You are on page 1of 21

OVERVIEW OF BASIC SCIENCE SECTION 1

Immunology
Thomas Schwarz
4
enzymatic amplifying cascade (see Ch. 60)4. Three pathways can trigger
Key features this cascade. The classical pathway is stimulated by antigen–antibody
■ The major purpose of the immune system is protection against complexes, the alternative pathway by polysaccharides derived from
harmful organisms. This is achieved by a rapid “primitive” reaction, microbial cell walls, and the later identified lectin pathway by the
called the innate immune response, and a more highly developed interaction of microbial carbohydrates with mannose-binding proteins.
specific reaction, called the adaptive immune response All three pathways lead to activation of the central C3 component and,
■ The characteristics of an adaptive immune response are specificity finally, the generation of a number of immunologically active sub-
and the accumulation of memory, thus enabling improvement stances. For example, C3b, the cleavage product of C3, binds to the
with each successive encounter with a particular antigen surface of microbes. Since phagocytic cells express receptors for C3b,
phagocytosis of the microorganisms is enhanced. In addition, comple-
■ The key event in an adaptive immune response is antigen
ment components bind to antigen–antibody immune complexes, which
presentation, yielding either a cell-mediated or a humoral
help complement receptor-bearing antigen-presenting cells to target
response. The cellular response involves primarily T cells, whereas
these immune complexes.
the humoral response involves B cells that ultimately mature into
C5a is a powerful attractant for neutrophils. C3a, C4a and C5a, also
antibody-secreting plasma cells
called anaphylatoxins, induce the release of inflammatory mediators
■ Immune responses are not always protective and can even be from mast cells. This increases vascular permeability, thereby enabling
pathogenic if the response induces severe tissue destruction or is proteins (e.g. antibodies) to enter the tissue. Assembly of the comple-
directed against an autoantigen ment components C5b, C6, C7, C8 and C9 forms the membrane-
■ As a barrier organ to the external environment, the skin is attack complex (MAC), which generates pores in cell membranes,
endowed with the capacity and the necessary cellular components causing death by osmotic lysis. Human cells are much less susceptible
to mount an immune response to killing by complement than are microbes, since human cells express
the complement receptor type 1 (CR1, CD35), decay-accelerating factor
(DAF, CD55), and membrane cofactor protein (MCP, CD46), which
inhibit C3 convertase and thereby block progression of the complement
INTRODUCTION cascade. CD59 is a protein that binds to C8 and inhibits insertion of
C9 into the cell membrane.
The skin is in a sense a defense organ, since it represents a major
barrier against the outside environment. As such, it is constantly con-
fronted with microbial, chemical, and physical insults. Within the past Toll-Like Receptors
four decades, there has been a greater appreciation for the fact that the Innate immunity serves to recognize invading microorganisms and then
skin not only functions as a mechanical barrier to the outside world, induce a host defense response. Several families of pattern recognition
but also uses the immune system for protection. Accordingly, the skin receptors (PRRs) mediate responses to pathogen-associated molecular
is endowed with the capacity to generate an immune response, which patterns (PAMPs) that are conserved among microorganisms. Toll-like
gave rise to the term “skin-associated lymphoid tissues” (SALT)1. receptors (TLRs; the mammalian homologs of the Toll receptors identi-
The classical immune response, also referred to as the adaptive fied in Drosophila) are one such family of PRRs. Ten TLRs have been
immune response, is characterized by specificity that is due to immu- identified to date5 (Fig. 4.1), with the following specificities: TLR2 (in
nologic memory (specific immunity)2. Innate immunity is a more association with either TLR1 or TLR6), recognition of lipoproteins and
primitive defense system that acts in a rapid but less specific manner. peptidoglycans; TLR4, lipopolysaccharide; TLR5, flagellin (a compo-
Both types of responses can be generated in the skin. Adaptive immune nent of bacterial flagella); and TLR9, bacterial CpG DNA sequences.
responses in the skin, however, are not always protective but can also TLRs may also be involved in the recognition of viral components.
be harmful in nature, e.g. allergic or autoimmune reactions. Numerous The signaling pathway of TLRs is highly homologous to that of the
skin diseases are caused by T lymphocytes and are therefore immuno- receptor for interleukin-1 (IL-1). Upon interaction with myeloid dif-
logically mediated. Consequently, many dermatoses respond favorably ferentiation factor 88 (MyD88), IL-1 receptor-associated kinase (IRAK)
to immunosuppressive therapy administered either systemically or is recruited, ultimately leading to activation of the transcription factor
topically. NF-κB (see Fig. 4.1). Activation of TLRs can also result in the release
of interferons (IFNs) via activation of interferon regulatory factor 3
(IRF3).
INNATE IMMUNE RESPONSE Dendritic cells express several types of TLRs. Upon activation of
these receptors by microbial components, the dendritic cells mature
Innate immune responses are characterized by a lack of immunologic
and migrate to the lymph nodes, where they present pathogen-derived
memory. These immune reactions are less complicated than adaptive
antigens to naive T cells and induce an adaptive immune response.
responses and developed earlier in evolution3. Nevertheless, failures in
TLRs thereby bridge the gap between the innate and adaptive immune
these “primitive” immune responses may be associated with severe,
systems6. These two systems constantly interact in the skin, and the
even fatal, health problems. Essential components of the innate
innate immune system represents a potential target for modulating
response are neutrophils, eosinophils, natural killer cells, mast cells,
adaptive immune responses7. Cutaneous dendritic cells that are stimu-
cytokines, complement, and antimicrobial peptides. The innate
lated by the innate immune system not only instruct T cells to respond
response is more rapid and less controlled than the adaptive immune
but also tell them how and where. Different PAMPs and danger signals
response.
polarize dendritic cells, giving them the ability to produce certain cyto-
kines and to induce T cells to differentiate into particular subtypes.
Complement Many of these danger signals are provided by keratinocytes, which also
The complement system plays an important role in innate immunity. express PRRs. In addition, TLRs expressed in the skin direct control of 81
It consists of at least 20 serum glycoproteins that are activated by an pathogens by the epithelium.
non-print metadata
ABSTRACT KEYWORDS:
The major purpose of the immune system is to provide protection adaptive immunity,
against harmful agents. This is achieved by a rapid “primitive” reaction, cutaneous immunology,
CHAPTER
called the innate immune response, and a more highly developed spe- antigen presentation,
cific reaction, called the adaptive immune response. The characteristics
of an adaptive immune response are specificity and the accumulation
B cells,
cytokines,
4
of memory, thus enabling improvement with each successive encounter dendritic cells,

Immunology
with a particular antigen. The key event during an adaptive immune innate immunity,
response is antigen presentation, yielding either a cell-mediated or a Langerhans cells,
humoral response. The cellular response involves primarily T cells, T cells,
whereas the humoral response involves B cells that ultimately mature Th17
into antibody-secreting plasma cells. More recently it has become clear
that there is a close crosstalk between the innate and adaptive immune
systems. Immune responses are not always protective and can even be
pathogenic if the response induces severe tissue destruction or is
directed against an autoantigen. As a barrier organ to the external
environment, the skin is endowed with the capacity and the necessary
cellular components to mount an immune response.

81.e1
Fig. 4.1 Toll-like receptors, their ligands and
TOLL-LIKE RECEPTORS, THEIR LIGANDS AND SIGNALING PATHWAYS signaling pathways. Toll-like receptors (TLRs)
recognize pathogen-associated molecular patterns
and/or synthetic compounds in a specific fashion. A
SECTION Triacylated Diacylated TLR2/TLR1 dimer recognizes triacylated lipoproteins,

1 and a TLR2/TRL6 dimer interacts with diacylated


lipoproteins lipoproteins Flagellin LPS ?
Imidazo- CpG lipoproteins. TLR5 recognizes flagellin and TLR4
quinolines ssRNA DNA dsRNA recognizes lipopolysaccharide (LPS). These TLRs are
OVERVIEW OF BASIC SCIENCE

located on the cell membrane and are internalized


TLR1 TLR2 TLR2 TLR6 TLR5 TLR4 TLR10
upon ligand interaction. TLR3, TLR7, TLR8 and TLR9
are located on intracellular membranes of
endosomes and lysosomes. TLR3 recognizes viral
double-stranded RNA (dsRNA); TLR7 and TLR8 viral
TIRAP MyD88 TIRAP MyD88 MyD88 TLR7 TLR8 TLR9 TLR3 TRIF MyD88 single-stranded RNA (ssRNA); and TLR9 bacterial and
viral hypomethylated DNA (CpG motifs). TLR7 and
?
TLR8 also bind to synthetic compounds
(imidazoquinolones). All TLRs except TLR3 utilize
MyD88 MyD88 MyD88 TRIF Endosome myeloid differentiation factor 88 (MyD88) for
signaling, while TLR2 and TLR6 also require Toll-
interleukin-1 receptor domain-containing adaptor
protein (TIRAP). MyD88 signaling via IL-1 receptor-
IRAK4 associated kinase-4 (IRAK-4) and tumor necrosis
TRAF6 factor receptor-activated factor-6 (TRAF-6) mostly
results in activation of nuclear factor κB (NF-κB),
ultimately inducing transcription of genes encoding
immunomodulatory and proinflammatory molecules.
IRF3 IFN-!/" TLR3 and TLR4 signal via TIR-domain-containing
adapter-inducing interferon-β (TRIF). The TRIF
pathway induces the production of interferons (IFNs)
Cytoplasm Immunomodulatory Nucleus via interferon regulatory factor-3 (IRF3). Both the
NF-#B
genes ligand and signaling pathway of TLR10 are still
unknown. Adapted from Miller LS. Toll-like receptors in skin. Adv
Dermatol. 2008;24:71–87 and from McInturff JE, Modlin RL, Kim J. The
role of toll-like receptors in the pathogenesis and treatment of
dermatological disease. J Invest Dermatol. 2005;125:1–8.

Inflammasomes peptides can be induced by bacteria, bacterial products, or proinflam-


matory cytokines via TLRs and other mechanisms. Enhanced and
Closely related to these processes are inflammasomes, innate immune
reduced production of these peptides in psoriasis and atopic dermatitis,
complexes which sense intracellular danger-associated molecular pat-
respectively, may explain why superinfections are so rare in the former
terns (DAMPs) or PAMPs. Four types of inflammasomes have been
disease and common in the latter9,11. However, others have found
identified: Aim2 (absent in melanoma 2), the pyrin domain-containing
enhanced expression of antimicrobial peptides in atopic skin, which
NLRP1 (nucleotide-binding domain leucine-rich repeat-containing
may reflect disruption of the epidermal barrier12. Ultraviolet (UV) B
receptor 1, also called NALP), NLRP3, and NLRC4 (Nod-like receptor
radiation has been shown to induce expression of antimicrobial pep-
CARD domain-containing 4)8. Aim2 is activated by double-stranded
tides, potentially explaining the lack of UVB-related bacterial infections
DNA resulting from either viral or bacterial intracellular pathogens,
despite its immunosuppressive effects (see Ch. 86)13.
NLRP1 by muramyl dipeptide, and NLRC4 by flagellin. NLRP3 is trig-
Beta-defensins can also attract immature dendritic cells and memory
gered by numerous PAMPs and DAMPs and is thus the most impor-
T cells via the chemokine receptor (CCR)-6, illustrating another link
tant inflammasome (Fig. 4.2). The ultimate result of activation of
between innate epithelial defense and adaptive immunity14. The anti-
inflammasomes is the cleavage of pro-IL-1β into active IL-1β, a highly
microbial peptide LL-37 (also known as cathelicidin antimicrobial
potent inflammatory mediator. NLRPs are large backbone proteins of
peptide [CAMP]) mediates dendritic cell activation in psoriasis by
the complex which, upon assembly of the inflammasome complex, bind
binding self-DNA and forming structures that stimulate TLR9 and
to ASC (apoptosis-associated speck-like protein containing a caspase
thereby induce IFN production15.
recruitment domain [CARD]). The latter interacts with caspase-1,
resulting in its activation. Active caspase-1 cleaves and activates pro-IL-
1β, as well as the proinflammatory cytokine pro-IL-18. Dysregulation Cytokines
of inflammasomes is the cause of a group of inherited autoinflamma- Cytokines are a large, heterogeneous family of low-molecular-weight
tory diseases called cryopyrin-associated periodic syndromes (CAPS) messenger substances that play a crucial role in intercellular commu-
which are associated with recurrent episodes of fever, urticarial skin nication. Cytokines can be secreted by almost any cell type, and they
lesions, arthritis, and systemic inflammation (see Ch. 45). These dis- may act in an autocrine, paracrine, or endocrine manner. Cytokines
orders respond very well to IL-1 blocking drugs (see Fig. 4.2). exert their biologic activities by binding to specific cell surface recep-
tors16. Although the vast majority of cytokines occur in a soluble form,
some can be membrane-bound, making the differentiation between
Antimicrobial Peptides cytokine and receptor difficult. Cytokines influence the proliferation,
To cope with an environment that is full of microorganisms, plants and differentiation, and activation of cells. Each cytokine exhibits multiple
invertebrates produce a variety of highly effective antimicrobial pro- activities, a fact that complicates strict categorization.
teins. Human epithelia, including the epidermis, secrete such anti- Cytokines that are produced by leukocytes and exert effects prefer-
microbial peptides as a mechanism of innate defense. The first entially on other white blood cells are called interleukins (IL). Colony-
antimicrobial peptide to be found in human skin (specifically from stimulating factors (CSFs) are mediators that induce differentiation
psoriatic scales) was human β-defensin-2 (hBD-2)9. A number of other and proliferation of hematopoietic progenitor cells, while IFNs interfere
antimicrobial peptides have subsequently been isolated (Table 4.1). In with viral replication. Cytokines that have chemoattractant activity are
addition to antibacterial properties, some of these peptides possess termed chemokines, and they play a crucial role in leukocyte migration.
antimycotic and likely antiviral activities. As demonstrated for psoria- The main subgroups of chemokines are differentiated according to the
82 sin, which prevents Escherichia coli infection10, these peptides may position of two cysteine (C) residues compared with the other amino
protect the skin from bacterial infections. Expression of antimicrobial acid residues (X), CXC (α-chemokines) and CC (β-chemokines)17.
Fig. 4.2 NLRP3 inflammasome
NLRP3 INFLAMMASOME AND ITS RELATIONSHIP TO SEVERAL HEREDITARY PERIODIC FEVER SYNDROMES and its relationship to several
AND AUTOINFLAMMATORY DISORDERS
hereditary periodic fever
syndromes and autoinflammatory
Lipopolysaccharide disorders. Upon release of CHAPTER

T
autoinhibition, cryopyrin interacts
with apoptosis-associated
speck-like protein with a CARD
4
Stimulus
L

Immunology
(e.g. bacterial domain (ASC) and CARD-inhibitor
R
4
RNA) of NF-κB-activating ligand
(CARDINAL) within an
IL-1R inflammasome, leading to
CAPS: PAPA activation of caspase-1 and
Muckle Wells syndrome syndrome generation of mature interleukin
Familial cold autoinflammatory (IL)-1β. Pyrin can competitively
syndrome Familial bind ASC and (pro)-caspase-1,
NOMID/CINCA syndrome PSTPIP1 Mediterranean
thereby preventing them from
fever
being incorporated into the NLRP3
Cryopyrin
Pyrin inflammasome. Proline–serine–
(inactive)
threonine phosphatase-interacting
protein 1 (PSTPIP1) binds (and
perhaps inhibits) pyrin, and this
interaction is strengthened by
Domains the PSTPIP1 mutations that
B30.2 underlie PAPA syndrome.
CAPS, cryopyrin-associated
CARD
periodic syndromes; CARD,
Caspase-1 caspase-recruitment domain;
CINCA, chronic infantile neurologic,
Coiled coil cutaneous and articular syndrome;
Leucine-rich IL-1R, interleukin-1 receptor; NF-κB,
Cryopyrin nuclear factor-κB; NLRP3, NOD-like
repeats
(active) receptor pyrin domain-containing
NOD
NF-κB protein 3; NOD, nucleotide-binding
Pyrin
oligomerization domain; NOMID,
Zinc finger neonatal-onset multisystem
NLRP3 inflammasome
Proteins inflammatory disease; PAPA,
pyogenic arthritis, pyoderma
ASC
gangrenosum and acne; RIP2,
CARDINAL receptor-interacting protein-2; TLR,
Pro- Toll-like receptor; TNF, tumor
TNF-α necrosis factor. Courtesy, Julie V Schaffer,
Pro-IL-1β caspase-1
MD.
IL-1R antagonist
(anakinra)
Pro-IL-1β IL-1!
IL-1-TRAP
(rilonacept)
Nucleus Lysosome
mAb anti-IL-1β
(canakinumab)

Chemokines that recruit leukocytes are termed inflammatory chemo- macrophages possess receptors for antibodies and complement. Hence,
kines, whereas those that regulate trafficking within lymphoid tissues microorganisms that are coated with antibodies and/or complement are
are called lymphoid chemokines. more readily phagocytosed18. After phagocytosis, the microorganisms
Early innate immune responses are dominated by cytokines with are exposed to a variety of toxic intracellular molecules, including
inflammatory (e.g. IL-1, IL-6, IL-18, tumor necrosis factor-α [TNF-α], superoxide anions, hydroxyl radicals, hypochlorous acid, nitric oxide,
inflammatory chemokines) and antiviral (e.g. IFN-α, IFN-β) capacities. lysozyme, and antimicrobial cationic proteins. Macrophages can also
Induction of adaptive immune responses is critically dependent on present processed antigens to T and B cells. However, their T-cell
cytokines with immunomodulatory capacities (e.g. IL-2, IL-4, IL-12, stimulatory capacity is less effective than that of dendritic cells.
IL-13, IL-17, IL-22, IL-23, IFN-γ). However, since most of these media- Activated macrophages release granulocyte colony-stimulating factor
tors exhibit multiple and sometimes overlapping activities, a strict (G-CSF) and granulocyte–macrophage colony-stimulating factor (GM-
separation into inflammatory and immunomodulatory cytokines is not CSF). These two cytokines induce the division of myeloid precursors
possible. Due to structural similarities, some cytokines are grouped in the bone marrow, releasing millions of neutrophils into the circula-
into families, e.g. the IL-6 family (IL-6, IL-11, oncostatin M, leukemia tion. Under normal conditions, neutrophils circulate in the blood-
inhibitory factor [LIF]), the IL-10 family (IL-10, IL-19, IL-20, IL-22, stream, some rolling along the vascular endothelium19. To enter the
IL-24, IL-26), and the IL-12 family (IL-12, IL-23, IL-27). site of an infection, neutrophils utilize a complex process that involves
proinflammatory mediators, adhesion molecules, chemoattractants,
and chemokines (see Ch. 26). The recruited neutrophils phagocytose
Macrophages and Neutrophils organisms and kill them within phagolysosomes by using oxygen-
Macrophages, phagocytic cells derived from blood-borne monocytes, dependent and oxygen-independent mechanisms. The former, called
carry receptors for carbohydrates that are usually not expressed on the respiratory burst, involves the production of hydrogen peroxide,
vertebrate cells (e.g. mannose). Through this mechanism, macrophages hydroxyl radicals, and singlet oxygen. The oxygen-independent method 83
can discriminate between “foreign” and “self” molecules. Furthermore, utilizes highly toxic cationic proteins and enzymes, such as
SKIN-DERIVED ANTIMICROBIAL PEPTIDES

Antimicrobial activity
SECTION
Inducibility
Size Bacteria Fungi (e.g. by bacteria,
1 Name (kD) Cellular source Gram+ Gram− cytokines)
Antileukoprotease (ALP) 11.7 Keratinocytes ++ ++ ++ −
OVERVIEW OF BASIC SCIENCE

Airway epithelia
Dermcidin (DCD)-1 4.7 Sweat glands +++ +++ ++ −
Human β-defensin (HBD)-2 4.3 Keratinocytes (+)* +++ ++ +
Airway epithelia
Intestinal tract
HBD-3 5.2 Keratinocytes +++ +++ +++ +
Airway epithelia
HBD-4 6.0 Keratinocytes ++ ++ + +
Airway epithelia (mRNA)
LL-37/cathelicidin antimicrobial peptide (CAMP)/human cationic 4.5 Keratinocytes ++ ++ ++ +
antimicrobial peptide 18 (hCAP18) Airway epithelia
Urogenital tract
Granulocytes
Lysozyme 14.7 Keratinocytes ++ ++ − −
Airway epithelia
Psoriasin 11.4 Keratinocytes (+)* ++† (+)* +
Sebocytes
RNase 7 14.5 Keratinocytes +++ +++ +++ +
Airway epithelia

*† In high concentrations.
E. coli, others in high concentrations.
Table 4.1 Skin-derived antimicrobial peptides. In the skin, antileukoprotease (ALP) is also referred to as SKALP, skin-derived ALP.

myeloperoxidase (MPO) and lysozyme. Organisms that are coated with Basophils and Mast Cells
antibodies or complement components bind to Fc and complement
Basophils (found in the blood) and mast cells (located within tissues)
receptors, respectively, on neutrophils (as well as macrophages) and are
exhibit similar functional and morphologic characteristics22. At least
more effectively phagocytosed and killed.
two populations of mast cells exist, which can be differentiated by the
Ectosomes are vesicles released from the plasma membrane of neu-
enzymes they contain and by their tissue location. Mucosal mast cells
trophils upon their stimulation (e.g. by bacteria)20. They carry mem-
contain only trypsin, while connective tissue mast cells contain both
brane surface receptors (e.g. CD15, L-selectin), phosphatidylserine, and
trypsin and chymotrypsin (see Ch. 118). In contrast to pulmonary,
granule proteins (e.g. CD66b, CD87, MPO, elastase, proteinase 3,
uterine and tonsillar mast cells, cutaneous mast cells express the recep-
defensins, lactoferrin, collagenase I). Depending on the composition,
tor for C5a (CD88), which implies that triggering of mast cells through
ectosomes can induce a variety of responses in their target cells which
the anaphylatoxin C5a results in cutaneous, but not systemic, reac-
include endothelial cells, platelets, natural killer cells, and dendritic
tions23. Basophils and mast cells express high-affinity receptors for IgE
cells.
(FcεRI) that avidly bind IgE (see Ch. 18).
In addition, neutrophils can release neutrophilic extracellular traps
When a specific antigen binds to mast cell-bound IgE, the FcεRI
(NETs) and thereby develop into anuclear cytoplasts20. NETs are com-
becomes activated, which leads to degranulation and release of pre-
posed of extracellular strands of DNA bound to neutrophil-derived
formed mediators, including histamine and serotonin. Other mediators
antimicrobial peptides and proteins. Because NETs can entrap bacteria,
such as prostaglandins, leukotrienes (B4, C4, D4 and E4), and platelet-
fungi and viruses, they may offer some antimicrobial protection. On
activating factor are also released, and they enhance vascular permeabil-
the other hand, NETs can induce autoimmunity by stimulating immune
ity, bronchoconstriction, and induction of an inflammatory response
responses against the NET-associated nuclear antigens (see Fig. 41.1).
(see Ch. 18). Hence, basophils and mast cells play an important role
Whether formation of NETs contributes to the exacerbation of auto-
in immediate-type allergic reactions such as urticaria and angioedema.
immune diseases by infections remains to be determined.
There is also evidence that mast cells are involved in contact hyper-
sensitivity reactions.
Eosinophils
The major function of eosinophils is most likely to protect the host Natural Killer Cells
from infections by parasites, particularly nematodes. Infections with The major task of natural killer (NK) cells is to eliminate virally
these organisms are associated with the production of antigen-specific infected or malignant cells24. NK cells can recognize their targets in
IgE antibodies that coat the parasite. Via their low-affinity recep- two ways. Since they express Fc receptors that bind IgG (FcγRIII,
tors (FcεRII, CD23), eosinophils bind to IgE antibodies and become CD16), NK cells can adhere to and kill target cells that are coated with
activated. In contrast to macrophages and neutrophils, eosinophils IgG. This killing process is referred to as antibody-dependent cellular
are only weakly phagocytic. They harbor large granules that contain cytotoxicity (ADCC).
major basic protein, eosinophilic cationic protein, eosinophil peroxi- The second recognition system involves killer-activating and killer-
dase, and eosinophil-derived neurotoxin (see Ch. 25). Upon activa- inhibitory receptors. Killer-activating receptors recognize molecules
tion, eosinophils release these toxic products, which can kill parasites, that are expressed by nucleated cells. This provides a signal for the NK
together with prostaglandins, leukotrienes, and various cytokines21. cell to kill the target cell by secretion of perforins, which make holes
84 Eosinophils also play an important role in the pathogenesis of allergic in the cell membrane through which granzymes are injected. Gran-
reactions. zymes lyse target cells by activating the apoptotic caspase cascade. In
addition, NK cells carry inhibitory receptors (KIR) on their surface that Activated LCs elongate their dendrites which can penetrate keratino-
recognize major histocompatibility complex (MHC) class I self- cyte tight junctions and survey the area just beneath the stratum
molecules. KIRs shut off the killer signal and thus prevent autolysis of corneum for antigens31. These penetrating dendrites take up antigens
the host. Tumor cells and viruses often down-regulate MHC class I which co-localize with langerin/Birbeck granules. By forming new tight
CHAPTER
molecules to escape recognition by cytotoxic T cells. Paradoxically, this junctions, keratinocytes rapidly close the defects produced by the pen-
mechanism renders “MHC class I-low” cells susceptible to recognition
by NK cells.
etrating dendrites, thereby maintaining skin integrity during antigen
uptake.
4
Histochemically, human LCs can be visualized by staining for

Immunology
adenosine triphosphatase (ATPase), a membrane-bound, formalin-
ADAPTIVE IMMUNE RESPONSE resistant, sulfhydryl-dependent enzyme. In addition to langerin
(CD207), antigenic moieties present on human LCs include the
The characteristic features of an adaptive immune response are its panhematopoietic marker CD45, MHC class II antigens (HLA-DR),
specificity and enhancement with each successive antigen encounter CD1a, the S100 protein, and vimentin. CD1a is a useful marker
owing to the accumulation of “memory”2,25. A crucial event during the for LCs, since within the epidermis (normal or inflamed) it is exclu-
generation of an adaptive immune response is antigen presentation. sively expressed on LCs, whereas HLA-DR antigens are expressed on
keratinocytes in inflamed skin (Table 4.2). Since CD1a does not exist
Antigen-Presenting Cells in the murine system, staining for MHC class II antigens is often
used for detection of murine LCs in non-perturbed skin32 (Fig. 4.3). In
Various cells can present antigens, depending on how and where the
addition, human LCs express the high-affinity IgE receptor (FcεRI)33,
antigen first encounters cells of the immune system. Interdigitating
which was initially thought to be exclusively expressed on mast cells
dendritic cells (DCs) located in the T-cell areas of the spleen and lymph
and basophils.
nodes are the most effective antigen-presenting cells (APCs). Within
The density of murine LCs is dependent on a variety of factors,
the epidermis, Langerhans cells (LCs) are key APCs. Therefore, the
including strain, age, sex, and anatomic location. LCs are almost absent
following section will focus primarily on LCs.
in the tail region, the pouch, and the cornea. In humans, the numbers
of LCs are reduced on the palms and soles, genitalia, and buccal
Langerhans cells mucosa. In addition, the density of LCs decreases with age and is
In 1868, Paul Langerhans26 first described Langerhans cells as DCs reduced in chronically UV-exposed skin32.
located within the epidermis26. Because of their dendritic shape, Lang-
erhans thought that these cells might be of neural origin. More than Ontogeny of Langerhans cells
100 years later, it was shown that LCs are derived from bone marrow27. Bone marrow chimera experiments showed that LCs are derived and
However, in the 1990s it was observed that LCs are intimately associ- constantly replenished from the bone marrow27. However, according to
ated with nerve fibers and that nerves, through the release of neuro- a more recent hypothesis30, murine LCs are derived from radioresistant
peptides such as calcitonin gene-related peptide (CGRP), can modulate hematopoietic precursor cells that reside in the skin during embryonic
LC function28. development (Fig. 4.4). The formation of LCs depends on transforming
growth factor-β1 (TGF-β1) and macrophage colony-stimulating factor
Morphology of Langerhans cells receptor (M-CSFR) ligands (M-CSF, IL-34), which are provided in an
LCs cannot be identified in routinely fixed and stained histologic sec- autocrine fashion. LCs that are depleted during the steady state or after
tions; their recognition requires electron microscopy or histochemical minor injuries are repopulated locally, independent of circulating pre-
analysis. Ultrastructurally, LCs have rod-shaped organelles termed cursor cells. In lethally irradiated mice reconstituted with congenic
Birbeck granules (see Fig. 91.5). A Ca2+-dependent lectin with mannose- bone marrow cells, half of the LCs are eliminated in the first week after
binding specificity called langerin is associated with and responsible for transplantation, but LCs repopulate locally within 3 weeks. After UVB
the formation of Birbeck granules29. Birbeck granules are thought to irradiation, which does not affect the dermis or the hair follicle, LCs
result from the antigen-capture function of langerin, which routes appear to repopulate from the hair follicle30. By producing different
antigen into these organelles and provides access to a non-classical chemokines, distinct subpopulations of hair follicle keratinocytes can
antigen-processing pathway. However, langerin is not completely spe- promote (via release of CCL2 and CCL20) or inhibit (via release of
cific for LCs and can also be expressed by a certain type of dermal DC30. CCL8) repopulation of LCs34.

PHENOTYPIC MARKERS OF RESIDENT VERSUS MIGRATING


HUMAN LANGERHANS CELLS VERSUS HUMAN LANGERIN−
DERMAL DENDRITIC CELLS

Resident Migrating Langerin−


LC LC dermal DC
Birbeck granules ++ ++ −
Langerin (CD207) +++ ++ −
MHC class II ++ +++ ++
CD45 + + +
CD1a +++ +++ ++
CD11c + + +
CD11b +/− +/− ++
E-cadherin ++ + −
Epithelial cell adhesion + + −
molecule (EpCAM)
CCR6 + − −
CCR7 − + + Fig. 4.3 Langerhans cells express MHC class II molecules. In a sheet
preparation of murine epidermis, numerous Langerhans cells can be visualized
Table 4.2 Phenotypic markers of resident versus migrating human by staining with an antibody against MHC class II molecules (Ia antigen). Note
Langerhans cells (LCs) versus human langerin− dermal dendritic cells (DCs). the dendritic shape of Langerhans cells. Courtesy, N Romani, Department of Dermatology, 85
CCR, CC-chemokine receptor. Adapted from ref. 30. University of Innsbruck.
ONTOGENY AND PHENOTYPE OF ANTIGEN-PRESENTING CELLS IN MURINE SKIN

SECTION

1
OVERVIEW OF BASIC SCIENCE

TGF-β1 MHC class II+


CD11c+
M-CSF CD11b+
Epidermis
IL-34 EpCAMhigh
Radioresistant CD103–
precursor cell CCR7 LC F4/80+

Migratory LC Langerin+ DC Langerin – DC Macrophage


MHC class II+ MHC class II+ MHC class II+ MHC class II low
CD11c+ CD11c+ CD11c+ CD11c –
CD11b+ CD11b low CD11b high CD11b high
EpCAMhigh EpCAM – EpCAM – EpCAM –
CD103– CD103 + CD103– CD103–
F4/80+ F4/80 – F4/80+ F4/80+

CCR7 CCR7 CCR2


E-selectin
P-selectin Dermis

Blood vessel

Draining = Birbeck granule


lymph node Precursor cell

Fig. 4.4 Ontogeny and phenotype of antigen-presenting cells in murine skin. Langerhans cells (LCs) are derived from radioresistant hematopoietic precursor
cells in the skin in the presence of transforming growth factor-β1 (TGF-β1), macrophage colony-stimulating factor (M-CSF), and interleukin (IL)-34. LCs migrate to
the draining lymph nodes in a CC-chemokine receptor 7 (CCR7)-dependent fashion. The dermis harbors dermal langerin+ dendritic cells (DCs), which differentiate
from radiosensitive circulating precursor cells in a CCR2-, E-selectin- and P-selectin-dependent manner. LCs on the way to the lymph nodes can be distinguished
from dermal langerin+ DCs by the differential expression of CD11b, epithelial cell adhesion molecule (EpCAM), and CD103. Two additional types of APCs are present
in the dermis, dermal langerin− DCs and dermal macrophages, and each has a particular surface marker expression pattern. Markers in italics differ from those in
LCs. Adapted from Merad M, Ginhoux F, Collin M. Origin, homeostasis and function of Langerhans cells and other langerin expressing dendritic cells. Nat Rev Immunol. 2008;8:935–47.

As noted above, langerin is not an exclusive marker for epidermal the bone marrow or spleen, while human DCs are almost exclusively
LCs but can also be expressed by dermal DCs (see Fig. 4.4)30. Langerin+ generated from peripheral blood. Furthermore, many markers/antibodies
dermal DCs differentiate from radiosensitive circulating precursor cells, exist in the human but not the murine system, and vice versa. Whether
independent of TGF-β1 and M-CSFR ligands but dependent on each of the different types of DCs has a distinct immune function is
CC-chemokine receptor 2 (CCR2), E-selectin, and P-selectin. LCs that still a matter of debate36.
emigrate from the epidermis to the lymph nodes in a CCR7-dependent Concerning DC function, there is evidence in the murine system that
manner can also be detected in the dermis but can be distinguished spleen-derived CD8α+ DCs induce T helper 1 (Th1) responses, while
from langerin+ dermal DCs by the differential expression of CD11b, CD8α− DCs favor T helper 2 (Th2) responses37. In contrast, in the
epithelial-cell adhesion molecule (EpCAM), and CD103 (see Fig. 4.4). human system, lymphoid/plasmacytoid DCs were found to induce Th2
Two additional types of APCs can be found in the dermis, langerin− responses, and myeloid DCs generated Th1 reactions38. Consequently,
dermal DCs and dermal macrophages, and each has a particular surface myeloid DCs were referred to as DC1 and lymphoid/plasmacytoid DCs
marker expression pattern. as DC2. Activation of TLR7 via the immunomodulator imiquimod, an
In humans, the situation is less clear than in mice, since only studies imidazoquinoline (see Fig. 4.1), drives myeloid DCs to express perforin
with tissue explants are available. Subtle differences in the surface and granzyme B and plasmacytoid DCs to express TNF-related
marker expression of resident LCs, migrating LCs, and langerin− dermal apoptosis-inducing ligand (TRAIL). This enables killing of tumor cells,
DCs have been described (see Table 4.2). indicating that both myeloid and plasmacytoid DCs are directly involved
in imiquimod-induced destruction of skin cancer39.
Other dendritic cells However, the type of immune response that is generated also criti-
DCs are defined as professional APCs that display an extraordinary cally depends on the state of maturation of the stimulating DC at the
capacity to stimulate naive T cells and initiate a primary immune time of antigen presentation. For the induction of Th1 responses, the
response35. This capacity was initially described for the interdigitating presence of IL-12 is crucial. DCs tend to produce IL-12 directly after
DCs in the spleen. It has become apparent that the DC system is very an activating step in their maturation, driving T cells into a Th1 phe-
complex, and many questions still remain unanswered. notype if they meet the DCs at this stage40. At later time points, the
This complexity is based on the fact that DCs can arise from several production of IL-12 decreases, thereby favoring the development of a
types of progenitor cells, and a variety of functional phenotypes of DCs Th2 response. This process is also heavily influenced by the innate
can also be generated from the same precursor cell36. In addition, dif- immune system (e.g. stimulation by TLRs; see above).
86 ferences exist between experimental systems used to study human and Taken together, the parameters that are responsible for the type of
murine DCs. In the murine system, DCs are primarily obtained from Th cell differentiation that is induced by DCs remain to be fully
defined. On the other hand, there is agreement that immature DCs The release of proteolytic enzymes like matrix metalloproteinase-9
induce tolerance due to incomplete T-cell activation, resulting in T cells (type IV collagenase) may enable their penetration through the base-
of the regulatory type that suppress immune responses41. ment membrane. Their dendricity becomes more pronounced, and
surface molecules necessary for antigen presentation and T-cell priming
CHAPTER
are upregulated (e.g. MHC class I, MHC class II, CD40, CD54, CD58,
Antigen presentation
Antigen presentation cells: activation and migration
CD80, CD86). At this stage, LCs emigrating from the epidermis are
almost indistinguishable from DCs obtained from lymphoid organs46.
4
To initiate sensitization, antigens must be presented to T cells by APCs. The same phenotypic and functional changes may occur in other APCs

Immunology
For many years, LCs were thought to be the most important APC in of the skin that have important roles in antigen presentation.
the skin since contact sensitization could not be induced in sites that
were naturally devoid of LCs (e.g. murine tail skin) or in which LCs Antigen presentation to T cells
had been depleted (e.g. by UV radiation)42. However, transgenic mice In contrast to B cells, T cells cannot recognize soluble protein antigens
in which LCs are completely depleted via the diphtheria toxin receptor per se; instead, the T-cell receptor (TCR) recognizes antigen-derived
technique (i.e. short-term, inducible ablation) demonstrated variably peptides bound to MHC locus-encoded molecules expressed on APCs.
diminished but not completely abrogated sensitization responses43; in CD4+ T cells recognize antigens in association with MHC class II
one such model, the sensitization response was completely normal44. molecules, while CD8+ T cells, the majority of which become cytotoxic,
In a different knockout mouse model characterized by constitutive and recognize antigens in association with MHC class I molecules25
durable absence of epidermal LCs45, an enhanced sensitization response (Fig. 4.5).
was actually observed, suggesting that LCs may have regulatory func- Two pathways exist by which antigens can be “loaded” onto MHC
tions. This “LC paradigm” proposes that LCs may be tolerogenic when molecules. If the antigen has been produced endogenously within the
they present antigens under steady-state non-inflammatory conditions, cell (e.g. viral or tumor proteins), it is complexed with MHC class I
but sensitizing upon stimulation by inflammatory mediators. Which of molecules through intracellular processing pathways (Fig. 4.6)49. The
these activities is the main function of LCs remains to be determined, proteasome degrades cytosolic antigens produced by the cell. The
and there is accumulating evidence that dermal DCs are equally if not resulting peptides (which consist of 8 to 12 amino acid residues) are
even more important than LCs in presenting antigens. imported into the endoplasmic reticulum (ER) in a TAP (transporter
Cutaneous APCs actively take up antigens in the skin, but antigen associated with antigen processing)-dependent manner and loaded onto
presentation to lymphocytes takes place in the regional lymph nodes. MHC class I molecules50. After binding to the MHC class I–β2 micro-
In the presence of inflammation, the APCs become activated and leave globulin complex, these peptides are transported to the cell surface via
the skin, migrating to the draining lymph nodes. During emigration, the Golgi apparatus. Alternative pathways by which exogenous proteins
they change their phenotypic and functional behavior and develop into are phagocytosed and the phagosome fuses to the ER may also exist.
mature DCs. For example, molecules involved in antigen uptake and The proteins would subsequently be retransported out of the ER into
processing (Birbeck granules, Fc receptors) are down-regulated on acti- the cytoplasm (by an as yet unclear mechanism) and then degraded
vated LCs46. In addition, expression of E-cadherin, which mediates the by the proteasome. The degraded peptides could then enter the pathway
attachment of LCs to neighboring keratinocytes, is reduced, thereby that is normally employed for endogenous proteins via the TAP protein.
enabling the emigration of LCs. As the vast majority of nucleated cells express MHC class I molecules,
CD44, a hyaluronic acid receptor involved in the tissue homing of many cell types can serve as APCs for MHC class I-restricted antigen
leukocytes, is upregulated on activated LCs. The splice variant CD44v6 presentation in a secondary immune response.
supports binding of LCs to T-cell-rich areas of lymph nodes47. Further- In contrast, MHC class II-dependent antigen presentation is critically
more, the integrins α6β1 and α6β4, which exhibit affinity to the base- dependent on DCs, B cells, and monocytes/macrophages. MHC class
ment membrane zone, are induced on the surface of emigrating LCs48. II-associated antigen presentation targets primarily exogenous (Fig. 4.7)

TYPES OF ANTIGEN PRESENTATION

Vα Jα Cα Vβ Dβ Jβ Cβ

Recombination Recombination
CD4+ CD8+
T cell T cell

α β α β
CD3 CD3
C C C C
CD4 CD8
J J J J
D D
V V V V
Antigenic peptide Antigenic peptide
(15-22aa) (8-12aa)

MHC MHC
class II β2m class I

APC APC

Fig. 4.5 Types of antigen presentation. Antigens are presented by antigen-presenting cells to the T-cell receptor of CD4+ or CD8+ T cells in association with either
MHC class II or MHC class I molecules. The diversity of the T-cell receptors is generated by gene rearrangement. For reasons of clarity, the simplified gene
rearrangement of the α-chain is shown only in the CD4+ T cell and that of the β-chain in the CD8+ T cell. aa, amino acids; APC, antigen-presenting cell; β2m,
β2-microglobulin; V, variable; D, diversity; J, joining; C, constant. Adapted from Modlin RL. Lymphocytes. In: Freedberg IM, Eisen AZ, Wolf K, et al. (eds). Fitzpatrick’s Dermatology in General Medicine, 87
vol. 1. New York: McGraw-Hill, 1999;32:400–5.
Fig. 4.6 The pathway of endogenous antigen
THE PATHWAY OF ENDOGENOUS ANTIGEN DELIVERY TO MHC CLASS I MOLECULES delivery to class I MHC molecules. Newly
synthesized MHC class I molecules are stabilized by
calnexin. When β2-microglobulin (β2m) binds to the
SECTION complex, calnexin dissociates. This complex

1 Cell
membrane Cytoplasm
associates with the TAP (transporter associated with
antigen processing) protein waiting for a suitable
peptide. Endogenous antigens are degraded by the
OVERVIEW OF BASIC SCIENCE

proteasome and transported via TAP into the


endoplasmic reticulum (ER), where they bind to the
MHC class I/β2m complex. Finally, the peptide/MHC
complex is transported through the Golgi apparatus
to the cell surface. Not pictured is a possible
Endoplasmic alternate pathway in which exogenous proteins are
reticulum phagocytosed and the phagosome fuses to the ER.
The proteins are then retransported out of the ER
into the cytoplasm and degraded by proteasomes.
The degraded peptides can now enter this pathway
via the TAP protein. Adapted from Parkin J, Cohen B. An overview
of the immune system. Lancet. 2001;357:1777–89. With permission from
Elsevier.
Calnexin

MHCI Golgi
apparatus
β m
2

TAP-1 and
TAP-2

Endogenous protein Proteasome Digested protein MHCI


(viral, tumor)

β m, β -microglobulin
2 2 Antigen
TAP, transporter associated with antigen processing

Fig. 4.7 The pathway of exogenous antigen


THE PATHWAY OF EXOGENOUS ANTIGEN DELIVERY TO MHC CLASS II MOLECULES delivery to class II MHC molecules. Exogenous
antigens are taken up into the cell by endosomes.
The protein is cleaved into peptides within the
Cell membrane endosomes, which become increasingly acidic. In the
Exogenous endoplasmic reticulum (ER), newly synthesized MHC
protein
class II molecules bind to an invariant chain, which
Cytoplasm inhibits their disassociation while empty. During this
assembly, the complex is stabilized by calnexin. The
invariant chain transports the MHC class II molecule
from the ER (via the Golgi apparatus) into the
endosomal compartment, where the MHC class II
complex meets the peptides. The invariant chain is
cleaved, leaving a small fragment (CLIP) in the groove
CLIP of the MHC class II molecule. Finally, CLIP is replaced
Endosome by the antigenic peptide. The complex is transported
to the cell membrane and expressed on the cell
surface. CLIP, class II-associated invariant peptide.
Endoplasmic Adapted from Parkin J, Cohen B. An overview of the immune system.
reticulum Lancet. 2001;357:1777–89. With permission from Elsevier.

Calnexin

Invariant
chain

MHCII

Peptide
88 antigen
and less frequently endogenous antigens51. Exogenous antigens are class II molecules, whereas CD8+ T cells are usually cytotoxic and
taken up via macro- or micropinocytosis or by receptor-mediated endo- recognize antigens in association with MHC class I molecules (see Fig.
cytosis. One example of the latter is the DEC-205 receptor (CD205), 4.5). During their early stage of development within the thymus, T
which guides antigens into endocytic vesicles that reside deeper inside cells express both CD4 and CD858. Upon expression of an appropriate
CHAPTER
the cell and contain MHC class II molecules. As a consequence of this TCR, these immature T cells exhibit the capacity to recognize antigenic
unique intracellular targeting, antigens endocytosed by the DEC-205
receptor stimulate respective T cells up to 500-fold greater than anti-
peptides associated with either MHC class I or MHC class II molecules,
since they still express both CD4 and CD8 (double-positive stage). In
4
gens taken up by pinocytosis or other receptors. Protein degradation the subsequent process of maturation, one of these surface markers is

Immunology
eventually takes place in the endo-/lysosomes, yielding peptides with a lost, resulting in single-positive T cells expressing either CD4 or CD8.
typical length of 15 to 22 amino acid residues. These peptide fragments These cells are specialized for peptides presented only by MHC class
enter specialized endosomal compartments containing MHC class II II or MHC class I molecules, respectively.
molecules that are generated in the ER52.
Newly synthesized MHC class II molecules are associated with an T-cell receptor
invariant chain, which inhibits dissociation of empty MHC class II One major feature of an adaptive immune response is the recognition
molecules and transports the MHC class II complex from the ER to of specific antigens. This is achieved either by the TCR during a T-cell-
the specialized endosomal compartments where the molecules can mediated response or by antibodies during a B-cell response. TCRs are
interact with antigen peptide fragments (see Fig. 4.7). In this compart- transmembrane molecules consisting of α/β (vast majority of T cells)
ment, the invariant chain is cleaved by proteases, leaving a small frag- or γ/δ (<10% of T cells) heterodimers59. Antigen recognition by γ/δ T
ment called CLIP (class II-associated invariant peptide) that binds to cells appears to be independent of classic MHC molecules, instead
the class II molecule. Upon interaction with the antigenic peptides, the utilizing other MHC molecules (e.g. CD1) that preferentially present
CLIP fragment is released from the complex. The MHC class II mol- certain lipid and glycolipid antigens60.
ecule with the bound antigen peptide is subsequently expressed on the
cell surface, allowing antigen recognition by T cells carrying the appro- T-cell receptor diversity
priate TCR. An amazing feature of the immune system is that it provides specific
Classically, MHC class I molecules present self- or pathogen-derived receptors for every possible antigen. B cells are capable of producing
antigens that are synthesized within the cell to CD8+ T cells (see Fig. approximately 1016 different antibody variable regions, and T cells
4.6), whereas exogenous antigens derived via endocytic uptake are generate a comparable number of TCR variable regions. Remarkably,
loaded onto MHC class II molecules for presentation to CD4+ T cells these numerous proteins are encoded by fewer than 400 genes. This
(see Fig. 4.7). However, it has become evident that some DCs are also tremendous diversity is created via a unique recombination process that
able to process exogenous antigens into the MHC class I pathway for cuts, splices, and modifies genes in the variable region61.
presentation to CD8+ T cells. This mechanism, referred to as cross- Four segments of genes are involved in TCR formation: the variable
presentation53, allows DCs to induce either tolerance (to self-antigens) (V), diversity (D), joining (J), and constant (C) regions (see Fig. 4.5). In
or immunity (to exogenous pathogens). contrast to the TCRβ and TCRδ loci (both on chromosome 7), the
TCRα and TCRγ loci (both on chromosome 14) do not contain D seg-
T Cells ments. The segments are cut out by nucleases and spliced together by
ligases, ultimately forming the final gene sequence that encodes the
T-cell precursors continuously migrate from the bone marrow to the receptor molecule. The tremendous diversity is explained by the mul-
thymus, where T cells develop54. In the thymus, T cells bearing the tiplicity of all these regions within the genome (e.g. ~70–80 V genes
α/β TCR are subjected to a complex selection process. In contrast to and ~60 J genes for the TCRα alone), but only one of each type is used
antibodies, which represent the antigen receptor on B cells and recog- for a particular TCR.
nize antigens in their naive form, the α/β TCR recognizes only short Each lymphocyte utilizes a different combination of V, D, and J gene
peptide fragments that are generated during antigen processing within segments to form the genetic code of its antigen receptor. Any one of the
APCs (see above). These processed antigens are presented to the TCR genes can join with any other one to form the final VDJ region, thereby
by MHC molecules on the cell surface. The amino acid sequences creating tremendous diversity. In addition, the recombination process
recognized by the TCR include those from both the MHC molecule is subject to inaccuracies during splicing, which cause slight variations
and the antigenic peptide. Consequently, the TCR recognizes a combi- at the VDJ junctions, and additional nucleotides can be inserted by the
nation of “self” MHC molecules (which are highly polymorphic) and enzyme deoxyribonucleotidyl transferase at the spliced regions. These
“foreign” peptides. T cells that recognize “self” MHC molecules but not two events increase the diversity even further. Of note, only a tiny
“self” peptides are useful for immune defense and do not lead to unde- minority of these cells will become functional over a lifetime, with the
sirable autoimmunity. This requirement is achieved by a complex vast majority dying without ever having met their antigen.
process that involves both positive and negative selection within the Defects in the recombination-activating genes RAG1 and RAG262,
thymus. which encode two of the enzymes that mediate the recombination of
variable-region genes in both B cells and T cells, cause a form of severe
T-cell development combined immunodeficiency (see Ch. 60). Affected individuals are
Developing T cells within the cortex of the thymus engage MHC com- unable to produce lymphocytes bearing functional antigen receptors63.
plexes on thymic DCs. When the T cells are able to recognize these
MHC molecules via their TCR, they receive a survival signal (positive T-cell receptor signaling
selection). Otherwise, the T cells undergo apoptotic cell death55. At this TCRs are associated with the CD3 complex, which transmits signals
stage, more than 95% of the developing T cells die in the thymus upon antigen binding. CD3 consists of CD3γ, CD3δ, two molecules of
because they are not selected due to their uselessness (i.e. inability to CD3ε, and a disulfide-linked CD3ζ homodimer64. Upon binding to the
recognize “self” MHC molecules)56. In addition, T cells that express a peptide–MHC complexes, TCRs become cross-linked. Aggregation of
TCR with a very high affinity for the complex of a “self” peptide plus the TCRs causes phosphorylation of tyrosines in the cytoplasmic tails
a “self” MHC molecule are eliminated by apoptosis, since they are of the CD3 complex, which contain immunoreceptor tyrosine-based
potentially harmful. This process, called negative selection, takes place activation motifs (ITAMs). Phosphorylation involves kinases such as
in the thymic medulla and involves DCs and macrophages that process p56lck (which also binds to the cytoplasmic tails of CD4 and CD8),
and present a spectrum of “self” antigens. Positive and negative selec- p59fyn, and ZAP7065 (Fig. 4.8). These events ultimately lead to the
tion allow the survival of just those T cells that recognize foreign (but transcriptional activation of genes that encode cytokines and induce
not “self”) peptides in the context of “self” MHC molecules and thus cellular proliferation and differentiation.
are useful for immune defense without causing auto-attack57.
During thymic education, a variety of surface molecules are switched Costimulatory signals
on and off. Components of the CD3/TCR complex, together with CD4 Signaling through the TCR complex alone does not suffice to activate
or CD8 molecules, have roles in this process. In general, CD4+ T cells T cells. The presence of costimulatory signals is needed for T cells to 89
function as helper T cells and recognize antigens presented by MHC undergo antigen-specific clonal expansion66. Hence, development of a
T-CELL RECEPTOR-MEDIATED SIGNAL TRANSDUCTION COSTIMULATORY MOLECULES AND THEIR LIGANDS*

Co-receptor
SECTION Receptor molecule Expression† molecule Expression†

1 Antigen-presenting cell
CD80 (B7-1) APC CD28, CD152
(CTLA-4)
T
OVERVIEW OF BASIC SCIENCE

ICAM-1 B7-1 (CD80) CD86 (B7-2) APC CD28, CD152 T


LFA-3 (CD54) B7-2 (CD86) (CTLA-4)
CD275 (ICOS-L, APC CD278 (ICOS) T
LFA-1 CD28 B7H2, B7h, B7RP1)
CD2
MHC-II CD274 (PD-L1, APC CD279 (PD-1) T, B
B7H1)
CD4 CD273 (PD-L2, APC CD279 (PD-1) T, B
Ag ζ B7DC)

ζ
2 PKC
Vα PIP DAG CD40 APC, B CD154 (CD40L) T
Cβ fyn
CD3 γ 9 CD134 (OX40) T CD252 (OX40L) APC, B
ε Cα p5
δ Cγ
lck PL
ε 6 CD27 APC, T CD70 (CD27L) T, B
p5 0
β P7
α ZA CDw137 (4-1BB) T, B 4-1BBL APC, B
TCR CD265 (RANK) APC CD254 (RANKL, APC, T
TRANCE)
Gene
transcription CD30 T, B CD153 (CD30L) T, B, APC
CD58 (LFA-3) APC CD2 T
CD4+ T lymphocyte Nucleus
*† Only important costimulatory molecules and those with known ligands are included.
Expression refers only to T cells (T), B cells (B), and antigen-presenting cells (APC) including
Fig. 4.8 T-cell receptor-mediated signal transduction. Activation of the T-cell dendritic cells and Langerhans cells.
receptor (TCR) by presentation of the appropriate antigen by antigen- B7 family TNF receptor family adhesion molecules
presenting cells in association with MHC molecules induces a complex signal
transduction cascade. The T-cell receptor-associated signal transduction is Table 4.3 Costimulatory molecules and their ligands. CTLA-4, cytotoxic
primarily mediated by the CD3 complex and the ζ chain. Upon activation, the T-lymphocyte-associated antigen-4; ICOS, inducible costimulator; LFA-3,
cytoplasmic tails of these molecules become phosphorylated by protein lymphocyte function antigen-3; PD-L1, programmed death ligand 1; TNF, tumor
kinases (p56lck, p59fyn, ZAP70). This causes downstream signaling which necrosis factor.
ultimately leads to transcriptional activation of particular genes. Additional
stimuli are provided by the signaling of costimulatory molecules (CD2, LFA-1,
CD28). DAG, diacylglycerol; PIP2, phosphatidylinositol 4,5-bisphosphate; PKC,
protein kinase C; PLC, phospholipase C. productive T-cell immune response requires exposure of these cells to
at least two types of stimuli. The first signal is the interaction of the
TCR with peptide–MHC complexes presented by APCs, which deter-
mines the specificity of the immune response. The second signal
involves surface molecules and cytokines, which determine the clonal
expansion of specific T cells and their differentiation into effector and
memory cells. Without these costimuli, signaling by the antigen recep-
tors alone will either cause anergy (non-reactivity) or apoptotic cell
death (Fig. 4.9). The major receptors and ligands expressed on APCs
and T cells that play a role in activation are listed in Table 4.3.
ROLE OF COSTIMULATORY MOLECULES DURING T-CELL ACTIVATION The most relevant accessory molecules belong to the B7-CD28 super-
family which includes the following receptor : ligand pairs – CD28/
CTLA-4 : CD80/CD86, ICOS : ICOS-L, and PD-1 : PD-L1/PD-L267. The
APC APC APC
best-investigated costimulators for T cells are B7-1 (CD80) and B7-2
(CD86)68. Both are induced on immature, resting APCs by various TLR
C C C ligands or by cytokines (e.g. TNF-α, IL-1). The CD28 receptor on T
S S S cells recognizes the B7 molecules and delivers activating signals, includ-
M MHC M MHC M MHC
Signal 2

Signal 1

Signal 1

ing the expression of antiapoptotic genes and the production of cyto-


C Ag Ag kines such as IL-2 (see Fig. 4.8). Interaction of ICOS (inducible T-cell
S
TCR M TCR TCR co-stimulator; CD278) with ICOS-L (CD275) induces less IL-2 but
promotes T helper cell differentiation and effector function through the
T cell T cell T cell production of IL-10, -4, -17, -21 and IFN-γ67. The ICOS : ICOS-L signal-
ing pathway is also crucial for T-cell-dependent B-cell responses.
No response
Antigen-specific Anergy/ Cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4 [CD152])
response tolerance also binds to the B7 molecules, with even higher avidity. Unlike CD28
binding, cross-linking of CTLA-4 to B7 down-regulates IL-2 production
Fig. 4.9 Role of costimulatory molecules during T-cell activation. Presentation and cell cycle progression, whereas blockade of CTLA-4 signaling pro-
of the antigen by antigen-presenting cells to the T-cell receptor in association longs T-cell activation. Hence, CTLA-4 is regarded as a T-cell-associ-
with MHC molecules delivers the first signal necessary for T-cell activation. ated co-receptor with negative regulatory effects69. PD-L1 (CD274) and
The second signal is provided by the interaction of costimulatory molecules PD-L2 (CD273) also deliver a co-inhibitory signal to T cells via PD-1
present on antigen-presenting cells and T cells. An antigen-specific response is
(programmed death-1; CD279) and thus inhibit T-cell proliferation and
only induced when signal 1 and signal 2 are provided. Presentation of the
antigen in the absence of signal 2 does not lead to an antigen-specific cytokine production. Through the expression of CTLA-4, PD-Ll and
response but induces anergy and tolerance. APC, antigen-presenting cell; PD-L2, tumor cells may escape an immune response. Thus neutraliza-
90 CSM, costimulatory molecule; MHC, major histocompatibility complex; TCR, tion of these inhibitory molecules by antibodies (e.g. anti-CTLA-4 [ipi-
T-cell receptor. limumab], anti-PD-1 [nivolumab, pembrolizumab]) has proven to be
an effective therapy for a variety of malignancies including metastatic Associated inflammation causes nonspecific recruitment of T cells
melanoma (see Ch. 128). Many of the associated side effects such as from the bloodstream via endothelial cell activation; the few antigen-
colitis, hepatitis and hypophysitis are due to autoimmune phenom- specific T cells recruited into the skin in this manner also contribute
ena. For two other inhibitory members of this superfamily, B7-H3 and to the clearance of the pathogen. In addition, DCs carry the antigen to
CHAPTER
B7-H4, the human receptors have not yet been identified. the skin-draining lymph nodes and present it to TCM. This gives rise
Cytokines, especially inflammatory mediators like IL-1, IL-6 and
TNF-α, also provide costimulatory signals by themselves and, in addi-
to new populations of skin-homing TEM which migrate to the skin
and also participate in the clearance of the antigenic attack.
4
tion, upregulate costimulatory molecules. Hence, a T cell is much more

Immunology
likely to be activated if it meets its specific antigen via an APC that has Effector functions of T cells
been exposed to an inflammatory environment. Two major types of effector T cells have been identified: T helper (Th)
cells, which are CD4+, and cytotoxic T (Tc) cells, which are CD8+.
Clonal expansion CD4+ Th cells recognize foreign antigen peptides bound to MHC class
If a T cell recognizes its specific antigen peptide in association with the II and activate/modulate important components of the cell-mediated
appropriate MHC molecules and becomes activated by costimulatory immune response, mainly through cytokine production. One essential
signals, division and clonal expansion take place. Memory T cells have function of CD4+ Th cells is the activation of B cells. CD8+ Tc cells are
a much longer lifespan than effector cells and are characterized by the crucial to antiviral and antitumor responses73.
expression of the surface molecule CD45RO, while naive T cells express
CD45RA. Two functionally distinct subsets of human memory T cells T helper cells
have been described. CCR7+ memory T cells (central memory T cells CD4+ Th cells recognize “their” antigenic peptides in association with
[TCM]) express lymph node homing receptors and thus stay in the MHC class II molecules. Based primarily on differences in their cytokine
lymph nodes. These cells lack immediate effector function; however, secretion patterns, several types of Th cells have been described74. Naive
they efficiently stimulate DCs to produce IL-12 and can differentiate precursor Th cells only produce IL-2. Subsequently, preactivated precur-
into CCR7− effector cells upon secondary stimulation. In contrast, sor cells, called Th0, can release a wide variety of cytokines (e.g. IL-2,
CCR7− memory T cells (effector memory T cells [TEM]) express recep- IFN-γ, TNF-β, IL-3, IL-4, IL-5, IL-6, IL-9, IL-10, IL-13, GM-CSF, TNF-
tors for migration to inflamed tissues and have immediate effector α). As they develop into Th1, Th2 or Th17 cells, the cytokine secretion
functions70. pattern becomes more restricted and specific (see below), leading to
Memory T cells react in a more rapid fashion upon re-exposure to the generation of different responses. More recently described Th9 and
their specific antigen. The vast majority of these progenitor cells rep- Th22 cells may represent separate types. Morphologically, the cells in
resent effector cells with upregulated receptors that enable them to the various Th subsets are almost indistinguishable, although surface
leave the lymphoid tissue. Organ-specific adhesion molecules guide differences (especially in chemokine receptors) have been described75.
these cells to various locations, including the skin (see below)19,70. Th1 cells: Typical Th1 cytokines are IFN-γ, TNF-β, and IL-2. Produc-
Most of the T cells found in human skin reside in the dermis, with tion of IL-2 by Th1 cells induces proliferation of CD4+ cells in an
a minority in the epidermis (2% to 3% of all CD3+ cells present in autocrine manner and affects the cytokine differentiation pattern;
normal human skin). The majority of human epidermal T cells express however, it also stimulates CD8+ T-cell division and cytotoxicity,
the α/β TCR and either CD8 or CD4. In addition, skin-homing T cells thereby providing “help” for CD8+ Tc cells.
express CD2, CD5, and the skin-homing receptor CLA and they are of The major Th1-derived cytokine is IFN-γ, which activates macro-
the memory phenotype CD45RO+/CD45RA−71. It is estimated that phages to kill intracellular pathogens (e.g. mycobacteria, fungi, pro-
human skin contains twice as many T cells as the blood (~20 billion tozoa) and stimulates NK-cell cytotoxicity. Therefore, Th1 cytokines
T cells). These T cells are referred to as resident memory T cells (TRM). preferentially induce a cell-mediated inflammatory response, such as
They play a major role in host defense against previously encountered the granulomatous lesions of tuberculosis or leprosy. Macrophages
antigens and are more effective than circulating T cells in this respect72. stimulated by IFN-γ release huge amounts of IL-12, the major cyto-
Upon antigen encounter, TRM proliferate and clear the pathogen. kine that drives Th0 cells towards a Th1 phenotype (Fig. 4.10)76.

Fig. 4.10 Development of murine


DEVELOPMENT OF MURINE TH1, TH2 AND TH17 CELLS Th1, Th2 and Th17 cells. Th1, Th2
and Th17 cells differentiate under
the influence of cytokines, which
are secreted by various bystander
Mph cells and can act in a stimulatory
(green arrow) and inhibitory (red
IL-12 line) fashion. The interaction of
IL-12 T-bet IFN-! dendritic cells (DC) with naive CD4+
STAT4 Th1 IL-2 T cells in the various cytokine
NK milieus induces certain
IFN-γ transcription factors in the T cells
IL-12R
IFN-γ (T-bet, STAT4, ROR-γt, STAT3,
IL-23,
GATA3, STAT6), which direct
IL-21,
differentiation. Th1 cells are crucial
IL-23 TGF-β IL-6 IL-6
for T-cell-mediated immunity, Th2
Naive ROR-γt cells support the development
Th17 IL-17, IL-21, IL-22, IL-25
CD4+ STAT3 of humoral immunity, and Th17
TGF-β cells are involved in chronic
DC
IL-23R inflammatory and autoimmune
reactions. Eo, eosinophil; IL,
IL-4 IL-5 IgE
Eo interleukin; IFN-γ, interferon-γ; MC,
IL-4
IL-4 mast cell; Mph, macrophage; NK,
natural killer cell; PC, plasma cell;
Mast cell GATA3
Th2 IL-4, B PC ROR-γt, retinoid acid receptor-
STAT6
T cell IL-21, 22 related orphan receptor-γt;
STAT3/4/6, signal transducer and
IL-4R activator of transcription 3/4/6;
IL-10
Mph TGF-β, transforming growth
factor-β. Adapted from Miossec P, et al.
Interleukin-17 and type 17 helper T cells. N Engl 91
J Med. 2009;361:888–98.
The second most important cytokine in promoting Th1 differentiation plus IL-21 appear to be involved in the differentiation of Th17 cells
is IFN-α. by inducing the expression of ROR-c, the human analogue of murine
A Th1 response is crucial for the host to control the replication of ROR-γt.
intracellular pathogens. In addition, Th1 cells promote the isotype Th17 cells are rapidly induced in response to infectious agents – in
SECTION
switch of B cells to produce complement-binding antibodies. Augment- particular, bacteria and fungi. Patients with a defective Th17 response
1 ing the antitumor response of Th1 cells represents a potential target
of cancer immunotherapy. On the other hand, Th1 cells contribute
due to mutations in the gene encoding signal transducer and activator
of transcription 3 (STAT3), another transcription factor involved in
to the pathogenesis of autoimmune diseases such as rheumatoid Th17 cell development (see Fig. 4.10), have hyperimmunoglobulin E
OVERVIEW OF BASIC SCIENCE

arthritis, multiple sclerosis, allergic contact dermatitis and, perhaps, (hyper-IgE) syndrome and suffer from recurrent Candida albicans and
psoriasis. Staphylococcus aureus infections of the skin and lung (see Ch. 60)83.
Th2 cells: Th2 cells produce primarily IL-4, IL-5, IL-6 and IL-10. Th17 responses have important roles in chronic inflammation and in
These cytokines favor production of non-complement-binding antibod- mediating autoimmune reactions. There is evidence that Th17 cells
ies. IL-4 switches B cells to produce IgE, and IL-5 promotes the growth are involved in rheumatoid arthritis, psoriasis, multiple sclerosis, and
of eosinophils. Therefore, Th2 responses are often associated with inflammatory bowel diseases82. Genetic studies have linked variants of
allergic diseases. Comparable to the role of IL-12 in inducing Th1 the IL-23 and IL-23 receptor genes to susceptibility for psoriasis and
responses, IL-4 is the key cytokine in driving Th2 responses. In addi- psoriatic arthritis84. Since IL-23 enhances IL-17 production, inhibition
tion, IL-4 suppresses Th1 and Th17 differentiation (see Fig. 4.10). of IL-23 is a target for therapeutic intervention. Indeed, a monoclonal
However, IL-4 seems to exert opposing effects on CD4+ T cells and antibody (ustekinumab) directed against the common p40 chain,
DCs, since it can stimulate DCs to produce IL-12 and thus paradoxi- thereby blocking signaling by both IL-12 and IL-23, represents an effec-
cally promote Th1 development77. IL-10, another characteristic Th2 tive treatment for psoriasis85 as do guselkumab and tildrakizumab
cytokine, also inhibits the development of a Th1 response. which target the p19 chain of IL-23; the same applies for agents block-
Immune responses generally tend to drift into a Th1, Th2 or Th17 ing either IL-17A (ixekizumab, secukinumab) or the IL-17 receptor
(see below) pattern. The Th1/Th2 decision is crucial for effective (brodalumab) (see Fig. 128.9).
immunity and thus is regulated by a variety of factors78. The cytokine Th22 cells: A subset of human CD4+ Th cells characterized by the
profile and balance induced by the antigen is critical. For example, IL-12 secretion of IL-22 and TNF-α (in the absence of IFN-γ, IL-4 or IL-17)
is a potent priming stimulus for CD4+ T cells to produce IFN-γ and, have been referred to as Th22 cells86. IL-22, a member of the IL-10
thus, for Th1 cellular differentiation79. Many TLR signals and infec- cytokine family, binds to a heterodimeric receptor which is expressed
tions are strong inducers of IL-12 and therefore favor a Th1 response. by non-hematopoietic cells including keratinocytes. IL-22 induces the
Other TLR signals to DCs and early production of IL-4 favor the gen- expression of antimicrobial peptides (e.g. S100 proteins, defensins) as
eration of Th2 cells. Additional factors that influence the Th1/Th2 well as inflammatory chemokines and cytokines including IL-6. IL-22
decision include the dose of the antigen, the APCs involved36, the is also involved in tissue repair by inducing epithelial cell proliferation
cytokines secreted (by APCs and bystander cells), the genetic back- and enhancing cell survival. IL-22 and Th22 cells appear to play an
ground of the host, and the presence of costimulatory molecules. Of important role in psoriasis since IL-22 is highly expressed in psoriatic
note, many immune responses are not strongly polarized in the Th1 plaques and neutralization of IL-22 prevented the development of
or Th2 direction. lesions in a murine model of psoriasis81.
Th1 and Th2 responses differ greatly in their protective roles, and
the same applies with regard to their contribution to immunopathology. Cytotoxic T cells
Acute allograft rejection, allergic contact dermatitis, and multiple scle- Cytotoxic T (Tc) cells are CD8+, recognize “their” antigenic peptides in
rosis are characterized by dominant Th1 responses, and Th1 clones association with MHC class I molecules, and can directly lyse their
have been isolated from patients suffering from these diseases. On the targets (e.g. virally infected or tumor cells). For example, virally infected
other hand, T-cell clones isolated from patients suffering from atopic cells present peptides derived from intracellular viral proteins on MHC
dermatitis or systemic lupus erythematosus typically exhibit a Th2 class I molecules, and Tc cells that recognize this viral peptide–MHC
profile. complex are stimulated to kill the infected cell. Tc cells have at least
Th3 cells: Th3 cells are a poorly characterized type of CD4+ Th cell three different pathways of killing87, two of which involve direct contact
that primarily secretes TGF-β, provides help for IgA production, and with the target cell. In one method, Tc cells insert perforins into the
has suppressive properties against both Th1 and Th2 cells80. membrane of the target cell; this creates pores in the membrane,
Th9 cells: In response to TGF-β and IL-4, both human and murine through which granzymes are released from the Tc cell into the target
CD4+ T cells secrete IL-981. Th9 cells, the development of which is cell. Granzymes then activate caspases, which are proteolytic enzymes
critically dependent upon the IL-4-activated transcription factor STAT6, that induce apoptosis in the target cell. In a second mechanism, Tc cells
induce inflammation in a T-cell transfer colitis model and in experi- activate the death receptor Fas (CD95) on the target cell by expressing
mental autoimmune encephalomyelitis. They also contribute to aller- the cognate death ligand FasL (CD95L). The activated Fas also triggers
gic diseases, since IL-9 is highly expressed in the lungs of asthmatic apoptosis in the target cell. The third pathway is mediated by cytokines,
patients and there is greater expression by T cells from atopic children. including TNF-α and IFN-γ, which are released as long as TCR stimula-
It is also thought that Th9 cells play a role in autoimmunity. tion continues. These mediators can affect distant cells as well as the
Th17 cells: CD4+ T cells that produce IL-17, but neither IFN-γ target cell87.
nor IL-4 (and therefore do not fit into the Th1–Th2 spectrum), are Naive CD8+ T cells (Tc0 cells) can develop into Tc1 and Tc2 cells88.
referred to as Th17 cells82. Besides IL-17A, these cells also secrete Comparable to Th1 and Th2 cells, Tc1 and Tc2 cells are differentiated
IL-17F, IL-21 and IL-22. Whereas the heterodimeric cytokine IL-12 based on their different cytokine secretion patterns. However, their
(consisting of a p35 chain and a p40 chain) is crucially involved in the functional roles still remain to be determined. Even though there is
development of Th1 cells, the IL-12-related cytokine IL-23 (consisting evidence that some CD8+ T cells exhibit suppressor functions89, most
of a p19 chain and the same p40 chain) is essential for the genera- CD8+ T cells develop a cytotoxic phenotype.
tion of Th17 cells. Naive T cells do not express the IL-23 receptor.
In mice, a combination of TGF-β and IL-6 induces expression of the Regulatory T cells
transcription factor retinoid acid receptor-related orphan receptor-γt More than three decades ago, it was postulated that a subpopulation of
(ROR-γt), which stimulates transcription of the IL-17 gene and expres- T cells could suppress immune responses. Consequently, these cells
sion of the IL-23 receptor (see Fig. 4.10). Of note, in the absence of were designated as T suppressor cells. The most convincing evidence
IL-6, TGF-β drives naive T cells into the regulatory phenotype by for their existence was provided by adoptive transfer experiments in
inducing the transcription factor forkhead box P3 (FoxP3; see below). which suppression could be transferred in an antigen-specific manner.
IL-6 and IL-23 enhance the expression of IL-17 and IL-22 and sup- However, the phenotype of these hypothetical cells and the molecular
press production of IL-10 and IFN-γ, thereby stabilizing the Th17 mechanisms underlying suppressive phenomena were poorly character-
phenotype. IL-21 is also produced in large amounts by mature Th17 ized. Consequently, the term “T suppressor cells” was almost banned
92 cells and amplifies Th17-cell differentiation82. In human T cells, com- and the entire concept of suppression drawn into question. Passive
binations of TGF-β plus IL-21, TGF-β plus IL-6 and IL-23, or IL-6 mechanisms such as clonal deletion, clonal anergy, and immunologic
ignorance were evoked to explain immunologic unresponsiveness and
peripheral tolerance90. DEVELOPMENT AND FUNCTION OF REGULATORY T CELLS
Nevertheless, the concept of T suppressor cells was persistently
pursued in the field of photoimmunology. Application of contact aller-
CD4+ CHAPTER
gens onto the skin of mice exposed to UV radiation not only resulted Development FoxP3+
in a failure of sensitization but also induced antigen-specific tolerance,
since at a later time these animals could not be resensitized to the same
CD8–
4
allergen42 (see Ch. 86). Adoptive transfer of splenocytes from animals Thymus

Immunology
tolerized in such a manner rendered naive recipients unresponsive to
the specific allergen. Depending on the model used (local or systemic Periphery
immunosuppression, type of allergen, UV dose), different subtypes of naive
TGF-β CD4+
T cells appeared to be responsible for the transfer of suppression91. Due IL-10
to the lack of specific surface markers, to date these cells have not been IL-10
isolated or cloned; nevertheless, the adoptive transfer experiments
clearly indicated that active suppression exists. CD4+ CD4+
Interest in the entire field was rekindled by the observation that iTreg CD25 +
Tr1
CD25 + nTreg
chronic activation of both human and murine CD4+ T cells in the FoxP3+ Immature FoxP3+
presence of IL-10 induced CD4+ T-cell clones with a low proliferative
capacity that produced high levels of IL-10, low levels of IL-2, and no
IL-492. These antigen-specific T-cell clones suppressed the proliferation IL-10
of CD4+ T cells in response to antigen and prevented T-cell-mediated
colitis in SCID mice. This CD4+ T-cell subset was designated T regula-
tory cells 1 (Tr1).
Another subset of CD4+ regulatory T cells (Tregs) is characterized by Mode of action
IL-2
the constitutive expression of the α-chain of the IL-2 receptor (CD25).
Some CD4+CD25+ Tregs are generated in the thymus and become Induction of 1
activated upon re-encountering their antigen in the periphery. Thymec- apoptosis
CD4+ CD25 + IL-2 deprivation
tomized mice that do not develop CD4+CD25+ T cells develop auto- FoxP3+
immune diseases93. In turn, reinjection of such cells prevents the
development of the autoimmune phenomena, indicating that the Perforin Treg cAMP 2
Granzymes
CD4+CD25+ cells prevent autoimmunity and thus can act in a sup-
pressive fashion. FoxP3, a transcription factor whose inherited dysfunc- CD39
Adenosine
tion leads to an autoimmune and inflammatory syndrome in humans IL-10 CD73
(IPEX: immune dysregulation, polyendocrinopathy, enteropathy, IL-35 3
X-linked) and mice (Scurfy strain), is specifically expressed in naturally TGF-β ATP Metabolic
LAG-3 CTLA-4
arising CD4+CD25+ Tregs94. Retroviral gene transfer of Foxp3 converts alterations
naive T cells toward a Treg phenotype. Thus, Foxp3 is a key regulatory MHCII CD80/86 (1−3)
gene in the development of Tregs.
Natural Tregs (nTregs) are produced in the thymus as a functionally Production of
mature subpopulation of T cells95. These CD4+CD25+ T cells consti- suppressive
cytokines IDO
tute ~10% of all murine peripheral CD4+ T cells. However induced
Tregs (iTregs) can also develop from naive T cells in the periphery
through several mechanisms (Fig. 4.11). Immature DCs appear to be Modulation
involved in the generation of human iTregs. Upon repeated stimulation of DCs
by immature DCs, allogeneic CD4+ T cells have been shown to: express
the negative regulatory molecule CTLA-4; lose their capacity to produce Fig. 4.11 Development and function of regulatory T cells. Natural regulatory
IFN-γ, IL-2 or IL-4; and differentiate into non-proliferating, IL-10- T cells (nTregs), which express CD4, CD25 and FoxP3, develop from CD4+CD8− T
producing T cells41. In co-culture experiments, these T cells inhibited cells in the thymus. In the periphery, naive conventional CD4+ T cells (in the
the antigen-driven proliferation of Th1 cells in a cell contact-dependent presence of interleukin [IL]-10 and/or transforming growth factor-β [TGF-β])
manner. The same may apply for damaged APCs, since the migration can develop into FoxP3− regulatory type 1 cells (Tr1) or induced regulatory T
of UV-damaged LCs is required for the generation of Tregs by UV cells (iTreg) that express CD24, CD25, and FoxP3. Tregs exert their inhibitory
radiation96. functions in several ways. They release inhibitory cytokines (e.g. IL-10, IL-35,
Together, these findings show that immature and mature DCs induce TGF-β), induce apoptosis of their target effector T cells via release of granzymes
different types of T-cell responses: (1) mature, IL-12-producing DCs and perforin, and interfere with metabolic functions of their target T cells. The
latter can be accomplished by: (1) IL-2 deprivation, which triggers apoptosis; (2)
result in inflammatory Th1 cells; (2) less well-defined “DC2s” result
transferring cAMP, which inhibits proliferation and IL-2 production, into target
in Th2 cells; and (3) immature or damaged DCs result in IL-10-pro- cells; and (3) increasing pericellular levels of cytotoxic adenosine via conversion
ducing iTregs. These observations provide evidence for constant control of ATP to adenosine by CD39 and CD73, two ectonucleotidases expressed on
of peripheral tolerance to self-antigens97. In this scenario, immature the surface of Tregs. Tregs also modulate dendritic cell (DC) maturation and
DCs constantly take up proteins from normal cells as they undergo function via interaction of cytotoxic T-lymphocyte-associated antigen-4
regular cell turnover. In the absence of inflammatory signals, DCs do (CTLA-4) and lymphocyte-activation gene-3 (LAG-3) on Tregs with their ligands
not mature and therefore enter the regional lymph nodes in an imma- CD80/CD86 and MHC class II, respectively, on DCs and induction of the
ture stage. Self-antigens expressed on immature DCs lead to the devel- tryptophan-degrading enzyme indoleamine 2,3-dioxygenase [IDO] in DCs.
opment of Tregs, which then migrate to the respective organ where they Adapted from Workman CJ, Szymczak-Workman AL, Collison LW, et al. The development and function of
regulatory T cells. Cell Mol Life Sci. 2009;66:2603–22 and from Sakaguchi S, et al. Regulatory T cells and
exhibit suppressive effects. During an infection, immature DCs take
immune tolerance. Cell. 2008;133:775–87.
up self-antigens but mature in the presence of TLR and inflammatory
signals. In the regional lymph nodes, the mature DCs prime naive
CD4+ or CD8+ T cells, which become effector T cells. These cells
re-migrate to the initial site of inflammation, where they encounter the can induce apoptosis of target effector T cells via perforin and gran-
inhibitory effects of Tregs that are already present. This process nor- zymes. Tregs also interfere with metabolic functions in their target T
mally prevents the development of autoimmunity under both normal cells by depriving them of IL-2 (which results in apoptosis), increasing
and inflammatory conditions. their intracellular levels of cAMP (which inhibits T-cell proliferation
Tregs exert their suppressive functions in several ways95 (see Fig. and IL-2 production), and increasing pericellular levels of adenosine 93
4.11). They release inhibitory cytokines (e.g. IL-10, IL-35, TGF-β) and (which is cytotoxic). The latter is achieved by the conversion of ATP to
adenosine via CD39 and CD73, two ectonucleotidases expressed on the surface molecule L-selectin (CD62L) allows naive T cells to attach
the Treg membrane. Furthermore, negative regulation of the matura- to and roll on the inner surface of the high endothelial venules, special-
tion and function of APCs occurs through the interaction of CTLA-4 ized postcapillary venules within the lymph nodes. The venules express
and lymphocyte-activation gene-3 (LAG-3) on Tregs with CD80/CD86 a chemokine, called the secondary lymphoid-tissue chemokine (SLC/
SECTION
and MHC class II ligands, respectively, on APCs. Induction of the CCL21)104, which activates T cells via interaction with the chemokine
1 tryptophan-degrading enzyme indoleamine 2,3-dioxygenase (IDO) in
DCs has been suggested as another mechanism by which Tregs exert
receptor CCR7. Subsequently, T cells adhere tightly to the venules via
interaction of endothelial intercellular adhesion molecule-1 (ICAM-1,
suppression. CD54) with lymphocyte function antigen-1 (LFA-1, CD11a). Finally, T
OVERVIEW OF BASIC SCIENCE

cells extravasate through the high endothelial venules into the lymph
Natural killer T cells node and accumulate in the T-cell-rich areas. They can return into the
NK T cells are a distinct subset of thymocytes and peripheral T cells bloodstream by leaving the lymph node via the efferent lymphatics.
that have characteristics of both T cells and NK cells98. They express Since naive T cells lack the necessary expression of a specific combina-
the NK marker NK1.1 and an invariant TCR restricted to CD1d. NK tion of adhesion molecules and chemokine receptors, they cannot enter
T cells produce large amounts of various cytokines, including IFN-γ, extranodal tissues from the blood, forcing them back into the lymph
IL-4, and IL-10. Since they are present at the early stages of ontogeny nodes.
in the thymus, they are thought to be involved in the differentiation of Upon activation by APCs presenting specific antigenic peptides, T
various T-cell subsets, especially Th2 cells. They may also play a role cells start to proliferate and begin to express activation molecules,
in the control of autoimmunity, since NK T cells are numerically and thereby developing into memory T cells. During this process, they
functionally deficient in mice with autoimmune diseases. NK T cells express new surface molecules that allow them to exit the blood vessels
also seem to contribute to UV-induced immunosuppression, and they and enter extranodal tissues105. A hallmark of memory T cells is their
down-regulate immune responses to UV-induced tumors99. ability to “remember” the anatomic location that the lymph node was
draining. Due to the expression of specific adhesion molecules, they
γ/δ T cells exhibit the capacity to migrate primarily into the sites where the
A small proportion of T cells carry a TCR composed of γ and δ (rather antigen was first encountered by the host. Hence, lymph nodes draining
than α and β) protein chains. Under normal conditions, the great different organs generate distinct, “tissue-specific” memory T cells.
majority of T cells detected in the skin belong to the α/β type. However, Memory T cells entering the skin express cutaneous lymphocyte
in certain infectious diseases such as leprosy and leishmaniasis, γ/δ T antigen (CLA), whereas memory T cells found in other tissues are
cells can account for almost one-third of the T cells infiltrating the primarily CLA-negative. CLA is generated by modification of the pre-
skin. The exact functional role of γ/δ T cells remains to be determined, existing P-selectin glycoprotein ligand-1 (PSGL-1, CD162) by glycosyl-
but they may be involved in the recognition of non-peptide moieties ation enzymes106. The function of CLA (recognized by the antibody
expressed by microbial pathogens100. HECA-452) is to allow the early tethering of T cells to the endothelium
of cutaneous postcapillary venules. This step leads to the subsequent
Dendritic epidermal T cells slowing, arrest, and then extravasation of the T cells. The endothelial
CD3+ lymphocytes found in murine epidermis are dendritic in shape ligand for CLA is E-selectin (CD62E), which is constitutively expressed
and are therefore designated as dendritic epidermal T cells (DETCs)101. at low levels on cutaneous microvessels. Its expression is strongly
These murine DETCs express a γ/δ type of TCR with limited diver- upregulated by inflammatory stimuli. Although interaction between
sity. The same TCR configuration is detected in early thymocytes, CLA and E-selectin is an important initial step, it does not suffice for
giving rise to speculation that fetal thymocytes are the progenitors of transmigration. Interactions of LFA-1 and the β-integrin very late
DETCs. Alternatively, maturation could take place in the skin. Despite antigen-4 (VLA-4) on T cells with ICAM-1 (CD54) and VCAM-1
extensive studies, the functional role of DETCs could not be clarified (CD106), respectively, on endothelial cells produces the firm adhesion
completely. It was proposed that DETCs protect stressed keratinocytes that is required for extravasation of T cells from the blood into the skin
and exhibit NK function, but also down-regulate T-cell responses. (see Ch. 102).
To date, a human equivalent of the rodent DETC has not been
identified.
B Cells
Innate lymphoid cells The major task of B cells is the production of immunoglobulins (anti-
Innate lymphoid cells (ILC) are lymphoid progenitor-derived leukocytes bodies) that bind to specific antigens. Antibodies also interact with
which lack T-cell and B-cell receptors and do not express conventional components of the innate immune system by preventing foreign organ-
hematopoietic lineage markers102. ILC require the transcription factor isms from adhering to mucosal surfaces, activating complement, and
Id2 and the cytokine IL-7 for their development. Three subgroups have opsonizing bacteria for phagocytosis. Immunoglobulins are secreted
been defined based upon their cytokine and transcription factor expres- products of mature B cells. However, during early B-cell development,
sion profiles. ILC1 rely on the T-bet transcription factor and secrete immunoglobulins function as antigen-specific B-cell receptors on the
type 1 cytokines such as IFN-γ and TNF-α in response to intracellular cell membrane, equivalent to the TCR on T cells. CD19, CD20, and
pathogens. ILC2 are under the control of GATA3 and ROR-α transcrip- CD22 are the main markers currently used to detect B cells. Activation
tion factors and produce type 2 cytokines (e.g. IL-5, IL-9, IL-13) in of B cells results in cell division and maturation into plasma cells,
response to extracellular parasite infections. ILC3 are under the control which secrete the specific antibodies.
of the ROR-γ transcription factor and produce IFN-γ, IL-17, and IL-22.
The exact role of ILC in physiologic and pathologic processes still Immunoglobulins
remains to be determined which is rather tricky since they represent
Immunoglobulins consist of two identical heavy chains and two identi-
just a tiny minority of hematopoietic cells and there are currently no
cal light chains, linked together by disulfide bonds107 (Fig. 4.12). The
specific markers which would allow exact identification.
N-terminus of each chain contains a variable domain that binds the
ILC3 may play a role in triggering psoriatic flares since an increase
antigen through three hypervariable complementarity-determining
in NKp44+ILC3 that release IL-17 and IL-22 have been found in the
regions (CDRs). The C-terminal domains of the heavy and light chains
circulation and skin of psoriatic patients. Their potential pathogenic
form the constant regions. Five classes of immunoglobulin (IgG, IgA,
role is also supported by a significant reduction in NKp44+ILC3 after
IgM, IgD and IgE) are defined by the amino acid sequences of the con-
successful treatment with adalimumab.
stant regions of the heavy chains, and the designation of κ or λ depends
on the constant region of the light chain. The class/subclass and titer
Lymphocyte recruitment of the antibody that is formed depend on: the physical and chemical
A major challenge for lymphocytes is to be at the right place at the properties of the antigen; the site, duration, repetition, and amount of
right time to meet “their” antigen. Naive lymphocytes encounter their antigen exposure; and the individual’s propensity to recognize the
antigens for the first time in lymphoid organs, while memory cells have antigen as foreign.
94 their encounters in the periphery. There is continuous recirculation of Digestion of immunoglobulins with the enzyme papain yields an
naive T cells between the blood and lymphoid organs103. Expression of antigen-binding fragment termed Fab and a fragment that spontaneously
Immunoglobulin G
STRUCTURE OF PROTOTYPE IMMUNOGLOBULIN IgG is the most abundant immunoglobulin, accounting for ~75% of
the total amount of serum immunoglobulin, and the major immuno-
Antigen-
globulin of the secondary immune response. Four subclasses (IgG1,
CHAPTER
IgG2, IgG3, IgG4) are defined by the amino acid sequences of their
4
binding Variable
site region constant region. They differ in their ability to activate complement.
Heavy N-terminus
While IgG1 and IgG3 are potent activators of the classical complement
chain
pathway, IgG2 is less effective and IgG4 lacks such ability. In addition,

Immunology
Constant
VH
S region
S IgG1 and IgG3 bind more avidly to mononuclear cells, i.e. they are
S S more cytophilic. Most of the autoimmune dermatoses caused by auto-
Fab S S
S CH1 S antibodies are mediated by IgG, most often IgG4.
region VL S
S S
S Immunoglobulin A
S S
S S
CL S S
IgA is the predominant immunoglobulin present in mucosal surfaces,
S S where it protects the host from invasion of microorganisms. IgA can
Light
chain
activate the complement system via the alternative (but not the clas-
Hinge
region
sical) pathway. Two subclasses of IgA exist, IgA1 and IgA2. IgA mol-
ecules can be joined by a J chain; this dimer form is mostly found in
secretions, while in the serum, IgA circulates primarily as a monomer.
Complement
IgA molecules can be involved in the pathogenesis of bullous auto-
immune diseases.
SS

SS

binding CH2 CH2


region
Fc Immunoglobulin E
region IgE is the classic anaphylactic antibody that mediates most immediate
allergic and anaphylactic reactions. Although in normal serum IgE is
present in only very small amounts (10–70 mcg/100 ml), just a few
molecules appear to be required for sensitization. Mast cells and baso-
SS

SS

CH3 CH3
phils express high-affinity receptors for the Fc portion of IgE (FcεRI).
Antigens, anti-IgE antibodies, or other substances that cross-link at
least two IgE molecules bound on mast cells induce the release of
C-terminus mediators such as histamine, serotonin, leukotrienes, and prostaglan-
dins (see Ch. 18). The second IgE receptor, FcεRII (CD23), exhibits
Fig. 4.12 Structure of a prototype immunoglobulin. The basic weaker binding affinity and is expressed on macrophages, eosinophils,
immunoglobulin structure consists of two identical light polypeptide chains platelets, and particular subtypes of T and B cells. Macrophages stimu-
and two identical heavy polypeptide chains, which are linked together by lated via activation of FcεRII exhibit increased phagocytic and cytotoxic
disulfide bonds. The antigen-binding site is at the N-terminal end. activity; secrete prostaglandins, leukotrienes and lysosomal enzymes;
and generate superoxides.
In the past, it was believed that mast cells and basophils were the
crystallizes with time and thus is called the Fc portion. Immunoglobu- only cells that express the high-affinity FcεRI receptor. However, it
lins are divalent, since twice as many Fab fragments than Fc portions became clear that LCs33, dermal DCs, and peripheral blood DCs as
are obtained upon cleavage with papain. Digestion of immunoglobulins well as monocytes from atopic individuals can bind monomeric IgE via
with pepsin provides Fab fragments that are disulfide bond-linked and the high-affinity FcεRI108. FcεRI, both quantitatively and qualitatively,
thus called F(ab) fragments. is the crucial serum IgE-binding structure on APCs of atopic individuals
Through their CDRs, antibodies recognize the conformational struc- and functions as an allergen-focusing molecule. Allergens are more
ture of their epitopes. No antigen processing is required. As for the efficiently taken up, processed, and presented to T cells following tar-
TCR, extensive gene rearrangement involving V, D, and J regions allows geting to APCs via the FcεRI. Thus, FcεRI-IgE-dependent allergen pre-
the generation of a nearly infinite variety of immunoglobulins specific sentation may lower the threshold to elicit allergen-specific T-cell
for any putative antigen61 (see T-cell receptor diversity above). An responses in atopic individuals104. This may perpetuate allergen-specific
even greater repertoire of B-cell receptors than TCRs is produced, IgE production and even modulate T-cell-mediated delayed-type hyper-
since further gene rearrangement of immunoglobulins occurs during sensitivity reactions in allergen-exposed tissues.
B-cell division after antigen stimulation, a process called somatic
Immunoglobulin D
hypermutation.
The function of IgD still remains mysterious. More recent evidence
The ability to induce effector functions resides in the Fc portion of
suggests that IgD participates in respiratory immune defense109. It
the heavy chain. The Fc portion of IgG contains domains called CH1,
binds to basophils and mast cells, stimulating their production of anti-
CH2 and CH3 for IgG. Complement activation is mediated via the CH2
microbial factors. IgD may also exert proinflammatory functions, as
domain, while binding to macrophages or monocytes is mediated via
illustrated by the hyperimmunoglobulinemia D with periodic fever
the CH3 domain. Binding of the Fc portion of IgG to phagocytic cells
syndrome (HIDS; see Table 45.2).
via their Fcγ receptors promotes phagocytosis of antibody–antigen com-
plexes. Three receptors for human IgG exist. FcγRI (CD64) binds
monomeric IgG with a high degree of affinity. FcγRII (CD32) is broadly
B-cell activation
distributed on cells and binds only complexed IgG with a low degree of B cells can respond to some antigens in a T-cell-independent manner110.
affinity. FcγRIII (CD16) is expressed on macrophages, NK cells and Such antigens have numerous repeating epitopes (mainly polysaccha-
some T cells, and it interacts with monomeric as well as complexed rides), which bind and cross-link multiple B-cell receptors and activate
IgG. IgA binds only to the FcαR (CD89), while IgE can bind to three the B cells directly to secrete IgM. However, this response is limited to
surface molecules: FcεRI, FcεRII (CD23), and the IgE-binding protein. IgM and is of short duration and poor specificity due to the lack of
germinal center formation, affinity maturation, Ig class switching, and
Immunoglobulin M memory.
With a size of approximately 900 Da, IgM is the largest immunoglobu- Most B-cell responses are activated in a T-cell-dependent manner.
lin. IgM molecules are pentamers that (in addition to light and heavy Upon recognition by the specific IgM on the B-cell surface, the antigen
chains) contain one J chain. IgM is the major immunoglobulin pro- is internalized, processed, and re-expressed within the MHC class II
duced in the primary immune response. Upon binding to its antigen, molecule on the surface. As a result, the B cell can present the antigen
IgM induces agglutination and activates the classical complement to a specific T cell. Primed Th cells, in turn, secrete cytokines (IL-2, 95
pathway. IL-4, IL-5, IL-6, IFN-γ) that function as B-cell growth factors, inducing
proliferation, isotype switch, and maturation to plasma cells. Activa-
T-CELL-DEPENDENT B-CELL ACTIVATION tion of CD40 (which is expressed on B cells) by CD40 ligand (CD154)
expressed on T cells is needed for isotype switching from the initial IgM
to an IgG response (Fig. 4.13). Once the switch from IgM to another
SECTION
Ag isotype has occurred, some of the activated cells become long-lived
1 T cell IgM B cell memory cells that can rapidly produce antibodies upon re-challenge with
the same antigen. Mutations in the genes encoding CD40 ligand and
CD40 are associated with forms of primary immunodeficiency known
OVERVIEW OF BASIC SCIENCE

Ag processing
as hyper-IgM syndromes, which are characterized by low or undetect-
TCR MHC II able levels of IgG, IgA, and IgE but elevated levels of IgM (see Ch. 60).
Upon subsequent antigen exposure, B cells become activated by fol-
MHC licular DCs, which are located in the germinal centers of the lymph
CD154/CD40L CD40 class II
nodes. Follicular DCs bear Fc and complement receptors and bind
Isotype immune complexes. In contrast to “conventional” DCs, follicular DCs
CD28 CD80/86 switching do not endocytose and process antigens. Instead, they trap antigen–
antibody complexes and provide intact antigen for interaction with the
B-cell receptor. This interaction stimulates B-cell activation and dif-
ferentiation. In addition, follicular DCs form immune complex-coated
bodies (iccosomes) that are taken up by B cells. Upon processing, B
IgG
cells present this follicular DC-derived antigen to T cells and thus may
obtain T-cell assistance111.

Allergic Contact Hypersensitivity


Allergic contact hypersensitivity (CHS) is highly relevant for dermatolo-
Fig. 4.13 T-cell-dependent B-cell activation. B cells present the antigen in gists, since it is the pathogenic basis for allergic contact dermatitis, a
association with MHC II molecules to CD4+ T cells. This results in the frequent inflammatory dermatosis. In addition, important basic immu-
upregulation of CD154/CD40 ligand which interacts with CD40 expressed on B nologic discoveries have been made utilizing the model of CHS112.
cells. CD40–CD40 ligand interaction causes isotype switching of
immunoglobulins and upregulation of CD80/86 which interacts with CD28
expressed on T cells. This results in further activation of T cells. Ag, antigen;
Induction of CHS
CD40L, CD40 ligand; Ig, immunoglobulin; MHC II, major histocompatibility Most contact allergens are low-molecular-weight chemicals that, after
complex II. penetrating the skin, have to couple with host proteins to be able to
act as full antigens. This process is called haptenization, and the low-
molecular-weight allergens are referred to as haptens. Upon epicutane-
ous application of a hapten in a naive host, APCs in the skin take up
the hapten, migrate to the regional lymph nodes and present it to naive
T cells (Fig. 4.14). Although this was previously thought to be done

Fig. 4.14 Induction of contact hypersensitivity.


INDUCTION OF CONTACT HYPERSENSITIVITY Application of contact allergens (Ag) induces the
release of cytokines by keratinocytes (KCs),
Langerhans cells (LCs), and other cells within the
Ag
skin. These cytokines in turn activate LCs and/or
dermal dendritic cells (dDCs), which uptake the
antigen and emigrate into the regional lymph nodes.
LC
During this process, they develop into mature
antigen-presenting cells. In addition, the antigen is
Ag IL-1β processed, re-expressed on the surface, and, finally,
IL-1, TNF presented to naive T cells in the regional lymph
KC
GM-CSF node. Upon antigen presentation, T cells bearing the
Ag appropriate T-cell receptor clonally expand and
uptake become effector T cells. These alter their migratory
behavior due to the expression of specific surface
molecules like cutaneous lymphocyte antigen (CLA).
Emigration Effector T cells recirculate into the periphery, where
dDC
they may later meet the antigen again.
Ag processing
Maturation

Regional
lymph Cutaneous
T naive
node postcapillary
venule
Afferent lymph vessel

Ag
presentation E-selectin

Clonal
expansion

Effector CLA
T cell

96
Fig. 4.15 Elicitation of contact hypersensitivity.
ELICITATION OF CONTACT HYPERSENSITIVITY Application of contact allergens (Ag) onto a
sensitized individual causes the release of cytokines
by keratinocytes (KCs) and Langerhans cells (LCs).
These cytokines induce the expression of adhesion CHAPTER
Ag
molecules and activation of endothelial cells, which
ultimately attracts leukocytes to the site of antigen
application. Among these cells, T effector cells are
4

Immunology
present, which are now activated upon antigen
LC presentation either by LCs, dermal dendritic cells
(dDCs), or infiltrating macrophages (Mphs). Antigen-
KC Inflammation specific T-cell activation again induces the release of
cytokines by T cells. This causes the attraction of
Ag presentation LC other inflammatory cells, including granulocytes and
dDC macrophages, which ultimately cause the clinical
manifestation of contact dermatitis. CLA, cutaneous
Cytokine lymphocyte antigen.
release Mph
Mediator
release

T cells

Inflammation-
induced
recruitment

CLA+ Upregulation
T effector cell of E-selectin
Neutrophils Mast cells Macrophages
(Mph)

only by LCs, it has been established that other cutaneous APCs (e.g. The earliest histopathologic findings during a CHS response are mast
dermal DCs) can present antigens, and the role of LCs as the primary cell degranulation, vasodilation, and an influx of neutrophils, which is
APC in the skin is currently under debate (see Antigen presentation followed by infiltration of monocytes and T cells (Fig. 4.15). However,
above). the pathophysiologic events that result in allergic contact dermatitis
During the emigration from the skin, APCs convert from a “resting” are clearly T-cell-dependent, since T-cell-deficient mice are unable to
into an “activated” functional state. This process is initiated by keratin- mount a CHS response. Furthermore, low doses of hapten that are
ocytes, which secrete inflammatory cytokines as a result of hapten sufficient to stimulate hapten-specific T cells are insufficient to elicit
application; triggering of TLRs and direct effects of haptens on the a CHS response. This indicates that the elicitation of a CHS response
APCs may also be involved. Furthermore, the inflammasomes within requires, in addition to hapten-specific recognition, a proinflammatory
keratinocytes become activated by contact sensitizers resulting in the stimulus that can be provided in a dose-dependent manner by the
release of biologically active IL-1β and IL-18. It follows that hapten itself114.
inflammasome-deficient mice (ASC- and NALP3-knockout mice)
display an impaired response to contact allergens113. Induction of IL-1β Keratinocytes as immune targets and initiators
appears to be specific for haptens, since it is not observed with irritants In addition to the capacity to secrete a wide variety of proinflammatory
or tolerogens. In addition, other cytokines, including chemokines, and anti-inflammatory cytokines (e.g. IL-1 and IL-10, respectively),
TNF-α and GM-CSF, may also contribute to APC activation and migra- keratinocytes express immunologically relevant surface molecules.
tion. Hence, the hapten itself, through its capacity to provoke a specific Thus, keratinocytes function not only as effectors, but also as targets
cytokine pattern, seems to be the initial trigger that activates APCs and for immune reactions. Like other nucleated cells, keratinocytes express
induces sensitization (see Fig. 4.14). MHC class I molecules and therefore can be attacked by CD8+ Tc
Presentation of the hapten by APCs in the regional lymph nodes cells, particularly after viral infection. MHC class I-restricted T-cell-
causes activation of naive T cells carrying the appropriate TCR and mediated cytolysis appears to be involved in CHS, lichen planus,
results in the generation of effector cells. In contrast to other types of fixed drug eruptions, cutaneous GVHD, and herpes simplex virus
delayed-type hypersensitivity responses, which are mediated by CD4+ infections.
T cells, most haptens induce a T-cell response of predominantly CD8+ Although keratinocytes do not express MHC class II molecules under
effector T cells112. In addition, Treg populations that inhibit the CHS normal conditions, they can be induced to do so in inflammatory
response are induced. The balance between effector T cells and Tregs settings115. The major inducer of MHC class II expression on keratin-
seems to depend on the dose of the antigen applied, since application ocytes is IFN-γ. Accordingly, MHC class II expression by keratinocytes
of extremely low doses of the hapten does not result in sensitization, is frequently observed in association with pronounced lymphocytic
but rather in tolerance89. infiltrates. Several studies have shown that MHC class II-bearing
keratinocytes can induce proliferation of allogeneic CD4+ T-cell lines,
Elicitation of CHS but not of resting T cells. Nevertheless, it is important to note that
T cells that have been primed in the draining cutaneous lymph nodes despite their capacity to express MHC class II molecules, keratinocytes
express the skin-homing marker CLA and thereby exhibit the capacity are not able to induce a primary T-cell response, a major difference
to enter the skin71. These T cells become activated when they encoun- from APCs.
ter their relevant hapten presented by APCs within the skin. However, On the other hand, upon overexpression of immunologically relevant
in contrast to the sensitization phase, antigen presentation can now be molecules, keratinocytes may play a pathogenic role. Transgenic mice
effectively performed by other cells (including keratinocytes, dermal that overexpress IFN-γ (a cytokine not derived from keratinocytes)
mast cells, and macrophages) that are capable of presenting antigen at within the basal layer of the epidermis develop an autoimmune phe-
least in a MHC class I-restricted fashion112. Alternatively, inflamma- notype116. Autoimmunity is also induced when LCs are constantly
tory cells that infiltrate the site of hapten application very early during activated by keratinocytes through CD40/CD40L interactions. Trans- 97
the response may function as APCs. genic mice overexpressing CD40L in the basal epidermis demonstrate
a dramatic decrease in the number of LCs within the epidermis that only serve as targets of the immune system, but under certain con-
may be due to emigration of CD40-activated LCs117. These mice ditions also directly modulate the initiation and course of immune
suffer from chronic inflammation of the skin and, over time, develop responses.
systemic lupus-like features (e.g. lymphadenopathy, antinuclear anti-
SECTION
bodies, proteinuria). These models indicate that keratinocytes not For additional online figure visit www.expertconsult.com
1
OVERVIEW OF BASIC SCIENCE

REFERENCES
1. Streilein JW. Skin-associated lymphoid tissues (SALT): calcitonin gene-related peptide. Nature 48. Price AA, Cumberbatch M, Kimber I, Ager A. Alpha 6
origins and functions. J Invest Dermatol 1993;363:159–63. integrins are required for Langerhans cell migration
1983;80(Suppl.):12s–6s. 29. Valladeau J, Ravel O, Dezutter-Dambuyant C, et al. from the epidermis. J Exp Med 1997;186:1725–35.
2. Parkin J, Cohen B. An overview of the immune system. Langerin, a novel C-type lectin specific to Langerhans 49. Margulies DH. Interactions of TCRs with MHC-peptide
Lancet 2001;357:1777–89. cells, is an endocytic receptor that induces the complexes: a quantitative basis for mechanistic
3. Medzhitov R, Janeway C Jr. Innate immunity. N Engl J formation of Birbeck granules. Immunity models. Curr Opin Immunol 1997;9:390–5.
Med 2000;343:338–44. 2000;12:71–81. 50. Pamer E, Cresswell P. Mechanisms of MHC class
4. Walport MJ. Complement. First of two parts. N Engl J 30. Merad M, Ginhoux F, Collin M. Origin, homeostasis and I-restricted antigen processing. Annu Rev Immunol
Med 2001;344:1058–66. function of Langerhans cells and other langerin- 1998;16:323–58.
5. Akira S, Takeda K. Toll-like receptor signalling. Nat Rev expressing dendritic cells. Nat Rev Immunol 51. Watts C. Capture and processing of exogenous
Immunol 2004;4:499–511. 2008;8:935–47. antigens for presentation on MHC molecules. Annu
6. Akira S, Takeda K, Kaisho T. Toll-like receptors: critical 31. Kubo A, Nagao K, Yokouchi M, et al. External antigen Rev Immunol 1997;15:821–50.
proteins linking innate and acquired immunity. Nat uptake by Langerhans cells with reorganization of 52. Watts C. Antigen processing in the endocytic
Immunol 2001;2:675–80. epidermal tight junction barriers. J Exp Med compartment. Curr Opin Immunol 2001;13:26–31.
7. McInturff JE, Modlin RL, Kim J. The role of toll-like 2009;206:2937–46. 53. Guermonprez P, Amigorena S. Pathways for antigen
receptors in the pathogenesis and treatment of 32. Stingl G, Maurer D, Hauser C, Wolff K. The epidermis: cross presentation. Springer Semin Immunopathol
dermatological disease. J Invest Dermatol an immunologic microenvironment. In: Freedberg IM, 2005;26:257–71.
2005;125:1–8. Eisen AZ, Wolff K, et al., editors. Fitzpatrick’s 54. Kruisbeek AM. Regulation of T cell development by
8. Contassot E, Beer HD, French LE. Interleukin-1, dermatology in general medicine, vol. 1. New York: the thymic microenvironment. Semin Immunol
inflammasomes, autoinflammation and the skin. Swiss McGraw-Hill; 1999. p. 343–70. 1999;11:1–2.
Med Wkly 2012;142:w13590. 33. Wang B, Rieger A, Kilgus O, et al. Epidermal 55. Fink PJ, Bevan MJ. Positive selection of thymocytes.
9. Harder J, Schröder JM. Psoriatic scales: a promising Langerhans cells from normal human skin bind Adv Immunol 1995;59:99–133.
source for the isolation of human skin-derived monomeric IgE via Fc epsilon RI. J Exp Med 56. Anderson G, Moore NC, Owen JJT, Jenkinson EJ.
antimicrobial proteins. J Leukoc Biol 2005;77:476–86. 1992;175:1353–65. Cellular interactions in thymocyte development. Annu
10. Gläser R, Harder J, Lange H, et al. Antimicrobial 34. Nagao K, Kobayashi T, Moro K, et al. Stress-induced Rev Immunol 1996;14:73–99.
psoriasin (S100A7) protects human skin from production of chemokines by hair follicles regulates 57. Kruisbeek AM, Amsen D. Mechanisms underlying T-cell
Escherichia coli infection. Nat Immunol 2005;6:57–64. the trafficking of dendritic cells in skin. Nat Immunol tolerance. Curr Opin Immunol 1996;8:233–44.
11. Ong PY, Ohtake T, Brandt C, et al. Endogenous 2012;13:744–52. 58. Ellmeier W, Sawada S, Littman DR. The regulation of
antimicrobial peptides and skin infections in atopic 35. Banchereau J, Steinman RM. Dendritic cells and the CD4 and CD8 coreceptor gene expression during T
dermatitis. N Engl J Med 2002;347:1151–60. control of immunity. Nature 1998;392:245–52. cell development. Annu Rev Immunol 1999;17:523–54.
12. Harder J, Dressel S, Wittersheim M, et al. Enhanced 36. Ardavin C, Martinez del Hoyo G, Martin P, et al. Origin 59. Garcia KC, Teyton L, Wilson IA. Structural basis of T cell
expression and secretion of antimicrobial peptides in and differentiation of dendritic cells. Trends Immunol recognition. Annu Rev Immunol 1999;17:369–97.
atopic dermatitis and after superficial skin injury. J 2001;22:691–700. 60. Born W, Cady C, Jones-Carson J, et al.
Invest Dermatol 2010;130:1355–64. 37. Maldonado-Lopez R, De Smedt T, Michel P, et al. Immunoregulatory functions of gamma delta T cells.
13. Gläser R, Navid F, Schuller W, et al. UV-B radiation CD8α+ and CD8α- subclasses of dendritic cells direct Adv Immunol 1999;71:77–144.
induces the expression of antimicrobial peptides in the development of distinct T helper cells in vivo. J 61. Tonegawa S. Somatic generation of antibody diversity.
human keratinocytes in vitro and in vivo. J Allergy Clin Exp Med 1999;189:587–92. Nature 1983;302:575–81.
Immunol 2009;123:1117–23. 38. Rissoan MC, Soumelis V, Kadowaki N, et al. Reciprocal 62. Agrawal A, Schatz DG. RAG1 and RAG2 form a stable
14. Yang D, Chertov O, Bykovskaia SN, et al. Beta- control of T helper cell and dendritic cell postcleavage synaptic complex with DNA containing
defensins: linking innate and adaptive immunity differentiation. Science 1999;283:1183–6. signal ends in V(D)J recombination. Cell
through dendritic and T cell CCR6. Science 39. Stary G, Bangert C, Tauber M, et al. Tumoricidal activity 1997;89:43–53.
1999;286:525–8. of TLR7/8-activated inflammatory dendritic cells. J Exp 63. Schwarz K, Gauss GH, Ludwig L, et al. RAG mutations
15. Lande R, Gregorio J, Facchinetti V, et al. Plasmacytoid Med 2007;204:1441–51. in human B cell-negative SCID. Science 1996;274:97–9.
dendritic cells sense self-DNA coupled with 40. Langenkamp A, Messi M, Lanzavecchia A, Sallusto F. 64. Malissen B, Ardouin L, Lin SY, et al. Function of the
antimicrobial peptide. Nature 2007;449:564–9. Kinetics of dendritic cell activation: impact on priming CD3 subunits of the pre-TCR and TCR complexes
16. Thomson AW. The cytokine handbook. 2nd ed. of TH1, TH2 and nonpolarized T cells. Nat Immunol during T cell development. Adv Immunol
London: Academic Press; 1996. 2000;1:311–16. 1999;72:103–48.
17. Cyster JG. Chemokines and cell migration in 41. Jonuleit H, Schmitt E, Schuler G, et al. Induction of 65. van Leeuwen JE, Samelson LE. T cell antigen-receptor
secondary lymphoid tissue. Science interleukin 10-producing, nonproliferating CD4(+) T signal transduction. Curr Opin Immunol
1999;286:2098–102. cells with regulatory properties by repetitive 1999;11:242–8.
18. Aderem A, Underhill DM. Mechanisms of phagocytosis stimulation with allogeneic immature human 66. Tamada K, Chen L. T lymphocyte costimulatory
in macrophages. Annu Rev Immunol 1999;17:593–623. dendritic cells. J Exp Med 2000;192:1213–22. molecules in host defense and immunologic diseases.
19. von Andrian UH, Mackay CR. T-cell function and 42. Toews GB, Bergstresser PR, Streilein JW. Epidermal Ann Allergy Asthma Immunol 2000;85:164–75.
migration. Two sides of the same coin. N Engl J Med Langerhans cell density determines whether contact 67. Leitner J, Grabmeier-Pfistershammer K, Steinberger P.
2000;343:1020–34. hypersensitivity or unresponsiveness follows Receptors and ligands implicated in human T cell
20. Nauseef WM, Borregaard N. Neutrophils at work. Nat skin painting with DNFB. J Immunol 1980;124: costimulatory processes. Immunol Lett
Immunol 2014;15:602–11. 445–53. 2010;128:89–97.
21. Robinson DS, Kay AB, Wardlaw AJ. Eosinophils. Clin 43. Bennett CL, van Rijn E, Jung S, et al. Inducible ablation 68. Greenfield EA, Nguyen KA, Kuchroo VK. CD28/B7
Allergy Immunol 2002;16:43–75. of mouse Langerhans cells diminishes but fails to costimulation: a review. Crit Rev Immunol
22. Abraham SN, Arock M. Mast cells and basophils in abrogate contact hypersensitivity. J Cell Biol 1998;18:389–418.
innate immunity. Semin Immunol 1998;10:373–81. 2005;169:569–76. 69. Salomon B, Bluestone JA. Complexities of CD28/B7:
23. Fureder W, Agis H, Willheim M, et al. Differential 44. Kissenpfennig A, Henri S, Dubois B, et al. Dynamics CTLA-4 costimulatory pathways in autoimmunity and
expression of complement receptors on human and function of Langerhans cells in vivo: dermal transplantation. Annu Rev Immunol 2001;19:225–52.
basophils and mast cells. Evidence for mast cell dendritic cells colonize lymph node areas distinct 70. Sallusto F, Lenig D, Förster R, et al. Two subsets of
heterogeneity and CD88/C5aR expression on skin from slower migrating Langerhans cells. Immunity memory T lymphocytes with distinct homing
mast cells. J Immunol 1995;155:3152–60. 2005;22:643–54. potentials and effector functions. Nature
24. Raulet DH. Development and tolerance of natural 45. Kaplan DH, Jenison MC, Saeland S, et al. Epidermal 1999;401:708–12.
killer cells. Curr Opin Immunol 1999;11:129–34. Langerhans cell-deficient mice develop enhanced 71. Robert C, Kupper TS. Inflammatory skin diseases, T
25. Delves PJ, Roitt IM. The immune system. First of two contact hypersensitivity. Immunity 2005;23: cells, and immune surveillance. N Engl J Med
parts. N Engl J Med 2000;343:37–49. 611–20. 1999;341:1817–28.
26. Langerhans P. Über die Nerven der menschlichen 46. Schuler G, Steinman RM. Murine epidermal 72. Clark RA. Skin-resident T cells: the ups and downs of
Haut. Virchows Arch A Pathol Pathol Anat 1868;44: Langerhans cells mature into potent on site immunity. J Invest Dermatol 2010;130:
325. immunostimulatory dendritic cells in vitro. J Exp Med 362–70.
27. Katz SI, Tamaki K, Sachs DH. Epidermal Langerhans 1985;161:526–46. 73. Delves PJ, Roitt IM. The immune system. Second of
cells are derived from cells originating in bone 47. Weiss JM, Sleeman J, Renkl AC, et al. An essential role two parts. N Engl J Med 2000;343:108–17.
marrow. Nature 1979;282:324–6. for CD44 variant isoforms in epidermal Langerhans 74. Mosmann TR, Sad S. The expanding universe of T-cell
98 28. Hosoi J, Murphy GF, Egan CL, et al. Regulation of cell and blood dendritic cell function. J Cell Biol subsets: Th1, Th2 and more. Immunol Today
Langerhans cell function by nerves containing 1997;137:1137–47. 1996;17:138–46.
Online only content

CHAPTER

Immunology
eFig. 4.1 Electron microscopic picture of a Langerhans cell. Arrows indicate
the Birbeck granules, rod-shaped organelles specific for Langerhans cells. They
are said to resemble tennis rackets. Courtesy, N Romani, Department of Dermatology,
University of Innsbruck.

98.e1
75. Austrup F, Vestweber D, Borges E, et al. P- and 89. Steinbrink K, Sorg C, Macher E. Low zone tolerance to 105. Picker LJ, Kishimoto TK, Smith CW, et al. ELAM-1 is an
E-selectin mediate recruitment of T-helper-1 but not contact allergens in mice: a functional role for CD8+ T adhesion molecule for skin-homing T cells. Nature
T-helper-2 cells into inflamed tissues. Nature helper type 2 cells. J Exp Med 1996;183:759–68. 1991;349:796–9.
1997;385:81–3. 90. Miller JF, Basten A. Mechanisms of tolerance to self. 106. Fuhlbrigge RC, Kieffer JD, Armerding D, Kupper TS.
76. Hsieh CS, Macatonia SE, Tripp CS, et al. Development Curr Opin Immunol 1996;8:815–21. Cutaneous lymphocyte antigen is a specialized form
CHAPTER
of TH1 CD4+ T cells through IL-12 produced by 91. Beissert S, Schwarz A, Schwarz T. Regulatory T cells. J of PSGL-1 expressed on skin-homing T cells. Nature
Listeria-induced macrophages. Science
1993;260:547–9.
Invest Dermatol 2006;126:15–24.
92. Groux H, O’Garra A, Bigler M, et al. A CD4+ T-cell
1997;389:978–81.
107. Davies DR, Metzger H. Structural basis of antibody 4
77. Biedermann T, Zimmermann S, Himmelreich H, et al. subset inhibits antigen-specific T-cell responses and function. Annu Rev Immunol 1983;1:87–117.

Immunology
IL-4 instructs TH1 responses and resistance to prevents colitis. Nature 1997;389:737–42. 108. Stingl G, Maurer D. IgE-mediated allergen presentation
Leishmania major in susceptible BALB/c mice. Nat 93. Sakaguchi S. Naturally arising CD4+ regulatory T cells via Fc epsilon RI on antigen-presenting cells. Int Arch
Immunol 2001;2:1054–60. for immunologic self-tolerance and negative control Allergy Immunol 1997;113:24–9.
78. O’Garra A, Arai N. The molecular basis of T helper 1 of immune responses. Annu Rev Immunol 109. Chen K, Xu W, Wilson M, et al. Immunoglobulin D
and T helper 2 cell differentiation. Trends Cell Biol 2004;22:531–62. enhances immune surveillance by activating
2000;10:542–50. 94. Hori S, Nomura T, Sakaguchi S. Control of regulatory T antimicrobial, proinflammatory and B cell-stimulating
79. Trinchieri G. Interleukin-12: a cytokine at the interface cell development by the transcription factor Foxp3. programs in basophils. Nat Immunol 2009;10:
of inflammation and immunity. Adv Immunol Science 2003;299:1057–61. 889–98.
1998;70:83–243. 95. Workman CJ, Szymczak-Workman AL, Collison LW, 110. Mond JJ, Vos Q, Lees A, Snapper CM. T cell
80. Weiner HL. Induction and mechanism of action of et al. The development and function of regulatory T independent antigens. Curr Opin Immunol
transforming growth factor-beta-secreting Th3 cells. Cell Mol Life Sci 2009;66:2603–22. 1995;7:349–54.
regulatory cells. Immunol Rev 2001;182:207–14. 96. Schwarz A, Maeda A, Kernebeck K, et al. Prevention of 111. Tew JG, Wu J, Fakher M, et al. Follicular dendritic cells:
81. Cosmi L, Maggi L, Santarlasci V, et al. T helper cells UV radiation-induced immunosuppression by IL-12 is beyond the necessity of T-cell help. Trends Immunol
plasticity in inflammation. Cytometry A 2014;85: dependent on DNA repair. J Exp Med 2005;201: 2001;22:361–7.
36–42. 173–9. 112. Grabbe S, Schwarz T. Immunoregulatory mechanisms
82. Miossec P, Korn T, Kuchroo VK. Interleukin-17 and type 97. Roncarolo MG, Levings MK, Traversari C. Differentiation involved in elicitation of allergic contact
17 helper T cells. N Engl J Med 2009;361:888–98. of T regulatory cells by immature dendritic cells. J Exp hypersensitivity. Immunol Today 1998;19:37–44.
83. Milner JD, Brenchley JM, Laurence A, et al. Impaired Med 2001;193:F5–9. 113. Watanabe H, Gaide O, Pétrilli V, et al. Activation of the
T(H)17 cell differentiation in subjects with autosomal 98. Bendelac A, Rivera MN, Park SH, Roark JH. Mouse IL-1beta-processing inflammasome is involved in
dominant hyper-IgE syndrome. Nature CD1-specific NK1 T cells: development, specificity, and contact hypersensitivity. J Invest Dermatol
2008;452:773–6. function. Annu Rev Immunol 1997;15:535–62. 2007;127:1956–63.
84. Cargill M, Schrodi SJ, Chang M, et al. A large-scale 99. Moodycliffe AM, Nghiem D, Clydesdale G, Ullrich SE. 114. Grabbe S, Steinert M, Mahnke K, et al. Dissection of
genetic association study confirms IL12B and leads to Immune suppression and skin cancer development: antigenic and irritative effects of epicutaneously
the identification of IL23R as psoriasis-risk genes. Am J regulation by NKT cells. Nat Immunol 2000;1:521–5. applied haptens in mice. Evidence that not the
Hum Genet 2007;80:273–90. 100. Stenger S, Modlin RL. T cell mediated immunity to antigenic component but nonspecific
85. Leonardi CL, Kimball AB, Papp KA, et al. Efficacy and Mycobacterium tuberculosis. Curr Opin Microbiol proinflammatory effects of haptens determine the
safety of ustekinumab, a human interleukin-12/23 1999;2:89–93. concentration-dependent elicitation of allergic
monoclonal antibody, in patients with psoriasis: 101. Payer E, Elbe A, Stingl G. Epidermal T lymphocytes contact dermatitis. J Clin Invest 1996;98:1158–64.
76-week results from a randomised, double-blind, – ontogeny, features and function. Springer Semin 115. Volc-Platzer B, Majdic O, Knapp W, et al. Evidence of
placebo-controlled trial (PHOENIX 1). Lancet Immunopathol 1992;13:315–31. HLA-DR antigen biosynthesis by human keratinocytes
2008;371:1665–74. 102. Montaldo E, Vacca P, Moretta L, Mingari MC. in disease. J Exp Med 1984;159:1784–9.
86. Eyerich S, Eyerich K, Pennino D, et al. Th22 cells Development of human natural killer cells and other 116. Seery JP, Carroll JM, Cattell V, Watt FM. Antinuclear
represent a distinct human T cell subset involved in innate lymphoid cells. Semin Immunol autoantibodies and lupus nephritis in transgenic mice
epidermal immunity and remodeling. J Clin Invest 2014;26:107–13. expressing interferon gamma in the epidermis. J Exp
2009;119:3573–85. 103. Butcher EC, Picker LJ. Lymphocyte homing and Med 1997;186:1451–9.
87. Andersen MH, Schrama D, Thor Straten P, Becker JC. homeostasis. Science 1996;272:60–6. 117. Mehling A, Loser K, Varga G, et al. Overexpression of
Cytotoxic T cells. J Invest Dermatol 2006;126:32–41. 104. Campbell JJ, Bowman EP, Murphy K, et al. 6-C-kine CD40 ligand in murine epidermis results in chronic
88. Mosmann TR, Li L, Sad S. Functions of CD8 T-cell (SLC), a lymphocyte adhesion-triggering chemokine skin inflammation and systemic autoimmunity. J Exp
subsets secreting different cytokine patterns. Semin expressed by high endothelium, is an agonist for the Med 2001;194:615–28.
Immunol 1997;9:87–92. MIP-3β receptor CCR7. J Cell Biol 1998;141:1053–9.

99

You might also like