Sie sind auf Seite 1von 21

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/269178869

Immunology and Skin in Health and Disease

Article  in  Cold Spring Harbor Perspectives in Medicine · December 2014


DOI: 10.1101/cshperspect.a015339 · Source: PubMed

CITATIONS READS

12 99

2 authors:

Jillian Richmond John E Harris


University of Massachusetts Medical School University of Massachusetts Medical School
32 PUBLICATIONS   453 CITATIONS    56 PUBLICATIONS   870 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Simvastatin Prevents and Reverses Depigmentation in a Mouse Model of Vitiligo View project

T cells and CTCL View project

All content following this page was uploaded by John E Harris on 02 December 2015.

The user has requested enhancement of the downloaded file.


Immunology and Skin in Health and Disease

Jillian M. Richmond and John E. Harris


Department of Medicine, Division of Dermatology, University of Massachusetts Medical School, Worcester,
Massachusetts 01605
Correspondence: john.harris@umassmed.edu

The skin is a complex organ that, in addition to providing a strong barrier against external
insults, serves as an arena for a wide variety of inflammatory processes, including immunity
against infections, tumor immunity, autoimmunity, and allergy. A variety of cells collaborate
to mount functional immune responses, which are initiated by resident populations and
evolve through the recruitment of additional cell populations to the skin. Inflammatory
responses are quite diverse, resulting in a wide range of signs and symptoms that depend
on the initiating signals, characteristics of the infiltrating cell populations, and cytokines that
are produced (cytokines are secreted protein that allows for cell –cell communication;
usually refers to communication between immune–immune cells or stromal–immune
cells). In this work, we will review the skin architecture and resident and recruited cell
populations and discuss how these populations contribute to inflammation using human
diseases and treatments when possible to illustrate their importance within a clinical context.
www.perspectivesinmedicine.org

issues at the interface of the host and the where sterile inflammation, including tumor
T environment, including the skin, gut, and
other mucosal surfaces, present the first line of
immunity, allergy, and autoimmune responses,
may participate in disease. Tumor immunity
defense against pathogens. The barrier function is defective in organ transplant patients who
of the skin is of critical importance, which is are immunosuppressed, but is co-opted dur-
evident when this barrier is disrupted following ing imiquimod treatment of various skin can-
injury, or in atopic dermatitis, ichthyosis, or cers and warts. Allergic responses, which likely
irritant contact dermatitis. Once the barrier is evolved to protect against parasitic invasion,
disrupted, the rapid but nonspecific innate im- may cause disease when directed against innoc-
mune response is recruited in defense, a process uous foreign materials, as in allergic contact der-
that relies on detection of both self and foreign matitis. Autoimmunity, possibly caused by anti-
“danger signals” as the initial alarm. Next, the pathogen or antitumor immunity misdirected
slower, but specific adaptive immune response against self, causes a wide range of pathology
may be required for definitive clearance of a in the skin, including vitiligo, lupus, psoriasis,
pathogen. and other diseases.
In addition to providing protection against Appropriate functioning of the skin in these
invading pathogens, the skin is also an arena roles requires close communication and collab-

Editors: Anthony E. Oro and Fiona M. Watt


Additional Perspectives on The Skin and Its Diseases available at www.perspectivesinmedicine.org
Copyright # 2014 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 10.1101/cshperspect.a015339
Cite this article as Cold Spring Harb Perspect Med 2014;4:a015339

1
J.M. Richmond and J.E. Harris

oration among a number of various cell types, through receptors called pattern-recognition
including stromal cells (keratinocytes, fibro- receptors (PRRs are intracellular or cell surface
blasts, endothelial cells, and adipocytes) as well receptors activated by DAMPs to induce in-
as those derived from the bone marrow (den- flammation). Adaptive immune populations
dritic cells, macrophages, natural killer cells, form slower, but pathogen-specific, responses
mast cells, T cells, and others). Of the bone mar- to infection through specialized and unique
row– derived cells that can be found in the antigen-specific receptors formed via genetic
skin, some are resident cell populations that rearrangement (an antigen is any molecule ca-
migrate to the skin where they terminally dif- pable of inducing an antibody response or T-
ferentiate and primarily reside there, some re- cell response; it can be protein, lipid, or carbo-
circulate continuously and perform a surveil- hydrate). These receptors permit immune cells
lance role, some are recruited to fight infection to mount a more targeted attack on invaders,
for the short term, and others are recruited and and these cells can then become long-lived and
maintained as memory cells to protect against capable of a rapid, specific response against
future reinvasion. Bone marrow– derived cells reinvasion of a pathogen, known as a memory
can be further subdivided into innate and adap- response.
tive immune populations (see Fig. 1). Innate Initiation of an adaptive immune response is
cells form rapid, but nonspecific, responses performed by antigen-presenting cells (APCs),
to infection. They generally recognize non- which efficiently take up ( phagocytose) pro-
self-molecular patterns on pathogens or path- teins, process them into recognizable peptides
ogen-associated molecular patterns (PAMPs), (antigens), and present them to T cells on sur-
www.perspectivesinmedicine.org

Skin-resident cells
Innate immune cells Adaptive immune cells Nonhematopoietic origin

Keratinocyte
Langerhans cell Macrophage T cell
(usually memory, Fibroblast
NKT, or γδ)

Endothelial cell
Dermal dendritic cell Mast cell

Recruited immune cells


Innate immune cells Adaptive immune cells
Granulocytes

NK cell Monocyte T cell B cell


Neutrophil Eosinophil

Figure 1. Immune populations in the skin. (Top) Langerhans cells, dermal dendritic cells, macrophages, other
innate cells (mast cells, NK cells, NKT cells, gd-T cells), and memory T cells comprise the skin-resident immune
system under the steady state. Langerhans cells, dermal dendritic cells, macrophages, and gd-T cells first sense
infection or injury and initiate a rapid, innate response that includes the recruitment of effector cells. Innate
effectors (NK populations) provide a rapid, antigen-nonspecific response, whereas memory T cells provide a
rapid, antigen-specific memory response to previously encountered pathogens. Stromal cell populations, such
as keratinocytes, fibroblasts, and endothelial cells, also participate in immune responses by sensing tissue
damage and producing inflammatory cytokines. (Bottom) On activation of the skin-resident immune system,
additional immune cells are recruited to help contain and fight infection and/or to remove cellular debris to aid
in the healing process. These include additional innate cells like neutrophils and eosinophils, as well as adaptive
populations like naı̈ve or central memory T cells and B cells.

2 Cite this article as Cold Spring Harb Perspect Med 2014;4:a015339


Skin Immunology

face human leukocyte antigen (HLA) I and HLA how the immune system works within a clinical
II molecules (human leukocyte antigen class I context.
and II; typically, class I presents intracellular an-
tigens and class II presents extracellular anti-
NORMAL SKIN IMMUNE SYSTEM
gens). All nucleated cells in the body express
HLA I, providing an important mechanism for Tissue Architecture and Composition
detection of viral infections as well as malignant
Normal skin architecture includes the epider-
transformation through direct communication
mis, dermis, and subcutaneous fat (see Fig. 2).
with immune cells. HLA I presents antigens that
The resident cell populations that make up these
are produced within the cell and permits their
strata can be broadly divided into immune and
recognition by antigen-specific T cells through
nonimmune cells. Nonimmune cell popula-
their T-cell receptor, making the cells susceptible
tions are important for the structure and func-
to cytotoxic T-cell-mediated killing. Typically,
tion of the skin, but also contribute to skin im-
if those antigens are from normal self-proteins,
munity, as they first provide a general barrier
they will be spared, a process called immunolog-
to invasion by foreign materials. They also
ical tolerance. However, if those antigens are de-
directly participate in inflammation, shaping
rived from intracellular pathogens (like viruses),
the immune response as it develops within the
or if the antigen is an abnormal self-protein
tissue.
generated following malignant transformation
of the cell, then that cell may be killed to pre-
vent further injury to the host. Unlike HLA I, Epidermis
HLA II is typically only expressed on APCs,
The epidermis is comprised primarily of kera-
and is capable of presenting antigens derived
www.perspectivesinmedicine.org

tinocytes, which are tightly connected to one


from extracellular proteins acquired through
another and act similarly to a brick wall to limit
phagocytosis (cell eating; the process by which
access to the internal environment. As a conse-
antigen-presenting cells and other phagocytic
quence, these tight connections limit move-
cells take up pathogens or other debris). Den-
ment within this layer and therefore other cell
dritic cells (DCs) are recognized as professional
types that reside there, which include both stro-
APCs because their main function is to take up
mal and bone marrow– derived cells, are pri-
antigens, thereby linking innate and adaptive
marily fixed in position. However, when dam-
immune responses. If DCs phagocytose a path-
age occurs and immune cells sense danger, they
ogen in the presence of a PAMP that alerts them
release proinflammatory mediators to recruit
to danger, they then produce proinflammatory
innate effector cells, and then exit to draining
mediators to recruit innate immune cells to ini-
lymph nodes where they encounter T cells and
tiate the defense, and then typically migrate out
other members of the adaptive immune system.
of the skin through lymphatic channels into
draining lymph nodes. There they position
themselves to directly encounter T cells, and Cellular Components of the Epidermis
each T cell will briefly probe the DC to deter-
† Keratinocytes maintain tight junctions and
mine whether it is presenting an antigen that the
form the stratum corneum, which is critical
T cell recognizes. If so, the T cell will become
for the barrier function of the epidermis (see
activated and initiate a search for the location
Box 1). They may also contribute to inflam-
of the infection.
mation, as they can express HLA II and se-
In this article, we will describe how all of the
crete cytokines.
cell populations within the skin contribute to
immune responses, and the general principles † Melanocytes are pigment-producing cells in
and tools required for effective immunity. We the skin. The melanin pigment they produce
will use specific examples of common skin dis- and distribute to keratinocytes helps to shield
eases and treatments when possible to illustrate DNA from ultraviolet radiation, which can

Cite this article as Cold Spring Harb Perspect Med 2014;4:a015339 3


J.M. Richmond and J.E. Harris

Epidermis

Merkel cell

Extracellular matrix
protein
Dermis

Nerve bundle

Blood vessel

Adipocyte
Subcutaneous fat

Figure 2. Location of immune cells within normal skin architecture. Distinct populations of immune cells
inhabit local niches within the skin. The epidermis contains Langerhans cells to provide immune surveillance.
Memory T cells are also retained in the epidermis, presumably for early detection and control of re-encountered
pathogens. Keratinocytes may sense pathogens or other damage-associated signals and communicate this to the
immune system through cytokines. The dermis contains dermal dendritic cells, T cells, and fibroblasts. In this
example, we depict a T cell being recruited out of the blood and into the dermis and potentially the epidermis,
depending on the site of infection/injury. Recruitment of cells to peripheral tissues requires production of
chemoattractants. Endothelial cells, which form the walls of the vasculature, may present chemokines and/or
adhesion molecules to immune cells to direct their transmigration into the skin.
www.perspectivesinmedicine.org

induce DNA damage. Melanocytes are also that radiate in all directions, allowing them to
capable of expressing MHC II, and are the probe throughout the entire epidermis. The
targets of autoimmunity in vitiligo. exact role of LHCs is not entirely clear, al-
though they may promote tolerance to envi-
† Merkel cells are specialized cells that commu-
ronmental antigens, including commensal
nicate with cutaneous neurons in skin sen-
bacteria and fungi, and help to polarize T cells
sation. A clear contribution of Merkel cells
into a particular inflammatory response as
to cutaneous immunity has not yet been de-
described below. Although keratinocytes
scribed.
and melanocytes may also be capable of acting
as APCs, the implication of this function is
Bone Marrow– Derived Cells of the not yet clear.
Epidermis
† Memory T cells may reside within the epi-
† Langerhans cells (LHCs) are dendritic cells dermis for very long periods of time. Where-
(DCs) that spend the majority of their time as neutrophils and T cells can infiltrate the
in the epidermis. LHCs are tightly connected epidermis under some circumstances (atopic
to keratinocytes through dendritic processes and contact dermatitis, cutaneous T-cell

BOX 1. SKIN BARRIER DYSFUNCTION

Filaggrin is a protein that multimerizes, binds to keratin fibers, and strengthens connective tissues,
helping to form a tight barrier against exposure to environmental agents. It is essential to barrier
function within the epidermis. In atopic dermatitis and ichthyosis, filaggrin mutations cause barrier
dysfunction, resulting in chronic inflammation and opportunistic infection.

4 Cite this article as Cold Spring Harb Perspect Med 2014;4:a015339


Skin Immunology

lymphoma, psoriasis, and vitiligo), they are with neurons to regulate innate immunity
typically excluded. (Rosas-Ballina et al. 2011). Neuroimmunol-
ogy is a relatively new field, and the relation-
Dermis ship between skin neurons and immune cells
is not yet fully appreciated.
The dermis is primarily comprised of extracel-
lular matrix proteins that give the skin structure
and elasticity. Unlike the epidermis, it permits Bone Marrow – Derived Cells of the Dermis
the free migration of cell populations. The der- Innate populations of the dermis:
mis and epidermis are separated by the base-
ment membrane, a thin, tight sheet of extracel- † Dermal DCs (dDCs) and plasmacytoid DCs
lular matrix proteins that regulates movement (pDCs), two distinct populations of den-
of cells and proteins in between these two layers. dritic cells, are located in the dermis. They
have fewer dendrites but increased motility
compared with LHCs, and are capable of mi-
Cellular Components of the Dermis grating on collagen paths to monitor the der-
mis. Each DC population has been character-
† Fibroblasts produce structural proteins,
ized by the production of different cytokines,
which in addition to providing a supporting
and may initiate distinct inflammatory re-
scaffold, serve as highway systems for migra-
sponses following activation. For example,
tory immune cells. As in the lymph node,
pDCs have been reported to initiate antifun-
these highways ensure that immune cells
gal immunity (Ramirez-Ortiz et al. 2011),
frequently contact each other, which is impor-
whereas dDCs initiate antiviral immunity
www.perspectivesinmedicine.org

tant in communication during immune re-


(Kaplan 2010). These roles are not likely to
sponses. Like keratinocytes, fibroblasts are ca-
be exclusive, such that different DC popula-
pable of producing cytokines.
tions may contribute to different immune
† Endothelial cells form the innermost layer of responses in multiple ways.
the blood vessels in the skin, and regulate the
† Macrophages are skin-resident immune cells
passage of immune cells into the skin
with high phagocytic capacity and motility.
through the production of adhesion mole-
Although they are less likely than DCs to pre-
cules, cytokines, and chemokines (a cytokine
sent antigen to T cells, due to relative cell
that acts as a chemoattractant to induce cell
numbers, they are capable of activating im-
migration) (see also Box 2).
mune responses through PRRs and cytokine
† Neurons form nerve bundles in the skin, secretion. They also “clean up” debris from
which allow for sensation. Recently, it has dead or dying cells, invading pathogens,
been shown that memory T cells can interact or environmental insults, including tattoo

BOX 2. LFA-1 AND LEUKOCYTE ADHESION

LFA-1 is expressed by endothelial cells and promotes adhesion of leukocytes, permitting their mi-
gration into peripheral tissues. A defect in the adhesion molecule CD18, a component of LFA-1,
results in leukocyte adhesion deficiency (LAD). LAD is characterized by chronic bacterial skin
infections owing to impaired neutrophil chemotaxis. In addition, the CD11a component of LFA-1
was the target of the drug efalizumab for psoriasis, which presumably functioned by inhibiting
immune cell migration into the skin. Predictable side effects were infections of multiple organs,
including a potentially lethal JC virus infection within the brain, which led to its withdrawal from the
market in 2009.

Cite this article as Cold Spring Harb Perspect Med 2014;4:a015339 5


J.M. Richmond and J.E. Harris

ink. For example, “melanophages” represent teins. Basophils, which also degranulate in
macrophages that have phagocytosed mela- this manner, can act as potential APCs.
nocyte fragments and melanin released into Mast cells are typically skin-resident granulo-
the dermis following epidermal inflamma- cytes that mediate immune responses against
tion. parasites following binding and cross-linking
of IgE antibodies bound to parasitic invad-
† Monocytes are a type of immature macro- ers, followed by degranulation of histamines
phages that are usually found in the circula- and other proinflammatory proteins. They
tion. They can be recruited to the skin to have also been implicated in the pathogenesis
maintain homeostasis or in response to in- of allergic responses when IgE antibodies are
fection/injury, where they receive cues to dif- produced against innocuous environmental
ferentiate into macrophages or myeloid DCs, antigens, like dust mite proteins and animal
a DC population that is not well understood. dander.
† Granulocytes include neutrophils (also called † Natural killer (NK) cells are classified as in-
polymorphonuclear cells or PMNs), eosino- nate cells because of their pattern-recogni-
phils, basophils, and mast cells. These innate tion functions, but may also confer memory,
immune cell populations can be recruited to like adaptive populations. NK cells detect the
the skin following activation of a tissue-resi- level of self-HLA I expressed on the surface
dent cell and subsequent release of chemo- of cells, and are activated when expression
kines and activation of the endothelium. levels are too low, which occurs when either
Granulocytes are named for their cytoplas- malignant or virally infected cells impair the
mic granules that are filled with proteases, expression of HLA I to escape immune de-
www.perspectivesinmedicine.org

vasoactive peptides, and antimicrobial pep- tection by T cells. NK cells can also perform
tides, which are released during degranula- antibody-dependent cellular cytotoxicity
tion. Neutrophils are typically the first cell (ADCC) by binding IgG-coated pathogens
type recruited to the skin following activation and cells, releasing granules containing per-
of dendritic cells and/or macrophages in re- forin and granzyme.
sponse to PAMPs encountered during infec-
tion, and are capable of efficiently phagocy- Adaptive populations of the dermis:
tosing and killing pathogens (see Box 3).
Neutrophils have also recently been shown † Most of the skin-resident adaptive cells are
to make “sticky” extracellular traps (NETs) T lymphocytes (T cells) of different subsets.
by expelling the DNA from their nucleus, ef- There are approximately 20 billion T cells pre-
fectively trapping pathogens like flies caught sent in the skin, nearly twice the number of T
in a spider web. Eosinophils and basophils cells in the blood (Clark 2010) (a lymphocyte
contribute to antiparasitic and allergic re- is a type of adaptive immune cell including T
sponses, although we are just beginning to and B cells, capable of genetically rearranging
understand their unique roles in immunity. antigen-specific receptors). CD8þ cytotoxic
Eosinophils degranulate when their IgE re- T cells (CTLs) are effector cells that recognize
ceptors become cross-linked, and they release a specific antigen presented on a cell surface
proteases, chemokines, and vasoactive pro- by HLA I, and subsequently kill that cell.

BOX 3. CHRONIC GRANULOMATOUS DISEASE

Neutrophils kill pathogens through the use of a “respiratory burst,” which depends on NADPH
oxidase to generate oxygen free radicals. This gene is defective in chronic granulomatous disease
(CGD), and patients with CGD develop chronic skin bacterial and fungal infections.

6 Cite this article as Cold Spring Harb Perspect Med 2014;4:a015339


Skin Immunology

CD4þ helper T cells (TH cells) “help” effector the circulation. These antibodies may medi-
cell populations like CTLs and B cells through ate either infectious or autoimmune re-
the production of cytokines and promatura- sponses, depending on their specificity.
tion signals to DCs, which both license the Whereas B cells and plasma cells can occa-
effector cells to carry on an attack, and steer sionally be found in the skin (syphilitic infec-
the response in a particular direction, either tion, lupus, and other diseases), the reason
antiviral, antitumor, antibacterial, or anti- for this localization to the skin is unknown.
parasitic, as described in more detail below.
This TH-cell help provides an important
Subcutaneous Fat
check to the development of an immune re-
sponse, essentially licensing other antigen- The subcutaneous fat layer is mainly comprised
specific T and B cells before allowing the re- of adipocytes, but also contains nerves, blood,
sponse to develop. T-regulatory cells (Tregs) and lymphatic vessels. Adipocytes are fat cells
are typically CD4þ and express FoxP3, a crit- that sequester potentially inflammatory fatty
ical transcription factor for their develop- acids in the form of lipids. They are also capable
ment (see Box 4). Their role is to suppress of producing proinflammatory cytokines.
immune responses as a major contributor
to peripheral tolerance, helping to prevent
autoimmunity and to resolve inflammation Soluble Proteins of the Skin Immune System
once a threat has been controlled. As men- † There are several families of proteins that are
tioned above, any of these cell populations important for skin immunity. Complement is
may be temporary, migrating through the a family of soluble plasma proteins that can
skin for surveillance, or may remain long
www.perspectivesinmedicine.org

bind to pathogens, self-catalyze to form solu-


term as skin-resident memory T cells (TRM) ble chemoattractants and membrane-bound
to prevent reinfection. opsonins that promote phagocytosis, and
† gd-T cells, which have a limited repertoire of membrane attack complexes that can punc-
T-cell receptors (TCRs), and natural killer T ture bacteria (see Box 5). Complement fixa-
cells (NKTs), which are classified by their tion to the bacterium can occur by three
expression of both NK cell receptors and different pathways: classical, alternative, or
the universal T-cell marker CD3, are other lectin. Classical activation of complement is
T-cell populations of the dermis. Both pop- catalyzed by antibodies bound to the patho-
ulations respond to lipid antigens. gen, alternative activation occurs through di-
rect binding of complement to the pathogen,
† Once activated, B lymphocytes (B cells) ma- and lectin activation is catalyzed by mannose
ture into antibody-producing plasma cells binding lectin (MBL) bound to the pathogen.
that usually reside in the lymph nodes and Phagocytes express complement receptors
bone marrow, producing antibodies that be- that recognize complement bound to an in-
come passively delivered to the skin through vading organism, which facilitates phagocy-

BOX 4. IPEX SYNDROME

Immunodysregulation polyendocrinopathy enteropathy X-linked syndrome (IPEX) is a genetic dis-


ruption of the FoxP3 transcription factor, which is important for the generation of Tregs. Tregs are
critical for the maintenance of peripheral tolerance to self-tissues and thus in the prevention of
autoimmunity. Without this, patients develop multiple autoimmune diseases, including enteropathy;
insulin-dependent diabetes mellitus, thyroid disease, and other endocrinopathies; and skin disease
including eczema, psoriasiform dermatitis, urticaria, and alopecia universalis, among others.

Cite this article as Cold Spring Harb Perspect Med 2014;4:a015339 7


J.M. Richmond and J.E. Harris

BOX 5. COMPLEMENT ABNORMALITIES

Genetic defects in complement can result in too much or too little complement activity. Loss-of-
function mutations ultimately result in recurrent infections because of decreased opsonization and/
or decreased lytic activity. Common infections resulting from complement deficiency include recur-
rent meningococcal infection (Neisseria subspecies), Streptococcus pneumoniae infection, or other
encapsulated bacterial infections. Hereditary or autoimmune angioedema results from too much
complement activity caused by a loss of the C1 esterase inhibitor. As a result, complement activation
occurs spontaneously, which induces production of bradykinin. Bradykinin is a vasoactive peptide
that results in blood vessel dilation, thereby causing the edema and other symptoms of the disease.

tosis of the invader regardless of how comple- molecular patterns alert immune cells to dan-
ment was first fixed to the surface (a form of ger. These patterns are typically present only
opsonization, which is the process of coating when normal cellular processes have been dis-
with proteins that facilitate phagocytosis). rupted, as seen in infection, malignancy, or
necrotic cell death. These patterns are broad-
† Antimicrobial peptides are another family of
ly referred to as danger-associated molecular
proteins that contribute to skin immunity.
patterns (DAMPs; can be pathogen-derived or
Examples include b-defensins, psoriasin,
altered-self signals), microbe-associated molec-
and lysozyme, which have varying but direct
ular patterns (MAMPs), viral-associated molec-
antimicrobial activity against bacteria and
ular patterns (VAMPs), or pathogen-associated
fungi. Keratinocytes are one of the main
molecular patterns (PAMPs), depending on
www.perspectivesinmedicine.org

sources of antimicrobial peptides, which are


from where they are derived. Examples include
induced by TLR ligation and/or cytokine
lipopolysaccharide (LPS), flagellin, and pepti-
production.
doglycan from bacteria, double-stranded RNA
† Interferons (IFN) belong to a family of cyto- from viruses, or mislocalized (extranuclear)
kines that are proinflammatory and also di- double-stranded DNA during necrotic cell
rectly “interfere” withviral replication through death. The receptors that recognize these pat-
reducing protein translation and increasing terns include Toll-like receptors (TLRs) and
p53 levels, which promotes apoptosis. NOD-like receptors (NLRs), and are expressed
by DCs, other immune cells, and sometimes
† Antibodies bind with high specificity and af-
stromal cells. There are both surface and intra-
finity to both foreign proteins during an in-
cellular PRRs to allow for both extra- and intra-
fectious immune response, or self-proteins in
cellular recognition of pathogens. Activation of
autoimmunity. They can injure cells by pro-
these receptors by DAMPs promotes antigen
moting phagocytosis through opsonization,
processing and presentation, up-regulation of
complement-mediated lysis, activation of
costimulatory receptors for T-cell activation,
signaling cascades, and through neutraliza-
and secretion of proinflammatory cytokines in-
tion of surface proteins.
cluding IL-6, IL-1b, TNF-a, and others. There-
fore, it is these “danger signals” that initiate a
proinflammatory response.
INITIATION AND EVOLUTION OF AN
On activation, tissue-resident cells secrete
IMMUNE RESPONSE IN THE SKIN
proinflammatory cytokines that promote the
DCs regularly take up proteins within the skin, recruitment of innate effector cells, including
but must distinguish whether they are presented neutrophils, monocytes, and NK cells. These
in a dangerous context like infection or malig- first responders can begin the attack using the
nancy, or a safe context like normal cell turn- mechanisms described above. However, an ef-
over during tissue homeostasis. A number of fective immune response usually requires the

8 Cite this article as Cold Spring Harb Perspect Med 2014;4:a015339


Skin Immunology

subsequent involvement of the adaptive im- TH17 cells produce IL-17, IL-21, and IL-22; and
mune system, and so DCs must interact with a TH2 cells produce IL-4, IL-5, and IL-13. These
multitude of T cells to identify the correct cells different cytokine patterns are usually associat-
with the capacity to recognize the specific invad- ed with recruitment of slightly different types
ing pathogen. Because very few T cells are capa- of immune effector populations. For example,
ble of responding to any particular challenge, TH2 responses recruit basophils, eosinophils,
DCs must migrate to a draining lymph node, and mast cells to coordinate an antiparasitic
the equivalent of Grand Central Station in response, whereas TH1 responses result in re-
New York, to most efficiently survey the millions cruitment of CTLs for an antiviral or antitumor
of potential responders and find those that can response, and TH17 promotes an antibacterial
participate in any particular response. or antifungal response through the recruitment
Following the successful activation of an an- of neutrophils and production of cytokines and
tigen-specific T cell, that cell will proliferate, exit antimicrobial peptides. TH1 and TH17 respons-
the lymph node into the blood stream, and es may also promote autoimmunity, whereas
search for the location in the body that contains TH2 responses may mediate allergy (see also
its target. The DC will help to direct the T cell to the section on Diseased Skin). For efficiency
the skin through a mechanism that is not fully and efficacy, immune responses often polarize
understood, called “imprinting”; however, it ap- toward a single specific pathway; however,
pears that vitamins play a role. Vitamin D is mixed responses may also occur, creating a sig-
produced primarily within the skin following nificant level of complexity that has yet to be
exposure to UV light, and therefore skin DCs fully understood for all such responses in vivo.
contain large amounts of this vitamin. When T The subpopulation of DC that initiates the
cells are activated by DCs containing vitamin D, immune response and the signals present dur-
www.perspectivesinmedicine.org

the T cells express skin-homing receptors, in- ing the initiation can influence the nature of that
cluding CCR4, CCR10, and CLA. A similar T- response. For example, LHC may suppress im-
cell educational program occurs via intestinal mune responses, acting as tolerogenic media-
DCs that contain vitamin A, which is acquired tors because they regularly sample foreign pro-
through the diet, inducing a6b4 protein, a gut- teins and organisms present on the intact skin
homing receptor. Once the T cells are in the surface, which are primarily nonthreatening.
bloodstream and express skin-homing recep- When LHC-deficient mice are exposed to a con-
tors, the initial cytokines and adhesion mole- tact allergen, the response is exacerbated, sup-
cules on endothelial cells at the site of inflam- porting this suppressive role of LHCs in the
mation help to guide those cells to the correct skin. In contrast, dDC-deficient mice elicit a
site. Although activated B cells, which produce dampened response, suggesting this dDC pop-
antibodies, may also be recruited to the skin ulation is proinflammatory in this context. This
during inflammation, they more often remain role is intuitive, because exposure of dermal
within the lymph nodes or bone marrow, secret- DCs to a foreign antigen would require disrup-
ing the antibody into the blood where it is pas- tion of the epidermis, which is a potentially
sively carried to the skin and contributes to the dangerous event. Recent studies also support a
immune response. nuanced proinflammatory role for these two
As described above, T-cell subsets have spe- populations. Whereas activated LHCs seem to
cialized functions, including those that are di- promote an antibacterial/antifungal immune
rectly cytotoxic, others that suppress responses, response through the production of TH17-spe-
and still others that oversee and help to shape cific cytokines, dDC promote antiviral respons-
the response through the secretion of cytokines. es through the production of IFNs and other
TH subsets are usually designated with a number TH1-specific cytokines. This may be because of
to delineate which types of cytokines they pro- the fact that viruses often enter the skin systemi-
duce. For example, TH1 cells produce interfer- cally or through epidermal disruption and
on-g (IFN-g) and tumor necrosis factor (TNF); therefore primarily enter the dermis, whereas

Cite this article as Cold Spring Harb Perspect Med 2014;4:a015339 9


J.M. Richmond and J.E. Harris

bacteria most often enter through the epider- proliferation and stimulate fibroblast produc-
mis. Little is known about the mechanism by tion of extracellular matrix proteins (see Box
which DCs distinguish these stimuli, although 6). In both the skin and the gut, Tregs have
it is likely mediated through PRRs. been shown to play an important role in main-
taining tolerance to normal bacterial flora. It has
recently been postulated that Treg sensing of flo-
Turning off Inflammation and Initiating ra is also important for normal wound healing
Wound Healing (Chen et al. 2013).
An important part of normal immune responses
is turning off inflammation once the infection is
DISEASED SKIN
cleared or injury is healed. Skin-resident Treg
populations have been shown to be activated Proper Immune Responses in Disease—
by epidermal LHC (Seneschal et al. 2012), and Infectious Immunity
play an important role in dampening inflamma-
Infectious Immunity: Part I. Examples of
tion. There are several different Treg populations
Bacterial Immunity
that are usually subdivided into central and pe-
ripheral subtypes. Whereas central Tregs develop Staphylococcus aureus is a prevalent human skin
in the thymus, peripheral Tregs are generated commensal and pathogen that can cause super-
during immune responses in lymphoid organs ficial skin infections (impetigo and exacerba-
and/or tissues, where asymmetric division of tion of atopic dermatitis), infection of the hair
effectorcells allows for generation of a small pop- follicle (folliculitis and furunculosis), as well as
ulation of Tregs. Examples of these include TH3 deep infections (ecthyma and abscesses). Like
www.perspectivesinmedicine.org

and TR1 cells, which produce the anti-inflam- most other bacterial infections of the skin, these
matory cytokine IL-10. Both central and periph- conditions are characterized clinically by ten-
eral Tregs are capable of down-modulating in- der, red, inflamed pustules and abscesses that
flammation via cytokine production, uptake, form as a result of cytokine expression, neutro-
and removal of the T-cell growth factor IL-2, phil recruitment, and an epidermal response,
and through DC interactions. Interleukin-10 including keratinocyte proliferation and pro-
(IL-10) is an anti-inflammatory cytokine that duction of antimicrobial peptides. Twenty per-
will down-regulate the expression of other cyto- cent of the population is colonized with S. au-
kines, MHC II, and costimulatory molecules. reus, and infections like those described above
Tregs express the high-affinity IL-2 receptor, are quite common.
CD25, and will therefore preferentially bind Methicillin-resistant S. aureus (MRSA) is a
and “sop up” IL-2 in the milieu, thereby remov- particularly recalcitrant infection due both to
ing the T-cell growth and survival signal. Tregs its antibiotic resistance as well as multiple viru-
are also capable of directly interacting with DCs lence factors (a component of a pathogen that
and inducing down-regulation of costimulatory permits infection and survival; can be protein,
molecules through cell – cell interactions. Tregs lipid, carbohydrate, or nucleic acid) (reviewed
also produce transforming growth factor-b by Foster 2005). S. aureus is a Gram-positive
(TGF-b), which can both inhibit immune cell coccus, which has an outer cell wall. Innate im-

BOX 6. IMIQUIMOD TREATMENT OF WARTS

Imiquimod is a synthetic TLR-7/8 agonist that can be used as adjuvant therapy. In the case of HPV,
imiquimod can induce production of type I IFN, IL-6, and TNF to overcome the lack of inflammatory
mediator production and actively suppress Treg function. An additional approach to treating warts is
intradermal injection of Candida antigens, which are natural TLR ligands.

10 Cite this article as Cold Spring Harb Perspect Med 2014;4:a015339


Skin Immunology

mune receptors and PRRs important in the de- host phagocytes, S. aureus can also scavenge free
tection of Gram-positive bacteria include Toll- radicals via superoxide dismutases. The unique
like receptor 2 (TLR-2), and mannose-binding structure of its wall is resistant to degradation by
lectin (MBL), which are capable of detecting lysozyme. These factors may in part explain why
the sugars and lipids that comprise bacterial it is so difficult to induce good adaptive immune
cell walls. As described in detail above, ligation responses to S. aureus, as there could be less ef-
of PRRs associated with skin-resident immune ficient antigen generation and presentation.
cells induces activation and maturation of DCs, S. aureus produces the chemotaxis inhibi-
keratinocyte proliferation and antimicrobial tory protein of staphylococci (CHIPS), which
peptide production, and the production of cy- can bind to and inhibit chemoattractant recep-
tokines and chemokines to promote the recruit- tors on the surface of neutrophils. It also pro-
ment of neutrophils and macrophages to the site duces Eap, a protein that disrupts LFA-1 and
of infection. Once in the skin, neutrophils and ICAM-1 interactions that are required for neu-
macrophages phagocytose and destroy the bac- trophils to adhere to endothelial cells and trans-
teria. As mentioned above, phagocytosis is aided migrate into the skin (see also Box 2). The bac-
by opsonization with antibodies and the binding teria are also capable of preventing complement
of complement. Once bacteria are taken into the fixation through production of a Staphylococcus
phagosome, it fuses with the lysosome resulting complement inhibitor (SCIN), Efb, or through
in acidification, activation of proteases, and pro- Staphylokinase, an enzyme that promotes deg-
duction of reactive oxygen species, all of which radation of complement, antibodies, and clots.
are capable of breaking down the pathogens. As mentioned above, complement is important
This process also results in the generation of not only for phagocytosis of bacteria, but also
antigens, which can be presented on HLA II to for neutrophil chemotaxis. By inhibiting com-
www.perspectivesinmedicine.org

activate the adaptive immune response. During plement fixation, therefore, the bacteria are not
this process, some macrophages and DCs will only avoiding complement-mediated lysis and
migrate to the draining lymph node to activate opsonization, but also potentially reducing the
Tand B cells via the HLA II– antigen complexes number of neutrophils that are recruited to the
for initiation of the adaptive immune response. skin by the complement proteins C3a and C5a.
Activation of T cells and TH17 skewing seems S. aureus is capable of expressing nucleases,
to be particularly important for antibacterial which can cut neutrophilic NETs comprised of
immunity. This is attributable to the functions DNA discussed above, in addition to lipases and
of TH17-associated cytokines: IL-17, 22, and 23 proteases, which supports their dissemination.
can promote thickening of the epidermis, pro- Another way that S. aureus can avoid the
duction of antimicrobial peptides, and recruit- host immune response is by invading epithelial
ment of neutrophils, which further enhances and endothelial cells and living inside them in
bacterial clearance. a semi-dormant state. Intracellular pathogens
S. aureus has developed several mechanisms usually require cell-mediated immunity to clear
to avoid the host immune response. It can avoid infection. However, S. aureus has also developed
phagocytosis through the virulence factors Pro- mechanisms to evade T cell and other leukocyte
tein A (interferes with IgG binding to its FcgR), responses, namely through production of tox-
ClfA ( promotes coating with fibrinogen, which ins and superantigens. The a-toxin is capable of
outcompetes the binding of opsonins includ- inserting into host membranes and multimeriz-
ing complement), and capsule formation ( pro- ing to form a pore, much like the way comple-
motes biofilm formation via polysaccharide ment can form a pore in bacterial cell walls.
intercellular adhesion), which inhibits comple- Other toxins include the Panton – Valentine leu-
ment and antibody binding. It can also prevent kocidin (PVL), which can lyse leukocytes, and
phagolysosomal fusion via the virulence fac- the leukocidin D/E, leukocidin M/F, and the g-
tor SarA, thereby evading death and surviving hemolysin (Hlg), which can lyse erythrocytes
within the immune cells themselves. Inside the and leukocytes. Recurrent furunculosis and

Cite this article as Cold Spring Harb Perspect Med 2014;4:a015339 11


J.M. Richmond and J.E. Harris

other conditions have been associated with PVL machinery to manufacture progeny viruses. The
expression by S. aureus. S. aureus also produces HPV replication cycle is tied to keratinocyte dif-
one of the best-characterized superantigens, ferentiation, in which HPV early proteins (“E”
which nonspecifically activates T cells via strong 1 – 7) are produced in undifferentiated keratino-
binding of both the MHC II and the T-cell re- cytes and late proteins (“L” 1 and 2), which are
ceptor. Toxic shock syndrome toxin-1 (TSST-1, involved in capsid formation, are produced in
which can initiate tampon-associated toxic more superficial cells to promote sloughing of
shock syndrome) prevents the generation of virus. Some HPV strains can be transmitted sex-
normal T-cell responses against the bacterium ually and cause genital warts, whereas some can
and also affects the ability to generate antibody cause warts on other parts of the skin. Different
responses by B cells, which often need T cell help HPV strains are adapted to infect specific ana-
for appropriate class switching and activation. tomic locations. For example, strain 1 usually
In light of all of the ways in which S. aureus infects the soles of the feet, strain 2 the palms
can evade immune responses, it is easy to see of the hands, strains 6 and 11 are associated with
why drug-resistant strains pose a threat. Immu- genital warts, and strains 16 and 18 can cause
nocompromised patients are especially vulner- cervical cancer. Recently, two vaccines were de-
able (see Box 3), making it of utmost impor- veloped against HPV: Gardasil (types 6, 11, 16,
tance to understand skin immunity to bacteria. and 18) and Cervarix (types 16 and 18) in hopes
of preventing most cervical cancers (90% con-
tain HPV DNA).
Infectious Immunity: Part II. Examples
HPV produces proteins E6 and E7 that in-
of Viral Immunity
hibit host p53 and Rb, respectively, which nor-
Antiviral immunity largely depends on NK and mally are involved in sensing DNA damage
www.perspectivesinmedicine.org

CTL responses. Both NKs and CTLs kill their and repair mechanisms to halt cell-cycle progres-
targets via perforin/granzyme-induced apopto- sion. This is accomplished via ubiquitin-medi-
sis. The inflammatory response to most viral ated proteasomal degradation. P53 and Rb are
infections of the skin consists of minimal in- known as the “guardians of the genome” be-
flammation and symptoms caused by the target- cause the DNA damage-sensing mechanism is
ed destruction of virally infected cells and there- important for cells to maintain the health of the
fore usually lack redness, swelling, or pus that are genome and to avoid improper growth. Because
characteristic of bacterial or fungal infections. viruses need to use the host cell machinery to
TH1-type cytokines are important for antiviral replicate, overriding these proteins allows them
immunity because they drive CTL responses to replicate more efficiently. A byproduct of this
and antiviral mechanisms through production is the hyperproliferation of keratinocytes that
of IFN-g. We will discuss human papilloma vi- characterizes a wart. HPV can also avoid IFN-
rus (HPV) as an example of antiviral immunity mediated antiviral responses through E7, which
in the skin. HPV is the most common STD; it is can bind to and inhibit the promoters of type-I
estimated that 50% of sexually active men and IFN-related genes. Treatment with IFN-a can be
women get infected. The majority of people who used for genital warts, although patients with
contract HPV clear the infection within 2 yr, and higher E7 levels tend to respond less well than
not everyone that has HPV will develop cancer, patients with low E7. E6 can down-regulate IL-
as this is strain dependent. In this section, we will 18 expression, which is important for the gen-
highlight concepts of antiviral immunity and eration of TH1 and CTL responses.
ways in which HPV can subvert the host im- Productive antiviral immunity depends on
mune response. NK cell responses, CTL responses, and antibody
More than 70 different strains of HPV exist. production. NK cells look for the presence or
HPV belongs to the papillomaviridae family of absence of self HLA I on the surface of cells,
nonenveloped DNA viruses. HPV productively whereas CTLs respond to specific HLA I– pep-
infects keratinocytes, and takes over the cellular tide complexes. To avoid detection by CTLs,

12 Cite this article as Cold Spring Harb Perspect Med 2014;4:a015339


Skin Immunology

many viruses have developed the ability to fore lacks stimulation of APCs to mature, secrete
down-regulate HLA I. For example, the E5 pro- proinflammatory cytokines or promote effector
tein of HPV is capable of down-regulating HLA I T-cell activation. Several HPV proteins mimic
expression via inhibition of tapasin and HLA I host proteins and therefore can be tolerogenic.
promoter binding by transcription factors. Yet (Subversion of host immunity by HPV is nicely
even though the chance of activating a CTL is summarized by Tindle [2002].)
decreased by E5, an NK cell could still kill an
HPV-infected target cell. NK cells receive acti-
Infectious immunity: Part III. Examples
vating signals through killer activating receptors
of fungal and yeast immunity
(KARs), whereas inhibitory signals are transmit-
ted through killer inhibitory receptors (KIRs), Antifungal immunity relies heavily on the innate
which detect presence of HLA I on the cell’s immune system, and protective immunity de-
surface. Therefore, it is the balance of positive pends on both TH1 and TH17 responses as well
and negative signals that the NK cell receives that as antibody production. Like S. aureus, Candida
will determine whether or not it will kill the albicans colonizes much of the population,
target cell. However, not many NK cells are re- but is primarily pathogenic in immunocompro-
cruited to sites of HPV infection, as the virus mised patients, where disease incidence is 24
has devised other mechanisms to subvert in- cases per 100,000. Treatments for Candida are
flammatory responses. Part of this is because antifungal drugs, such as miconazole and flu-
of the sequestration of the virus inside primarily conazole, which inhibit formation of fungal cell
differentiated keratinocytes, which are located membranes. Several antifungal vaccines have
high in the epidermis. HPV induces the recruit- made it to phase I clinical trials, however achiev-
ment of CD4þCD25þFoxP3þ Tregs via CCL17 ing both efficacy and safety has proven difficult.
www.perspectivesinmedicine.org

and CCL22 production by LHC and macrophag- PRRs that are important for recognition of
es within the wart. Consistent with their func- Candida include TLRs 2, 4, and 6, as well as
tion discussed above, these Tregs can dampen lectin-like receptors including dectins, galectins,
inflammation and promote viral survival. In ad- DC-SIGN and the mannose receptor. All of these
dition, Tregs specific for HPVantigens have been PRRs recognize sugar and lipid components of
found in cervical cancer patients (see Box 6). the fungal cell wall, such as b-glucan, zymosan,
Antibody responses to viruses like HPV help and chitin. Complement receptor 3 (CR3) is
prevent initial infection or spread of infection important for recognizing opsonized Candida
via neutralization of binding of the virus to the and other pathogens. TLR-9, an intracellular
host cells. Yet the availability of HPV antigens PRR, is important for recognition of fungal
during infection is often low owing to the fact DNA, which is hypomethylated compared with
that, unlike other viruses, HPV does not induce mammalian DNA (recognition of Candida is
lysis of its target cells to allow for release of vi- summarized by Netea et al. 2008). Skin-resident
rions thereby producing free antigens. Addi- macrophages and DCs use these PRRs to detect
tionally, the L proteins are most immunogenic Candida, and initiate the recruitment of neutro-
but are primarily produced in superficial kera- phils and monocytes via cytokine and chemo-
tinocytes, thereby allowing the virus to avoid kine production. Recruited populations of neu-
detection by most LHC, which reside closer to trophils and monocytes phagocytose Candida
the basement membrane. Therefore, antibody and kill it via generation of reactive oxygen spe-
responses to HPV are often delayed and inade- cies. Extracellular killing is also induced, al-
quate. Some LHC and other APCs are able to though the mechanism for this is not yet known.
take up HPV antigens either through phagocy- As described above, skin-resident LHCs
tosis of cellular debris or possibly through exo- have been reported to initiate TH17 cell respons-
somes, which are nanometer-sized structures es. Interestingly, ligation of different PRRs
secreted by the cell. However, HPV infection is generates different TH profiles: ligation of the
not a highly inflammatory process, and there- mannose receptor usually results in IFN-g pro-

Cite this article as Cold Spring Harb Perspect Med 2014;4:a015339 13


J.M. Richmond and J.E. Harris

duction and TH1 responses, chitin recognition cuss skin immune responses to the hookworm
and fungal DNA recognition results in IL-4 and as an example of antiendoparasite immunity
IL-13 production and TH2 responses, and hy- (see Box 7 for a note on ectoparasites). Hook-
phae recognition by dectin 1 results in IL-17 pro- worms are nematodes that belong to the fam-
duction and TH17 responses. Treg responses can ily Ancylostomatidae, and infect approximately
also be generated through tolerogenic DC pop- 20% of the world population. The larvae burrow
ulations and TRIF-dependent signaling down- into the skin, often through the feet that come
stream of some PRRs. Most of these associations into contact with contaminated soil or water.
have been determined using PRR knockout As they mature, they travel through the blood
mouse models of Candidiasis. However, little to the lungs and eventually the intestines, where
is known about how these mixed T-cell respons- they feed off of blood and reproduce. Hook-
es ultimately influence the host’s ability to clear worm eggs are secreted in feces, and embryos
the pathogen, although it seems that early gen- develop and hatch in the soil or water where
eration of Treg responses allows Candida to sub- they consume bacteria until they develop into
vert host immunity. It is possible that confusing the infective worm stage, L3, thus completing
the immune system by inducing these mixed T- the life cycle (reviewed by Loukas and Prociv
cell responses also dampens host immunity ow- 2001).
ing to a lack of positive-feedback loops. One On entering the skin, L3 hookworm larvae
possible mechanism for this differential induc- shed their outer cuticle and begin expressing
tion of immune responses would be the mor- enzymes that permit their movement through
phological changes that occur when Candida tissues. Cuticle or sheath antigens can be taken
transitions from yeast to hyphae, which results up by APCs and presented to T and B cells.
in differences in bioavailability of PAMPs from Initially following skin invasion, zoonotic spe-
www.perspectivesinmedicine.org

the cell wall. Understanding this modulation of cies elicit inflammatory responses that cause
DCs by different fungal epitopes is important a creeping eruption or ground itch, whereas
for the design of effective antifungal vaccines. anthropophilic species can do so silently. Anti-
body responses have been detected to the cuticle
as well as the exsheathing fluid, and it has been
Infectious Immunity: Part IV. Examples
suggested that this allows the worm to misdirect
of Parasite Immunity
the immune response, similar to countermea-
Immunityagainst parasites is mediated bya mix- sure decoys to distract a heat-seeking missile.
ture of innate and adaptive immune responses Antibody responses to fluid and cuticle proteins
that rely on IgE, granulocytes, and TH2 cells. may be used clinically as a diagnostic tool for
Parasites present challenges to the immune sys- detecting infection. Although T-cell responses
tem because of several factors, including their seem to be weak and their exact specificities are
size and complexity, migration to different tis- unknown, TH2-cell responses against hook-
sues within the body, and ability to subvert the worms result in production of IL-4 and IL-13,
host immune response. These immune evasion which induce B-cell class switching to IgE. In-
mechanisms have made it notoriously difficult terleukin-5 (IL-5), a major eosinophil survival
to develop vaccines against parasites. We will dis- and activation factor, is also produced by TH2

BOX 7. ARTHROPOD BITES AND IMMUNITY TO VECTOR-BORNE DISEASES

Many insects have specialized salivary proteins, which allow them to consume a blood meal without
inducing coagulation, and some have developed proteins that inhibit immune responses. Research is
being conducted to examine the effects of an insect bite on cytokine and chemokine production in
the skin.

14 Cite this article as Cold Spring Harb Perspect Med 2014;4:a015339


Skin Immunology

cells. Eosinophils are recruited to sites of infec- Improper Immune Responses in Disease
tion and are major players in antiparasite im-
Improper Immune Responses: Part I. Allergy
munity. They bind IgE-opsonized larvae via the
Fc1R, causing them to degranulate. Enzymes, Allergy is an improper immune response to an
and the reactive oxygen species that they release, otherwise innocuous antigen. The incidence of
degrade larvae. Once the larvae matures into an allergic disease is rising in developed countries,
adult worm in the intestine, immune responses and treatments cost $400 million annually in the
seem to become elevated most likely in response United States alone (see Box 8). Allergic contact
to the greater availability of worm antigens. dermatitis occurs following chemical or envi-
Coinciding with this, peripheral eosinophilia ronmental exposure, resulting in generation of
is often seen in patients with hookworm infec- neoantigens via haptenization of self-molecules
tions. Tissue-resident mast cells also degranu- (reviewed by Kaplan et al. 2012). Haptens them-
late on encountering a larvae or worm, which selves can cause oxidative stress in keratino-
may result in the recruitment of more eosino- cytes, resulting in release of reactive oxygen
phils. Their mechanism of activation is slightly species and danger signals, such as ATP. Neo-
different from that of eosinophils in that they antigens produced in response to hapten ex-
may have IgE preloaded in their Fc1Rs. IL-9 posure, such as byproducts of hyaluronic acid
seems to be important for mast-cell activation degradation, can activate TLRs, resulting in pro-
and production of proteases. duction of proinflammatory cytokines by skin-
In addition to misdirecting antibody re- resident immune cells. Studies in mice have indi-
sponses to their cuticle, hookworms have de- cated that recognition of neoantigens is mediated
veloped several other mechanisms to subvert by TLR-2 and/or TLR-4 in IL-12-dependent and
the host immune response. They produce a neu- -independent mechanisms. It is also thought that
www.perspectivesinmedicine.org

trophil inhibitory factor protein (NIF), which NLRs and the inflammasome are capable of
interferes with neutrophil recognition of opson- recognizing haptens and neoantigens, although
ized parasites; C-type lectins that mimic those many ligand-receptor partners have yet to be de-
of the host, which interfere with immune re- termined. Repeated exposure induces sensitiza-
cognition and coagulation; metalloproteinases tion and subsequent type IV hypersensitivity
and peptides that interfere with coagulation and (see Box 9). This response is characterized by an
permit feeding; cysteine proteases that cleave influx of T cells to the skin, resulting in tissue
Igs and the low-affinity IgE receptor; and aspar- damage and an inflammatory eruption.
tic proteases, which help them digest hemoglo- Tissue penetration of haptens is crucial for
bin but also may be involved in cleaving Igs and induction of allergic contact dermatitis. Close
complement. They also produce protease in- to 3000 compounds have been discovered that
hibitors and antioxidants that help them with- are capable of inducing contact dermatitis.
stand the effects of degranulation by eosinophils Small molecular weight compounds can diffuse
and mast cells. Hookworms also produce ace- through the epidermis, although disruption of
tylcholinesterases, which are thought to both the barrier function of keratinocytes is thought
inhibit gut peristalsis and interfere with im- to speed up the sensitization process (see Box
mune function. 1). Examples of compounds that can elicit al-

BOX 8. HYGIENE HYPOTHESIS

The hygiene hypothesis, which was first proposed by Strachan in 1989, states that exposure to
pathogens in childhood helps to develop normal, balanced immune responses. Growing up in
ultraclean environments, therefore, results in the failure to appropriately modulate Th2 responses
in childhood, thereby creating a predisposition to allergic immune responses.

Cite this article as Cold Spring Harb Perspect Med 2014;4:a015339 15


J.M. Richmond and J.E. Harris

BOX 9. HYPERSENSITIVITY REACTIONS

† Type I hypersensitivity—immediate hypersensitivity (15 –30 min, but can be delayed up to 10–
12 h); IgE-mediated, involves mast cells, basophils, and eosinophils (e.g., asthma).
† Type II hypersensitivity—cytotoxic hypersensitivity (min-h); IgG and IgM antibody plus comple-
ment-mediated (e.g., blood group incompatibility).
† Type III hypersensitivity—immune-complex hypersensitivity (3–10 h); circulating IgG com-
plexed with antigen deposits on basement membranes (e.g., arthus reaction/serum sickness).
† Type IV hypersensitivity—delayed type hypersensitivity ( peaks at 48 h); memory T-cell mediated
(e.g., contact dermatitis).

lergic contact dermatitis include nickel, which is and, potentially, organ failure. There are check-
the most common contact allergen and can points to prevent the immune system from tar-
bind directly to TLR-4 (see Box 10 and Schmidt geting self-tissues; and, therefore, autoimmune
et al. 2010), and the dust mite allergens Der p 2 disease is believed to require multiple hits that
and Der f 2, which are homologs of the en- deactivate those checkpoints (similar to the
dogenous TLR-4-binding protein MD2. Dust multihit hypothesis of cancer development).
mites, or Dermatophagoides subspecies, con- These mechanisms include central tolerance
sume sloughed keratinocytes, and are often through deletion of autoreactive T and B cells
found in clothing, bedding, and on the skin. in the thymus (see Box 11), peripheral tolerance
www.perspectivesinmedicine.org

Dust mites often cause allergic responses attrib- through the action of CD25þ Tregs (see Box
utable to the type of immune responses they 4), production of anti-inflammatory cytokines,
elicit: IgG and IgE responses confer immunity such as IL-10 and TGF-b, and down-modula-
but also hypersensitivity reactions. Tissue-resi- tion of proinflammatory cytokine production
dent mast cells also play a role in skin allergy, by ligation of certain PRRs on innate and tis-
caused by their ability to degranulate after their sue-resident immune cells, such as the phospha-
surface-loaded IgE is cross-linked on an aller- tidyl serine receptor on macrophages, which
gen encounter. Whereas systemic and topical aids in uptake of apoptotic cells in a physiologic
steroids are sometimes used to manage acute context. Because autoimmunity is a destructive
symptoms, the preferred treatment for contact process and does not appear to be beneficial for
dermatitis is avoidance of the allergen. survival, these responses likely evolved as over-
zealous anti-infectious or antitumor responses.
For example, Japanese have a very low incidence
Improper Immune Responses: Part II.
of psoriasis compared with U.S. or European
Autoimmunity
populations, whereas their risk for tuberculosis
Autoimmunity results when the immune sys- is much higher, suggesting that their genetic
tem targets self-tissues, resulting in destruction, makeup puts them at low risk for psoriasis but

BOX 10. NICKEL ALLERGY

One of the most common triggers of contact hypersensitivity is nickel, which is present in jewelry,
orthopedic materials, and coins. Schmidt et al. (2010) showed that nickel is an inorganic ligand for the
TLR-4–MD2 complex. On binding to histidine residues in human TLR-4, nickel can induce cross-
linking of the receptor that induces a proinflammatory signal via the MyD88 adapter protein. This
leads to subsequent cytokine production and elicitation of contact hypersensitivity.

16 Cite this article as Cold Spring Harb Perspect Med 2014;4:a015339


Skin Immunology

BOX 11. APECED SYNDROME

Autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED) syndrome results


from a genetic disruption of the transcription factor autoimmune regulator (AIRE). AIRE is expressed
by specialized cells in the thymus and induces the expression of tissue-specific self-antigens from
peripheral tissues to allow for negative selection of autoreactive T cells as they develop. Without
AIRE, patients are unable to delete autoreactive T cells, resulting in development of multiple auto-
immune diseases including autoimmune skin diseases, such as alopecia, vitiligo, nail dystrophy, and
enamel hypoplasia.

high risk for tuberculosis. IFIT1 is a component Keratinocytes produce significant levels of an-
of interferon signaling, and is a risk allele for timicrobial peptides, and the cytokines IL-23,
type I diabetes, but the high-risk allele for dia- IL-17, IL-6, and IL-22 appear to play a promi-
betes is protective against coxsackie viral infec- nent role in pathogenesis. These characteristics
tion, suggesting that this allele evolved to pro- mirror those seen in an antibacterial response
tect from infection. Native Americans were and, in fact, lesions of psoriasis rarely become
devastated by tuberculosis infection on arrival superinfected, unlike in atopic dermatitis. This
of Europeans—the descendants of those who resistance to infection is probably caused by this
survived now have a very high risk for rheuma- overzealous antibacterial-like response.
toid arthritis, suggesting that tuberculosis sur- Treatment for psoriasis includes local, gene-
vivors possessed aggressive immune responses ral immunosuppressive medications like topical
www.perspectivesinmedicine.org

that now promote autoimmunity. These exam- steroids and calcineurin inhibitors, systemic ge-
ples support the hypothesis that autoimmu- neral immunosuppressants like cyclosporine A
nity developed as a consequence of evolving and methotrexate, as well as newer, more target-
potent anti-infectious responses. Therefore, we ed systemic biologic medications like TNF-a
will categorize autoimmune responses accord- inhibitors, p40 (a component of IL-23) inhibi-
ingly. In addition to improving our understand- tors, and IL-17 inhibitors. Although these treat-
ing of pathogenesis, an advantage to thinking ments are likely to have improved safety profiles
about autoimmunity in this way is that treat- over more general immunosuppressants, they
ments used to interfere with autoimmunity are still have rare, but predictable, side effects based
likely to increase the risk for infections con- on their ability to interfere with the antibacteri-
trolled by that response. al response, including an increased incidence of
In this section, we will discuss both T-cell- bacterial and fungal infections. A more detailed
driven autoimmune disease and B-cell-driven discussion concerning psoriasis and its treat-
autoantibody-mediated disease in the context ments is addressed in the literature.
of improper antiviral/tumor, or antibacterial/
fungal immune responses. Antiviral-Like Autoimmunity
Vitiligo is an autoimmune disease of the skin
Antibacterial-Like Autoimmunity
that results in the loss of melanocytes from the
Psoriasis, which afflicts 2% – 3% of the world epidermis, which can be quite psychologically
population, is a highly inflammatory disease devastating for patients. It affects 0.5% of the
of the skin that presents with pruritus, pain, population worldwide, without preference for
erythema, occasional pustules, and thickened race or gender, and targets all areas of the skin,
scales. Histologically, it is characterized by epi- with a preference for the face and genitals great-
dermal acanthosis, vascular proliferation, and a er than hands and feet greater than trunk and
significant inflammatory infiltrate consisting of proximal extremities. The patchy depigmenta-
neutrophils, T cells, DCs, and other populations. tion is typically symmetrical except in the case of

Cite this article as Cold Spring Harb Perspect Med 2014;4:a015339 17


J.M. Richmond and J.E. Harris

segmental vitiligo, in which unilateral depig- go using anti-TNF-a inhibitors have been disap-
mentation typically remains localized and lim- pointing, and no other systemic therapies for
ited by the midline. Segmental vitiligo affects vitiligo are widely available. Typical treatments
5% of all vitiligo patients but is responsible for vitiligo include topical steroids and calci-
for a larger component of childhood vitiligo, neurin inhibitors, as well as narrow-band UVB
affecting 20%. Patients with vitiligo are typi- light therapy. New approaches to treatment will
cally asymptomatic, with only 20% complain- likely benefit from targeting IFN-g and other
ing of mild itching in lesional skin. components of the TH1 inflammatory pathway.
On physical exam, there is typically no ery-
thema or scale, signs seen only in the rare in-
Antibody-Mediated Autoimmunity
flammatory subtype. Histological examination
may reveal a subtle infiltrate made up of CD4þ The skin is affected by a number of autoimmune
and CD8þ T cells in the absence of neutrophils diseases that are clearly mediated by antibodies.
or epidermal proliferation. Often, lesional skin These diseases can be subdivided according to
contains few T cells. Because keratinocytes re- whether they are tissue specific, or tissue non-
tain the pigment that they acquire from mela- specific. Tissue-specific diseases are relative-
nocytes as they differentiate through the layers ly straightforward, as the antibodies interfere
of the epidermis until they are sloughed off, with protein –protein binding, and the symp-
melanocyte destruction does not result in visi- toms develop from a lack of protein func-
ble depigmentation until complete turnover of tion. Tissue-specific antibody-mediated auto-
the overlying keratinocytes, a process that re- immune diseases of the skin include pemphigus
quires 14– 48 d. During this time, destructive vulgaris, pemphigus foliaceus, bullous pemphi-
T cells likely migrate laterally through the skin goid, epidermolysis bullosa acquisita, linear
www.perspectivesinmedicine.org

to perpetuate disease, and therefore are not IgA, and others. Pemphigus vulgaris is mediated
present once the depigmented epidermis be- by antibody production against desmoglein 3,
comes clinically visible. Thus, biopsy recommen- which is predominantly expressed in the basal
dations to “catch” an immune infiltrate include epidermis and acts as a glue to hold keratino-
selecting perilesional skin up to 1 cm beyond cytes together. The result is separation of the
the visible border, a slightly hyperpigmented epidermis just superior to the basal keratinocyte
region beyond the border, or better, an elliptical layer, which then accumulates fluid and forms a
biopsy radiating outward from the lesion, in- bulla on the surface of the skin. In contrast, pem-
cluding the border itself and up to 1 cm beyond. phigus foliaceus is mediated by desmoglein 1
Vitiligo is mediated by CTLs and IFN-g antibodies, forming bullae in the superficial lay-
production, reflecting a TH1 immune response ers of the epidermis and mucosae, because
within the skin. Unlike some autoimmune dis- of predominant expression of desmoglein 1 at
eases where it has been difficult to determine that location. Because these diseases are primar-
which antigens the autoreactive T cells are re- ily antibody mediated, they require little contri-
sponding to, the TCR specificities in vitiligo bution from immune cells during the effector
have been determined for a number of peptides, phase apart from the plasma cells that produce
including MART-1/Melan-A (melanoma anti- the antibodies. As mentioned above, despite be-
gen recognized by T cells), tyrosinase (an enzyme ing a B-cell-driven autoimmune disease of the
required for melanin production), and gp100 skin, B cells are not typically found within the
(also known as premelanosome protein, a trans- skin, but reside within the lymph nodes and
membrane protein expressed on the surface of bone marrow, secreting antibody into the blood
pigment-producing cells in the skin and eye). stream, which is then delivered passively to the
This clinical, histological, and mechanistic pic- skin and sterically disrupts protein – protein
ture is similar towhat is seen in response to aviral binding. Consequently, there is little inflamma-
infection or antitumor response, in contrast to tion present within lesional skin, and topical
psoriasis. Consequently, attempts to treat vitili- immunosuppression has limited efficacy. Treat-

18 Cite this article as Cold Spring Harb Perspect Med 2014;4:a015339


Skin Immunology

ment is aimed at systemic general immunosup- self-peptides on HLA I that can be recognized
pression, including prednisone, mycophenylate by CTLs. For in-depth discussions of malignan-
mofetil, azathioprine, cyclophosphamide, and/ cies of the skin, please refer to the chapters cover-
or cyclosporine, which ultimately results in de- ing melanoma and cutaneous T-cell lymphoma.
creased antibody production. Recent studies
suggest that rituximab, an antibody that targets
B cells but not plasma cells, may be an effective CONCLUDING REMARKS
treatment, so it is not yet clear exactly how it Skin structural, stromal, and hematopoietic
modulates this plasma cell-driven disease. cells are crucial for protective immunity to a
Tissue nonspecific antibody-mediated au- multitude of pathogens. Genetic susceptibility
toimmune diseases are best characterized by or subversion of the host immune response can
the connective tissue diseases. Lupus erythema- lead to chronic, continued infections. Similarly,
tosus is probably the best understood of these genetic susceptibility in addition to environ-
diseases, yet mechanisms of pathogenesis are far mental triggers can lead to development of con-
from clear. Autoantibodies in lupus appear to ditions like allergy and autoimmunity. Under-
target self-proteins that are not limited to a spe- standing the relationships between the cells and
cific tissue, unlike the tissue-specific diseases. proteins that confer protective immunity in the
Common targets include DNA, RNA, and their skin will ultimately shed light on new potential
associated proteins. It is likely that necrotic cell treatments for a myriad of diseases.
death initiates the inflammatory response fol-
lowing release of nuclear contents, a process that
is prevented during programmed cell death, ACKNOWLEDGMENTS
or apoptosis. Sun exposure may contribute to
www.perspectivesinmedicine.org

pathogenesis through damage of keratinocytes J.E.H. is supported by National Institute of Ar-


and subsequent release of nuclear contents. thritis and Musculoskeletal and Skin Diseases,
Antibody – protein complexes form within the part of the National Institutes of Health, under
vasculature and precipitate onto small vessel en- Award Number AR061437 and research grants
dothelium in specific organs, including the skin, from the Charles H. Hood Foundation, Vitiligo
kidney, joints, and others. Therefore, inflamma- Research Foundation, and Dermatology Foun-
tion may appear in any of these organs, resulting dation. Conflicts of interest: J.E.H. has received
in a wide variety in clinical presentation and grant support from Combe Inc., Abbvie, and
symptoms, even within the same individual. Re- Sanofi-Genzyme.
cent data suggest that nuclear components act as
self-DAMPs (see above) and initiate immune
REFERENCES
responses through TLRs and NLRs within the
tissues. More research will be required to under- Chen L, Guo S, Ranzer MJ, Dipietro LA. 2013. Toll-like
stand the detailed pathogenesis of these complex receptor 4 has an essential role in early skin wound heal-
ing. J Invest Dermatol 133: 258– 267.
systemic diseases. Clark RA. 2010. Skin-resident T cells: The ups and downs of
on site immunity. J Invest Dermatol 130: 362 –370.
Foster TJ. 2005. Immune evasion by staphylococci. Nat Rev
Malignancy Microbiol 3: 948–958.
Proper immune responses to combat malig- Kaplan DH. 2010. In vivo function of Langerhans
cells and dermal dendritic cells. Trends Immunol 31:
nancy are similar to antiviral responses, often 446 –451.
involving NK cells and CTLs, which are capable Kaplan DH, Igyarto BZ, Gaspari AA. 2012. Early immune
of killing altered-self cells. Often, tumors down- events in the induction of allergic contact dermatitis. Nat
Rev Immunol 12: 114 –124.
regulate HLA I expression and avoid CTL-me-
Loukas A, Prociv P. 2001. Immune responses in hookworm
diate killing, yet they then become susceptible infections. Clin Microbiol Rev 14: 689– 703.
to NK cells. They are also often stressed because Netea MG, Brown GD, Kullberg BJ, Gow NA. 2008. An
of hyperproliferation, and can present altered integrated model of the recognition of Candida albicans

Cite this article as Cold Spring Harb Perspect Med 2014;4:a015339 19


J.M. Richmond and J.E. Harris

by the innate immune system. Nat Rev Microbiol 6: Schmidt M, Raghavan B, Muller V, Vogl T, Fejer G,
67–78. Tchaptchet S, Keck S, Kalis C, Nielsen PJ, Galanos C, et
Ramirez-Ortiz ZG, Lee CK, Wang JP, Boon L, Specht CA, al. 2010. Crucial role for human Toll-like receptor 4 in the
Levitz SM. 2011. A nonredundant role for plasmacytoid development of contact allergy to nickel. Nat Immunol
dendritic cells in host defense against the human fungal 11: 814– 819.
pathogen Aspergillus fumigatus. Cell Host Microbe 9: Seneschal J, Clark RA, Gehad A, Baecher-Allan CM, Kupper
415–424. TS. 2012. Human epidermal Langerhans cells maintain
Rosas-Ballina M, Olofsson PS, Ochani M, Valdes-Ferrer SI, immune homeostasis in skin by activating skin-resident
Levine YA, Reardon C, Tusche MW, Pavlov VA, Andersson regulatory T cells. Immunity 36: 873– 884.
U, Chavan S, et al. 2011. Acetylcholine-synthesizing T Tindle RW. 2002. Immune evasion in human papillo-
cells relay neural signals in a vagus nerve circuit. Science mavirus-associated cervical cancer. Nat Rev Cancer 2:
334: 98–101. 59– 65.
www.perspectivesinmedicine.org

20 Cite this article as Cold Spring Harb Perspect Med 2014;4:a015339

View publication stats

Das könnte Ihnen auch gefallen