into apparently indistinguishable cells. By contrast, corresponding populations in the oral mucosa appear to originate from pre-DCs, with a contribution from monocyte-derived cells, though they too express a very similar transcriptomic signature to those in skin (155).
While the LCs of adult mouse and human skin have been associated with tolerogenic and immune functions (see above), the functions of the yolk sac-derived LCs within the embryo are unknown. These cells populate the epidermis even before the endothelium develops at around E9 and later differentiates into lymphatic and venous components at E11.5; hence, these LCs are present in the skin before the blood and lymph circulatory systems have developed. Potentially they may have specialized homeostatic functions within this specialized “mucosal-like” environment (cf. fish and amphibian tadpoles above). Curiously, however, the establishment of this population coincides with development of the umbilical link between fetus and mother. The only immunological cells of relevance at this stage would seem to be maternally transferred lymphocytes. In principle at least, maternal T cells could conceivably gain access to fetal skin perhaps in response to the endogenous chemokine CXCL12 (stromal cell-derived factor 1 [SDF-1]). This plays important roles in the movement of embryonic cells during morphogenesis and is a ligand for the CXCR4 receptor on mature T cells. Fetal engraftment by maternal T cells has in fact been demonstrated in some cases of SCID, for example (156, 157). Such maternally transferred T cells could be detrimental to the fetus (through recognition of paternal antigens) or beneficial if they were Tregs. Hence, it is an open question whether embryonic epidermal LCs might be involved in the induction or maintenance of tolerance, through local interactions or even after migration from fetus to mother (158, 159).

Other “Subsets” of DCs

Numerous publications have postulated the existence of other presumptive DC subsets in addition to the five types discussed here. Perhaps the most persuasive (or at least suggestive) evidence is for a subdivision of mouse DCII cells in SLOs into two further subsets. This is based on their apparent dependence on the transcription factors Notch2 or klf4 for development (79). These putative subsets have also been associated with Th17 and Th2 responses, respectively. Moreover, there is undoubted phenotypic heterogeneity of DCII cells within mouse SLOs based on differential expression of molecules such as Clec4a4 (CDIR2; 33D1; see above), CD4, and ESAM. As another example, subsets of “double-negative” DCs that do not express “selected” DCI or DCII markers have been reported in various tissues. These include the dermis of skin (XCR1CD11b) (160), spleen (CD4CD8) (161), Peyer’s patches (CD8CD11b) (162, 163), and mesenteric lymph nodes (MLNs) (CD103CD11b) (164). These double-negative cells have variably been reported to be IRF4 dependent, to secrete IL-12, and/or to express CD207 (Langerin) or CX3CR1. In addition, a double-positive population has been reported in gut (CD103+CX3CR1+) (164), and there are undoubtedly other examples. However, the expression of different transcription factors can be induced or extinguished because of intrinsic developmental programs or extrinsic microenvironmental influences, and can also be modulated during cellular responses. It is therefore possible that at least some of the additional DC subsets that have been proposed, such as those noted above, might represent early stages during the development of progenitors, such as pre-DCs, or of monocytes adopting different cell fates within tissues (compare Tip-DCs and LysoDCs above). Given the general preoccupation with three main types of polarized CD4+ T-cell responses (Th17, Th1, and Th2), it is perhaps not surprising that some might expect the existence of three dedicated subsets of classical DCs (see “Immunity”). There are almost certainly two subsets of classical DCs; there may be three; many more seem like fashion accessories.

DCs IN TISSUES

This section provides an overview of the dynamic anatomy of DCs within tissues. It focuses on their distributions in anatomical compartments, including their migration from NLTs to SLOs, and their behavior within microenvironments under physiological conditions and after inflammation or infection. Other local or migrant cell populations will be noted, with an emphasis on macrophage-like cells. Unless otherwise stated, two subsets of classical DCs are present in all, and moDCs may also develop under inflammatory conditions. DCs within the thymus are discussed in “Tolerance.”

Steady State versus Homeostatic Inflammation

The term “steady state” is often used to describe conditions in tissues under physiological conditions in the absence of overt infection. However, the presence of commensal organisms on external stratified epithelia such as the epidermis of skin, and at luminal surfaces within mucosal tracts such as that of the gut, is continuously sensed by the immune system. Indeed, the microbiota also shape adaptive immunity, and a state of mutualism is established and maintained between them and their hosts (165169). The microbiota induce a state of “homeostatic inflammation,” also termed “tonic stimulation,” which is physiological and tightly regulated and controlled (170). However, depending on the relative loads of commensals, the levels of homeostatic inflammation may vary, perhaps being highest in gut and lowest in organs and tissues such as heart and skeletal muscle, for example. This should be borne in mind when considering the relative complexity of DCs and other populations in different tissues. It is also a general, though not entirely accurate, rule that naive T cells are excluded from NLTs. However, populations of effector and/or memory T cells may or may not constitutively traffic into different tissues, which adds a further layer of complexity to the physiological conditions within each. Hence, in this review the term “homeostatic conditions” is often used for those in normal tissues in general, while the term “inflammation” relates to conditions that prevail after infection or tissue damage.

The Skin and Peripheral Lymph Nodes

Skin

The skin has two distinct anatomical compartments: the epidermis and the dermis, separated by a basement membrane. The outer epidermis is a stratified squamous epithelium that is avascular and lacks its own blood supply. The connective tissue of the dermis is penetrated by a rich network of blood vessels and capillaries as well as blind-ended, fenestrated capillaries; nerves also traverse the dermis, as in all tissues, and some extend into the epidermis. The extravascular fluid of the dermis drains into blind-ended lymphatics and flows as afferent lymph into regional lymph nodes, which are often connected to each other in chains. The skin contains one population of LCs within the epidermis and the two subsets of classical DCs, mDCI and mDCII, in the dermis; a third, double-negative population has also been reported (see “Lineages”). The functions of these DC populations are tightly knitted with those of other cell types that exist within the skin. Collectively, they play crucial roles during immune surveillance for pathogens (160, 171, 172) and continuously maintain states of mutualism with commensals (173) and tolerance to self-antigens of the tissue (174).
Epidermal LCs are localized with their cell bodies mostly positioned within the spinous layer (stratum spinosum) just above the basement membrane (stratum basale). They are intimately associated with keratinocytes, with which they closely interact (see “Lineages”). For example, they can deliver topically applied carcinogens to neighboring keratinocytes, which subsequently undergo malignant transformation, but this does not occur in LC-depleted skin (175). It has been claimed that each LC is separate and does not communicate directly with any other, but instead extends its dendritic processes around its “own group” of keratinocytes. These express PRRs, and their inflammatory responses may stimulate LC migration from the tissue. More-recent findings show that in fact the dendritic processes of LCs can extend upwards through the tight junctions of the keratinocytes in the uppermost layers, and just below the outermost stratum corneum, which comprises dead keratinocytes (147). Here they can sample antigens and contribute to the induction of immune responses against them. LCs were, for example, found to be essential for the induction of IgG against the exfoliative toxin of Staphylococcus aureus and could prophylactically protect mice from subsequent disease (176). In this respect, LCs may resemble the “antigen-sampling” cells within simple epithelia of other tissues such as the gut and airways (see below). However, LCs may have very limited ability to phagocytose bacteria (177). Whether LCs may also extend dendritic processes from the epidermis through the basement membrane below and into the dermis, perhaps to communicate directly or indirectly with dermal DCs, deserves further study.
Within the dermis, the mDCI and mDCII subsets reciprocally express CD103 and CD11b, respectively. Both dermal classical DC subsets are believed to undergo “homeostatic maturation” and hence migrate constitutively from the dermis into SLOs, where they may contribute to induction or maintenance of peripheral tolerance. The dermis also contains other populations such as mast cells close to the blood vessels and innate lymphoid cells. During infection and inflammation, there is a complex interplay between these and other cells in skin that ultimately leads to maturation of the classical DCs and migration to the regional peripheral lymph nodes (pLNs). During inflammation, monocytes may enter and develop into moDCs, and pDCs may also be present in pathological settings. It is, however, quite clear that dermal mDCI cells also express CD207 (178), although they appear not to possess Birbeck granules. This is an important point to note in studies that have endeavored to trace the migration of LCs into pLNs, for example (179). It also raises the issue, which does not yet seem to have been addressed, as to whether or not CD207 might also induce development of Birbeck granules in these cells within pLNs. This seems to be especially pertinent since, in contrast, earlier studies clearly documented the loss of Birbeck granules by LCs during maturation in culture (see “History”). In fact, other studies have also shown that after migration into the dermis, LCs can undergo apoptosis (150). The apoptotic contents were then acquired by DCs, particularly the mDCI subset, which subsequently migrated to the pLNs, where they (rather than LCs) stimulated antigen-specific responses. Hence, some immunological responses that have been associated with LCs may in fact not be induced directly by these cells. Perhaps it is time for a much closer examination of these issues.

Peripheral lymph nodes

The pLNs are specialized SLOs that monitor peripheral tissues and integrate the different types of information derived from them under homeostatic and inflammatory conditions. Subsequently, tolerogenic or immunogenic adaptive responses can be initiated within them. Lymph nodes are surrounded by a dense fibrous capsule that is penetrated by multiple afferent lymphatics leading from the tissues, such as dermis of skin, which during infection and inflammation contains antigens and proinflammatory mediators and chemokines. The afferent lymphatics also provide the route by which migrating DCs that have acquired antigens, as well as necrotic or apoptotic cells and debris, can enter pLNs and subsequently interact with T cells. Hence, three populations of peripheral DCs are likely to enter pLNs from skin: the epidermal LCs and both subsets of dermal DCs. Presumably, moDCs that develop in response to inflammation may also migrate to nodes via afferent lymphatics, although inflammatory monocytes may also enter directly from blood (see below).
After entering a pLN, the afferent lymph drains into a narrow subcapsular space immediately beneath the capsule and surrounding much of the underlying cortex and medulla. The “floor” of this space is populated by macrophages above the tissue below. The cortex is penetrated by trabeculae through which the lymph enters and is distributed through the lymphatic sinuses or medullary cords. These then drain into usually a single efferent lymphatic, and the lymph leaves the lymph node at the hilum. Ultimately, the efferent lymph is collected by the thoracic duct and drains back to the blood. Blood vessels enter the node at the hilum and run upwards mostly into the cortex (which may not be directly accessible to lymph). Here there are multiple arteriovenous communications surrounded by specialized endothelial cells that comprise the HEVs. Lymphocytes enter the lymph node from the blood via the HEVs and generally segregate within the cortex. The B cells become localized to follicles, with T cells in the interfollicular areas between them. (In fact, entry of T cells into the lymph nodes via HEVs is regulated by DCs [180].) This latter region is distinct from the more superficial parafollicular areas adjacent to the trabeculae. Pre-DCs also enter the nodes from the blood via HEVs, as do monocytes and pDCs during inflammation (181).
Lymph nodes also contain several networks of reticular cells with specialized functions in different anatomical compartments (182). These include a fibroblastic reticular cell (FRC) network that extends from the subcapsular space, ramifies through the cortex, and connects to the HEV. The fibers of the FRC network contain a central core of collagen bundles that is surrounded by extracellular matrix components that are synthesized by the FRCs that closely surround them (183, 184). These structures are otherwise known as conduits, and have also been identified in the spleen and more recently the thymus (see “Tolerance”), although there may be molecular differences in their composition. At the distal ends of these pLN conduits, a specialized protein, PLVAP (plasmalemma vesicle-associated protein), of lymphatic endothelial cells forms diaphragms that act as molecular sieves (185). These permit small (<70-kDa) molecules to enter the conduits, from where they can be delivered deep into the cortex to the HEVs, but excludes all those that are larger. It is now clear that low-molecular-weight antigens and chemokines, and potentially cytokines, can pass directly through the central core of the conduits from peripheral regions such as the subcapsular sinus (186, 187). DCs remodel the FRC network over which they migrate, and both may promote the survival of T cells within the tissue (187189). A population of CD11b+ presumptive DCs is closely associated with the FRC network. Intermittent gaps, or “windows,” between the FRCs enable these cells to insert dendritic processes into the conduits and directly to sample antigens (190). Whether these cells are in fact classical DCs, possibly resident cDCII cells, or moDCs requires further investigation. In addition, chemokines can also be delivered via conduits directly to HEVs, where, under inflammatory conditions, they may recruit monocytes from the blood into the node (181).
Under homeostatic conditions, it is likely that the majority of DCs in pLNs are resident populations, with relatively few immigrants present (191). More recently, detailed information on the localization of DCs in pLNs has been gleaned from studies using multiphoton microscopy and a more recent analytical technique termed histo-cytometry (192). These have visualized distinct subsets of DCs within different microenvironments of the pLNs. The resident cDCI subset is mostly localized centrally within the T-cell zones. In contrast, the cDCII subset resides within the medullary lymphatic zone close to the surrounding subcapsular sinus. Here, these cells have been observed to capture particles from the lymph (193). The mDCI subset also localizes deep within the paracortex, whereas the mDCII subset tends to be positioned in the outer paracortical regions. Interestingly, under homeostatic conditions, the presumptive DCII subsets were found to express retinoic acid and be capable of inducing Tregs (features actually associated with the converse subsets in gut MLNs) (194). However, in models of skin inflammation, it was shown that the migratory DCs subsequently homed into the outer cortex, in the T-cell zone but adjacent to B-cell follicles. In contrast, presumptive LCs were observed to migrate from the skin much more slowly into lymph nodes, and were later detected within the inner paracortex, separated from the migratory DCs (179).
It is clear, even from the few examples above, that lymph nodes are highly dynamic structures and that DCs within them perform an extremely complex choreography. Nevertheless, some functional consequences for the initiation of CD4+ T-cell responses are becoming apparent (195). For example, it has been shown that antigens injected into the skin can be very rapidly acquired by resident lymph node DCs, which then induce the early stages of CD4+ T-cell activation (196). However, robust T-cell responses were only induced after the entry of antigen-bearing DCs that migrated from the site of antigen injection much later. It may be that conduit-associated DCs that have sampled small antigens also mature in response to the chemokines (some of which have proinflammatory activities) these structures may deliver, though whether this also includes proinflammatory cytokines requires further study. What these and other studies highlight is the fact that T-cell activation occurs much faster after antigen delivery than was previously appreciated. Moreover, many CD4+ T-cell responses may require serial engagement with different DC subsets to become fully effective (see below). Presumably, the cortex of the lymph nodes is relatively isolated and “sterile,” much like within the thymus (see earlier). Otherwise it would seem that uncontrolled access of immunomodulatory agents to DCs would presumably further modulate their functions and lead to dysregulated T-cell responses; for example, functional cell-cell communication may need cytokine gradients to be established between them (197). This is an interesting aspect of SLO structure and function that has been relatively little investigated.
Typically, activation of many CD8+ T-cell responses requires “help” delivered by CD4+ T cells. Other studies have visualized the sequential interactions of T cells with DCs during helper-dependent CD8+ T-cell activation (198, 199). These have revealed that a series of spatiotemporally distinct cellular interactions occur within different microenvironments of a lymph node, and that these are required to generate a robust CD8+ T-cell response. They also highlight serial engagements between different subsets of DCs and CD4+ and CD8+ T cells. Such findings are reminiscent of an earlier model proposed for helper-dependent CD8+ T-cell responses (200). It seems possible that antigen-specific CD4+ T cells are first activated by migratory DCs. These activated T cells then home toward cDCI cells and stimulate (“license”) them to increase their expression of costimulatory molecules sufficiently for CD8+ T-cell activation (roughly speaking, this is a “higher” level than is required by CD4+ T cells). These cDCI cells may then activate CD8+ T cells that subsequently engage with them. However, what does not seem to have been well defined is the precise source of antigen for the cDCI cells, which seem relatively sequestered deep within the T zone. Interestingly, they may communicate with each other over large distances through the formation of “tunneling nanotubules” between them, particularly after CD40 ligation, which could be provided by activated T cells (201).
Studies of other cell populations within “steady-state” pLNs have demonstrated clear differences in cellular compartmentalization (183). For example, NK cells, specialized invariant NKT cells, and unconventional γδ T cells are present in the interfollicular regions and medullary lymphatic zones, to which mDCII and cDCII cells, respectively, home. However, elucidation of potential roles of these cells in modulating DC responses, and possibly subsequent αβ T-cell responses, awaits further investigation. Furthermore, different types of macrophages populate the subcapsular space and medullary sinuses. Two of these, for example, express CD169 (sialoadhesin) and F4/80, respectively, in mice. The CD169 macrophages appear to comprise part of the “firewall” preventing access of lymph-derived pathogens into downstream tissues since, if they are depleted, infectious agents can spread to other connecting lymph nodes (202). They have also been implicated in the generation of CTL responses (203). Potentially, too, small-molecular mediators produced by these macrophages could also be delivered into the tissue via conduits.

The GI Tract, MLNs, and GALTs

Lymph from large regions of the gastrointestinal (GI) tract probably drains to the MLNs, though some areas drain into other regional lymph nodes (see below). Throughout the small and large intestine there are also distinct types of gut-associated lymphoid tissues (GALTs) that play specialized roles in tolerance and immunity (204). Descending from the duodenum, the jejunum and ileum contain variable numbers of Peyer’s patches, which, in humans, reach a peak in the third decade of life but then decline in number (205). These are embedded within the mucosal propria and submucosal tissue. They have specialized, sieve-like M cells within a follicle-associated epithelium, juxtaposed between the intestinal epithelial cells. Descending further into the cecum is the appendix, which may have specialized functions yet to be discovered. There are also increasing numbers of “solitary isolated lymphoid tissues,” such as isolated lymphoid follicles, descending from the ileum to the colon. These most probably represent tertiary lymphoid structures. They have much less well-defined structures than SLOs, lacking defined follicles and T-cell zones, for example, and are not developmentally controlled but are locally induced by a subset of ILCs (206). DCs populate the lamina propria and submucosal tissue of the gut wall and GALTs.

GI tract

The relative distributions of the two subsets of classical DCs within the lamina propria and submucosa of the GI tract may differ (29, 207). Their relative proportions also differ between the small intestine and colon, with a predominance of cDCII cells in the former and of cDCI cells in the latter. These cells are generated from pre-DCs that home to the gut through expression of the α4β7 integrin. Precisely how and where the homing of these progenitors is determined is not clear, and presumably different homing molecules may be required for migration to other NLTs. The two subsets of classical DCs in the gut have characteristic expression of CD11b (which is expressed by mDCII but not by mDCI cells), but both express CD103. The reason for this is not clear, though in other settings expression of CD103 can be induced by GM-CSF. Similar subsets have been identified in gut-derived pseudoafferent lymph in rats, indicating that they constitutively migrate into the MLNs (208). Under homeostatic conditions, the cDCI subset in afferent lymph was found to contain apoptotic cell inclusions, and it seems likely that these cells may contribute to tolerance induction against cell-derived components (see “Tolerance”). However, the flux of both subsets is dramatically increased in inflammatory settings.
In mice, the macrophage populations within the lamina propria of the gut most likely originate from fetal liver-derived monocytes, but are supplemented by bone marrow-derived monocytes after birth when the gut microbiota first becomes established (68). There is a general consensus that gut macrophages express high levels of CX3CR1, as well as CD11b. Hence, high-level expression of CX3CR1 or CD103 has been used to distinguish phenotypically between gut macrophage and DC populations, respectively, in a number of studies (87, 204, 209212). However, variant populations, including a double-positive subset of cells that expresses both CX3CR1 and CD103, have also been described (106, 164, 211), and the situation is further complicated by the fact that moDCs can express intermediate levels of CX3CR1 (see “Lineages”). Overall, therefore, multiple phenotypic subsets of presumptive DCs (or macrophages) have been identified, particularly in the gut, it seems. Possible explanations for this have been noted (see “Lineages”). Nevertheless, there appear to be clear differences in the subanatomical localization of the major populations of presumptive CX3CR1+ macrophages and CD103+ classical DCs in the small intestine, and most likely their functions.
Under homeostatic conditions, CX3CR1+ cells have been identified within the gut epithelium. It is now generally accepted that these cells express tight junction proteins that enable them to penetrate the epithelial cell layer, through which they extend dendritic processes to sample luminal contents (212, 213). However, this appears to be an inducible behavior since it is enhanced by TLR-dependent responses of the epithelial cells (213) and does not occur in germ-free gnotobiotic mice, though whether it occurs at all has been questioned (214). Nevertheless, under these circumstances, the CX3CR1+ cells may contribute to a state of tolerance or unresponsiveness to innocuous soluble food antigens. It has been shown they can acquire such antigens from the lumen and transfer them to lamina propria CD103+ cells, with subsequent induction of Tregs and tolerance, presumably after migration of the latter to MLNs (210, 215). The transfer of antigens is dependent on the formation of gap junctions between the CX3CR1+ and CD103+ cells, since genetic ablation of connexin-43 prevented the generation of Tregs and induction of oral tolerance. It has also been shown that soluble antigens can be transferred directly through mucus-secreting goblet cells to CD103+ cells in the lamina propria (214).
The capacity of CX3CR1+ cells to promote tolerance induction under homeostatic conditions may be due to their apparently constitutive, anti-inflammatory properties. These cells seem relatively refractile to stimulation by TLR agonists, can secrete IL-10, and also inhibit T-cell proliferation in a contact-dependent manner (210). They also appear to be specialized for the uptake of soluble antigens, and may have little or no capacity to phagocytose particulates. It is generally accepted that these cells are sessile and do not migrate from the tissue to the MLNs, though contrary evidence has been put forward (216). However, during experimental Salmonella infection of the gut, the CX3CR1+ cells may secrete chemokines to recruit CD103+ cells from the lamina propria into the epithelium above the basement membrane (217). Here, the CD103+ cells were also observed to extend processes into the lumen, capture bacteria, and rapidly retract their processes. This behavior was associated with the subsequent induction of CD8+ antigen-specific T-cell proliferation within the MLNs. Under homeostatic conditions, rare CD103+ cells were also observed “patrolling” the epithelium (217). It is possible that, under these circumstance, these cells might also sample innocuous luminal contents, migrate to the MLNs, and facilitate tolerance induction, though this requires further investigation.
In the gut, the specialized anti-inflammatory subset of CX3CR1+ macrophages may be required to dampen DC responses and help maintain tolerance to innocuous food antigen; otherwise homeostatic inflammation might promote sufficient DC maturation to enable them to induce immunity, although this aspect deserves further investigation. It is quite evident, however, that tolerance needs to be maintained even during times of infection, when DCs become able to stimulate TD primary responses. It is known that CD103+ DCs can produce the vitamin A derivative retinoic acid and anti-inflammatory TGF-β, and can promote the generation of FoxP3+ Tregs within the MLNs (217220). Locally, gut epithelial cells also produce thymic stromal lymphopoietin, which drives DCs toward a “regulatory” phenotype, and the latter can also produce indoleamine 2,3-dioxygenase, which depletes tryptophan and inhibits T-cell proliferation. In addition, the mDCI subset, through secretion of IL-12 and IL-15, can induce secretion of anti-inflammatory IFN-γ by T cells in the colon (221). Precisely how this balancing act is achieved remains a major puzzle in mucosal immunology and more generally in adaptive immunity as a whole.

Mesenteric lymph nodes

The MLNs that receive efferent lymph from the GI tract are developmentally distinct from the majority of “peripheral” lymph nodes that drain other tissues. They are described as mucosal lymph nodes, and also include the cervical lymph nodes that drain the nasopharynx, for example, and the sacral lymph nodes associated with the rectum. Development of the MLNs as well as Peyer’s patches requires key members of the lymphotoxin and lymphotoxin receptor family, which are related to the TNF family and its receptors (222). Nevertheless, at a gross level, the overall structure of the MLNs is similar to that of pLNs. However, there are likely to be functional differences due to the additional influence of gut-derived immunomodulatory molecules draining from afferent lymph (above) and perhaps produced locally. These may also contribute to tolerance under homeostatic conditions, for example, by inducing pTregs (223) (see “Tolerance”).
The MLNs play a central role in the induction of tolerance to food antigens, particularly in soluble form (224, 225). Oral tolerance cannot be established in mice that developmentally lack MLNs (as well as pLNs). The MLNs have been described as a firewall that helps to prevent commensals that may have invaded the gut lamina propria and traveled in lymph from entering the blood. Because of this, it is believed that the systemic immune system normally remains “ignorant” of the gut microbiota. Mice lacking MLNs develop fatal splenomegaly and lymphadenopathy. Likewise, the liver, which receives the entire output of intestinal venous blood through the portal vein, acts as an additional firewall to capture and clear gut commensals that may enter the blood in more-pathological settings (226). Damage to the liver impairs its function and can also lead to fatal infection with commensal organisms.
The MLNs contain the two resident subsets of classical DCs as well as corresponding subsets that migrate from the gut wall. As noted above, the migratory DCs may play important roles in the induction or maintenance of tolerance within the gut, although it is not clear if this is primarily against normal gut tissue antigens or against antigens associated with commensals or food. Certainly it seems likely that the induction of oral tolerance against food antigens is intimately associated with the migration of DCs to the gut since this is induced in a CCR7-dependent manner. During immunity to gut pathogens, the MLNs have been particularly associated with Th17 responses, which play particularly important protective roles in gut immunity, as well as Th1 responses. The MLNs also contain pDCs, though these appear to arrive from the blood rather than after traveling in lymph (227). Their precise contributions to tolerance and immunity are not precisely defined, though they are presumably important in responses against enteric viruses (228). It also seems likely that moDCs, developing during gut infections, migrate to MLNs and also contribute to T-cell responses.

Peyer’s patches

Peyer’s patches are specialized SLOs that monitor the intestinal lumen. They possess sieve-like M cells that are in direct contact with the lumen, and through which contents may be sampled (205). Below the follicle-associated epithelium, with its juxtaposed M cells, is the subepithelial dome (SED) region. Below this, the B cells and T cells are segregated into follicles (or, more usually, in this tissue, germinal centers [229]) and the interfollicular regions, respectively. Lymphatics do not appear to have been well described in Peyer’s patches, but if they exist, they would presumably represent efferents that drain to the MLNs. Different subsets of DCs have been characterized within the Peyer’s patches in generally discrete subanatomical compartments (230). The DCI cells occupy the interfollicular regions among the T cells, whereas DCII cells are located in the SED region. It seems likely that most or all are resident DC subsets (i.e., cDCI and cDCII) derived from the pre-DCs. Some contribution from migratory DCs derived from the gut lamina propria and/or submucosa is possible, but alternatively they may be excluded from this tissue. Within the interfollicular and SED regions, both pDCs and a subset of double-negative cells has also been identified that lacks expression of CX3CR1 and CD8 (see “Lineages”). The latter have not yet been identified within MLNs, suggesting that they do not migrate from the tissue, though relevant molecular expression could conceivably be induced if they do. Two populations of monocyte-derived cells have also been well characterized within the Peyer’s patches (230, 231). One has been termed a lysozyme-containing DC (LysoDC) and is likely to represent a form of moDC (cf. Tip-DC earlier), while the other has been termed a lysozyme-containing macrophage.
It seems generally assumed that soluble molecules may traverse the M cells. If so, they do not seem to be involved in the induction of oral tolerance, since this is not impaired in mice with developmental deficits of Peyer’s patches. What seems clear is that pathogenic bacteria and inert particulates can be captured by the LysoDCs that extend dendrites through the M-cell pores into the gut lumen (232). While these CX3CR1+ cells somewhat resemble those described within the gut epithelium (above), they appear to be distinct in that they do not express IL-10 and can secrete proinflammatory cytokines in responses to TLR7 agonists. They have been described as short-lived cells and associated with induction of Th17 responses. In contrast, the cDCI subset can secrete IL-12 in response to pathogenic bacteria and may be involved in induction of Th1 responses. It is possible that the cDCII subset may contribute to the initiation of TD B-cell responses and production of protective IgA in response to gut pathogens. This may be in contrast to the IgA that is secreted in a T-independent manner and that plays essential roles in confining commensals to the gut lumen (233). Nevertheless, in other settings, the Peyer’s patches may represent specialized inductive sites for Th2 responses, for example, in response to helminth infections. Collectively, therefore, DCs within the lamina propria and submucosa of the gut may facilitate the concomitant induction of tolerance to food antigens and polarized immune responses against pathogens. At the same time, those within Peyer’s patches may additionally maintain responses that help to contain commensal organisms to the lumen, but induce differently polarized responses against other pathogens.

Other Mucosal Tissues and MALTs

Distinct subsets of DCs also populate the lamina propria of all other mucosal tissues and their respective MALTs. Here, the focus will be on the lung and airways and their associated MALTs, though the urogenital tract will be mentioned briefly.

Lung and airways

Within the lung and airways, the mDCI subset is localized close to or within the pulmonary epithelium, while the mDCII subset tends to be confined within the lamina propria; different populations of moDCs or macrophages have also been identified (234, 235). There is evidence that a subset of presumptive DCs within alveoli can extend dendrites into the airspace and sample antigens, presumably for subsequent induction of tolerance or immunity (236, 237). In contrast, the alveolar macrophages remain relatively sessile and apparently focus on noninflammatory clearance of foreign particulates. The precise subset concerned with airway sampling awaits further characterization. However, evidence has also been presented for a two-step mechanism by which presumptive DCs may be recruited to the epithelium from the lamina propria in a chemokine-dependent manner (237). This is reminiscent of that described within the intestine whereby CX3CR1+ cells recruit CD103+ DCs to sample particulates (see above). It may also help explain the apparent contribution of CX3CR1 cells to antiviral responses in the lung, for example (238). The mDCI subset expresses tight junction proteins (236), which may enable it to penetrate the epithelium for direct sampling of the airspace, though this has not been directly proven. This subset selectively transports apoptotic cell contents to the regional pLNs under homeostatic conditions (239). However, during enteric viral infections, the same subset has also been associated with cross-presentation of antigens and the induction of CD8+ T-cell responses. Antigen-sampling cells have also been identified within human nasal mucosa (240). In addition, pDCs can be detected within the lamina propria of the airways, where they presumably contribute to antiviral responses through production of IFN-I, which has been shown also to protect classical DCs from viral infection (241). Finally, DCs have also been identified throughout the urogenital tract (242). It would perhaps not be surprising if some of these cells, too, had the capacity to sample luminal antigens similarly to those in other epithelial sites.

Waldeyer’s ring, BALTs, and NALTs

The DCs of the lung and airways and of the urogenital tract can migrate into regional pLNs, which are unlike the MLNs associated with the gut. In addition, specialized lymphoid tissues are, or can be, associated with the nasopharynx and the airways. The tonsils are situated within Waldeyer’s ring of the pharynx in humans; some animals also have additional tonsils. Mice, but not humans, have nasal-associated lymphoid tissue (NALT), which is developmentally controlled, like other SLOs. Furthermore, bronchial-associated lymphoid tissues (BALTs) may be present, particularly in pathological settings, though this might represent tertiary lymphoid tissue. Classical DCs have been identified in all these tissues, and in addition, pDCs may be present in large numbers in pathological circumstances. It is not yet clear whether or not classical DCs can contribute jointly to tolerance against innocuous antigens and the containment of commensals (particularly in the upper regions of the airways and nasopharynx), as well as immunity against pathogens, though this seems likely. Specialized mechanisms by which they may do so at these sites, however, awaits further study.

Fully Vascularized Tissues and the Spleen

Early studies demonstrated that DCs could be isolated from the blood, where they appeared to circulate in both relatively immature and mature forms (37). It was believed that the former represented DCs en route to the tissues, while the latter represented those migrating from these sites. However, this interpretation needs to be revisited in the light of our current understanding that classical DCs are generated locally in tissues from the pre-DCs.

The heart and nonmucosal tissues

Two subsets of classical DCs have been identified in all vascularized tissues that have been closely studied. Such tissues include the heart (243); cardiac valves and aorta (244, 245); skeletal muscle (178); kidney (246, 247); and tissues such as pancreas (248). Interestingly, antigen-sampling CD11c+ cells have been described within the subintimal space of the aorta, where they appeared to probe the vascular lumen (245). The macrophage populations in at least some of these tissues, such as heart and arterial walls, originate from both yolk sac-derived progenitors and fetal liver monocytes, but may be supplemented from bone marrow-derived monocytes later in life (125, 249). It is generally presumed that DCs from the above sites can all traffic into regional pLNs. What is not at all clear is whether many can also travel in blood.
As discussed earlier (see “History”), there seems to be good evidence that DCs can migrate from the heart via the blood into the spleen and colocalize with CD4+ T cells (38, 39, 250). Here, it was proposed, they initiated transplant rejection of cardiac allografts. And yet the heart also possesses abundant pericardial lymphatics (251), and it seems generally assumed that DCs migrate from the heart into the regional pLNs. It seems completely unknown why there should be two DC “outputs” from the heart, one via the blood and one via the lymph. However, an alternative (and speculative) explanation might be that DCs within different anatomical regions of the heart differentially migrate to the pLNs or spleen (for example, from the outer epicardium in the former case, but the myocardium and/or endocardium in the latter). It might also be the case that DCs from epithelial sites such as skin and mucosal tissues migrate preferentially via the afferent lymph to regional lymph nodes, while those from nonmucosal or fully vascularized sites migrate to the spleen. This seems to be an absolutely fundamental issue that does not appear to have been clearly raised, and almost certainly not addressed.
The migration of DCs from peripheral tissues via the blood is a really important aspect that deserves much further investigation. The reasons for this include the following. First, rare DCs can be detected in efferent and thoracic duct lymph, from which they might be transported, via the thoracic duct, into the blood; this would provide an alternative explanation for DCs migrating to spleen and other tissues, rather than direct entry into the blood (91, 252). Second, there is evidence that DCs from the oral submucosa may migrate to distant lymph nodes associated with the genital mucosa (253). Third, both DCI and DCII cells can migrate via the blood to the bone marrow, where they can elicit recall responses of memory T cells that appear to home to this tissue (252). Finally, cells that closely resemble the DCII subset can migrate from peripheral tissues into the thymus (see “Tolerance”). Hence, these issues are extremely important in terms of subsequent induction of both immunity and tolerance. There seems little doubt that further insights into the immunobiology of DCs may come from much more detailed examination of their migration patterns, and particularly via blood.

The spleen (and liver)

The spleen is an encapsulated, blood-filtering organ that lacks an afferent lymphatic supply and does not possess HEVs. It is anatomically divided into white pulp and red pulp. These are separated by the marginal zone, and in some species by a marginal sinus as well. Arterial blood enters the spleen via trabecular arteries, which then become “central arteries” surrounded by the so-called periarteriolar lymphoid sheaths (PALS) of the white pulp. Within the PALS, the B cells are segregated within follicles, surrounded by the T-cell zone. Small capillaries arise from the central vein and traverse the PALS to enter the marginal zone, which is generally considered the splenic equivalent of the subcapsular sinus of lymph nodes, though containing blood rather than afferent lymph. The lymphocytes enter the white pulp from the marginal zones, rather than via HEVs as in lymph nodes, before becoming segregated in their respective areas. The blood from the central veins and marginal zones is then collected into penicillar arterioles, which become capillaries entering the red pulp. The blood in some capillaries is transported directly through these vessels out of the tissue. However, others are open-ended and “dump” the blood into the red pulp, from where it travels through splenic cords before leaving the tissue. (As the blood enters the splenic cords, aging and effete erythrocytes that lack the elasticity to pass through are retained in the red pulp and eliminated by splenic macrophages.) There are, however, structural differences in the spleen between different species, and the functional implications of these are not well understood (254).
It is presumed that pre-DCs enter the tissue of the spleen via the marginal zone and generate the two subsets of classical DCs locally. In some studies in mice, these have been characterized according to their expression of CD8 or CD4, by DCI or DCII cells, respectively; a double-negative population in other respects resembling the latter has also been described (see “Lineages”). It is generally believed that the cDCI subset is localized within the T-cell zones of the PALS. The spleen also has so-called bridging channels, which traverse the marginal sinus and connect the outer region of the white pulp to that of the red pulp. Though this has yet to be clearly demonstrated, these may represent counterparts to the medulla of the lymph nodes. If so, by analogy, the DCs within the bridging channels might be predicted to contain resident cDCII cells, which, in lymph nodes, are located within lymphatic sinuses from where they may sample the lymph. A subset of cells resembling the DCII subset is indeed present within the bridging channels, from where they may sample particulates from the blood (255). The localization of this subset has been shown to be dependent on a specific chemotactic receptor (EBI2), and these cells seem essential for antigen-specific CD4+ T-cell activation and antibody responses (256). In addition, retinoic acid may be essential for development for some of these (Notch-dependent) DC cells (161). Much remains to be learned about the regulation of DC functions within the spleen in relation to their potential roles in tolerance and immunity, though some interesting insights have been obtained (257).
Under homeostatic conditions, “nests” of DCs were originally described penetrating the marginal zone (see “History”). These cells can phagocytose apoptotic cells and subsequently home into the T-cell zones, presumably for induction of tolerance (258). These findings are reminiscent of those showing similar nests of cells penetrating the subcapsular sinus of lymph nodes. It is tempting to speculate that they may represent migratory DCs that may transiently reside in these sites before migrating constitutively in small numbers into the T zones for induction of tolerance. Conceivably, however, after infection, their homing to these sites could be accelerated and they may acquire the capacity to activate T cells rapidly against antigens they captured from lymph. Within the marginal zone there are also specialized macrophage populations that include the same CD169+ subset present in the subcapsular sinus of lymph nodes (above); if these are depleted, pathogens can spread systemically (202). In addition, fibroblastic reticular networks have been identified within the spleen, similar to those in nodes, including one ramifying through the white pulp from the marginal zone.
It might be expected that that the spleen is important for the induction of robust immunity against blood-borne antigens. However, genetically asplenic and splenectomized individuals have increased susceptibility only to encapsulated bacterial infections. This is most likely because of the absence or loss of a specialized subset of B-1 cells, and perhaps accessory marginal-zone macrophages, that can produce T-independent antibody responses against such pathogens. Defects of T-cell-dependent immunity have not generally been reported, presumably because such responses can be induced at other sites. Importantly, the venous blood from the spleen is transported via the hepatic arteries into the liver, where the two subsets of classical DCs also reside. Relatively little is known about the immunological functions of the liver, though presumably these DCs may migrate to regional lymph nodes by the same route that appears to be taken by presumptive moDCs that have captured particulates from the blood (143); it has even been suggested that the liver acts as a “biological concentrator” for blood DCs, to direct them to hepatic nodes (142). Nevertheless, in transplant settings the liver appears to be a relatively nonimmunogenic organ, so much so that liver allografts can be transplanted with little or no immunosuppression of the recipients. It has been suggested that this is due to its large bulk of poorly immunogenic hepatocytes and a relative paucity of DCs, though other factors contributing to this may yet be discovered.

Common Themes in Tissues

Some common themes emerge. The increasing number of reports of antigen-sampling cells in NLTs is provocative (see above). Epidermal LCs in skin sample antigens from under the stratum corneum impregnated with skin oils and surrounded by air (or water). Within mucosal tissues, CX3CR1+ “macrophages” of the gut sample the lumen under the mucus, while perhaps related cells of the lung sample the airspaces under mucus or airway lubricants. Perhaps they exist in the urogenital tract. Within nonmucosal and fully vascularized tissues, similar cells in contact with blood appear to do likewise. Perhaps these are also present in other tissues, such as kidney, where they may sample filtrates. It might be speculated that the function of these macrophage-like populations may be to monitor “danger.” If none is detected, they may regulate the populations of classical DCs that populate the dermis of skin, lamina propria of the mucosal tissues, and connective tissue of other organs and tissues. Hence, antigen-sampling LCs and macrophages may control the homeostatic maturation of classical DCs and/or induce regulatory functions that promote tolerance when the latter migrate into SLOs.
If danger is sensed by the antigen-sampling macrophage-like cells, they may recruit classical DCs, such as DCI cells, to sample its source for themselves. These then transport the antigen into SLOs to initiate protective immunity. Conceivably, the CDII cells are coopted if the danger spreads into the tissues. Hence, innocuous antigens such as from food or aerosols may generally result in tolerance, or perhaps “ignorance.” Commensals may be contained by barrier defenses and homeostatic “background immunity,” which includes T-independent production of IgA (these responses generally lack memory, to enable the microbiota to evolve). However, pathogens lead to the initiation of protective immunity, which may include TD IgA responses against commensals that invade the lamina propria. At the tissue and organ level, firewalls also exist. The MLNs and the liver, respectively, provide firewall protection for the lymph and blood derived from the gut. Likewise, the pLNs and the spleen may play comparable roles for the lymph and blood derived from all other peripheral tissues and organs. Presumably, related mechanisms may also be involved in protection of the brain, though much of this tissue seems very different (see below).

The Central Nervous System, Brain, and “Glymphatics”

It is generally accepted that DCs are absent from the parenchyma of the brain. Some CD11c+ populations have been identified in isolated areas such as the pituitary (259), although these could well represent monocyte-derived cells rather than classical DCs. Moreover, there is no evidence that the abundant microglial cells that are present throughout are any more than relatively sessile resident macrophages that remain within the tissues. However, cells that closely resemble classical DCs have been identified in sites such as the meninges and the choroid plexus in contact with the cerebrospinal fluid. Early studies demonstrated that small tracers could be transported from the brain into the deep cervical lymph nodes, and were believed to do so via drainage through the cribriform plate. However, the more recent finding of the existence of a specialized dural lymphatic system (sometimes termed “glymphatics”) that drains to these lymph nodes may provide an alternative explanation for these observations (260, 261). It may also provide a potential route by which DCs may migrate to SLOs, potentially to induce immunity against pathogens in the central nervous system, though this has yet to be studied in depth.
There are very close links between the central nervous system and the immune system. For example, it is quite clear that inflammation in peripheral tissues can be regulated via the hypothalamic-pituitary-adrenal axis (262). Furthermore, gut immunity can be regulated by the enteric nervous system and is controlled in part by the vagus nerve (263). Even the development of SLOs may be initiated by nerves that infiltrate tissues (222). Hence, the nervous system may regulate DC responses at least indirectly. However, both nociceptors (264) and neurotransmitters can directly influence DCs (265). Perhaps more remarkable, however, is the recent report that DCs (and macrophages) may be directly innervated in lymph nodes “by a mesh of filamentous neurofilament positive structures originating from single nerve fibers and covering each single APC similar to a glass fishing float” (266). There are also other highly provocative findings suggesting, for example, that nerve endings in spleen can form synapses with a subset of memory T cells that produce acetylcholine (262). Hence, investigation of the bipotential links between the central nervous system and the immune system, and of potential roles of DCs in them, may be a most fruitful field of study for the future.

DCs IN TOLERANCE

At a fundamental level, the immune system must discriminate with a high degree of precision between what is self and what is danger. In all settings, it is critical for survival of the host to maintain, and if necessary restore, the normal state of immunological unresponsiveness to self, or tolerance. (The converse state, immunological responsiveness to self, is, of course, termed autoreactivity.) Innate immunity is inherently self-tolerant. PRRs have evolved to recognize PAMPs and DAMPs but not normal components of self with exquisite sensitivity. (A pertinent example is the gene for TLR11, which is expressed in mice and can recognize protozoal profilin, but is a pseudogene in humans and not expressed because profilin-like molecules are synthesized [267, 268].) In contrast, a substantial proportion of the enormous repertoires of lymphocyte antigen receptors that are generated for adaptive immunity is inherently autoreactive. Moreover, nonfunctional receptors can be generated that are unable to recognize peptides in the context of MHC. This necessitates the existence of specialized mechanisms to promote and ensure the operational fitness of the total repertoire of TCRs and of the T-cell clones that express them (and similarly for BCRs and B cells).
T cells developing within the thymus are known as thymocytes. During their maturation, the developing thymocytes are subjected to the two critical selection processes of positive and negative selection, after which vast numbers will have been killed. Positive selection facilitates the further development of thymocytes that express TCRs that recognize peptide-MHC complexes, rather than either alone (though if the affinity is too high, they too are eliminated). It also controls whether the developing cells will eventually become mature CD4+ or CD8+ T cells. Negative selection involves the deletion through apoptosis of thymocytes that express TCRs that recognize self peptide-MHC complexes with a high affinity, and therefore contributes to tolerance. Within the thymus, there is also a third process whereby the fate of certain thymocytes, probably those of intermediate affinity, is redirected to generate populations of “thymus-derived” regulatory T cells (tTregs), which have counterregulatory (suppressive) functions (269, 270). These tTregs subsequently migrate from the thymus and seed the extrathymic tissues, where they contribute to the maintenance of peripheral tolerance. They are reinforced by additional populations of peripherally induced Tregs (pTregs) that are generated extrathymically and which most probably have high-affinity TCRs (271, 272). Hence, the relatively small proportion of remaining T cells that eventually emerge from the thymus have TCRs that may be able to recognize an epitope from an infectious agent that might be encountered in the future, purely by chance. (If any engineer were to design such a dodgy-looking system, they would be sacked on the spot.)

Central Tolerance

The thymus is an encapsulated and lobulated organ. Within the lobules, the tissue is broadly divided into an outer cortex and inner medulla by the corticomedullary junction, which is rich in blood vessels and afferent lymphatics. A conduit system has also been identified, akin to that described in SLOs (273). The conduits appear to originate at blood vessels, traverse the medulla, and may terminate at Hassall’s corpuscles, which develop from medullary thymic epithelial cells (mTECs) after they lose Aire expression (see below); alternatively, they may originate at the latter and extend to the blood vessels. The early thymic precursors migrate to the thymus from the blood via the corticomedullary junction and home to the outer cortex. As these progenitors develop into mature T cells, they are believed to travel through the cortex into the medulla. Finally, mature T cells leave the thymus, enter the blood, and begin their lymphocyte recirculation between different SLOs.
The thymic cortex and medulla contain dense networks of highly specialized epithelial cells that nurture thymocyte development and play largely distinct roles in thymocyte selection. The cortical thymic epithelial cells (cTECs) contain a distinct form of the proteasome termed the thymoproteasome, which generates peptides for MHC-I loading. They also express specialized lysosomal proteases, cathepsin L and thymus-specific serine protease (TSSP), which generate peptides for MHC-II loading (274). Hence, cTECs express a unique spectrum of self peptide–MHC-I and peptide–MHC-II complexes that are not likely to be generated in other tissues. These positively select CD8+ and CD4+ thymocytes, respectively. Mice lacking thymoproteasomes or the specialized lysosomal proteases have deficiencies of CD8+ and CD4+ T cells, respectively.
In contrast, mTECs have the remarkable capacity to express an enormously diverse spectrum of otherwise tissue-specific antigens (TSAs) that are normally expressed only in peripheral (extrathymic) tissues. Such ectopic expression within the thymus is regulated, in whole or in part, by a transcriptional regulator termed the autoimmune regulator (Aire) and the transcription factor Fezf2, which induce the expression of distinct subsets of TSAs (275282). Consequently, mTECs can generate tissue-specific peptide–MHC-I complexes that contribute to negative selection of CD8+ thymocytes. In addition, mTECs may undergo macroautophagy and generate tissue-specific peptide–MHC-II complexes that may contribute to negative selection of CD4+ thymocytes. Humans with mutations in Aire suffer from the devastating autoimmune disease autoimmune polyendocrinopathy-candidiasis ectodermal dystrophy (APECED), and different hypomorphic mutations may underlie many other autoimmune manifestations. Likewise, mice deficient in Aire manifest with autoimmune disease, although the condition is generally less severe than that in humans. In contrast, mice deficient in FezF2 also present with autoimmune disease, but typically affecting a different spectrum of tissues. Hence, Aire and Fezf2 control the expression of distinct subsets of TSAs.

DCs in central tolerance

The thymus contains distinct populations of DCs that are predominantly localized within the medulla, though some are also present in the cortex. Most attention has been paid to a resident cDCI population, which is most likely generated from the pre-DC progenitor, and mDCII and pDC populations, which appear to originate from extrathymic tissues and migrate to the thymus via the blood (283). The expression of Aire by mTECs also regulates the expression of chemokines that may recruit the three DC populations toward mTECs, namely XCR1 for DCI, CCL2 for DCII, and CCL25 for pDC (106, 279). While thymic DCs do not seem to express Aire, any or all of these populations may acquire ectopic TSAs, which are promiscuously expressed by the mTECs in their vicinity (284). In addition, B cells are present in the thymus, where they can be induced to express Aire and a different spectrum of TSAs in a cell context-dependent manner (285). Thymic macrophages appear primarily to be responsible for clearing the large number of thymocytes that undergo apoptosis, having failed positive or negative selection. Inflammatory cells are normally excluded from the thymic microenvironment. What seems clear is that thymic DCs of one type or another play essential roles in central tolerance (286), and that both they and the corresponding extrathymic populations have complementary roles in peripheral tolerance (see below) (287).
In mice, the expression of Aire in mTECs has been shown to be crucial for the development of a perinatal population of tTregs that persists into adult life (288). The TSAs that are generated presumably represent those that are expressed in subsets of normal tissues, or at least subsets of self components within them. It is also clear that Aire-dependent TSAs can be acquired from mTECs and presented by bone marrow-derived APCs (280, 284, 289). However, these respective cell types play distinct roles in shaping of the adult T-cell repertoire through both deletion of autoreactive T cells and the generation of distinct populations of tTregs. It has been estimated that approximately half of both the Aire-dependent deletion of autoreactive thymocytes and selection of tTregs may be controlled by bone marrow-derived APCs that acquire TSAs from mTECs (280). The APCs responsible for generation of tTregs are most likely cDCI cells (290), although they are present at much lower frequencies in the thymus of perinatal mice than adults. It has also been suggested that Aire induces apoptosis of mTECs, potentially providing an abundant source of antigens for the cross-presenting cDCI population. Direct presentation of TSAs by mTECs, primarily or exclusively, induces the deletion of autoreactive CD8+ thymocytes. In contrast, through their additional costimulatory activities, which may be required for robust generation of tTregs, the cDCI cells may be particularly adept at controlling the generation of tTregs. In the periphery, these may regulate the functions of the mDCI population within normal tissues. What does not seem to have been explained is why the intrathymic cDCI subset, which would otherwise preferentially present antigens to CD8+ T cells in the periphery, seems so important in selecting CD4+ tTregs.
It has been suggested that CD11c+MHC-II+ cells, and most likely a population of mDCII cells as defined by expression of CD11b and/or CD172 (SIRPα), can traffic from peripheral tissues via the blood into the thymus. This comes from studies in mice that have used, for example, adoptive cell transfers (291, 292), bone marrow chimeras and culture systems (290), and parabiosis models (291, 293, 294). In general, however, it is not always possible to exclude the trafficking of progenitors such as pre-DCs rather than fully differentiated cells, particularly after transfer of cells subjected to in vitro manipulations such as expansion with Flt3L. What is clear is that, after intravenous injection of labeled soluble tracers and antigens, labeled cells resembling mDCII can subsequently be detected within the thymus (290, 292, 295). Here they appear to induce both deletion of autoreactive CD4+ T cells and generation of CD4+ tTregs (292, 295). Importantly, however, there may also be a large population of resident cDCII cells in the thymus, derived from the pre-DCs that also generate the cDCI population. This possibility is generally overlooked but was in fact acknowledged in a well-cited study that nevertheless classified these cells as being of extrathymic origin “for convenience” (293), and is consistent with findings of others (295). It has also been shown that cells resembling DCs are closely associated with recently described medullary conduits, though their phenotype was not fully explored (273). These findings are reminiscent of those described for SLOs, in which presumptive cDCII cells are closely juxtaposed to conduits from where they may sample small soluble molecules. The possibility that such molecules may also gain access to a resident cDCII population via thymic medullary conduits thus deserves further study.
Other studies have clearly shown that if particulate tracers too large to enter the thymus are injected intravenously, they can subsequently be detected in cells resembling the mDCII subset within the thymus (293, 296). However, this is also reminiscent of other studies that have documented the capture of such tracers by monocytes in peripheral tissues, and their subsequent differentiation into monocyte-derived DCs that traffic to the lymph nodes (141, 143). Interestingly, one study has documented the perivascular capture of a soluble tracer by cells that subsequently migrated in a CCR2-dependent manner into the thymic cortex (rather than medulla), where they remained in close proximity to blood vessels (292). However, CCR2-dependent migration is typically associated with monocyte migration and does not seem to have been described as important for migration of classical DCs. The thymus, similar to any other tissue, presumably requires defense against infection. This could therefore reflect a mechanism that might be involved in induction of protective (thymic) immunity, rather than tolerance, for example, after further trafficking through lymphatics into the regional lymphatics. This too deserves further investigation.
Additional evidence for trafficking of a tolerogenic presumptive DC population from peripheral tissues into the thymus has come from other studies. For example, after skin painting with a fluorescently labeled contact-sensitizing agent, labeled CD11c+ cells were detected in the thymus, but their accumulation was inhibited by blockade of the α4 integrin of very late antigen-4 (VLA-4), which therefore seems to plays a central role in their trafficking to the thymus (291). Furthermore, transgenic expression of a membrane-bound antigen exclusively in cardiomyocytes resulted in thymic deletion of antigen-specific CD4+ T cells, and this too was prevented by similar blockade. The former could represent a peripheral DC population that was induced to migrate in response to “sterile” inflammation, perhaps for induction of intrathymic tolerance against “damaged” tissue antigens (possibly via induction of tTregs). In contrast, the latter may represent migration from a “steady-state” tissue for induction of deletional tolerance against normal tissue antigens. Further studies are required, however, to identify the precise cells involved and determine whether or not such differences exist.
It has also been found, using techniques noted above, that pDCs can migrate from the blood into the thymus. These cells may endocytose soluble tracers and antigens after intravenous or subcutaneous injection (291, 293, 295, 296) and migrate to the thymus, where they appear to delete antigen-specific CD4+ T cells and induce CD4+ tTregs (295, 296). The apparent capture by phagocytosis of intravenously or subcutaneously injected particulates by pDCs has also been demonstrated (296), with subsequent migration of particle-laden cells to the thymus in an α4 integrin-dependent manner (296). The migration of pDCs into the thymus was shown to be dependent on CCR9. Interestingly, pDCs that were stimulated by TLR9 agonists appear to be excluded from the thymic microenvironment (296). If so, this might be a mechanism to prevent the transport of infectious viruses or microbes into the thymus. It has also been shown that traffic of adoptively transferred mDCII cells is much decreased after maturation in response to a TLR4 agonist (291). Further investigation is needed to establish whether this is a general mechanism to ensure that DCs can only traffic to the thymus under homeostatic conditions, but are prevented from doing so from infected tissues.
In summary, Aire and Fezf2 control the expression of TSAs by mTECs that subsequently induce deletion of autoreactive CD8+ thymocytes. These TSAs can be acquired by thymic cDCI cells, which subsequently induce tTregs that may perhaps control the activity of the mDCI populations in the periphery. In addition, the cDCII and/or extrathymically derived mDCII populations, together with pDCs, may induce the deletion of CD4+ T cells and the induction of tTregs against additional peripheral tissue antigens. Collectively, these mechanisms can lead to the deletion of newly generated autoreactive CD4+ and CD8+ thymocytes and generate a diverse spectrum of tTregs specific for peripheral tissue antigens. Some of these may possibly have roles in regulating the homeostatic maturation of DCs (see below) or in dampening their maturation at sites of homeostatic inflammation (see “Tissues”).
Under physiological conditions, any DC expresses its own self peptide-MHC complexes, which represent the normal epitopes that can be generated from its own cellular and molecular components (i.e., those that make a DC a DC rather than any other cell type). It would therefore seem essential to ensure that autoreactive thymocytes that might be specific for such components, which might be termed DC-specific antigens (DCAs), are also deleted or regulated. For example, if the concept that classical DCs are essential for initiating primary TD responses is correct, as generally seems to be the case, then DCs would be able to readily activate any autoreactive T cells that were specific for their own DCAs. Potentially, this would be drastic, as it could ultimately result in elimination of all DCs from SLOs and NLTs. Hence, one could argue that, in addition to inducing tolerance to a diverse spectrum of TSAs, a crucial role of the thymus may be to induce tolerance to the specialized cells that can initiate primary T-cell-dependent responses, and particularly the thymic cDCI and cDCII subsets. In this respect, therefore, the landscape of immunostimulatory cells within the thymus may mirror that which exists in the periphery. A corollary of this hypothesis is that pDCs and/or B cells can also activate primary T-cell responses, which under certain circumstances is possible, or that they are present within the thymus for different functions (e.g., for presentation of peripheral or Aire-dependent antigens, respectively).

Peripheral Tolerance

Peripheral tissues contain different populations of Tregs (269, 297). The centrally generated tTregs home to these sites from the thymus and are supplemented by pTregs that are generated within the periphery; conversely, it seems likely that pTregs, as well as small numbers of naive T cells (298), may home back to the thymus, for reasons largely unknown. While tTregs develop from thymocytes during their differentiation, pTregs can develop from mature, naive T cells that have these regulatory functions “imposed” upon them. In both settings, the master transcription factor FoxP3 regulates their development and functions (299). Mice that lack expression of FoxP3 develop lymphoproliferative autoimmune disease but can be rescued if normal expression is restored. Rare humans lacking FoxP3 expression develop IPEX (immune dysregulation, polyendocrinopathy, enteropathy, X-linked). Typically, they suffer from severe gut and skin inflammation, as well as autoimmune-mediated damage to other tissues and organs such as the thyroid. (It is interesting that many autoimmune conditions resulting from defects in central tolerance involve the endocrine tissues; see below.)
The thymus is critically important particularly for generating tTregs around the perinatal period, which migrate to peripheral tissues. Those that were induced by thymic DCs may, in turn, be selected, expanded, and maintained by peripheral DCs that express TSAs (300). Neonatally thymectomized but not adult mice, as well as rare humans with thymic hypoplasia (DiGeorge syndrome), suffer from severe autoimmune disease caused by the early, or aberrant, populations of T cells they respectively develop. A major stimulus for recruitment of tTregs into some tissues is likely the early colonization with commensals. For example, a wave of Tregs is recruited into neonatal skin of mice in response to establishment of its microbiota (301). Peripheral tolerance in the intestine and lung is associated with an influx of tTregs, perhaps also in response to their distinct and diverse populations of commensals (302). Hence, it appears that tTregs may be particularly important in maintaining tolerance to the microbiota at such sites. However, other evidence indicates that the majority of pTregs in the intestine are generated against food antigens (303). Nevertheless, from TCR repertoire analysis it is quite clear that the populations of Tregs vary enormously between different tissues (thousands of different specificities have been detected in peripheral sites, compared to just hundreds in SLOs) (297). The largely Aire-dependent, and hence TSA-specific, population of tTregs might control the induction of autoimmune T-cell responses against TSAs that might be liberated, for example, during tissue damage caused by invading commensals. But if, as seems likely, classical DCs are crucial for T-cell activation, then this would need to be regulated at the level of DCs. Alternatively, or in addition, a hypothetical population of DCA-specific tTregs (above) might also regulate the responses of DCs that captured and expressed commensal or other antigens. (This is not, however, intended to imply that Tregs only act on DCs and may not also regulate the functions of other cell types.)
In contrast to skin and gut (see above), tTregs may not be recruited to tissues lacking commensals, such as the exocrine pancreas, to which conventional T cells may normally be “ignorant” (302). If so, this may pertain to other nonmucosal or fully vascularized organs and tissues, such as heart and skeletal muscle. Perhaps this necessitates additional mechanisms for the maintenance of tolerance under other circumstances, such as might be provided by pTregs in such sites. It is currently believed that the continuous, homeostatic maturation and migration of DCs from peripheral tissues into SLOs plays a central role in generating pTregs (see below). In addition, there is accumulating evidence that expression of FoxP3 and regulatory function can be induced or imposed on populations of effector and memory T cells (304, 305).
Aire can also be expressed in the periphery and may control expression of TSAs in a cell context-dependent manner, different from those generated by mTECs. So-called extrathymic Aire-expressing cells have been identified in mouse SLOs (277, 306). These are bone marrow-derived cells but seem to be distinct from DCs, although Aire expression has also been reported in splenic marginal-zone DCs (probably the cDCII subset) (307), as well as other cells such as CD14+ monocytes (308). There is evidence that extrathymic Aire-expressing cells can directly inactivate, or perhaps anergize, CD4+ T cells even under inflammatory conditions in the lymph node (rather than inducing deletional tolerance or Tregs) (277). Another transcription factor, deformed epidermal autoregulatory factor 1 (Deaf-1), also controls further ectopic TSA expression in the periphery (309). Deaf-1 is expressed by lymph node stromal cells (LNSCs) and/or FRCs and can also be expressed in the pancreas (310). Hence, it appears that Aire and Deaf-1 may regulate ectopic expression of distinct subsets of TSAs in the periphery (i.e., within both SLOs and at least some NLTs). Moreover, FRCs in lymph nodes may themselves delete autoreactive CD8+ T cells that recognize the TSAs they express (309). Whether or not such antigens can also be acquired by peripheral DCs, perhaps for generation of pTregs, awaits further investigation. However, a converse pathway has also been demonstrated, in which LNSCs acquire peptide-MHC complexes from DCs and subsequently delete CD4+ T cells (311). Whether they also strip other membrane molecules from DCs (or “cross-dress” in them [312]) remains to be seen, but could explain observations that LNSCs may stimulate CD4+ T-cell responses (313).

DCs in peripheral tolerance

Within peripheral tissues, DCs can control the homeostatic expansion of Tregs. These may include the tTregs recruited to these sites that were originally generated by DCs within the thymus (300). In turn, Tregs may control the maturation of DCs, which express tissue antigens and which might otherwise be sufficient for T-cell activation (314). Some of these interactions have been directly visualized in mouse lymph nodes, where, for example, clusters of Tregs and effector cells were found to be tightly associated with migratory DCs in paracortical regions (315). In general, persistent interactions between CD4+ Tregs and DCs are essential for maintaining peripheral tolerance. If these are experimentally disrupted, for example, by preventing the expression of MHC-II on DCs, severe disease is induced (316). This can involve severe gut inflammation that is driven by commensals and can be prevented by their removal. It also leads to a failure to control CD8+ T-cell tolerance and results in severe autoimmune tissue damage, not only in the intestine but also in sites such as the pancreas noted earlier (317). The normal control of CD8+ T-cell responses seems to be regulated by Tregs, which most likely act at the level of helper CD4+ T cells in a cytotoxic-T-lymphocyte-associated antigen 4 (CTLA-4)-dependent and/or programmed death 1 (PD-1)-dependent manner (318, 319). These and probably other coinhibitory interactions may also contribute to the capacity of Tregs to dampen DC responses sufficiently to maintain normal homeostasis, perhaps especially at sites of homeostatic inflammation driven by commensals. In addition, other mechanisms of regulation by Tregs may be through production of IL-10 and/or TGF-β, as well as perforin-mediated cytolysis of DCs. Nevertheless, there is likely to be a delicate balance between controlling the activation of DCs locally to prevent the induction of immunity and allowing their migration into SLOs to induce pTregs perhaps against the commensals and other antigens they acquired. What is not clear, however, is the relative importance of tTregs and pTregs in controlling these states (320). It would also seem critical to ensure that such regulatory interactions are normally maintained while immunological responses are being induced by other DCs at peripheral sites of infection.
Numerous studies have documented the constitutive migration of DCs from the tissues under normal, homeostatic conditions (29, 321). Both types of classical DCs migrate constitutively from tissues such as gut, skin, and liver in a relatively immature state. Most probably the DCI subset can transport apoptotic cell contents from the gut and the lung via the afferent lymph (56, 239). These cells express relatively low levels of costimulatory molecules that may be inadequate to initiate T-cell activation against the soluble or cell-derived antigens they present, but sufficient to drive the production of Tregs and/or anergy or deletion (88, 299, 322). The NF-κB transcription factor has been shown to be essential for both constitutive migration and the subsequent induction of tolerance against normal tissue antigens (321). There is also evidence from studies in mice lacking IRF4 that this transcription factor regulates the migration of cDCII cells from the tissues under both homeostatic and inflammatory conditions, as well as contributing to their development from progenitors and enhancing their MHC-II antigen presentation capacity (112). Such information is currently lacking for the mDCI population, which, in mice, expresses CD103. However, a natural ligand for CD103 is E-selectin, which plays an essential role in the turnover of apoptotic cells (323). In addition, disruption of E-cadherin-mediated intracellular interactions between mouse DCs in culture induces a transcriptionally distinct maturation process. During this, the cells increase expression of costimulatory molecules but fail to mature fully or to secrete proinflammatory cytokines (324). At least in part, this process is regulated by β-catenin, which can form complexes with E-cadherin and has also been shown to control the homeostatic migration of DCs from the intestine and the subsequent induction of pTregs. Hence, while constitutive migration of mDCII cells may be part of a normal, IRF4-dependent differentiation program, that of cDCI cells may be regulated by apoptotic turnover in tissues and/or through a CD103-dependent mechanism controlled by a different program.
If DCs are crucial for the induction and maintenance of tolerance, then at first sight one might predict that deletion of DCs, or selective populations of DCs, would lead to autoimmune disease. This has been demonstrated in one setting in which all CD11c+ cells were deleted, with resultant induction of devastating autoimmune disease (325). In a related model, which did not, however, delete pDCs and LCs, myeloproliferative disease resulted instead (326). Other evidence is indirect; for example, intrathymic expression of XCL1 is controlled by Aire apparently to recruit DCI cells toward mTECs, and deficiency leads to autoimmune disease (107). Nevertheless, some have suggested that DCs play only minimal roles, if any, in tolerance. In part, this is because selective depletion of the DCI or DCII populations in IRF8- or IRF4-deficient mice, for example, does not result in autoimmune manifestations. Crucially, however, these manipulations may deplete not only the DC populations that are required for induction and maintenance of tolerance but also the very same populations that are required for induction of immunity. One possibility is that there is redundancy in immunological functions, such that depletion of one DC population spares the capacity to induce tolerance to overlapping antigens by the other. Alternatively, it is conceivable that each DC population induces tolerance to a distinct and selective repertoire of self-derived antigens, which is perfectly mirrored by those it naturally presents in the periphery.

DCs IN IMMUNITY

This section focuses, mainly at a cellular level, on the role of DCs and conventional αβ T cells in defense against infection. In particular, it deals with the functions of DCs in driving the different stages of adaptive immune responses. These stages are broadly divided into (i) sensing of infection and tissue damage, (ii) the priming phase, (iii) the effector phase, and (iv) the memory phase. All are, however, closely interlinked.

Sensing of Infection and Tissue Damage

Any tissue monitors itself, or is monitored, to sense infection and damage. The PRRs of epithelial cells in mucosal tissues are generally localized on the tissue-facing, abluminal surfaces. Hence, they preferentially recognize invaders of the lamina propria rather than commensals contained within the lumen. In addition to specialized populations of macrophages, which may play homeostatic rather than primary defense roles, most NLTs contain resident cells such as mast cells within the connective tissue or mucosae. Tissues such as the dermis of skin, the lamina propria of the gut and lung, and the liver additionally harbor different populations of ILCs (327329). They are also present in SLOs such as the spleen, MLNs, and tonsils (330). ILCs are lymphoid cells in origin, deriving from the CLP, and they closely resemble T cells but produce innate-like responses and lack expression of TCRs. They include three different types that are classified as ILC1, ILC2, and ILC3 cells. In response to infection or tissue damage, these secrete polarized patterns of cytokines very similar to those produced by Th1, Th2, and Th17 cells, respectively. They also express the corresponding master transcription factors T-bet, GATA-3, and ROR-γ, respectively. Innate-like NK cells have also been assigned to the general class of ILCs and closely resemble CTLs in their cellular cytotoxicity and cytokine secretion profiles. ILCs may represent very ancient lineages that evolved before the emergence of conventional T cells and presumably classical MHC molecules. Their existence reinforces the common view that the immune system focuses on sensing general classes of infectious agents and generating characteristic immunological outputs to deal with each.
Within peripheral tissues, DCs coexist with diverse types of tissue-specific and resident cells and may be bathed in complement components (etc.) produced by resident macrophages; the latter indeed may also regulate DC functions (see “Tissues”). Collectively, however, these may constitute local immunological networks that monitor their environment for infection and damage, immediately mount and amplify innate effector responses, and rapidly trigger inflammation to recruit new effectors. The composition of the DC populations itself also varies between tissues: classical DCs, LCs, and/or pDCs under homeostatic conditions in some, with pDCs and/or moDCs during and after inflammation in these or others (the same will be true for regulatory, effector, and/or memory T cells). Any or all DCs may contribute to innate responses, but it may be the classical DCs that are also essential for initiating subsequent adaptive responses. First, each classical DC may sense alarmins, PAMPs, and/or DAMPs within its own tissue microenvironment. This type of information from many varied inputs is then likely to be integrated, and in turn, each cell may generate a finely tuned molecular and cellular response as an output. Second, any DC may internalize antigens (through pinocytosis, receptor-mediated endocytosis, phagocytosis, and/or macropinocytosis) and generate representative peptide-MHC complexes from them. Third, DCs then migrate from NLTs into SLOs, where they can select antigen-specific T cells, deliver costimulatory signals to activate them, and modulate the functions of activated T cells at least in part according to the information they received in their peripheral microenvironment.

Heterogeneity of DC responses

While the above may seem obvious to most in the field, it raises two very important and closely related issues that have not yet been adequately addressed. The first is the extent of heterogeneity of responses that can be generated by peripheral DCs, and the second is the resultant diversity of T-cell responses that are subsequently induced. In the first case, it seems likely that, at any point in time, DCs in peripheral tissues are at different stages of homeostatic maturation and therefore heterogeneous. Such heterogeneity may be reinforced by the particular microenvironment that any such DC senses during inflammatory responses. An important issue therefore is whether, in the latter case, all DCs are driven to homogeneity during maturation before the critical stage is reached when they egress from the tissue, or whether they migrate as a highly heterogeneous population (331). At a population level, core transcriptomic signatures of the two subsets of classical DCs and pDCs converge during maturation in response to viral infection, although other cell-specific characteristics are retained (331), but clearly this says nothing about diversity at a single-cell level. The issue of DC heterogeneity has only recently begun to be studied (in contrived experimental settings) but indicates that DCs can be diverse at the single-cell level (332, 333). It also seems likely that, during the priming phase, DCs may select T cells with TCRs of different affinity for the peptide-MHC complexes they present, and TCR affinity is directly linked to the subsequent (effector) responses of these T-cell clones (334). Moreover, because of the plasticity of T cells, they respond differentially depending on the extent of costimulatory and other signals they receive. Hence, it seems likely (though yet to be proven, or otherwise) that highly heterogeneous populations of migratory DCs can subsequently generate highly diverse populations of activated and, subsequently, effector T cells.

The Priming Phase

Primary T-cell responses are initiated within SLOs. There is abundant evidence that each subset of classical DCs has specialized capacities to activate naive, antigen-specific T cells (and hence TD responses), and this is essential for generating protective responses (see “Lineages” and “Tissues”). Other DC subsets can express MHC-II and costimulatory molecules, as may other lineages, such as B cells. It is, however, important to stress that expression of these molecules is also essential for the induction of Tregs (see “Tolerance”) and may regulate the differentiation of effector and/or memory T cells. Moreover, the exuberant expression of MHC or costimulatory molecules that can be driven by various stimuli in vitro does not necessarily translate to the in vivo setting, where it may be more tightly regulated and controlled. The evidence that DCs can initiate primary T-cell responses in vivo seems strongest for classical DCs, perhaps less so for moDCs, and least convincing for LCs and pDCs.

Segregation of DC responses

Conventional T-cell responses are highly segregated. CD8+ and CD4+ T cells are restricted to recognizing peptide–MHC-I or peptide–MHC-II complexes, respectively. Consequently, recognition is focused toward the respective sources of antigen from which peptides can be generated and loaded onto each type of MHC molecule. Hence, CD8+ T cells are focused toward recognition of intracellularly derived “endogenous” antigens (from the cytosol), while CD4+ T cells recognize extracellularly derived “exogenous” antigens (which are endosomal). However, mere recognition is not enough. To respond, each subset of T cells requires recognition of peptide-MHC complexes together with costimulation that is provided by DCs particularly after they recognize PAMPs and/or DAMPs. Therefore, an extreme view might hold that responses of the two subsets of classical DCs may likewise be segregated. They may have evolved to acquire antigens and to sense DAMPs and PAMPs, preferentially from the corresponding sources that T cells recognize. For DCI cells, these include dead, dying, or damaged cells; apoptotic and necrotic bodies; and infectious agents contained within them (e.g., cytosolic viruses). In contrast, DCII cells may selectively acquire extracellular antigens (e.g., phagocytosed bacteria) and sense the PAMPs associated with them, as well as DAMPs that are released or produced in soluble form. Hence, each may present antigen to T cells and express essential costimulatory molecules to activate them only if they have concomitantly sensed the PAMPs and DAMPs within each respective context.
The relative segregation of discrete antigen presentation and costimulation pathways between the two subsets of classical DCs could promote and ensure the differential activation, and hence segregation, of CD8+ versus CD4+ T-cell responses. This segregation may be further reinforced by the rapid shutdown of further antigen sampling once DC maturation has commenced. It is, however, clear that DCI cells can capture exogenous antigens and quite possible that DCII cells acquire cell-derived components. Potentially this may facilitate subsequent interactions with activated CD4+ and CD8+ T cells, respectively (since activated T cells are no longer dependent on primary costimulation). In this way, DCII cells may selectively activate CD4+ T cells and help to regulate the responses of activated CD8+ T cells, and vice versa for DCI cells (see below). It is also possible that these (converse) interactions may induce early T-cell activation, but that robust T-cell responses require subsequent interactions with the other “default” subset (see “Tissues”). It should be noted that human DCs may also selectively express CD1 molecules, which bind lipids much as classical MHC molecules bind peptides. DCs may therefore differentially present lipid-CD1 complexes to populations of specialized innate-like T cells that recognize them (335), and may regulate or be regulated by them.

Integration of DC responses

The two subsets of classical DCs may function synergistically to enable the adaptive immune system to sense peripheral infection and tissue damage qualitatively, quantitatively, and temporarily, through a continuous stream of short-lived migratory DCs arriving in the SLOs (each of which may have a snapshot of prevailing conditions in the periphery). The mechanism for this would involve multiple sequential interactions between any given T cell and different DCs (see below). In turn, the adaptive immune system may be able to continually fine-tune its responses through many different immunological outputs. For example, the differential expression of TLR3 and CLEC9A (DNGR-1) by mDCI cells, coupled with that of TLR5 and TLR7 by mDCII cells, could ultimately permit discrimination between peripheral infection by an RNA virus or a flagellated bacterium; between the different stages of an RNA viral infection, through sensing of double-stranded RNA replicative intermediates or of genomic RNA before infection; and between cytopathic or noncytopathic infections, through sensing of apoptotic or necrotic bodies. Diverse PRRs for PAMPs and DAMPs are selectively, differentially, or commonly expressed by classical DCs (336). Hence, it is possible that the adaptive immune system may ultimately discriminate between even closely related infectious agents with exquisite selectivity and sensitivity. Taking this one step further, it seems possible that it may also clearly differentiate not just between apoptosis and necrosis but also between different forms of regulated necroptosis (337340), and hence monitor different stages of peripheral tissue damage and healing.

Activation of T-cell responses

Migratory classical DCs have a relatively short life span within the SLOs, on the order of 1 to 3 days. The same is thought to be true for resident classical DCs, which are constantly regenerated from pre-DCs within these tissues. Migratory DCs die within SLOs, but presumably their contents could be internalized and recycled by other cells, including resident DCs, thus further modulating responses of the latter and perpetuating the T-cell response until the peripheral source of antigen is eliminated. It is estimated that naive lymphocytes transiently reside within SLOs for between 6 and 24 h before leaving to continue their recirculation between these tissues if not activated (183); CD4+ T cells may dwell for the shortest times, CD8+ T cells for longer, and B cells for the longest. The intricate architecture within the SLOs seems to have evolved to greatly increase the probability, and effectively ensure, that any antigen-specific T cell can be activated by a DC that expresses its cognate antigen as a peptide-MHC complex. In large part, this may result from the T cells and migratory DCs crawling (or gliding) along the fibrils of the FRC network rather than attempting to traverse the spaces between them (182, 341345). In this way it is estimated that a DC can sample ∼5,000 T cells every hour (183). When a T cell meets its cognate DC, they remain tightly associated for up to 16 to 20 h, though this can be as short as 3 to 4 h, during which time the T cell is activated. Subsequently, the activated T cell detaches from its priming DC but remains within the SLO for some time (see below).

The Effector Phase

Some of the complex choreographies that are executed by T cells and DCs within different microenvironments of SLOs during priming have been noted (see “Tissues”). It is quite clear that both activated CD4+ and CD8+ T cells can make further sequential interactions with other cognate DCs in the tissue. During these, they presumably start to express their effector programs, which may be serially modulated by the different sets of information they receive from successive DCs (for example, depending on the quantity and quality of costimulatory and/or coinhibitory signals each can deliver). The frequency of these interactions is further enhanced by the increased dwell times of activated T cells within the SLOs compared to their naive counterparts, which are controlled in part by IFN-I (such as might be secreted by pDCs) (344). T cells proliferate at an exceptionally fast rate compared to other eukaryotic cells, each cell cycle being completed within just 6 to 8 h. This leads to a rapid expansion of the antigen-specific clones, each of which may interact with migratory and/or resident DCs. In turn, the effector programs of each T cell may be further modulated depending on the particular microenvironments that migratory DCs experienced in the periphery and/or that resident DCs experience within the SLOs. Potentially, the responses of any DC itself may be modulated by small molecules delivered via conduits or mediators produced by monocytes (181), moDCs, or “swarms” of neutrophils (345) that enter SLOs (see “Tissues”). This may drive further diversification of T-cell responses, producing even more heterogeneity in early effector-T-cell populations.
Some T cells execute their effector functions within the SLOs. These include CD4+ T cells that activate CD8+ T cells and potentially drive the generation of CTLs locally or later within NLTs. They also include CD4+ T cells that migrate to the borders of T-cell zones and the follicles where they activate antigen-specific B cells. Some CD4+ T cells may alternatively differentiate into follicular helper T cells, which are involved in the germinal center reaction; precisely where this latter fate is decided is not entirely clear, though it may be controlled at least in part by moDCs (346). Other CD4+ effector T cells may instead develop into memory T cells (see below). However, some CD4+ T cells migrate from the SLOs and execute their effector functions instead within NLTs (see below). There is good evidence that homing of effector T cells to different NLTs can be dependent on where their antigen originated, and this is controlled at least in part by DCs; hence, effector T cells may selectively migrate to distinct sites such as the skin, gut, or lung (347349).

Polarization of T-cell responses

For decades, many in the field have focused on polarized types of T-cell effector functions, such as Th17, Th1, and Th2 responses, which have been well studied at the cell population level (348). There has also been a tendency by some (though not all) to correlate these responses with different DC subsets that might induce them. This type of view originated from the initial description of the Th1-Th2 paradigm followed by many early studies (noted elsewhere [61]) showing that adoptive cell transfer of different DC populations, or selective targeting of antigens to one or the other, resulted in polarized Th1 or Th2 responses (105). This general concept was extended following the discovery of Th17 cells. It now seems to be believed by many that Th1 responses are typically induced by DCI cells, while Th17 and Th2 responses are more generally induced by DCII cells (whether or not of different subsets). Such notions indeed seem consistent with known functional specializations of these subsets (see “Lineages”). And yet there have been many contradictory findings, depending on the precise experimental system that is studied. Moreover, the description of apparently different polarized subsets, such as Th9 and Th22 cells that may play specialized roles in defense of skin or gut (349), is hard to reconcile with this general line of thinking unless further DC subsets are invoked. Furthermore, during natural infections of mostly outbred species such as human, such highly polarized patterns of T-cell behavior have been hard to find. It is, however, now absolutely clear that T cells are highly plastic and that, in any given response, CD4+ T cells coexpressing different combinations of master transcription factors can be readily detected (350); responses of CD8+ T cells are likewise highly diverse (351).
Rather than invoking a “one DC subset, one T-cell subset” view, an alternative is that, at the single-cell level, the effector function of any given T cell represents a summation of all the influences to which it has been subject during its short-term (but adequately long-lived) residence in an SLO and/or later in NLTs. In the simplest case, the initial activation of a CD4+ T cell by a DCI cell followed by serial interactions with DCs of the same subset may bias responses toward Th1 polarization. A similar case might also be made for DCII cells and Th2 responses, while reciprocal interactions between the two DC subsets might result in others, such as Th17 responses. Nevertheless, the serial interactions of any given activated T cell with DCs are likely to be considerably more heterogeneous in nature. It therefore seems possible that, during the effector phase, many different permutations and combinations of effector functions are generated at a single-cell level (though at a population level they may seem biased). Hence, from this large effector cell pool, only some T-cell clones may have the precise sets of effector functions that can promote the elimination or control of any given infectious agent. This hypothesis predicts the existence of heterogeneity in antigen-specific effector T cells at a single-cell level.

Heterogeneity of T-cell effector responses

Recent studies have revealed a remarkable diversity of effector-T-cell functions at the single-cell level, even during apparently polarized responses to a defined infectious agent. Earlier work that studied individual CD8+ T cells responding to a given antigen by multiplex analysis demonstrated considerable heterogeneity within the population (352, 353), though 14 different types of overall response were initially proposed. (Good luck to those who would seek 14 new DC subsets to drive these responses.) Recent high-dimensional, multiparametric techniques have reinforced these findings, but also tend to suggest that T-cell effector responses are even more heterogeneous and perhaps more closely resemble a continuum (354). If the above hypothesis is correct, then presumably an adequate diversity of effector functions can be generated for those clones that remain in the SLOs to execute their responses. Conceivably, however, the effector functions of those clones that migrate to NLTs, and their clonal progeny, could be further diversified in the periphery. If so, an essential cell type in this respect may be the moDC (136, 145, 153), though any APC might of course also contribute. Through their capacity to capture antigens within the periphery, moDCs are likely to express peptide-MHC complexes that can drive further proliferation of effector T cells for as long as any infection persists. They certainly appear to be particularly important in the effector phases of CD4+ T-cell responses. Potentially, too, moDCs may be modulated by their microenvironments such that the population as a whole is highly diverse at a single-cell level (compare classical DCs above). While activated T cells are not dependent on primary costimulation, the expression of qualitatively and perhaps quantitatively different costimulatory (and/or coinhibitory) molecules by moDCs may further differentially modulate the effector functions of each T-cell clone with which they interact. If this is the case, then conceivably LCs may function similarly during T-cell effector phases within the skin and structurally similar sites. Possibly pDCs could also contribute to diversification within the mucosal tissues (and perhaps even at inflammatory sites and in cancers).

The Memory Phase

Memory T cells can persist for the lifetime of any mouse or human, though they decline in numbers with time. While much still remains to be discovered, it seems clear that CD4+ memory T cells can persist in the apparent absence of the antigen to which they were induced, and that CD4+ T cells are also essential for the generation and maintenance of CD8+ memory T cells. However, the latter are better understood primarily because of the relative ease of studying their function in in vitro assays. Nevertheless, based on their phenotype and function, CD4+ memory T cells have been divided into central memory (Tcm) and effector memory (Tem) subsets, while others that appear to reside in peripheral tissues are termed resident memory (Trm) cells (355). The former, Tcm cells, recirculate between SLOs and can rapidly reexpress molecules such as CD40L on restimulation; within SLOs they may be positioned most closely to potential sites of pathogen entry (356). In contrast, Tem cells tend to populate peripheral tissues and can rapidly reacquire effector cell functions such as polarized patterns of cytokine secretion. It has been suggested that Tcm cells can develop into Tem cells after restimulation.
There is still much to be learned about the factors that regulate the generation of CD4+ and CD8+ memory T cells. However, pDCs have been implicated particularly in this process. As noted earlier, these cells may play specialized roles in viral infections and perhaps particularly in the regulation of CD8+ T-cell and CTL responses (see “Lineages”). As was also noted, pDCs can secrete high levels of IFN-I. Importantly, at the tissue level, it is known that IFN-I can prevent the egress of lymphocytes from SLOs (344). This latter example may be most relevant to the phenomenon of lymph node shutdown, which is observed soon after infection and can be induced in regional lymph nodes after injection of IFN-I into the skin (G. G. Macpherson, personal communication). The markedly increased cellularity of SLOs during this period may increase the frequency and duration of interaction between T cells and their cognate DCs, and possibly enhance the diversification of effector cells. Because CD8+ T cells generally have a higher activation threshold than CD4+ T cells (200), it is possible that this could also apply to their development into memory T cells. Hence, in principle, the capacity of pDCs to contribute to lymph node shutdown and extend intercellular communications could directly or indirectly contribute to robust CD8 memory-T-cell generation. Whether or not there might be segregation between moDCs and pDCs for generation of CD4+ and CD8+ memory T cells remains to be seen. The relative contributions of the different subsets to reactivation of memory cells during secondary responses is unclear. However, there is good evidence at least that classical DCs can elicit such recall responses, presumably in SLOs and also in bone marrow and NLTs (104, 357, 358).

DISCUSSION AND CONCLUSION

Collectively, the different DC subsets that are the focus of this review may constitute an integrated cellular network that is essential for both tolerance and immunity in the adaptive immune system. In this final section, two main areas will briefly be discussed. The first provides some speculations on the evolution of DCs. The second revisits concepts of the generation of diversity and clonal selection, but within the context of DCs driving tolerance and immunity.

DCs in Evolution

From an evolutionary standpoint, and if ontogeny indeed recapitulates phylogeny, it is tempting to speculate about the origins of DCs. The embryonic, yolk sac-derived population of LCs may be the most ancient of all. All animals have hematopoietic tissues that can generate cells, such as the wandering amoebocytes observed by Metchnikoff in starfish larvae or, later in evolution, blood-circulating cells such as monocytes (5). It is possible to envisage that cells such as amoebocytes might first have evolved into resident macrophages within the tissues, including LCs in the epidermis of skin. Potentially the earliest LCs, generated by a form of primitive myelopoiesis well before lymphoid cells evolved, may have played homeostatic roles in the skin of ancient fish, but might have used phagocytosis in defense. From extant species we know that LCs are present in all jawed fish that have been studied, but this does not seem known for agnathans. During the earliest stages of adaptive evolution in jawed fish, LCs may then have evolved into essential immunostimulatory cells for immune responses within skin-associated lymphoid tissues (359) (unless they could migrate to the spleen). The later LC populations successively derived from fetal liver- and bone marrow-derived monocytes, as observed in mice, may conceivably represent examples of evolutionary convergence. Certainly, as a population, all LCs seem homogeneous in skin at a transcriptional level (including the oral mucosal LCs that are generated from pre-DCs; see “Tissues”).
Blood-circulating monocytes may have evolved before adaptive immunity. Monocytic cells can even be found in agnathans (360). They were presumably early inflammatory cells since their capacity to circulate meant that they could be rapidly recruited to sites of infection and damage. Here, their greater numbers might have helped to reinforce the defense and repair functions of the more ancient and sessile (amoebocyte-derived) macrophages and LCs. With the evolution of adaptive immunity, their functions may have also diverged to generate populations of moDCs as immunostimulatory cells. Again, their circulatory capacity may have enabled them now to migrate from the tissues into the newly evolved spleen. Hence, these may have represented early migratory DCs. In the natterjack toad (Bufo calamita), for example, migratory monocytes in the red pulp of spleen appear to differentiate into giant, dendritic-like cells after immunization (86).
Potentially, the phagocytic activity of early monocyte-derived cells may have enabled them to contribute particularly in early immunity against bacteria, which perhaps have the greatest diversity of PAMPs for any eukaryotic host. However, most viruses are too small to be phagocytosed and may have fewer PAMPs, in part because they can envelop themselves within host cell-derived membranes. This may have provided the evolutionary pressure for the emergence of pDCs. These cells can be produced from both myeloid and lymphoid lineages and have certain features of both. In particular, they contain RAG and partial TCR or BCR transcripts (116). This could suggest that pDCs evolved during the diversification of the myeloid and lymphoid lineages. It would therefore be interesting to investigate whether pDCs are also present in fish and amphibians, which this hypothesis would predict, in addition to warm-blooded animals such as rodents, pigs, and humans. The earliest functions of pDCs may have been for inducing antiviral resistance. However, these cells also had the capacity to circulate within the blood and therefore could travel to the spleen. Perhaps therefore they acquired specialized functions to facilitate CD8+ T-cell responses against viruses. In contrast, the moDCs were presumably more specialized for CD4+ T-cell responses against bacteria, and most likely protozoa and fungal spores since these can be readily phagocytosed. Potentially, therefore, moDCs and pDCs could have represented the earliest DC-like cells with diversified but synergistic functions that drove early CD4+ and CD8+ T-cell responses.
Classical DCs can apparently also be generated from both myeloid and lymphoid lineages in vivo. Those in lymphoid tissues can also express either CD4 or CD8, which are otherwise generally associated with conventional or specialized αβ T-cell populations. However, they do not contain RAG or the abortive transcripts of pDCs noted above. It is therefore possible that they evolved even later than pDCs, as is also suggested by the apparently late appearance of the pre-DCs in simple linear models of differentiation (see “Lineages”). These two subsets of CDI and CDII cells might therefore be viewed as being “superimposed” upon the pDC and moDC populations that already existed, with LCs perhaps confined to local defense of the skin. Instead of focusing on particular types of infectious agents, the classical DC may have evolved primarily to sense PAMPs and DAMPs derived from intracellular and extracellular sources in general. This may have enabled a fine-tuning of protective T-cell responses against the respective infectious agents noted above. These DCs may also have facilitated more-robust responses to be generated against infectious agents such as helminths, which are generally difficult to eliminate and often cohabit with humans. Helminths are eukaryotes, and some can mask themselves in host proteins. Hence, they may express many fewer PAMPs than other types of infectious agents but can cause barrier damage that generates DAMPs. Sensing of these by classical DCs may have enhanced the sensing of “danger” and the capacity to induce T-cell responses in general. Interdigitating cells resembling classical DCs have also been identified in the splenic white pulp of different fish, including sharks (85), trout (84), and zebrafish (361). It is not, however, clear if these are classical DCs or monocyte-derived cells. Transcriptional comparisons with those of more recently evolved species may therefore provide insights into whether one subset of classical pre-DCs evolved before another, or whether they arose even later in the evolution of jawed vertebrates.
There are many ways to build an immune system (362364). All jawed animals have skin and gut (and fish have gills, too), so it is important to endow them with local forms of defense at the very least (359, 365). The essential specialized tissues for adaptive immunity seem to be a thymus and spleen, but thereafter additional primary lymphoid tissues and SLOs can evolve in different species. The essential cellular building blocks are T cells, B cells to produce antibodies, and almost certainly DCs. The critical molecular building blocks include lymphocytes with highly diversified antigen receptor repertoires and MHC molecules. However, the genetic mechanisms for diversification can differ between species, including, for example, somatic recombination in humans and mice, gene conversion in chickens, and somatic hypermutation in sharks. And at the cellular and molecular level, some startling differences are also apparent. The cod, for example, lacks CD4+ T cells and all components associated with the peptide–MHC-II pathway (366). This was presumably not an act of wanton vandalism, but due to evolutionary pressures that are still not understood (though a possible third round of genome duplication in some bony fish may be relevant [367]). Studies of DCs in this species may prove particularly interesting.

DCs in Generation of Diversity and Clonal Selection

Two cornerstones in our understanding of adaptive immunity were laid decades ago. The first was the clonal selection theory proposed separately by Burnet and Talmage. This envisaged the existence of a vast number of clonally distributed lymphocyte receptors that could potentially recognize almost any antigen in the universe. The second, originating from the later work of Tonegawa, was the mechanistic insight into precisely how this remarkable generation of diversity could be accomplished, through rearrangement of germ line gene segments accompanied by additional mechanisms of diversification. Since then, our thinking about both processes has been dominated by the role of antigen, in terms of both the diversification of antigen receptors and the subsequent selection of lymphocyte clones by antigen in immunological responses.
There are, in fact, two well-recognized systems for receptor diversification and clonal selection in adaptive immunity that have also been studied for decades. The first was as originally conceived, and occurs within primary lymphoid tissues that have evolved in all jawed vertebrates. Through whichever genetic mechanisms are used in the species, enormous repertoires of BCRs and TCRs can be created. Functional clones of naive B cells and T cells are selected in the thymus through their capacities to recognize self-antigen, in its native form for B cells and as self peptide-MHC complexes for T cells. This generates the primary repertoires of mature T cells and B cells that can subsequently be selected by foreign antigen for protective immunity. During T-cell development in the thymus, DCs may facilitate deletion of autoreactive thymocytes. Crucially, however, they may also select and shape the precise repertoires of tTregs that are subsequently required to regulate their functions in peripheral tissues. During this process it seems likely that they select tTregs with diverse, though moderate, affinities. In turn, the affinity of TCRs may regulate the precise effector functions of these cells (272). Hence, DCs may clonally select and diversify these tTreg populations. Moreover, because it is not possible to ensure that all representations of self are available within the thymus, DCs in normal peripheral tissues generate further repertoires of pTregs that regulate their own functions (and perhaps of course those of other APCs) to ensure the maintenance of tolerance. Again, the same considerations apply. Therefore, at these levels, DCs may be responsible for both clonal selection and generation of diversity of Tregs in tolerance.
The second well-recognized system for clonal diversification and receptor selection occurs within germinal centers of SLOs and acts exclusively on B cells (368). In this case, the generation of diversity occurs through somatic hypermutation, a mechanism that exists in all jawed vertebrates, although germinal centers only evolved later in birds and mammals. During this process, random point mutations are introduced into the variable regions of BCRs in clones of B cells that were selected by foreign antigen and activated in primary responses. This process massively increases diversification of their BCRs, and these diversified clones then undergo repeated rounds of iterative selection against foreign antigen. This is retained for considerable periods of time and displayed on the follicular DC network within the germinal center. This results in the selection of B-cell clones with high-affinity BCRs (different from those they initially expressed) and from which memory B cells can be subsequently generated; all other clones are eliminated. It has been estimated that in some cases the germinal center reaction may select for survival as few as one B-cell clone out of the vast numbers that were originally generated. For B cells, antigen recognition is inseparable from their primary effector functions. These are mediated by soluble forms of their BCRs, antibodies, that are secreted after differentiation into plasma cells. Hence, the germinal center reaction effectively both diversifies and clonally selects for enhanced B-cell effector functions.
For T cells, the situation is entirely different. There is no known equivalent of the germinal center reaction for T cells and no mechanism for clonal diversification of TCRs (since T cells are not subject to somatic hypermutation in birds and mammals). Moreover, the effector functions of T cells are completely unrelated to their antigen specificities, even though TCRs directionally target these toward other cell types (for example, during formation of immunological synapses between helper T cells and B cells, or between CTLs and infected cells). Instead, the effector functions of T cells are mediated by the specialized membrane molecules and soluble mediators they can express or secrete, and which modulate the functions of other cell types, to help, suppress, or otherwise regulate them or to induce apoptosis to kill. Hence, the only way in which it possible to generate diversity of T-cell responses is to select antigen-specific clones and differentially modify their effector responses (or to recruit others into the response for the same purposes). This diversification may originate during priming itself within the T-cell zones of SLOs. Even at this stage, DCs are likely to select clones of T cells with TCRs of different affinities that directly regulate their effector functions (334). Subsequently, the activated T cells execute their complex choreographies as they serially engage with DCs in the T-cell zone, just as B cells do with follicular DCs within their separate follicular compartment (though they may dance together at the borders).
Distinct populations of DCs may be centrally involved in clonal selection and generation of diversity during the different stages of T-cell or TD responses. The classical DCs (which may themselves be highly diversified) may select heterogeneous populations of antigen-specific T cells to be activated within SLOs. Consequently, they generate diverse populations of effector T cells within them. Within NLTs, moDCs (and perhaps LCs) may select from these clones and further diversify their functions during clonal expansion. Meanwhile, pDCs may modulate the functions of classical DCs in SLOs, and possibly moDCs in NLTs where they are present, in order to help drive further diversification of effector T cells (and perhaps recruit additional clones within SLOs). These processes may continue until protective T-cell clones are generated, at which point antigen can be eliminated. Selection and diversification of effector-T-cell clones by all DCs then inevitably ceases (although potentially DCs might subsequently drive other responses that help dampen the response). However, many or all of the early effector-T-cell clones that are generated in response to infection have the potential to develop into memory-T-cell clones. Some of these become long-lived cells that can provide immunological memory for a lifetime. Hence, if the same antigen is encountered again, these memory T cells can be rapidly reactivated by classical (and perhaps other) DCs to provide almost immediate protection. Hence, DCs may drive the evolution of adaptive responses that are central to immunity and tolerance (see above).

Conclusion

There is continuing debate about whether it is possible to define “dendritic cells,” whether these are in fact “just another type of macrophage,” or whether they should be included in the mononuclear phagocyte system. From the viewpoints expressed in this review, the five main lineages of DCs that are discussed appear to comprise, at least in part, an integrated DC network. If more are identified, perhaps these too may be accommodated within the general framework of ideas that has been presented. The individual functions of each DC subset seem diverse and specialized, as are those of macrophage populations within different tissues. Collectively they seem to be synergistic; any DC therefore may be essential for adaptive immunity or tolerance, but none by itself is sufficient. Whether or not DCs should be included in the mononuclear phagocyte system seems mainly a semantic argument, for example, depending on whether a cutoff point is taken above or below monocytes during development or if all are simply grouped together. It matters not. All such categorizations are anthropomorphic constructs designed to help our understanding of systems whose complexity may be beyond mere human comprehension. It is with this in mind that this review ends.

ACKNOWLEDGMENTS

Sincere thanks are due to the numerous academic colleagues, guests, students, and friends whose intellectual input over the years has helped shape the ideas presented in this review. It was great fortune and a privilege to work with Siamon Gordon as his first graduate student, and subsequently with Ralph Steinman as his first postdoctoral fellow within Zanvil Cohn’s department. This review is offered in gratitude to Siamon and to the memory of Ralph and Zan.

REFERENCES

1.
Sompayrak L. 2016. How the Immune System Works, 5th ed. John Wiley & Sons, Chichester, United Kingdom.
2.
MacPherson GG, Austyn JM. 2012. Exploring Immunology: Concepts and Evidence, 1st ed. Wiley-VCH Verlag, Weinheim, Germany.
3.
Murphy KM, Weaver CT. 2016. Janeway’s Immunobiology, 9th ed. Garland Science, Taylor & Francis Group, New York, NY.
4.
Paul WE (ed). 2013. Fundamental Immunology, 7th ed. Lippincott Williams & Wilkins, Philadelphia, PA.
5.
Flajnik MF, Du Pasquier L. 2013. Evolution of the immune system, p 67–128. In Paul WE (ed), Fundamental Immunology, 7th ed. Lippincott Williams & Wilkins, Philadelphia, PA.
6.
Sewell AK. 2012. Why must T cells be cross-reactive? Nat Rev Immunol 12:669–677.
7.
Steinman RM, Cohn ZA. 1973. Identification of a novel cell type in peripheral lymphoid organs of mice. I. Morphology, quantitation, tissue distribution. J Exp Med 137:1142–1162.
8.
Zinkernagel RM, Doherty PC. 1974. Restriction of in vitro T cell-mediated cytotoxicity in lymphocytic choriomeningitis within a syngeneic or semiallogeneic system. Nature 248:701–702.
9.
Steinman RM, Witmer MD. 1978. Lymphoid dendritic cells are potent stimulators of the primary mixed leukocyte reaction in mice. Proc Natl Acad Sci U S A 75:5132–5136.
10.
Farrant J, Clark JC, Lee H, Knight SC, O’Brien J. 1980. Conditions for measuring DNA synthesis in PHA stimulated human lymphocytes in 20 microliters hanging drops with various cell concentrations and periods of culture. J Immunol Methods 33:301–312.
11.
Inaba K, Steinman RM, Van Voorhis WC, Muramatsu S. 1983. Dendritic cells are critical accessory cells for thymus-dependent antibody responses in mouse and in man. Proc Natl Acad Sci U S A 80:6041–6045.
12.
Austyn JM, Steinman RM, Weinstein DE, Granelli-Piperno A, Palladino MA. 1983. Dendritic cells initiate a two-stage mechanism for T lymphocyte proliferation. J Exp Med 157:1101–1115.
13.
Macatonia SE, Hosken NA, Litton M, Vieira P, Hsieh CS, Culpepper JA, Wysocka M, Trinchieri G, Murphy KM, O’Garra A. 1995. Dendritic cells produce IL-12 and direct the development of Th1 cells from naive CD4+ T cells. J Immunol 154:5071–5079.
14.
Veldman JE, Molenaar I, Keuning FJ. 1978. Electron microscopy of cellular immunity reactions in B-cell deprived rabbits. Thymus derived antigen reactive cells, their micro-environment and progeny in the lymph node. Virchows Arch B Cell Pathol 28:217–228.
15.
Veerman AJ. 1974. On the interdigitating cells in the thymus-dependent area of the rat spleen: a relation between the mononuclear phagocyte system and T-lymphocytes. Cell Tissue Res 148:247–257.
16.
Balfour BM, Drexhage HA, Kamperdijk EW, Hoefsmit EC. 1981. Antigen-presenting cells, including Langerhans cells, veiled cells and interdigitating cells. Ciba Found Symp 84:281–301.
17.
Metlay JP, Witmer-Pack MD, Agger R, Crowley MT, Lawless D, Steinman RM. 1990. The distinct leukocyte integrins of mouse spleen dendritic cells as identified with new hamster monoclonal antibodies. J Exp Med 171:1753–1771.
18.
Köhler G, Milstein C. 1975. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature 256:495–497.
19.
Crowley M, Inaba K, Witmer-Pack M, Steinman RM. 1989. The cell surface of mouse dendritic cells: FACS analyses of dendritic cells from different tissues including thymus. Cell Immunol 118:108–125.
20.
Landry D, Lafontaine M, Cossette M, Barthélémy H, Chartrand C, Montplaisir S, Pelletier M. 1988. Human thymic dendritic cells. Characterization, isolation and functional assays. Immunology 65:135–142.
21.
Inaba K, Hosono M, Inaba M. 1990. Thymic dendritic cells and B cells: isolation and function. Int Rev Immunol 6:117–126.
22.
Schuler G, Romani N, Steinman RM. 1985. A comparison of murine epidermal Langerhans cells with spleen dendritic cells. J Invest Dermatol 85(Suppl):99s–106s.
23.
Schuler G, Steinman RM. 1985. Murine epidermal Langerhans cells mature into potent immunostimulatory dendritic cells in vitro. J Exp Med 161:526–546.
24.
Reis e Sousa C, Stahl PD, Austyn JM. 1993. Phagocytosis of antigens by Langerhans cells in vitro. J Exp Med 178:509–519.
25.
Hart DN, Fabre JW. 1981. Demonstration and characterization of Ia-positive dendritic cells in the interstitial connective tissues of rat heart and other tissues, but not brain. J Exp Med 154:347–361.
26.
Hart DN, McKenzie JL. 1990. Interstitial dendritic cells. Int Rev Immunol 6:127–138.
27.
Austyn JM, Hankins DF, Larsen CP, Morris PJ, Rao AS, Roake JA. 1994. Isolation and characterization of dendritic cells from mouse heart and kidney. J Immunol 152:2401–2410.
28.
Steinman RM, Pack M, Inaba K. 1997. Dendritic cells in the T-cell areas of lymphoid organs. Immunol Rev 156:25–37.
29.
Milling S, Yrlid U, Cerovic V, MacPherson G. 2010. Subsets of migrating intestinal dendritic cells. Immunol Rev 234:259–267.
30.
Mason DW, Pugh CW, Webb M. 1981. The rat mixed lymphocyte reaction: roles of a dendritic cell in intestinal lymph and T-cell subsets defined by monoclonal antibodies. Immunology 44:75–87.
31.
Spry CJ, Pflug AJ, Janossy G, Humphrey JH. 1980. Large mononuclear (veiled) cells like ‘Ia-like’ membrane antigens in human afferent lymph. Clin Exp Immunol 39:750–755.
32.
Drexhage HA, Mullink H, de Groot J, Clarke J, Balfour BM. 1979. A study of cells present in peripheral lymph of pigs with special reference to a type of cell resembling the Langerhans cell. Cell Tissue Res 202:407–430.
33.
Thorbecke GJ, Silberberg-Sinakin I, Flotte TJ. 1980. Langerhans cells as macrophages in skin and lymphoid organs. J Invest Dermatol 75:32–43.
34.
Knight SC, Balfour BM, O’Brien J, Buttifant L, Sumerska T, Clarke J. 1982. Role of veiled cells in lymphocyte activation. Eur J Immunol 12:1057–1060.
35.
Larsen CP, Steinman RM, Witmer-Pack M, Hankins DF, Morris PJ, Austyn JM. 1990. Migration and maturation of Langerhans cells in skin transplants and explants. J Exp Med 172:1483–1493.
36.
Stoitzner P, Pfaller K, Stössel H, Romani N. 2002. A close-up view of migrating Langerhans cells in the skin. J Invest Dermatol 118:117–125.
37.
O’Doherty U, Peng M, Gezelter S, Swiggard WJ, Betjes M, Bhardwaj N, Steinman RM. 1994. Human blood contains two subsets of dendritic cells, one immunologically mature and the other immature. Immunology 82:487–493.
38.
Kupiec-Weglinski JW, Austyn JM, Morris PJ. 1988. Migration patterns of dendritic cells in the mouse. Traffic from the blood, and T cell-dependent and -independent entry to lymphoid tissues. J Exp Med 167:632–645.
39.
Austyn JM, Kupiec-Weglinski JW, Hankins DF, Morris PJ. 1988. Migration patterns of dendritic cells in the mouse. Homing to T cell-dependent areas of spleen, and binding within marginal zone. J Exp Med 167:646–651.
40.
Larsen CP, Morris PJ, Austyn JM. 1990. Migration of dendritic leukocytes from cardiac allografts into host spleens. A novel pathway for initiation of rejection. J Exp Med 171:307–314.
41.
Cumberbatch M, Dearman RJ, Kimber I. 1997. Interleukin 1 beta and the stimulation of Langerhans cell migration: comparisons with tumour necrosis factor alpha. Arch Dermatol Res 289:277–284.
42.
Kimber I, Cumberbatch M. 1992. Stimulation of Langerhans cell migration by tumor necrosis factor alpha (TNF-alpha). J Invest Dermatol 99:48S–50S.
43.
Roake JA, Rao AS, Morris PJ, Larsen CP, Hankins DF, Austyn JM. 1995. Dendritic cell loss from nonlymphoid tissues after systemic administration of lipopolysaccharide, tumor necrosis factor, and interleukin 1. J Exp Med 181:2237–2247.
44.
Bowers WE, Berkowitz MR. 1986. Differentiation of dendritic cells in cultures of rat bone marrow cells. J Exp Med 163:872–883.
45.
Inaba K, Inaba M, Romani N, Aya H, Deguchi M, Ikehara S, Muramatsu S, Steinman RM. 1992. Generation of large numbers of dendritic cells from mouse bone marrow cultures supplemented with granulocyte/macrophage colony-stimulating factor. J Exp Med 176:1693–1702.
46.
Caux C, Dezutter-Dambuyant C, Schmitt D, Banchereau J. 1992. GM-CSF and TNF-α cooperate in the generation of dendritic Langerhans cells. Nature 360:258–261.
47.
Siena S, Di Nicola M, Bregni M, Mortarini R, Anichini A, Lombardi L, Ravagnani F, Parmiani G, Gianni AM. 1995. Massive ex vivo generation of functional dendritic cells from mobilized CD34+ blood progenitors for anticancer therapy. Exp Hematol 23:1463–1471.
48.
O’Doherty U, Ignatius R, Bhardwaj N, Pope M. 1997. Generation of monocyte-derived dendritic cells from precursors in rhesus macaque blood. J Immunol Methods 207:185–194.
49.
Sallusto F, Lanzavecchia A. 1994. Efficient presentation of soluble antigen by cultured human dendritic cells is maintained by granulocyte/macrophage colony-stimulating factor plus interleukin 4 and downregulated by tumor necrosis factor α. J Exp Med 179:1109–1118.
50.
Romani N, Reider D, Heuer M, Ebner S, Kämpgen E, Eibl B, Niederwieser D, Schuler G. 1996. Generation of mature dendritic cells from human blood. An improved method with special regard to clinical applicability. J Immunol Methods 196:137–151.
51.
Peters JH, Ruhl S, Friedrichs D. 1987. Veiled accessory cells deduced from monocytes. Immunobiology 176:154–166.
52.
Palucka K, Banchereau J. 2012. Cancer immunotherapy via dendritic cells. Nat Rev Cancer 12:265–277.
53.
Elster JD, Krishnadas DK, Lucas KG. 2016. Dendritic cell vaccines: a review of recent developments and their potential pediatric application. Hum Vaccin Immunother 12:2232–2239.
54.
Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, Redfern CH, Ferrari AC, Dreicer R, Sims RB, Xu Y, Frohlich MW, Schellhammer PF, IMPACT Study Investigators. 2010. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med 363:411–422.
55.
Grouard G, Rissoan MC, Filgueira L, Durand I, Banchereau J, Liu YJ. 1997. The enigmatic plasmacytoid T cells develop into dendritic cells with interleukin (IL)-3 and CD40-ligand. J Exp Med 185:1101–1111.
56.
Huang FP, Platt N, Wykes M, Major JR, Powell TJ, Jenkins CD, MacPherson GG. 2000. A discrete subpopulation of dendritic cells transports apoptotic intestinal epithelial cells to T cell areas of mesenteric lymph nodes. J Exp Med 191:435–444.
57.
Tew JG, Phipps RP, Mandel TE. 1980. The maintenance and regulation of the humoral immune response: persisting antigen and the role of follicular antigen-binding dendritic cells as accessory cells. Immunol Rev 53:175–201.
58.
Aguzzi A, Kranich J, Krautler NJ. 2014. Follicular dendritic cells: origin, phenotype, and function in health and disease. Trends Immunol 35:105–113.
59.
Chen LL, Adams JC, Steinman RM. 1978. Anatomy of germinal centers in mouse spleen, with special reference to “follicular dendritic cells.” J Cell Biol 77:148–164.
60.
Heesters BA, Myers RC, Carroll MC. 2014. Follicular dendritic cells: dynamic antigen libraries. Nat Rev Immunol 14:495–504.
61.
Pulendran B. 2015. The varieties of immunological experience: of pathogens, stress, and dendritic cells. Annu Rev Immunol 33:563–606.
62.
Merad M, Sathe P, Helft J, Miller J, Mortha A. 2013. The dendritic cell lineage: ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed setting. Annu Rev Immunol 31:563–604.
63.
Miller JC, Brown BD, Shay T, Gautier EL, Jojic V, Cohain A, Pandey G, Leboeuf M, Elpek KG, Helft J, Hashimoto D, Chow A, Price J, Greter M, Bogunovic M, Bellemare-Pelletier A, Frenette PS, Randolph GJ, Turley SJ, Merad M, Immunological Genome Consortium. 2012. Deciphering the transcriptional network of the dendritic cell lineage. Nat Immunol 13:888–899.
64.
Schraml BU, Reis e Sousa C. 2015. Defining dendritic cells. Curr Opin Immunol 32:13–20.
65.
Reizis B, Bunin A, Ghosh HS, Lewis KL, Sisirak V. 2011. Plasmacytoid dendritic cells: recent progress and open questions. Annu Rev Immunol 29:163–183.
66.
Ginhoux F, Merad M. 2010. Ontogeny and homeostasis of Langerhans cells. Immunol Cell Biol 88:387–392.
67.
Guilliams M, Ginhoux F, Jakubzick C, Naik SH, Onai N, Schraml BU, Segura E, Tussiwand R, Yona S. 2014. Dendritic cells, monocytes and macrophages: a unified nomenclature based on ontogeny. Nat Rev Immunol 14:571–578.
68.
De Kleer I, Willems F, Lambrecht B, Goriely S. 2014. Ontogeny of myeloid cells. Front Immunol 5:423. doi:10.3389/fimmu.2014.00423.
69.
Lee J, Breton G, Oliveira TYK, Zhou YJ, Aljoufi A, Puhr S, Cameron MJ, Sékaly RP, Nussenzweig MC, Liu K. 2015. Restricted dendritic cell and monocyte progenitors in human cord blood and bone marrow. J Exp Med 212:385–399.
70.
Strobl H, Bello-Fernandez C, Riedl E, Pickl WF, Majdic O, Lyman SD, Knapp W. 1997. flt3 ligand in cooperation with transforming growth factor-β1 potentiates in vitro development of Langerhans-type dendritic cells and allows single-cell dendritic cell cluster formation under serum-free conditions. Blood 90:1425–1434.
71.
Guo X, Zhou Y, Wu T, Zhu X, Lai W, Wu L. 2016. Generation of mouse and human dendritic cells in vitro. J Immunol Methods 432:24–29.
72.
Helft J, Böttcher J, Chakravarty P, Zelenay S, Huotari J, Schraml BU, Goubau D, Reis e Sousa C. 2015. GM-CSF mouse bone marrow cultures comprise a heterogeneous population of CD11c+MHCII+ macrophages and dendritic cells. Immunity 42:1197–1211.
73.
Poltorak MP, Schraml BU. 2015. Fate mapping of dendritic cells. Front Immunol 6:199. doi:10.3389/fimmu.2015.00199.
74.
Bar-On L, Jung S. 2010. Defining dendritic cells by conditional and constitutive cell ablation. Immunol Rev 234:76–89.
75.
Bennett CL, Clausen BE. 2007. DC ablation in mice: promises, pitfalls, and challenges. Trends Immunol 28:525–531.
76.
Collin M, Bigley V, Haniffa M, Hambleton S. 2011. Human dendritic cell deficiency: the missing ID? Nat Rev Immunol 11:575–583.
77.
Liu K, Victora GD, Schwickert TA, Guermonprez P, Meredith MM, Yao K, Chu F-F, Randolph GJ, Rudensky AY, Nussenzweig M. 2009. In vivo analysis of dendritic cell development and homeostasis. Science 324:392–397.
78.
Sathe P, Metcalf D, Vremec D, Naik SH, Langdon WY, Huntington ND, Wu L, Shortman K. 2014. Lymphoid tissue and plasmacytoid dendritic cells and macrophages do not share a common macrophage-dendritic cell-restricted progenitor. Immunity 41:104–115.
79.
Murphy TL, Grajales-Reyes GE, Wu X, Tussiwand R, Briseño CG, Iwata A, Kretzer NM, Durai V, Murphy KM. 2016. Transcriptional control of dendritic cell development. Annu Rev Immunol 34:93–119.
80.
Naik SH, Perié L, Swart E, Gerlach C, van Rooij N, de Boer RJ, Schumacher TN. 2013. Diverse and heritable lineage imprinting of early haematopoietic progenitors. Nature 496:229–232.
81.
Liu K, Nussenzweig MC. 2013. Dendritic cells, p 381–384. In Paul WE (ed), Fundamental Immunology, 7th ed. Lippincott Williams & Wilkins, Philadelphia, PA.
82.
Robbins SH, Walzer T, Dembélé D, Thibault C, Defays A, Bessou G, Xu H, Vivier E, Sellars M, Pierre P, Sharp FR, Chan S, Kastner P, Dalod M. 2008. Novel insights into the relationships between dendritic cell subsets in human and mouse revealed by genome-wide expression profiling. Genome Biol 9:R17. doi:10.1186/gb-2008-9-1-r17.
83.
Crozat K, Guiton R, Guilliams M, Henri S, Baranek T, Schwartz-Cornil I, Malissen B, Dalod M. 2010. Comparative genomics as a tool to reveal functional equivalences between human and mouse dendritic cell subsets. Immunol Rev 234:177–198.
84.
Granja AG, Leal E, Pignatelli J, Castro R, Abós B, Kato G, Fischer U, Tafalla C. 2015. Identification of teleost skin CD8α+ dendritic-like cells, representing a potential common ancestor for mammalian cross-presenting dendritic cells. J Immunol 195:1825–1837.
85.
Rumfelt LL, McKinney EC, Taylor E, Flajnik MF. 2002. The development of primary and secondary lymphoid tissues in the nurse shark Ginglymostoma cirratum: B-cell zones precede dendritic cell immigration and T-cell zone formation during ontogeny of the spleen. Scand J Immunol 56:130–148.
86.
García Barrutia MS, Villena A, Gomariz RP, Razquin B, Zapata A. 1985. Ultrastructural changes in the spleen of the natterjack, Bufo calamita, after antigenic stimulation. Cell Tissue Res 239:435–441.
87.
Bogunovic M, Ginhoux F, Helft J, Shang L, Hashimoto D, Greter M, Liu K, Jakubzick C, Ingersoll MA, Leboeuf M, Stanley ER, Nussenzweig M, Lira SA, Randolph GJ, Merad M. 2009. Origin of the lamina propria dendritic cell network. Immunity 31:513–525.
88.
Hammer GE, Ma A. 2013. Molecular control of steady-state dendritic cell maturation and immune homeostasis. Annu Rev Immunol 31:743–791.
89.
Jiao Z, Bedoui S, Brady JL, Walter A, Chopin M, Carrington EM, Sutherland RM, Nutt SL, Zhang Y, Ko H-J, Wu L, Lew AM, Zhan Y. 2014. The closely related CD103+ dendritic cells (DCs) and lymphoid-resident CD8+ DCs differ in their inflammatory functions. PLoS One 9:e91126. doi:10.1371/journal.pone.0091126.
90.
Li C, Buckwalter MR, Basu S, Garg M, Chang J, Srivastava PK. 2012. Dendritic cells sequester antigenic epitopes for prolonged periods in the absence of antigen-encoding genetic information. Proc Natl Acad Sci U S A 109:17543–17548.
91.
Randolph GJ, Ochando J, Partida-Sánchez S. 2008. Migration of dendritic cell subsets and their precursors. Annu Rev Immunol 26:293–316.
92.
Sánchez-Sánchez N, Riol-Blanco L, Rodríguez-Fernández JL. 2006. The multiple personalities of the chemokine receptor CCR7 in dendritic cells. J Immunol 176:5153–5159.
93.
Ohl L, Mohaupt M, Czeloth N, Hintzen G, Kiafard Z, Zwirner J, Blankenstein T, Henning G, Förster R. 2004. CCR7 governs skin dendritic cell migration under inflammatory and steady-state conditions. Immunity 21:279–288.
94.
Smith TRF, Kumar V. 2008. Revival of CD8+ Treg-mediated suppression. Trends Immunol 29:337–342.
95.
Dzionek A, Fuchs A, Schmidt P, Cremer S, Zysk M, Miltenyi S, Buck DW, Schmitz J. 2000. BDCA-2, BDCA-3, and BDCA-4: three markers for distinct subsets of dendritic cells in human peripheral blood. J Immunol 165:6037–6046.
96.
Adams TE, Huntington JA. 2006. Thrombin-cofactor interactions: structural insights into regulatory mechanisms. Arterioscler Thromb Vasc Biol 26:1738–1745.
97.
Chistiakov DA, Sobenin IA, Orekhov AN, Bobryshev YV. 2015. Myeloid dendritic cells: development, functions, and role in atherosclerotic inflammation. Immunobiology 220:833–844.
98.
Uematsu S, Fujimoto K, Jang MH, Yang B-G, Jung Y-J, Nishiyama M, Sato S, Tsujimura T, Yamamoto M, Yokota Y, Kiyono H, Miyasaka M, Ishii KJ, Akira S. 2008. Regulation of humoral and cellular gut immunity by lamina propria dendritic cells expressing Toll-like receptor 5. Nat Immunol 9:769–776.
99.
Iyoda T, Shimoyama S, Liu K, Omatsu Y, Akiyama Y, Maeda Y, Takahara K, Steinman RM, Inaba K. 2002. The CD8+ dendritic cell subset selectively endocytoses dying cells in culture and in vivo. J Exp Med 195:1289–1302.
100.
Savill J, Hogg N, Ren Y, Haslett C. 1992. Thrombospondin cooperates with CD36 and the vitronectin receptor in macrophage recognition of neutrophils undergoing apoptosis. J Clin Invest 90:1513–1522.
101.
Urban BC, Willcox N, Roberts DJ. 2001. A role for CD36 in the regulation of dendritic cell function. Proc Natl Acad Sci U S A 98:8750–8755.
102.
Hanč P, Fujii T, Iborra S, Yamada Y, Huotari J, Schulz O, Ahrens S, Kjær S, Way M, Sancho D, Namba K, Reis e Sousa C. 2015. Structure of the complex of F-actin and DNGR-1, a C-type lectin receptor involved in dendritic cell cross-presentation of dead cell-associated antigens. Immunity 42:839–849.
103.
Cao L, Shi X, Chang H, Zhang Q, He Y. 2015. pH-dependent recognition of apoptotic and necrotic cells by the human dendritic cell receptor DEC205. Proc Natl Acad Sci U S A 112:7237–7242.
104.
Kim TS, Gorski SA, Hahn S, Murphy KM, Braciale TJ. 2014. Distinct dendritic cell subsets dictate the fate decision between effector and memory CD8+ T cell differentiation by a CD24-dependent mechanism. Immunity 40:400–413.
105.
Dudziak D, Kamphorst AO, Heidkamp GF, Buchholz VR, Trumpfheller C, Yamazaki S, Cheong C, Liu K, Lee H-W, Park CG, Steinman RM, Nussenzweig MC. 2007. Differential antigen processing by dendritic cell subsets in vivo. Science 315:107–111.
106.
Ohta T, Sugiyama M, Hemmi H, Yamazaki C, Okura S, Sasaki I, Fukuda Y, Orimo T, Ishii KJ, Hoshino K, Ginhoux F, Kaisho T. 2016. Crucial roles of XCR1-expressing dendritic cells and the XCR1-XCL1 chemokine axis in intestinal immune homeostasis. Sci Rep 6:23505. doi:10.1038/srep23505.
107.
Lei Y, Ripen AM, Ishimaru N, Ohigashi I, Nagasawa T, Jeker LT, Bösl MR, Holländer GA, Hayashi Y, Malefyt RW, Nitta T, Takahama Y. 2011. Aire-dependent production of XCL1 mediates medullary accumulation of thymic dendritic cells and contributes to regulatory T cell development. J Exp Med 208:383–394.
108.
Guilliams M, Bruhns P, Saeys Y, Hammad H, Lambrecht BN. 2014. The function of Fcγ receptors in dendritic cells and macrophages. Nat Rev Immunol 14:94–108.
109.
Atif SM, Uematsu S, Akira S, McSorley SJ. 2014. CD103CD11b+ dendritic cells regulate the sensitivity of CD4 T-cell responses to bacterial flagellin. Mucosal Immunol 7:68–77.
110.
Iwasaki A, Medzhitov R. 2015. Control of adaptive immunity by the innate immune system. Nat Immunol 16:343–353.
111.
Uto T, Fukaya T, Takagi H, Arimura K, Nakamura T, Kojima N, Malissen B, Sato K. 2016. Clec4A4 is a regulatory receptor for dendritic cells that impairs inflammation and T-cell immunity. Nat Commun 7:11273. doi:10.1038/ncomms11273.
112.
Vander Lugt B, Khan AA, Hackney JA, Agrawal S, Lesch J, Zhou M, Lee WP, Park S, Xu M, DeVoss J, Spooner CJ, Chalouni C, Delamarre L, Mellman I, Singh H. 2014. Transcriptional programming of dendritic cells for enhanced MHC class II antigen presentation. Nat Immunol 15:161–167.
113.
Persson EK, Uronen-Hansson H, Semmrich M, Rivollier A, Hägerbrand K, Marsal J, Gudjonsson S, Håkansson U, Reizis B, Kotarsky K, Agace WW. 2013. IRF4 transcription-factor-dependent CD103+CD11b+ dendritic cells drive mucosal T helper 17 cell differentiation. Immunity 38:958–969.
114.
Swiecki M, Colonna M. 2015. The multifaceted biology of plasmacytoid dendritic cells. Nat Rev Immunol 15:471–485.
115.
Reizis B, Colonna M, Trinchieri G, Barrat F, Gilliet M. 2011. Plasmacytoid dendritic cells: one-trick ponies or workhorses of the immune system? Nat Rev Immunol 11:558–565.
116.
Shigematsu H, Reizis B, Iwasaki H, Mizuno S, Hu D, Traver D, Leder P, Sakaguchi N, Akashi K. 2004. Plasmacytoid dendritic cells activate lymphoid-specific genetic programs irrespective of their cellular origin. Immunity 21:43–53.
117.
Tian J, Avalos AM, Mao SY, Chen B, Senthil K, Wu H, Parroche P, Drabic S, Golenbock D, Sirois C, Hua J, An LL, Audoly L, La Rosa G, Bierhaus A, Naworth P, Marshak-Rothstein A, Crow MK, Fitzgerald KA, Latz E, Kiener PA, Coyle AJ. 2007. Toll-like receptor 9-dependent activation by DNA-containing immune complexes is mediated by HMGB1 and RAGE. Nat Immunol 8:487–496.
118.
Gregorio J, Meller S, Conrad C, Di Nardo A, Homey B, Lauerma A, Arai N, Gallo RL, Digiovanni J, Gilliet M. 2010. Plasmacytoid dendritic cells sense skin injury and promote wound healing through type I interferons. J Exp Med 207:2921–2930.
119.
Cao W, Rosen DB, Ito T, Bover L, Bao M, Watanabe G, Yao Z, Zhang L, Lanier LL, Liu YJ. 2006. Plasmacytoid dendritic cell-specific receptor ILT7-FcεRIγ inhibits Toll-like receptor-induced interferon production. J Exp Med 203:1399–1405.
120.
Simmons DP, Wearsch PA, Canaday DH, Meyerson HJ, Liu YC, Wang Y, Boom WH, Harding CV. 2012. Type I IFN drives a distinctive dendritic cell maturation phenotype that allows continued class II MHC synthesis and antigen processing. J Immunol 188:3116–3126.
121.
Watowich SS, Liu YJ. 2010. Mechanisms regulating dendritic cell specification and development. Immunol Rev 238:76–92.
122.
Guéry L, Hugues S. 2013. Tolerogenic and activatory plasmacytoid dendritic cells in autoimmunity. Front Immunol 4:59. doi:10.3389/fimmu.2013.00059.
123.
Cervantes-Barragan L, Lewis KL, Firner S, Thiel V, Hugues S, Reith W, Ludewig B, Reizis B. 2012. Plasmacytoid dendritic cells control T-cell response to chronic viral infection. Proc Natl Acad Sci U S A 109:3012–3017.
124.
Jakubzick C, Gautier EL, Gibbings SL, Sojka DK, Schlitzer A, Johnson TE, Ivanov S, Duan Q, Bala S, Condon T, van Rooijen N, Grainger JR, Belkaid Y, Ma’ayan A, Riches DWH, Yokoyama WM, Ginhoux F, Henson PM, Randolph GJ. 2013. Minimal differentiation of classical monocytes as they survey steady-state tissues and transport antigen to lymph nodes. Immunity 39:599–610.
125.
Ensan S, Li A, Besla R, Degousee N, Cosme J, Roufaiel M, Shikatani EA, El-Maklizi M, Williams JW, Robins L, Li C, Lewis B, Yun TJ, Lee JS, Wieghofer P, Khattar R, Farrokhi K, Byrne J, Ouzounian M, Zavitz CCJ, Levy GA, Bauer CMT, Libby P, Husain M, Swirski FK, Cheong C, Prinz M, Hilgendorf I, Randolph GJ, Epelman S, Gramolini AO, Cybulsky MI, Rubin BB, Robbins CS. 2016. Self-renewing resident arterial macrophages arise from embryonic CX3CR1+ precursors and circulating monocytes immediately after birth. Nat Immunol 17:159–168.
126.
Jung S, Aliberti J, Graemmel P, Sunshine MJ, Kreutzberg GW, Sher A, Littman DR. 2000. Analysis of fractalkine receptor CX3CR1 function by targeted deletion and green fluorescent protein reporter gene insertion. Mol Cell Biol 20:4106–4114.
127.
Austyn JM, Gordon S. 1981. F4/80, a monoclonal antibody directed specifically against the mouse macrophage. Eur J Immunol 11:805–815.
128.
Lin HH, Stacey M, Stein-Streilein J, Gordon S. 2010. F4/80: the macrophage-specific adhesion-GPCR and its role in immunoregulation. Adv Exp Med Biol 706:149–156.
129.
Tamoutounour S, Guilliams M, Montanana Sanchis F, Liu H, Terhorst D, Malosse C, Pollet E, Ardouin L, Luche H, Sanchez C, Dalod M, Malissen B, Henri S. 2013. Origins and functional specialization of macrophages and of conventional and monocyte-derived dendritic cells in mouse skin. Immunity 39:925–938.
130.
Geissmann F, Jung S, Littman DR. 2003. Blood monocytes consist of two principal subsets with distinct migratory properties. Immunity 19:71–82.
131.
Geissmann F, Manz MG, Jung S, Sieweke MH, Merad M, Ley K. 2010. Development of monocytes, macrophages, and dendritic cells. Science 327:656–661.
132.
Schlitzer A, McGovern N, Ginhoux F. 2015. Dendritic cells and monocyte-derived cells: two complementary and integrated functional systems. Semin Cell Dev Biol 41:9–22.
133.
Espinosa V, Jhingran A, Dutta O, Kasahara S, Donnelly R, Du P, Rosenfeld J, Leiner I, Chen C-C, Ron Y, Hohl TM, Rivera A. 2014. Inflammatory monocytes orchestrate innate antifungal immunity in the lung. PLoS Pathog 10:e1003940. doi:10.1371/journal.ppat.1003940.
134.
Lauvau G, Loke P, Hohl TM. 2015. Monocyte-mediated defense against bacteria, fungi, and parasites. Semin Immunol 27:397–409.
135.
Hohl TM, Rivera A, Lipuma L, Gallegos A, Shi C, Mack M, Pamer EG. 2009. Inflammatory monocytes facilitate adaptive CD4 T cell responses during respiratory fungal infection. Cell Host Microbe 6:470–481.
136.
Plantinga M, Guilliams M, Vanheerswynghels M, Deswarte K, Branco-Madeira F, Toussaint W, Vanhoutte L, Neyt K, Killeen N, Malissen B, Hammad H, Lambrecht BN. 2013. Conventional and monocyte-derived CD11b+ dendritic cells initiate and maintain T helper 2 cell-mediated immunity to house dust mite allergen. Immunity 38:322–335.
137.
Soudja SM, Ruiz AL, Marie JC, Lauvau G. 2012. Inflammatory monocytes activate memory CD8+ T and innate NK lymphocytes independent of cognate antigen during microbial pathogen invasion. Immunity 37:549–562.
138.
Rotta G, Edwards EW, Sangaletti S, Bennett C, Ronzoni S, Colombo MP, Steinman RM, Randolph GJ, Rescigno M. 2003. Lipopolysaccharide or whole bacteria block the conversion of inflammatory monocytes into dendritic cells in vivo. J Exp Med 198:1253–1263.
139.
Randolph GJ, Beaulieu S, Lebecque S, Steinman RM, Muller WA. 1998. Differentiation of monocytes into dendritic cells in a model of transendothelial trafficking. Science 282:480–483.
140.
Randolph GJ, Sanchez-Schmitz G, Liebman RM, Schäkel K. 2002. The CD16+ (FcγRIII+) subset of human monocytes preferentially becomes migratory dendritic cells in a model tissue setting. J Exp Med 196:517–527.
141.
Randolph GJ, Inaba K, Robbiani DF, Steinman RM, Muller WA. 1999. Differentiation of phagocytic monocytes into lymph node dendritic cells in vivo. Immunity 11:753–761.
142.
Kudo S, Matsuno K, Ezaki T, Ogawa M. 1997. A novel migration pathway for rat dendritic cells from the blood: hepatic sinusoids-lymph translocation. J Exp Med 185:777–784.
143.
Matsuno K, Ezaki T, Kudo S, Uehara Y. 1996. A life stage of particle-laden rat dendritic cells in vivo: their terminal division, active phagocytosis, and translocation from the liver to the draining lymph. J Exp Med 183:1865–1878.
144.
Yu B, Ueta H, Kitazawa Y, Tanaka T, Adachi K, Kimura H, Morita M, Sawanobori Y, Qian HX, Kodama T, Matsuno K. 2012. Two immunogenic passenger dendritic cell subsets in the rat liver have distinct trafficking patterns and radiosensitivities. Hepatology 56:1532–1545.
145.
Qu C, Brinck-Jensen N-S, Zang M, Chen K. 2014. Monocyte-derived dendritic cells: targets as potent antigen-presenting cells for the design of vaccines against infectious diseases. Int J Infect Dis 19:1–5.
146.
de Jong A, Cheng TY, Huang S, Gras S, Birkinshaw RW, Kasmar AG, Van Rhijn I, Peña-Cruz V, Ruan DT, Altman JD, Rossjohn J, Moody DB. 2014. CD1a-autoreactive T cells recognize natural skin oils that function as headless antigens. Nat Immunol 15:177–185.
147.
Kubo A, Nagao K, Yokouchi M, Sasaki H, Amagai M. 2009. External antigen uptake by Langerhans cells with reorganization of epidermal tight junction barriers. J Exp Med 206:2937–2946.
148.
Flacher V, Tripp CH, Mairhofer DG, Steinman RM, Stoitzner P, Idoyaga J, Romani N. 2014. Murine Langerin+ dermal dendritic cells prime CD8+ T cells while Langerhans cells induce cross-tolerance. EMBO Mol Med 6:1191–1204.
149.
Seneschal J, Clark RA, Gehad A, Baecher-Allan CM, Kupper TS. 2012. Human epidermal Langerhans cells maintain immune homeostasis in skin by activating skin resident regulatory T cells. Immunity 36:873–884.
150.
Kim M, Truong NR, James V, Bosnjak L, Sandgren KJ, Harman AN, Nasr N, Bertram KM, Olbourne N, Sawleshwarkar S, McKinnon K, Cohen RC, Cunningham AL. 2015. Relay of herpes simplex virus between Langerhans cells and dermal dendritic cells in human skin. PLoS Pathog 11:e1004812. doi:10.1371/journal.ppat.1004812.
151.
Hoeffel G, Wang Y, Greter M, See P, Teo P, Malleret B, Leboeuf M, Low D, Oller G, Almeida F, Choy SHY, Grisotto M, Renia L, Conway SJ, Stanley ER, Chan JKY, Ng LG, Samokhvalov IM, Merad M, Ginhoux F. 2012. Adult Langerhans cells derive predominantly from embryonic fetal liver monocytes with a minor contribution of yolk sac-derived macrophages. J Exp Med 209:1167–1181.
152.
Schuster C, Mildner M, Mairhofer M, Bauer W, Fiala C, Prior M, Eppel W, Kolbus A, Tschachler E, Stingl G, Elbe-Bürger A. 2014. Human embryonic epidermis contains a diverse Langerhans cell precursor pool. Development 141:807–815.
153.
Ginhoux F, Jung S. 2014. Monocytes and macrophages: developmental pathways and tissue homeostasis. Nat Rev Immunol 14:392–404.
154.
Sheng J, Ruedl C, Karjalainen K. 2015. Most tissue-resident macrophages except microglia are derived from fetal hematopoietic stem cells. Immunity 43:382–393.
155.
Capucha T, Mizraji G, Segev H, Blecher-Gonen R, Winter D, Khalaileh A, Tabib Y, Attal T, Nassar M, Zelentsova K, Kisos H, Zenke M, Seré K, Hieronymus T, Burstyn-Cohen T, Amit I, Wilensky A, Hovav AH. 2015. Distinct murine mucosal Langerhans cell subsets develop from pre-dendritic cells and monocytes. Immunity 43:369–381.
156.
Liu C, Duffy B, Bednarski JJ, Calhoun C, Lay L, Rundblad B, Payton JE, Mohanakumar T. 2016. Maternal T-cell engraftment interferes with human leukocyte antigen typing in severe combined immunodeficiency. Am J Clin Pathol 145:251–257.
157.
Buckley RH, Schiff RI, Schiff SE, Markert ML, Williams LW, Harville TO, Roberts JL, Puck JM. 1997. Human severe combined immunodeficiency: genetic, phenotypic, and functional diversity in one hundred eight infants. J Pediatr 130:378–387.
158.
Dawe GS, Tan XW, Xiao ZC. 2007. Cell migration from baby to mother. Cell Adhes Migr 1:19–27.
159.
Nijagal A, Wegorzewska M, Jarvis E, Le T, Tang Q, MacKenzie TC. 2011. Maternal T cells limit engraftment after in utero hematopoietic cell transplantation in mice. J Clin Invest 121:582–592.
160.
Malissen B, Tamoutounour S, Henri S. 2014. The origins and functions of dendritic cells and macrophages in the skin. Nat Rev Immunol 14:417–428.
161.
Beijer MR, Molenaar R, Goverse G, Mebius RE, Kraal G, den Haan JM. 2013. A crucial role for retinoic acid in the development of Notch-dependent murine splenic CD8CD4 and CD4+ dendritic cells. Eur J Immunol 43:1608–1616.
162.
Iwasaki A, Kelsall BL. 2001. Unique functions of CD11b+, CD8α+, and double-negative Peyer’s patch dendritic cells. J Immunol 166:4884–4890.
163.
De Jesus M, Ostroff GR, Levitz SM, Bartling TR, Mantis NJ. 2014. A population of Langerin-positive dendritic cells in murine Peyer’s patches involved in sampling β-glucan microparticles. PLoS One 9:e91002. doi:10.1371/journal.pone.0091002.
164.
Cerovic V, Bain CC, Mowat AM, Milling SW. 2014. Intestinal macrophages and dendritic cells: what’s the difference? Trends Immunol 35:270–277.
165.
Belkaid Y, Segre JA. 2014. Dialogue between skin microbiota and immunity. Science 346:954–959.
166.
Belkaid Y, Naik S. 2013. Compartmentalized and systemic control of tissue immunity by commensals. Nat Immunol 14:646–653.
167.
Belkaid Y, Bouladoux N, Hand TW. 2013. Effector and memory T cell responses to commensal bacteria. Trends Immunol 34:299–306.
168.
Bessman NJ, Sonnenberg GF. 2016. Emerging roles for antigen presentation in establishing host-microbiome symbiosis. Immunol Rev 272:139–150.
169.
Cullender TC, Chassaing B, Janzon A, Kumar K, Muller CE, Werner JJ, Angenent LT, Bell ME, Hay AG, Peterson DA, Walter J, Vijay-Kumar M, Gewirtz AT, Ley RE. 2013. Innate and adaptive immunity interact to quench microbiome flagellar motility in the gut. Cell Host Microbe 14:571–581.
170.
Miyake K, Kaisho T. 2014. Homeostatic inflammation in innate immunity. Curr Opin Immunol 30:85–90.
171.
Heath WR, Carbone FR. 2013. The skin-resident and migratory immune system in steady state and memory: innate lymphocytes, dendritic cells and T cells. Nat Immunol 14:978–985.
172.
Nestle FO, Di Meglio P, Qin JZ, Nickoloff BJ. 2009. Skin immune sentinels in health and disease. Nat Rev Immunol 9:679–691.
173.
Naik S, Bouladoux N, Linehan JL, Han SJ, Harrison OJ, Wilhelm C, Conlan S, Himmelfarb S, Byrd AL, Deming C, Quinones M, Brenchley JM, Kong HH, Tussiwand R, Murphy KM, Merad M, Segre JA, Belkaid Y. 2015. Commensal-dendritic-cell interaction specifies a unique protective skin immune signature. Nature 520:104–108.
174.
Shklovskaya E, O’Sullivan BJ, Ng LG, Roediger B, Thomas R, Weninger W, Fazekas de St Groth B. 2011. Langerhans cells are precommitted to immune tolerance induction. Proc Natl Acad Sci U S A 108:18049–18054.
175.
Modi BG, Neustadter J, Binda E, Lewis J, Filler RB, Roberts SJ, Kwong BY, Reddy S, Overton JD, Galan A, Tigelaar R, Cai L, Fu P, Shlomchik M, Kaplan DH, Hayday A, Girardi M. 2012. Langerhans cells facilitate epithelial DNA damage and squamous cell carcinoma. Science 335:104–108.
176.
Ouchi T, Kubo A, Yokouchi M, Adachi T, Kobayashi T, Kitashima DY, Fujii H, Clausen BE, Koyasu S, Amagai M, Nagao K. 2011. Langerhans cell antigen capture through tight junctions confers preemptive immunity in experimental staphylococcal scalded skin syndrome. J Exp Med 208:2607–2613.
177.
van der Aar AM, Picavet DI, Muller FJ, de Boer L, van Capel TM, Zaat SA, Bos JD, Janssen H, George TC, Kapsenberg ML, van Ham SM, Teunissen MB, de Jong EC. 2013. Langerhans cells favor skin flora tolerance through limited presentation of bacterial antigens and induction of regulatory T cells. J Invest Dermatol 133:1240–1249.
178.
Langlet C, Tamoutounour S, Henri S, Luche H, Ardouin L, Grégoire C, Malissen B, Guilliams M. 2012. CD64 expression distinguishes monocyte-derived and conventional dendritic cells and reveals their distinct role during intramuscular immunization. J Immunol 188:1751–1760.
179.
Kissenpfennig A, Henri S, Dubois B, Laplace-Builhé C, Perrin P, Romani N, Tripp CH, Douillard P, Leserman L, Kaiserlian D, Saeland S, Davoust J, Malissen B. 2005. Dynamics and function of Langerhans cells in vivo: dermal dendritic cells colonize lymph node areas distinct from slower migrating Langerhans cells. Immunity 22:643–654.
180.
Moussion C, Girard JP. 2011. Dendritic cells control lymphocyte entry to lymph nodes through high endothelial venules. Nature 479:542–546.
181.
Palframan RT, Jung S, Cheng G, Weninger W, Luo Y, Dorf M, Littman DR, Rollins BJ, Zweerink H, Rot A, von Andrian UH. 2001. Inflammatory chemokine transport and presentation in HEV: a remote control mechanism for monocyte recruitment to lymph nodes in inflamed tissues. J Exp Med 194:1361–1373.
182.
Mueller SN, Germain RN. 2009. Stromal cell contributions to the homeostasis and functionality of the immune system. Nat Rev Immunol 9:618–629.
183.
Qi H, Kastenmüller W, Germain RN. 2014. Spatiotemporal basis of innate and adaptive immunity in secondary lymphoid tissue. Annu Rev Cell Dev Biol 30:141–167.
184.
Malhotra D, Fletcher AL, Turley SJ. 2013. Stromal and hematopoietic cells in secondary lymphoid organs: partners in immunity. Immunol Rev 251:160–176.
185.
Rantakari P, Auvinen K, Jäppinen N, Kapraali M, Valtonen J, Karikoski M, Gerke H, Iftakhar-E-Khuda I, Keuschnigg J, Umemoto E, Tohya K, Miyasaka M, Elima K, Jalkanen S, Salmi M. 2015. The endothelial protein PLVAP in lymphatics controls the entry of lymphocytes and antigens into lymph nodes. Nat Immunol 16:386–396.
186.
Gretz JE, Norbury CC, Anderson AO, Proudfoot AE, Shaw S. 2000. Lymph-borne chemokines and other low molecular weight molecules reach high endothelial venules via specialized conduits while a functional barrier limits access to the lymphocyte microenvironments in lymph node cortex. J Exp Med 192:1425–1440.
187.
Roozendaal R, Mempel TR, Pitcher LA, Gonzalez SF, Verschoor A, Mebius RE, von Andrian UH, Carroll MC. 2009. Conduits mediate transport of low-molecular-weight antigen to lymph node follicles. Immunity 30:264–276.
188.
Acton SE, Reis e Sousa C. 2016. Dendritic cells in remodeling of lymph nodes during immune responses. Immunol Rev 271:221–229.
189.
Acton SE, Farrugia AJ, Astarita JL, Mourão-Sá D, Jenkins RP, Nye E, Hooper S, van Blijswijk J, Rogers NC, Snelgrove KJ, Rosewell I, Moita LF, Stamp G, Turley SJ, Sahai E, Reis e Sousa C. 2014. Dendritic cells control fibroblastic reticular network tension and lymph node expansion. Nature 514:498–502.
190.
Sixt M, Kanazawa N, Selg M, Samson T, Roos G, Reinhardt DP, Pabst R, Lutz MB, Sorokin L. 2005. The conduit system transports soluble antigens from the afferent lymph to resident dendritic cells in the T cell area of the lymph node. Immunity 22:19–29.
191.
Jakubzick C, Bogunovic M, Bonito AJ, Kuan EL, Merad M, Randolph GJ. 2008. Lymph-migrating, tissue-derived dendritic cells are minor constituents within steady-state lymph nodes. J Exp Med 205:2839–2850.
192.
Gerner MY, Kastenmuller W, Ifrim I, Kabat J, Germain RN. 2012. Histo-cytometry: a method for highly multiplex quantitative tissue imaging analysis applied to dendritic cell subset microanatomy in lymph nodes. Immunity 37:364–376.
193.
Gerner MY, Torabi-Parizi P, Germain RN. 2015. Strategically localized dendritic cells promote rapid T cell responses to lymph-borne particulate antigens. Immunity 42:172–185.
194.
Guilliams M, Crozat K, Henri S, Tamoutounour S, Grenot P, Devilard E, de Bovis B, Alexopoulou L, Dalod M, Malissen B. 2010. Skin-draining lymph nodes contain dermis-derived CD103 dendritic cells that constitutively produce retinoic acid and induce Foxp3+ regulatory T cells. Blood 115:1958–1968.
195.
Catron DM, Itano AA, Pape KA, Mueller DL, Jenkins MK. 2004. Visualizing the first 50 hr of the primary immune response to a soluble antigen. Immunity 21:341–347.
196.
Itano AA, McSorley SJ, Reinhardt RL, Ehst BD, Ingulli E, Rudensky AY, Jenkins MK. 2003. Distinct dendritic cell populations sequentially present antigen to CD4 T cells and stimulate different aspects of cell-mediated immunity. Immunity 19:47–57.
197.
Thurley K, Gerecht D, Friedmann E, Höfer T. 2015. Three-dimensional gradients of cytokine signaling between T cells. PLoS Comput Biol 11:e1004206. doi:10.1371/journal.pcbi.1004206.
198.
Hor JL, Whitney PG, Zaid A, Brooks AG, Heath WR, Mueller SN. 2015. Spatiotemporally distinct interactions with dendritic cell subsets facilitates CD4+ and CD8+ T cell activation to localized viral infection. Immunity 43:554–565.
199.
Eickhoff S, Brewitz A, Gerner MY, Klauschen F, Komander K, Hemmi H, Garbi N, Kaisho T, Germain RN, Kastenmüller W. 2015. Robust anti-viral immunity requires multiple distinct T cell-dendritic cell interactions. Cell 162:1322–1337.
200.
Smith CM, Wilson NS, Waithman J, Villadangos JA, Carbone FR, Heath WR, Belz GT. 2004. Cognate CD4+ T cell licensing of dendritic cells in CD8+ T cell immunity. Nat Immunol 5:1143–1148.
201.
Zaccard CR, Watkins SC, Kalinski P, Fecek RJ, Yates AL, Salter RD, Ayyavoo V, Rinaldo CR, Mailliard RB. 2015. CD40L induces functional tunneling nanotube networks exclusively in dendritic cells programmed by mediators of type 1 immunity. J Immunol 194:1047–1056.
202.
Junt T, Scandella E, Ludewig B. 2008. Form follows function: lymphoid tissue microarchitecture in antimicrobial immune defence. Nat Rev Immunol 8:764–775.
203.
Bernhard CA, Ried C, Kochanek S, Brocker T. 2015. CD169+ macrophages are sufficient for priming of CTLs with specificities left out by cross-priming dendritic cells. Proc Natl Acad Sci U S A 112:5461–5466.
204.
Bain CC, Mowat AM. 2014. Macrophages in intestinal homeostasis and inflammation. Immunol Rev 260:102–117.
205.
Jung C, Hugot J-P, Barreau F. 2010. Peyer’s patches: the immune sensors of the intestine. Int J Inflamm 2010:823710.
206.
Kruglov AA, Grivennikov SI, Kuprash DV, Winsauer C, Prepens S, Seleznik GM, Eberl G, Littman DR, Heikenwalder M, Tumanov AV, Nedospasov SA. 2013. Nonredundant function of soluble LTα3 produced by innate lymphoid cells in intestinal homeostasis. Science 342:1243–1246.
207.
Mowat AM, Agace WW. 2014. Regional specialization within the intestinal immune system. Nat Rev Immunol 14:667–685.
208.
Pugh CW, MacPherson GG, Steer HW. 1983. Characterization of nonlymphoid cells derived from rat peripheral lymph. J Exp Med 157:1758–1779.
209.
Bain CC, Scott CL, Uronen-Hansson H, Gudjonsson S, Jansson O, Grip O, Guilliams M, Malissen B, Agace WW, Mowat AM. 2013. Resident and pro-inflammatory macrophages in the colon represent alternative context-dependent fates of the same Ly6Chi monocyte precursors. Mucosal Immunol 6:498–510.
210.
Mazzini E, Massimiliano L, Penna G, Rescigno M. 2014. Oral tolerance can be established via gap junction transfer of fed antigens from CX3CR1+ macrophages to CD103+ dendritic cells. Immunity 40:248–261.
211.
Scott CL, Bain CC, Wright PB, Sichien D, Kotarsky K, Persson EK, Luda K, Guilliams M, Lambrecht BN, Agace WW, Milling SW, Mowat AM. 2015. CCR2+CD103 intestinal dendritic cells develop from DC-committed precursors and induce interleukin-17 production by T cells. Mucosal Immunol 8:327–339.
212.
Rescigno M, Urbano M, Valzasina B, Francolini M, Rotta G, Bonasio R, Granucci F, Kraehenbuhl JP, Ricciardi-Castagnoli P. 2001. Dendritic cells express tight junction proteins and penetrate gut epithelial monolayers to sample bacteria. Nat Immunol 2:361–367.
213.
Chieppa M, Rescigno M, Huang AYC, Germain RN. 2006. Dynamic imaging of dendritic cell extension into the small bowel lumen in response to epithelial cell TLR engagement. J Exp Med 203:2841–2852.
214.
McDole JR, Wheeler LW, McDonald KG, Wang B, Konjufca V, Knoop KA, Newberry RD, Miller MJ. 2012. Goblet cells deliver luminal antigen to CD103+ dendritic cells in the small intestine. Nature 483:345–349.
215.
Shakhar G, Kolesnikov M. 2014. Intestinal macrophages and DCs close the gap on tolerance. Immunity 40:171–173.
216.
Diehl GE, Longman RS, Zhang J-X, Breart B, Galan C, Cuesta A, Schwab SR, Littman DR. 2013. Microbiota restricts trafficking of bacteria to mesenteric lymph nodes by CX3CR1hi cells. Nature 494:116–120.
217.
Farache J, Koren I, Milo I, Gurevich I, Kim K-W, Zigmond E, Furtado GC, Lira SA, Shakhar G. 2013. Luminal bacteria recruit CD103+ dendritic cells into the intestinal epithelium to sample bacterial antigens for presentation. Immunity 38:581–595.
218.
Coombes JL, Siddiqui KRR, Arancibia-Cárcamo CV, Hall J, Sun CM, Belkaid Y, Powrie F. 2007. A functionally specialized population of mucosal CD103+ DCs induces Foxp3+ regulatory T cells via a TGF-β and retinoic acid-dependent mechanism. J Exp Med 204:1757–1764.
219.
Coombes JL, Powrie F. 2008. Dendritic cells in intestinal immune regulation. Nat Rev Immunol 8:435–446.
220.
Travis MA, Reizis B, Melton AC, Masteller E, Tang Q, Proctor JM, Wang Y, Bernstein X, Huang X, Reichardt LF, Bluestone JA, Sheppard D. 2007. Loss of integrin αvβ8 on dendritic cells causes autoimmunity and colitis in mice. Nature 449:361–365.
221.
Muzaki ARBM, Tetlak P, Sheng J, Loh SC, Setiagani YA, Poidinger M, Zolezzi F, Karjalainen K, Ruedl C. 2016. Intestinal CD103+CD11b dendritic cells restrain colitis via IFN-γ-induced anti-inflammatory response in epithelial cells. Mucosal Immunol 9:336–351.
222.
van de Pavert SA, Mebius RE. 2010. New insights into the development of lymphoid tissues. Nat Rev Immunol 10:664–674.
223.
Tanoue T, Atarashi K, Honda K. 2016. Development and maintenance of intestinal regulatory T cells. Nat Rev Immunol 16:295–309.
224.
Worbs T, Bode U, Yan S, Hoffmann MW, Hintzen G, Bernhardt G, Förster R, Pabst O. 2006. Oral tolerance originates in the intestinal immune system and relies on antigen carriage by dendritic cells. J Exp Med 203:519–527.
225.
Macpherson AJ, Smith K. 2006. Mesenteric lymph nodes at the center of immune anatomy. J Exp Med 203:497–500.
226.
Balmer ML, Slack E, de Gottardi A, Lawson MAE, Hapfelmeier S, Miele L, Grieco A, Van Vlierberghe H, Fahrner R, Patuto N, Bernsmeier C, Ronchi F, Wyss M, Stroka D, Dickgreber N, Heim MH, McCoy KD, Macpherson AJ. 2014. The liver may act as a firewall mediating mutualism between the host and its gut commensal microbiota. Sci Transl Med 6:237ra66. doi:10.1126/scitranslmed.3008618.
227.
Yrlid U, Cerovic V, Milling S, Jenkins CD, Zhang J, Crocker PR, Klavinskis LS, MacPherson GG. 2006. Plasmacytoid dendritic cells do not migrate in intestinal or hepatic lymph. J Immunol 177:6115–6121.
228.
Yang JY, Kim MS, Kim E, Cheon JH, Lee YS, Kim Y, Lee SH, Seo SU, Shin SH, Choi SS, Kim B, Chang SY, Ko HJ, Bae JW, Kweon MN. 2016. Enteric viruses ameliorate gut inflammation via Toll-like receptor 3 and Toll-like receptor 7-mediated interferon-β production. Immunity 44:889–900.
229.
Reboldi A, Cyster JG. 2016. Peyer’s patches: organizing B-cell responses at the intestinal frontier. Immunol Rev 271:230–245.
230.
Bonnardel J, Da Silva C, Henri S, Tamoutounour S, Chasson L, Montañana-Sanchis F, Gorvel J-P, Lelouard H. 2015. Innate and adaptive immune functions of Peyer’s patch monocyte-derived cells. Cell Rep 11:770–784.
231.
Bonnardel J, Da Silva C, Masse M, Montañana-Sanchis F, Gorvel JP, Lelouard H. 2015. Gene expression profiling of the Peyer’s patch mononuclear phagocyte system. Genom Data 5:21–24.
232.
Lelouard H, Fallet M, de Bovis B, Méresse S, Gorvel J-P. 2012. Peyer’s patch dendritic cells sample antigens by extending dendrites through M cell-specific transcellular pores. Gastroenterology 142:592–601.e3. doi:10.1053/j.gastro.2011.11.039.
233.
Macpherson AJ, McCoy KD. 2015. Independence Day for IgA. Immunity 43:416–418.
234.
Cook PC, MacDonald AS. 2016. Dendritic cells in lung immunopathology. Semin Immunopathol 38:449–460.
235.
Kopf M, Schneider C, Nobs SP. 2015. The development and function of lung-resident macrophages and dendritic cells. Nat Immunol 16:36–44.
236.
Sung S-SJ, Fu SM, Rose CE Jr, Gaskin F, Ju S-T, Beaty SR. 2006. A major lung CD103 (αE)-β7 integrin-positive epithelial dendritic cell population expressing Langerin and tight junction proteins. J Immunol 176:2161–2172.
237.
Thornton EE, Looney MR, Bose O, Sen D, Sheppard D, Locksley R, Huang X, Krummel MF. 2012. Spatiotemporally separated antigen uptake by alveolar dendritic cells and airway presentation to T cells in the lung. J Exp Med 209:1183–1199.
238.
Bonduelle O, Duffy D, Verrier B, Combadière C, Combadière B. 2012. Cutting edge: protective effect of CX3CR1+ dendritic cells in a vaccinia virus pulmonary infection model. J Immunol 188:952–956.
239.
Desch AN, Randolph GJ, Murphy K, Gautier EL, Kedl RM, Lahoud MH, Caminschi I, Shortman K, Henson PM, Jakubzick CV. 2011. CD103+ pulmonary dendritic cells preferentially acquire and present apoptotic cell-associated antigen. J Exp Med 208:1789–1797.
240.
Takano K, Kojima T, Go M, Murata M, Ichimiya S, Himi T, Sawada N. 2005. HLA-DR- and CD11c-positive dendritic cells penetrate beyond well-developed epithelial tight junctions in human nasal mucosa of allergic rhinitis. J Histochem Cytochem 53:611–619.
241.
Helft J, Manicassamy B, Guermonprez P, Hashimoto D, Silvin A, Agudo J, Brown BD, Schmolke M, Miller JC, Leboeuf M, Murphy KM, García-Sastre A, Merad M. 2012. Cross-presenting CD103+ dendritic cells are protected from influenza virus infection. J Clin Invest 122:4037–4047.
242.
Abraham SN, Miao Y. 2015. The nature of immune responses to urinary tract infections. Nat Rev Immunol 15:655–663.
243.
Dieterlen MT, John K, Reichenspurner H, Mohr FW, Barten MJ. 2016. Dendritic cells and their role in cardiovascular diseases: a view on human studies. J Immunol Res 2016:5946807. doi:10.1155/2016/5946807.
244.
Busch M, Westhofen TC, Koch M, Lutz MB, Zernecke A. 2014. Dendritic cell subset distributions in the aorta in healthy and atherosclerotic mice. PLoS One 9:e88452. doi:10.1371/journal.pone.0088452.
245.
Choi JH, Do Y, Cheong C, Koh H, Boscardin SB, Oh YS, Bozzacco L, Trumpfheller C, Park CG, Steinman RM. 2009. Identification of antigen-presenting dendritic cells in mouse aorta and cardiac valves. J Exp Med 206:497–505.
246.
Rogers NM, Ferenbach DA, Isenberg JS, Thomson AW, Hughes J. 2014. Dendritic cells and macrophages in the kidney: a spectrum of good and evil. Nat Rev Nephrol 10:625–643.
247.
Gottschalk C, Kurts C. 2015. The debate about dendritic cells and macrophages in the kidney. Front Immunol 6:435. doi:10.3389/fimmu.2015.00435.
248.
Ferris ST, Carrero JA, Mohan JF, Calderon B, Murphy KM, Unanue ER. 2014. A minor subset of Batf3-dependent antigen-presenting cells in islets of Langerhans is essential for the development of autoimmune diabetes. Immunity 41:657–669.
249.
Epelman S, Lavine KJ, Beaudin AE, Sojka DK, Carrero JA, Calderon B, Brija T, Gautier EL, Ivanov S, Satpathy AT, Schilling JD, Schwendener R, Sergin I, Razani B, Forsberg EC, Yokoyama WM, Unanue ER, Colonna M, Randolph GJ, Mann DL. 2014. Embryonic and adult-derived resident cardiac macrophages are maintained through distinct mechanisms at steady state and during inflammation. Immunity 40:91–104.
250.
Larsen CP, Morris PJ, Austyn JM. 1990. Donor dendritic leukocytes migrate from cardiac allografts into recipients’ spleens. Transplant Proc 22:1943–1944.
251.
Aspelund A, Robciuc MR, Karaman S, Makinen T, Alitalo K. 2016. Lymphatic system in cardiovascular medicine. Circ Res 118:515–530.
252.
Cavanagh LL, Bonasio R, Mazo IB, Halin C, Cheng G, van der Velden AW, Cariappa A, Chase C, Russell P, Starnbach MN, Koni PA, Pillai S, Weninger W, von Andrian UH. 2005. Activation of bone marrow-resident memory T cells by circulating, antigen-bearing dendritic cells. Nat Immunol 6:1029–1037.
253.
Hervouet C, Luci C, Bekri S, Juhel T, Bihl F, Braud VM, Czerkinsky C, Anjuère F. 2014. Antigen-bearing dendritic cells from the sublingual mucosa recirculate to distant systemic lymphoid organs to prime mucosal CD8 T cells. Mucosal Immunol 7:280–291.
254.
Steiniger BS. 2015. Human spleen microanatomy: why mice do not suffice. Immunology 145:334–346.
255.
Yi T, Cyster JG. 2013. EBI2-mediated bridging channel positioning supports splenic dendritic cell homeostasis and particulate antigen capture. eLife 2:e00757. doi:10.7554/eLife.00757.
256.
Gatto D, Wood K, Caminschi I, Murphy-Durland D, Schofield P, Christ D, Karupiah G, Brink R. 2013. The chemotactic receptor EBI2 regulates the homeostasis, localization and immunological function of splenic dendritic cells. Nat Immunol 14:446–453.
257.
Yi T, Li J, Chen H, Wu J, An J, Xu Y, Hu Y, Lowell CA, Cyster JG. 2015. Splenic dendritic cells survey red blood cells for missing self-CD47 to trigger adaptive immune responses. Immunity 43:764–775.
258.
Qiu CH, Miyake Y, Kaise H, Kitamura H, Ohara O, Tanaka M. 2009. Novel subset of CD8α+ dendritic cells localized in the marginal zone is responsible for tolerance to cell-associated antigens. J Immunol 182:4127–4136.
259.
Glennon E, Kaunzner UW, Gagnidze K, McEwen BS, Bulloch K. 2015. Pituitary dendritic cells communicate immune pathogenic signals. Brain Behav Immun 50:232–240.
260.
Louveau A, Smirnov I, Keyes TJ, Eccles JD, Rouhani SJ, Peske JD, Derecki NC, Castle D, Mandell JW, Lee KS, Harris TH, Kipnis J. 2015. Structural and functional features of central nervous system lymphatic vessels. Nature 523:337–341.
261.
Aspelund A, Antila S, Proulx ST, Karlsen TV, Karaman S, Detmar M, Wiig H, Alitalo K. 2015. A dural lymphatic vascular system that drains brain interstitial fluid and macromolecules. J Exp Med 212:991–999.
262.
Chavan SS, Tracey KJ. 2013. Neurophysiologic reflex mechanisms in immunology, p 850–862. In Paul WE (ed), Fundamental Immunology, 7th ed. Lippincott Williams & Wilkins, Philadelphia, PA.
263.
Rhee SH, Pothoulakis C, Mayer EA. 2009. Principles and clinical implications of the brain-gut-enteric microbiota axis. Nat Rev Gastroenterol Hepatol 6:306–314.
264.
McMahon SB, La Russa F, Bennett DLH. 2015. Crosstalk between the nociceptive and immune systems in host defence and disease. Nat Rev Neurosci 16:389–402.
265.
Prado C, Contreras F, González H, Díaz P, Elgueta D, Barrientos M, Herrada AA, Lladser Á, Bernales S, Pacheco R. 2012. Stimulation of dopamine receptor D5 expressed on dendritic cells potentiates Th17-mediated immunity. J Immunol 188:3062–3070.
266.
Wülfing C, Günther HS. 2015. Dendritic cells and macrophages neurally hard-wired in the lymph node. Sci Rep 5:16866. doi:10.1038/srep16866.
267.
Yarovinsky F. 2014. Innate immunity to Toxoplasma gondii infection. Nat Rev Immunol 14:109–121.
268.
Balenga NA, Balenga NA. 2007. Human TLR11 gene is repressed due to its probable interaction with profilin expressed in human. Med Hypotheses 68:456.
269.
Abbas AK, Benoist C, Bluestone JA, Campbell DJ, Ghosh S, Hori S, Jiang S, Kuchroo VK, Mathis D, Roncarolo MG, Rudensky A, Sakaguchi S, Shevach EM, Vignali DAA, Ziegler SF. 2013. Regulatory T cells: recommendations to simplify the nomenclature. Nat Immunol 14:307–308.
270.
Caramalho Í, Nunes-Cabaço H, Foxall RB, Sousa AE. 2015. Regulatory T-cell development in the human thymus. Front Immunol 6:395. doi:10.3389/fimmu.2015.00395.
271.
Panduro M, Benoist C, Mathis D. 2016. Tissue Tregs. Annu Rev Immunol 34:609–633.
272.
Li MO, Rudensky AY. 2016. T cell receptor signalling in the control of regulatory T cell differentiation and function. Nat Rev Immunol 16:220–233.
273.
Drumea-Mirancea M, Wessels JT, Müller CA, Essl M, Eble JA, Tolosa E, Koch M, Reinhardt DP, Sixt M, Sorokin L, Stierhof YD, Schwarz H, Klein G. 2006. Characterization of a conduit system containing laminin-5 in the human thymus: a potential transport system for small molecules. J Cell Sci 119:1396–1405.
274.
Klein L, Kyewski B, Allen PM, Hogquist KA. 2014. Positive and negative selection of the T cell repertoire: what thymocytes see (and don’t see). Nat Rev Immunol 14:377–391.
275.
Kurd N, Robey EA. 2016. T-cell selection in the thymus: a spatial and temporal perspective. Immunol Rev 271:114–126.
276.
Takaba H, Morishita Y, Tomofuji Y, Danks L, Nitta T, Komatsu N, Kodama T, Takayanagi H. 2015. Fezf2 orchestrates a thymic program of self-antigen expression for immune tolerance. Cell 163:975–987.
277.
Gardner JM, Metzger TC, McMahon EJ, Au-Yeung BB, Krawisz AK, Lu W, Price JD, Johannes KP, Satpathy AT, Murphy KM, Tarbell KV, Weiss A, Anderson MS. 2013. Extrathymic Aire-expressing cells are a distinct bone marrow-derived population that induce functional inactivation of CD4+ T cells. Immunity 39:560–572.
278.
Lucas B, McCarthy NI, Baik S, Cosway E, James KD, Parnell SM, White AJ, Jenkinson WE, Anderson G. 2016. Control of the thymic medulla and its influence on αβT-cell development. Immunol Rev 271:23–37.
279.
Perry JS, Hsieh CS. 2016. Development of T-cell tolerance utilizes both cell-autonomous and cooperative presentation of self-antigen. Immunol Rev 271:141–155.
280.
Perry JS, Lio CW, Kau AL, Nutsch K, Yang Z, Gordon JI, Murphy KM, Hsieh CS. 2014. Distinct contributions of Aire and antigen-presenting-cell subsets to the generation of self-tolerance in the thymus. Immunity 41:414–426.
281.
Abramson J, Husebye ES. 2016. Autoimmune regulator and self-tolerance—molecular and clinical aspects. Immunol Rev 271:127–140.
282.
St-Pierre C, Brochu S, Vanegas JR, Dumont-Lagacé M, Lemieux S, Perreault C. 2013. Transcriptome sequencing of neonatal thymic epithelial cells. Sci Rep 3:1860. doi:10.1038/srep01860.
283.
Proietto AI, van Dommelen S, Wu L. 2009. The impact of circulating dendritic cells on the development and differentiation of thymocytes. Immunol Cell Biol 87:39–45.
284.
Taniguchi RT, DeVoss JJ, Moon JJ, Sidney J, Sette A, Jenkins MK, Anderson MS. 2012. Detection of an autoreactive T-cell population within the polyclonal repertoire that undergoes distinct autoimmune regulator (Aire)-mediated selection. Proc Natl Acad Sci U S A 109:7847–7852.
285.
Yamano T, Nedjic J, Hinterberger M, Steinert M, Koser S, Pinto S, Gerdes N, Lutgens E, Ishimaru N, Busslinger M, Brors B, Kyewski B, Klein L. 2015. Thymic B cells are licensed to present self antigens for central T cell tolerance induction. Immunity 42:1048–1061.
286.
Oh J, Shin JS. 2015. The role of dendritic cells in central tolerance. Immune Netw 15:111–120.
287.
Osorio F, Fuentes C, López MN, Salazar-Onfray F, González FE. 2015. Role of dendritic cells in the induction of lymphocyte tolerance. Front Immunol 6:535. doi:10.3389/fimmu.2015.00535.
288.
Yang S, Fujikado N, Kolodin D, Benoist C, Mathis D. 2015. Immune tolerance. Regulatory T cells generated early in life play a distinct role in maintaining self-tolerance. Science 348:589–594.
289.
Hubert FX, Kinkel SA, Davey GM, Phipson B, Mueller SN, Liston A, Proietto AI, Cannon PZ, Forehan S, Smyth GK, Wu L, Goodnow CC, Carbone FR, Scott HS, Heath WR. 2011. Aire regulates the transfer of antigen from mTECs to dendritic cells for induction of thymic tolerance. Blood 118:2462–2472.
290.
Proietto AI, van Dommelen S, Zhou P, Rizzitelli A, D’Amico A, Steptoe RJ, Naik SH, Lahoud MH, Liu Y, Zheng P, Shortman K, Wu L. 2008. Dendritic cells in the thymus contribute to T-regulatory cell induction. Proc Natl Acad Sci U S A 105:19869–19874.
291.
Bonasio R, Scimone ML, Schaerli P, Grabie N, Lichtman AH, von Andrian UH. 2006. Clonal deletion of thymocytes by circulating dendritic cells homing to the thymus. Nat Immunol 7:1092–1100.
292.
Baba T, Nakamoto Y, Mukaida N. 2009. Crucial contribution of thymic Sirpα+ conventional dendritic cells to central tolerance against blood-borne antigens in a CCR2-dependent manner. J Immunol 183:3053–3063.
293.
Li J, Park J, Foss D, Goldschneider I. 2009. Thymus-homing peripheral dendritic cells constitute two of the three major subsets of dendritic cells in the steady-state thymus. J Exp Med 206:607–622.
294.
Donskoy E, Goldschneider I. 2003. Two developmentally distinct populations of dendritic cells inhabit the adult mouse thymus: demonstration by differential importation of hematogenous precursors under steady state conditions. J Immunol 170:3514–3521.
295.
Atibalentja DF, Murphy KM, Unanue ER. 2011. Functional redundancy between thymic CD8α+ and Sirpα+ conventional dendritic cells in presentation of blood-derived lysozyme by MHC class II proteins. J Immunol 186:1421–1431.
296.
Hadeiba H, Lahl K, Edalati A, Oderup C, Habtezion A, Pachynski R, Nguyen L, Ghodsi A, Adler S, Butcher EC. 2012. Plasmacytoid dendritic cells transport peripheral antigens to the thymus to promote central tolerance. Immunity 36:438–450.
297.
Yadav M, Stephan S, Bluestone JA. 2013. Peripherally induced Tregs—role in immune homeostasis and autoimmunity. Front Immunol 4:232. doi:10.3389/fimmu.2013.00232.
298.
Bosco N, Kirberg J, Ceredig R, Agenès F. 2009. Peripheral T cells in the thymus: have they just lost their way or do they do something? Immunol Cell Biol 87:50–57.
299.
Josefowicz SZ, Lu LF, Rudensky AY. 2012. Regulatory T cells: mechanisms of differentiation and function. Annu Rev Immunol 30:531–564.
300.
Leventhal DS, Gilmore DC, Berger JM, Nishi S, Lee V, Malchow S, Kline DE, Kline J, Vander Griend DJ, Huang H, Socci ND, Savage PA. 2016. Dendritic cells coordinate the development and homeostasis of organ-specific regulatory T cells. Immunity 44:847–859.
301.
Scharschmidt TC, Vasquez KS, Truong HA, Gearty SV, Pauli ML, Nosbaum A, Gratz IK, Otto M, Moon JJ, Liese J, Abbas AK, Fischbach MA, Rosenblum MD. 2015. A wave of regulatory T cells into neonatal skin mediates tolerance to commensal microbes. Immunity 43:1011–1021.
302.
Legoux FP, Lim J-B, Cauley AW, Dikiy S, Ertelt J, Mariani TJ, Sparwasser T, Way SS, Moon JJ. 2015. CD4+ T cell tolerance to tissue-restricted self antigens is mediated by antigen-specific regulatory T cells rather than deletion. Immunity 43:896–908.
303.
Kim KS, Hong SW, Han D, Yi J, Jung J, Yang BG, Lee JY, Lee M, Surh CD. 2016. Dietary antigens limit mucosal immunity by inducing regulatory T cells in the small intestine. Science 351:858–863.
304.
Rosenblum MD, Way SS, Abbas AK. 2016. Regulatory T cell memory. Nat Rev Immunol 16:90–101.
305.
Sanchez Rodriguez R, Pauli ML, Neuhaus IM, Yu SS, Arron ST, Harris HW, Yang SH, Anthony BA, Sverdrup FM, Krow-Lucal E, MacKenzie TC, Johnson DS, Meyer EH, Löhr A, Hsu A, Koo J, Liao W, Gupta R, Debbaneh MG, Butler D, Huynh M, Levin EC, Leon A, Hoffman WY, McGrath MH, Alvarado MD, Ludwig CH, Truong HA, Maurano MM, Gratz IK, Abbas AK, Rosenblum MD. 2014. Memory regulatory T cells reside in human skin. J Clin Invest 124:1027–1036.
306.
Eldershaw SA, Sansom DM, Narendran P. 2011. Expression and function of the autoimmune regulator (Aire) gene in non-thymic tissue. Clin Exp Immunol 163:296–308.
307.
Lindmark E, Chen Y, Georgoudaki AM, Dudziak D, Lindh E, Adams WC, Loré K, Winqvist O, Chambers BJ, Karlsson MC. 2013. AIRE expressing marginal zone dendritic cells balances adaptive immunity and T-follicular helper cell recruitment. J Autoimmun 42:62–70.
308.
Suzuki E, Kobayashi Y, Kawano O, Endo K, Haneda H, Yukiue H, Sasaki H, Yano M, Maeda M, Fujii Y. 2008. Expression of AIRE in thymocytes and peripheral lymphocytes. Autoimmunity 41:133–139.
309.
Fletcher AL, Malhotra D, Turley SJ. 2011. Lymph node stroma broaden the peripheral tolerance paradigm. Trends Immunol 32:12–18.
310.
Yip L, Su L, Sheng D, Chang P, Atkinson M, Czesak M, Albert PR, Collier AR, Turley SJ, Fathman CG, Creusot RJ. 2009. Deaf1 isoforms control the expression of genes encoding peripheral tissue antigens in the pancreatic lymph nodes during type 1 diabetes. Nat Immunol 10:1026–1033.
311.
Dubrot J, Duraes FV, Potin L, Capotosti F, Brighouse D, Suter T, LeibundGut-Landmann S, Garbi N, Reith W, Swartz MA, Hugues S. 2014. Lymph node stromal cells acquire peptide-MHCII complexes from dendritic cells and induce antigen-specific CD4+ T cell tolerance. J Exp Med 211:1153–1166.
312.
Campana S, De Pasquale C, Carrega P, Ferlazzo G, Bonaccorsi I. 2015. Cross-dressing: an alternative mechanism for antigen presentation. Immunol Lett 168:349–354.
313.
Fletcher AL, Lukacs-Kornek V, Reynoso ED, Pinner SE, Bellemare-Pelletier A, Curry MS, Collier AR, Boyd RL, Turley SJ. 2010. Lymph node fibroblastic reticular cells directly present peripheral tissue antigen under steady-state and inflammatory conditions. J Exp Med 207:689–697.
314.
Scheinecker C, McHugh R, Shevach EM, Germain RN. 2002. Constitutive presentation of a natural tissue autoantigen exclusively by dendritic cells in the draining lymph node. J Exp Med 196:1079–1090.
315.
Liu Z, Gerner MY, Van Panhuys N, Levine AG, Rudensky AY, Germain RN. 2015. Immune homeostasis enforced by co-localized effector and regulatory T cells. Nature 528:225–230.
316.
Loschko J, Schreiber HA, Rieke GJ, Esterházy D, Meredith MM, Pedicord VA, Yao KH, Caballero S, Pamer EG, Mucida D, Nussenzweig MC. 2016. Absence of MHC class II on cDCs results in microbial-dependent intestinal inflammation. J Exp Med 213:517–534.
317.
Muth S, Schütze K, Schild H, Probst HC. 2012. Release of dendritic cells from cognate CD4+ T-cell recognition results in impaired peripheral tolerance and fatal cytotoxic T-cell mediated autoimmunity. Proc Natl Acad Sci U S A 109:9059–9064.
318.
Schildknecht A, Brauer S, Brenner C, Lahl K, Schild H, Sparwasser T, Probst HC, van den Broek M. 2010. FoxP3+ regulatory T cells essentially contribute to peripheral CD8+ T-cell tolerance induced by steady-state dendritic cells. Proc Natl Acad Sci U S A 107:199–203.
319.
Probst HC, McCoy K, Okazaki T, Honjo T, van den Broek M. 2005. Resting dendritic cells induce peripheral CD8+ T cell tolerance through PD-1 and CTLA-4. Nat Immunol 6:280–286.
320.
Shimoda M, Mmanywa F, Joshi SK, Li T, Miyake K, Pihkala J, Abbas JA, Koni PA. 2006. Conditional ablation of MHC-II suggests an indirect role for MHC-II in regulatory CD4 T cell maintenance. J Immunol 176:6503–6511.
321.
Baratin M, Foray C, Demaria O, Habbeddine M, Pollet E, Maurizio J, Verthuy C, Davanture S, Azukizawa H, Flores-Langarica A, Dalod M, Lawrence T. 2015. Homeostatic NF-κB signaling in steady-state migratory dendritic cells regulates immune homeostasis and tolerance. Immunity 42:627–639.
322.
Suffner J, Hochweller K, Kühnle MC, Li X, Kroczek RA, Garbi N, Hämmerling GJ. 2010. Dendritic cells support homeostatic expansion of Foxp3+ regulatory T cells in Foxp3. LuciDTR mice. J Immunol 184:1810–1820.
323.
Lubkov V, Bar-Sagi D. 2014. E-cadherin-mediated cell coupling is required for apoptotic cell extrusion. Curr Biol 24:868–874.
324.
Jiang A, Bloom O, Ono S, Cui W, Unternaehrer J, Jiang S, Whitney JA, Connolly J, Banchereau J, Mellman I. 2007. Disruption of E-cadherin-mediated adhesion induces a functionally distinct pathway of dendritic cell maturation. Immunity 27:610–624.
325.
Ohnmacht C, Pullner A, King SBS, Drexler I, Meier S, Brocker T, Voehringer D. 2009. Constitutive ablation of dendritic cells breaks self-tolerance of CD4 T cells and results in spontaneous fatal autoimmunity. J Exp Med 206:549–559.
326.
Birnberg T, Bar-On L, Sapoznikov A, Caton ML, Cervantes-Barragán L, Makia D, Krauthgamer R, Brenner O, Ludewig B, Brockschnieder D, Riethmacher D, Reizis B, Jung S. 2008. Lack of conventional dendritic cells is compatible with normal development and T cell homeostasis, but causes myeloid proliferative syndrome. Immunity 29:986–997.
327.
Eberl G, Colonna M, Di Santo JP, McKenzie ANJ. 2015. Innate lymphoid cells: a new paradigm in immunology. Science 348:aaa6566. doi:10.1126/science.aaa6566.
328.
McKenzie AN, Spits H, Eberl G. 2014. Innate lymphoid cells in inflammation and immunity. Immunity 41:366–374.
329.
Hazenberg MD, Spits H. 2014. Human innate lymphoid cells. Blood 124:700–709.
330.
Bar-Ephraïm YE, Mebius RE. 2016. Innate lymphoid cells in secondary lymphoid organs. Immunol Rev 271:185–199.
331.
Manh TP, Alexandre Y, Baranek T, Crozat K, Dalod M. 2013. Plasmacytoid, conventional, and monocyte-derived dendritic cells undergo a profound and convergent genetic reprogramming during their maturation. Eur J Immunol 43:1706–1715.
332.
Shalek AK, Satija R, Adiconis X, Gertner RS, Gaublomme JT, Raychowdhury R, Schwartz S, Yosef N, Malboeuf C, Lu D, Trombetta JJ, Gennert D, Gnirke A, Goren A, Hacohen N, Levin JZ, Park H, Regev A. 2013. Single-cell transcriptomics reveals bimodality in expression and splicing in immune cells. Nature 498:236–240.
333.
Shalek AK, Satija R, Shuga J, Trombetta JJ, Gennert D, Lu D, Chen P, Gertner RS, Gaublomme JT, Yosef N, Schwartz S, Fowler B, Weaver S, Wang J, Wang X, Ding R, Raychowdhury R, Friedman N, Hacohen N, Park H, May AP, Regev A. 2014. Single-cell RNA-seq reveals dynamic paracrine control of cellular variation. Nature 510:363–369.
334.
DuPage M, Bluestone JA. 2016. Harnessing the plasticity of CD4+ T cells to treat immune-mediated disease. Nat Rev Immunol 16:149–163.
335.
Mori L, Lepore M, De Libero G. 2016. The immunology of CD1- and MR1-restricted T cells. Annu Rev Immunol 34:479–510.
336.
Wang D, Sun B, Feng M, Feng H, Gong W, Liu Q, Ge S. 2015. Role of scavenger receptors in dendritic cell function. Hum Immunol 76:442–446.
337.
Vanden Berghe T, Linkermann A, Jouan-Lanhouet S, Walczak H, Vandenabeele P. 2014. Regulated necrosis: the expanding network of non-apoptotic cell death pathways. Nat Rev Mol Cell Biol 15:135–147.
338.
Pasparakis M, Vandenabeele P. 2015. Necroptosis and its role in inflammation. Nature 517:311–320.
339.
Yang WS, Stockwell BR. 2016. Ferroptosis: death by lipid peroxidation. Trends Cell Biol 26:165–176.
340.
Bergsbaken T, Fink SL, Cookson BT. 2009. Pyroptosis: host cell death and inflammation. Nat Rev Microbiol 7:99–109.
341.
Kumar V, Dasoveanu DC, Chyou S, Tzeng TC, Rozo C, Liang Y, Stohl W, Fu YX, Ruddle NH, Lu TT. 2015. A dendritic-cell-stromal axis maintains immune responses in lymph nodes. Immunity 42:719–730.
342.
Roozendaal R, Mebius RE. 2011. Stromal cell-immune cell interactions. Annu Rev Immunol 29:23–43.
343.
Turley SJ, Fletcher AL, Elpek KG. 2010. The stromal and haematopoietic antigen-presenting cells that reside in secondary lymphoid organs. Nat Rev Immunol 10:813–825.
344.
Shiow LR, Rosen DB, Brdicková N, Xu Y, An J, Lanier LL, Cyster JG, Matloubian M. 2006. CD69 acts downstream of interferon-α/β to inhibit S1P1 and lymphocyte egress from lymphoid organs. Nature 440:540–544.
345.
Hampton HR, Chtanova T. 2016. The lymph node neutrophil. Semin Immunol 28:129–136.
346.
Chakarov S, Fazilleau N. 2014. Monocyte-derived dendritic cells promote T follicular helper cell differentiation. EMBO Mol Med 6:590–603.
347.
Johansson-Lindbom B, Svensson M, Pabst O, Palmqvist C, Marquez G, Förster R, Agace WW. 2005. Functional specialization of gut CD103+ dendritic cells in the regulation of tissue-selective T cell homing. J Exp Med 202:1063–1073.
348.
Annunziato F, Romagnani C, Romagnani S. 2015. The 3 major types of innate and adaptive cell-mediated effector immunity. J Allergy Clin Immunol 135:626–635.
349.
Raphael I, Nalawade S, Eagar TN, Forsthuber TG. 2015. T cell subsets and their signature cytokines in autoimmune and inflammatory diseases. Cytokine 74:5–17.
350.
Weinmann AS. 2014. Roles for helper T cell lineage-specifying transcription factors in cellular specialization. Adv Immunol 124:171–206.
351.
Arens R, Schoenberger SP. 2010. Plasticity in programming of effector and memory CD8 T-cell formation. Immunol Rev 235:190–205.
352.
Peixoto A, Evaristo C, Munitic I, Monteiro M, Charbit A, Rocha B, Veiga-Fernandes H. 2007. CD8 single-cell gene coexpression reveals three different effector types present at distinct phases of the immune response. J Exp Med 204:1193–1205.
353.
Monteiro M, Evaristo C, Legrand A, Nicoletti A, Rocha B. 2007. Cartography of gene expression in CD8 single cells: novel CCR7 subsets suggest differentiation independent of CD45RA expression. Blood 109:2863–2870.
354.
Buchholz VR, Schumacher TN, Busch DH. 2016. T cell fate at the single-cell level. Annu Rev Immunol 34:65–92.
355.
Shin H, Iwasaki A. 2013. Tissue-resident memory T cells. Immunol Rev 255:165–181.
356.
Kastenmüller W, Brandes M, Wang Z, Herz J, Egen JG, Germain RN. 2013. Peripheral prepositioning and local CXCL9 chemokine-mediated guidance orchestrate rapid memory CD8+ T cell responses in the lymph node. Immunity 38:502–513.
357.
Alexandre YO, Ghilas S, Sanchez C, Le Bon A, Crozat K, Dalod M. 2016. XCR1+ dendritic cells promote memory CD8+ T cell recall upon secondary infections with Listeria monocytogenes or certain viruses. J Exp Med 213:75–92.
358.
Wakim LM, Waithman J, van Rooijen N, Heath WR, Carbone FR. 2008. Dendritic cell-induced memory T cell activation in nonlymphoid tissues. Science 319:198–202.
359.
Ángeles Esteban M. 2012. An overview of the immunological defenses in fish skin. ISRN Immunol 2012:1–29.
360.
Han Q, Das S, Hirano M, Holland SJ, McCurley N, Guo P, Rosenberg CS, Boehm T, Cooper MD. 2015. Characterization of lamprey IL-17 family members and their receptors. J Immunol 195:5440–5451.
361.
Lugo-Villarino G, Balla KM, Stachura DL, Bañuelos K, Werneck MBF, Traver D. 2010. Identification of dendritic antigen-presenting cells in the zebrafish. Proc Natl Acad Sci U S A 107:15850–15855.
362.
Flajnik MF, Kasahara M. 2010. Origin and evolution of the adaptive immune system: genetic events and selective pressures. Nat Rev Genet 11:47–59.
363.
Hirano M. 2015. Evolution of vertebrate adaptive immunity: immune cells and tissues, and AID/APOBEC cytidine deaminases. BioEssays 37:877–887.
364.
Sun JC, Ugolini S, Vivier E. 2014. Immunological memory within the innate immune system. EMBO J 33:1295–1303.
365.
Gross M, Salame TM, Jung S. 2015. Guardians of the gut—murine intestinal macrophages and dendritic cells. Front Immunol 6:254. doi:10.3389/fimmu.2015.00254.
366.
Star B, Nederbragt AJ, Jentoft S, Grimholt U, Malmstrøm M, Gregers TF, Rounge TB, Paulsen J, Solbakken MH, Sharma A, Wetten OF, Lanzén A, Winer R, Knight J, Vogel JH, Aken B, Andersen O, Lagesen K, Tooming-Klunderud A, Edvardsen RB, Tina KG, Espelund M, Nepal C, Previti C, Karlsen BO, Moum T, Skage M, Berg PR, Gjøen T, Kuhl H, Thorsen J, Malde K, Reinhardt R, Du L, Johansen SD, Searle S, Lien S, Nilsen F, Jonassen I, Omholt SW, Stenseth NC, Jakobsen KS. 2011. The genome sequence of Atlantic cod reveals a unique immune system. Nature 477:207–210.
367.
Kasahara M. 2007. The 2R hypothesis: an update. Curr Opin Immunol 19:547–552.
368.
De Silva NS, Klein U. 2015. Dynamics of B cells in germinal centres. Nat Rev Immunol 15:137–148.
369.
Dudda JC, Simon JC, Martin S. 2004. Dendritic cell immunization route determines CD8+ T cell trafficking to inflamed skin: role for tissue microenvironment and dendritic cells in establishment of T cell-homing subsets. J Immunol 172:857–863.

Information & Contributors

Information

Published In

cover image Microbiology Spectrum
Microbiology Spectrum
Volume 4Number 630 December 2016
eLocator: 10.1128/microbiolspec.mchd-0046-2016
Editor: Siamon Gordon, Oxford University, Oxford, United Kingdom

History

Received: 1 August 2016
Returned for modification: 22 September 2016
Published online: 2 December 2016

Contributors

Author

Jonathan M. Austyn
Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, United Kingdom

Editor

Siamon Gordon
Oxford University, Oxford, United Kingdom

Notes

Correspondence: Jonathan M. Austyn, [email protected]

Metrics & Citations

Metrics

Note:

  • For recently published articles, the TOTAL download count will appear as zero until a new month starts.
  • There is a 3- to 4-day delay in article usage, so article usage will not appear immediately after publication.
  • Citation counts come from the Crossref Cited by service.

Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. For an editable text file, please select Medlars format which will download as a .txt file. Simply select your manager software from the list below and click Download.

View Options

Figures and Media

Figures

Media

Tables

Share

Share

Share the article link

Share with email

Email a colleague

Share on social media

American Society for Microbiology ("ASM") is committed to maintaining your confidence and trust with respect to the information we collect from you on websites owned and operated by ASM ("ASM Web Sites") and other sources. This Privacy Policy sets forth the information we collect about you, how we use this information and the choices you have about how we use such information.
FIND OUT MORE about the privacy policy