You are on page 1of 7

Available online at www.sciencedirect.

com

Immune responses in the skin in old age


Milica Vukmanovic-Stejic1, Malcolm HA Rustin2, Janko Nikolich-Zugich3
and Arne N Akbar1

A marked increase in the susceptibility to cutaneous infections contributes to the increase in cutaneous malignancies
and malignancies has been observed in older humans [2,3]. This indicates that defective cutaneous immunity
indicating that cutaneous immunity becomes defective with develops during ageing and the purpose of this article to
age. In this review we will focus on recent developments in the assess recent data that clarify the nature of the defect
understanding of age-related changes in immune function of that occurs.
the skin with a particular emphasis on how alterations in the
interaction between cells involved in innate and adaptive The investigation of antigen-specific T cell
immunity leads to decreased cutaneous antigen-specific T cell responses in the skin
immunosurveillance. Since the induction and amplification of cutaneous immu-
Addresses nity is an integrated multistep process, in vivo experimen-
1
Division of Infection and Immunity, Department of Immunology, tal models are the most informative. A particularly useful
University College London, London W1T 4JF, United Kingdom experimental system is the delayed type hypersensitivity
2
Department of Dermatology, Royal Free Hospital, London NW3 2QG,
United Kingdom
(DTH) response, where recall antigens are injected intra-
3
Department of Immunobiology and the Arizona Center on Aging, dermally [4–6]. This is a classical model of an antigen-
University of Arizona College of Medicine, Tucson, AZ 85719, USA specific memory T cell (secondary) response since indi-
viduals (mice or humans) who have not been exposed to
Corresponding author: Akbar, Arne N (a.akbar@ucl.ac.uk)
the antigen do not respond to the antigen challenge [4–7].
The level of DTH reactions in humans is determined by
Current Opinion in Immunology 2011, 23:525–531 the diameter of cutaneous induration and the extent of
This review comes from a themed issue on
erythema at the site of antigen injection at 48 h after
Immune Senescence antigen injection or in mice by the amount of ear or
Edited by Beatrix Grubeck-Loebenstein and John Cambier footpad swelling at 24 h [4–7]. There are some differ-
ences in the kinetics and nature of the response in mice
Available online 22nd June 2011
and humans, possibly due to the different site of antigen
0952-7915/$ – see front matter injection nevertheless, broadly speaking, the cellular
# 2011 Elsevier Ltd. All rights reserved. infiltrates are similar in both species [4–7]. While other
experimental models for the study of cutaneous immu-
DOI 10.1016/j.coi.2011.05.008
nity in mice exist [8,9], in humans, the DTH response
provides an ethically acceptable model to study the
kinetics and regulation of a memory T cell response in
Skin and immunity vivo. In subsequent sections we will mainly discuss the
The skin forms the body’s largest interface with the human DTH response and how it has been used to study
environment and its principal function is that of a barrier. changes in cutaneous immunity during ageing.
It protects the organism by being impermeable to a multi-
tude of harmful exogenous substances and maintains From a histological perspective, antigen exposure and
internal homeostasis by preventing excessive water and trauma to the skin induce non-specific danger signals to
heat loss. In addition there is a highly specialised immune recruit and activate cells of the innate immune system
system consisting of leukocytes that are resident, recruited and at very early time points (4–6 h) the majority of
or re-circulate within the tissue (Table 1). These cells are infiltrating cells are neutrophils [5]. This cellular infiltra-
distributed in the epidermal and dermal layers of the skin tion is dependent on the production of pro-inflammatory
and participate in both adaptive and innate immune cytokines such as IFN-g and TNFa that stimulate
responses. They are also responsible for distinguishing expression of adhesion molecules on the endothelium
self from non-self which is of fundamental importance and increase permeability of the local blood vessel [10].
since the skin comes into daily contact with exogenous E-selectin is induced on capillary endothelium as early as
substances. Close interlinking between innate and adap- 1–2 h after injection and by 12 h adhesion molecules
tive pathways of immunity plays an important role in the ICAM-1 and VCAM-1 are also expressed [7]. These
initiation and amplification of immune responses in this molecules interact with LFA-1 and VLA-4 on monocytes
tissue. However, there is a decrease in cutaneous immune and lymphocytes allowing the accumulation of these cells
function in older humans that leads to increased bacterial in the dermis [7,11]. In parallel with the endothelial
(such as Streptococus and Staphylococus induced celluli- activation and conditioning at the site of antigen
tis) and fungal infections (often candidaiasis) [1] and exposure, antigen presenting cells from the skin transport

www.sciencedirect.com Current Opinion in Immunology 2011, 23:525–531


526 Immune Senescence

Table 1

Cells of the adaptive and innate immune system in the skin

Resident Recruited Recirculating


Innate Keratinocytes Monocytes Natural Killer Cells
Endothelial cells Granulocytes Dendritic cells
-Vascular -Basophilic
-Lymphatic -Eosinophilic
Dendritic cells -Neutrophilic
Mast cells Mast cells ?Promonocytes
Tissue macrophages Epitheloid cells
Adaptive T lymphocytes T lymphocytes T lymphocytes
B lymphocytes

? indicates that there is a possibility, in this case, rather than strong evidence.

antigen to the lymph node where they present it to and receptors (TLR) are one of the most important transdu-
activate memory T cells that subsequently migrate via cers of ‘danger signals’ [20], resulting from the presence of
blood into the site of inflammation in the skin [10]. The antigen and trauma to the skin. TLRs are expressed by
infiltration of T cells during a DTH response is biphasic, various cells of the innate immune systems including
comprising an early non-specific infiltration of these cells monocytes, macrophages (MF), dendritic cells (DC)
that appear around dermal blood vessels approximately and Langerhans cells (LC) as well as keratinocytes.
12 h after challenge [7] and a later peak of antigen- Evidence in humans and mice suggest that TLR expres-
specific T cell accumulation [4,5] that may be partly sion or TLR induced production of inflammatory
due to the proliferation of these cells in the skin mediators by cells of the innate immune system is
[12,13]. Maximal numbers of macrophages are present reduced during ageing (reviewed in [22]). Furthermore
at 24 h but by 48 h the majority of infiltrating cells are T a recent study demonstrated the decreased production of
cells [5,7,14]. pro-inflammatory cytokines such as TNF-a, IL-6 and IL-
12 from circulating DCs in old individuals stimulated
Recently elegant studies of cutaneous infection with with various TLR ligands [23]. It is possible therefore that
HSV-1 have suggested that HSV-specific cells that have one reason for defective cutaneous immunity during
migrated to the site of Ag exposure, remain resident in the ageing may be related to defective conditioning of the
skin following Ag-clearance [15,16]. These long-lived skin environment by innate cells resulting from defective
skin resident cells provide a rapid response to subsequent TLR signalling.
re-exposure to the same pathogen, presented by tissue
resident DC and could contribute to the initial steps of Skin APC and ageing
activation and to the initiation of the inflammatory cas- The main components of the innate immune system in
cade [15,16]. the skin are Langerhans cells, dermal dendritic cells and
skin resident macrophages. These antigen presenting
Older humans and mice have been shown to have defec- cells produce inflammatory mediators in response to
tive DTH responses [17–19]. At first glance this appears TLR signalling, present antigen and provide co-stimu-
to suggest that this may reflect the development of lation to T cells both in the skin and in the draining lymph
defective immune memory to antigen during ageing. node. A number of recent reviews have covered in detail
However as will be discussed later, the decreased mani- changes in the innate immune cells that occur with ageing
festation of antigen-specific cutaneous immunity is actu- [24,25], this section highlights some of the recent studies
ally due to altered coordination between innate and that are particularly relevant to the antigen presenting
acquired arms of the cutaneous immune system in older cells in the skin.
subjects [11]. In order to fully appreciate the implica-
tions of this, it is first necessary to consider the data In general, the number and phenotype of cutaneous DCs
available on defects of individual leukocyte populations are comparable in young and old subjects during ageing
that accumulate during ageing. however migration, phagocytosis and capacity to stimu-
late T cells may be decreased [24–26]. A study using a
Toll-like receptors and ageing mouse melanoma model by Grolleau-Julius et al. found
The endothelial activation and conditioning of the skin that aged DC have impaired capacity to migrate from the
environment at the site of antigenic challenge, which periphery into the LN despite expressing equivalent
enables the recruitment of leukocytes from the blood, is levels of CCR7 as young cells [27]. However even when
considered to be due to the secretion of proinflammatory the defective migration was corrected these aged DC
cytokines by cells of the innate immune system that are in were less efficient at inducing anti-tumour immunity
turn activated by ‘danger signals’ [10,20,21]. Toll like suggesting an additional functional defect that correlated

Current Opinion in Immunology 2011, 23:525–531 www.sciencedirect.com


Skin and old age Vukmanovic-Stejic et al. 527

with a selective reduced DC-SIGN expression [27]. Sim- Cutaneous T cells during ageing
ilarly human DC from elderly individuals showed Normal, healthy human skin contains large numbers of
impaired migration towards MIP-1b despite equivalent antigen experienced CD4+ and CD8+ T cells [42,43].
CCR7 expression [28]. According to some calculations there are approximately
20 billion skin-resident T cells in healthy human skin,
Plasmacytoid dendritic cells (PDC) are a unique dendritic nearly twice the number present in circulation [43].
cell subset, which is present in the normal skin in very small Schierli et al. suggested that the majority of human T
numbers. However, they play an important role in viral cells in healthy skin express CCR8 and act as immune
infection (for example VZV) and inflammatory skin dis- surveillance cells, which recognise peptide–MHC com-
orders (such as psoriasis, lupus erythematosus and lichen plexes on epidermal APC and trigger an immune defense
planus) [29]. PDCs have also been described in skin program involving monocytes, granulocytes, inflamma-
tumours (melanoma, BCC, SCC, CTCL) [29]. Skin injury tory T cells and antibodies [42]. In this scheme activated
(by tape striping or by chemical treatment) also results in immune surveillance T cells play a dual role. Through
rapid recruitment of PDCs [30] and in rapid and transient the secretion of TNF-a and IFN-g after antigen chal-
secretion of type I interferon that promotes wound healing lenge, they initiate inflammatory cascades that lead to the
[31]. In contrast to classical DCs, there is clear evidence recruitment of other leucocytes. Secondly, these cells
that PDC cytokine secretion is reduced with age [23,32]. may migrate to the draining LN where they may pro-
Jing et al. also observed that circulating PDC number and liferate themselves or accelerate the activation of other
IFN-a production decreased in older subjects [33]. The antigen-specific T and B cell responses. It is possible
evidence that older people often have problems with VZV that one of the reasons for impaired cutaneous immunity
reactivation, causing shingles, supports the possibility of in the elderly may be due to changes in the skin resident
the impaired function of PDC in the skin, either through T cells.
decreased migration, activation or cytokine secretion.
Repeated antigenic stimulation throughout life may com-
A number of macrophage functions decline with ageing, promise antigen-specific T cells in two ways [44]. First,
including diminished TLR expression (or reduced cyto- they may become functionally exhausted and lose essen-
kine secretion in response to TLR stimulation), reduced tial functional activity that is necessary for immune
phagocytic capacity and a decline in secretion of chemo- protection [45,46]. Functional exhaustion is a way of
kines and cytokines (reviewed in [24]). In the DTH limiting the magnitude of effector T cell responses and
model, there is decreased ability of macrophages in the is often associated with the increased expression of sur-
antigen-challenged skin of older humans to secrete face inhibitory receptors. Although this may safeguard
inflammatory cytokines such as TNF-a following antigen against autoimmune responses, it may also compromise
challenge [11]. effective immunity against infectious agents and tumours
[47]. Second, repeated T cell stimulation can lead to a loss
Langerhans cells are myeloid-derived dendritic cells that of replicative capacity of some antigen-specific T cell
reside in the epidermis, in close contact with keratino- populations as a result of telomere erosion and/or unre-
cytes. Until recently LC were considered to be the main paired DNA damage (a process known as replicative
antigen presenting cell in the skin, responsible for pro- senescence) [48,49]. Interestingly, accelerated telomere
moting immune response to invading pathogens [10]. erosion has been observed during immune response in
The classical view is that LC capture and process the skin, because of inhibition of the enzyme telomerase,
cutaneous antigens then migrate into the LN where they by type I interferon [12]. It is not clear at present if skin
present the antigen to T cells [10]. More recently it was resident T cells, or those that are recruited during anti-
reported that Langerhans cells play a crucial role in the genic challenge, may be compromised by either exhaus-
induction of IL-22 secreting T cells (Th22) [34,35] that tion or senescence in older humans and this requires
play an important role in cutaneous immune responses. further investigation.
Other studies have suggested that LC play an important
role in downregulating immune responses [36]. For Regulatory T cells accumulate in the skin
example, Langerhans cells are required for UV-radiation during ageing
induced suppression, and this immunological unrespon- Regulatory T cells play a crucial role in regulating the
siveness is mediated through the induction of Tregs [37– magnitude of an immune response. We and others have
39]. The ability of LC to migrate to draining lymph nodes shown that 5–10% of T cells resident in normal human
has been shown to decline with age [40,41]. Collectively skin express Foxp3 and have other characteristics of
these results suggest that a wide range of defects may Tregs [11,13,50]. These regulatory cells also proliferate
occur in innate populations of leucocytes in the skin in the skin during the course of a DTH response [13]. A
during ageing but it is not clear how this may lead to recent study has shown that Tregs (as well as responder T
the susceptibility of older humans or mice to skin infec- cells) circulate between the skin and LNs and vice versa,
tions or malignancy. in the steady state and during an immune response [9].

www.sciencedirect.com Current Opinion in Immunology 2011, 23:525–531


528 Immune Senescence

These Tregs can directly inhibit both T cells and antigen immuno histology. Since the DTH response is widely
presenting cells (APC) such as dendritic cells and macro- considered to reflect a secondary (memory) T cell
phages [51,52]. Using a mouse model of melanoma, response to antigen, these results were thought to
Richards et al. demonstrated that Tregs can suppress very indicate a general age-related decline in T cell-mediated
early stages of the inflammatory response after Ag chal- immunity [19,62]. In contrast, a recent study in humans
lenge [53]. Following depletion of Tregs, there was a has shown that DTH responses to bacterial, fungal and
significant increase in neutrophil infiltration, secondary to viral antigens are reduced with age, despite similar
increased concentrations of neutrophil chemoattractants responsiveness of circulating T cell to the same antigen
(CXCL1, CXCL2) [53]. in vitro [11]. This suggests that the decreased cutaneous
reactivity is a defect in local and not systemic immunity
Older mice and humans individuals have increased pro- and highlights the possibility of defective migration of
portion of Tregs in normal skin [11,54]. These Tregs may specific T cells into the skin after antigenic challenge. An
interfere with the effective initial proliferation/function of age associated defect in cutaneous antigen-specific T cell
resident antigen-specific T cells, or they could inhibit the migration is supported by studies in the mouse model of
activation of the innate immune response that may lead to accelerated ageing (SAMP1 mice) where Toichi et al. also
the decreased immunity in these individuals. The negative observed defective T cell migration [18].
effect of Treg accumulation on the induction of effective
immune responses in the skin is well documented in a The decreased ability of T cells to migrate into the skin
number of cancers. Increased numbers of Tregs have been after a DTH response in old humans was not found to be
observed in primary melanoma, melanoma metastasis and due to defective chemokine receptor or integrin expres-
basal cell carcinoma [55,56,57]. Furthermore, in human sion by circulating T cells [11]. Furthermore the T cells
squamous cell carcinoma of the skin, 50 percent of T cells from older humans also had unimpaired physical capa-
that are present are Foxp3+ [57]. Topical treatment with bility to migrate across monolayers of endothelial cells in
a TLR7 agonist imiquimod that has been shown to be an vitro [11]. Instead the reason for the reduced DTH
effective therapy in these patients, reduces the percentage clinical and cellular response appears to be lack of acti-
of Treg and their suppressive function [57]. In addition, vation of the endothelium. Activation of the endothelium
Klages et al. have shown that in the mouse model of results in the upregulation of E-selectin, VCAM and
melanoma, selective depletion of Foxp3+ cells synergises ICAM, adhesion molecules crucial for T cells transmigra-
with vaccination strategies to elicit anti-tumour responses tion into skin. TNF-a and IFN-g, are the main inducers
[58]. In a similar model Simon et al. demonstrated that of the endothelial activation [63] and in the DTH
Tregs suppressed innate immune response and that their response of young humans, these cytokines were mainly
removal improved tumour rejection [59]). secreted by macrophages [11]. The secretion of these
cytokines was significantly reduced at the site of antigen
The reasons for and the mechanism by which Tregs injection in the skin although macrophage numbers were
accumulate in the skin and other tissues of older humans normal in older humans after antigen challenge [11].
and mice is not clear [11,54,60]. It is known that Tregs The macrophages however were perfectly capable of
can be induced in the skin by UV irradiation, and that the secreting these cytokines when isolated from the skin
UV-induced Tregs then affect the function of dendritic and stimulated with TLR ligands in vitro, suggesting that
cells, through the secretion of IL-10, resulting in the they were being inhibited in vivo. The importance of
induction of more Tregs by these ‘tolerogenic’ DC TNF-a in effective cutaneous immunity is supported by
[52]. Alternatively, circulating Tregs may preferentially anecdotal evidence that rheumatoid arthritis patients on
home to the skin as a large proportion of them express anti-TNF-a therapy show increased susceptibility to skin
CLA. Collectively these data suggest that the accumu- infections and cancers [64].
lation of Tregs in the skin during ageing may contribute
to the defective cutaneous immunity in these individuals One explanation for the decreased secretion of pro-
by inhibiting either the T cells themselves or cells of the inflammatory cytokines after cutaneous antigenic chal-
innate immune system. lenge in old humans may be that the increased proportion
of Tregs that are found in the skin of these individuals
Reduction in human antigen-specific both before and after antigenic challenge may inhibit
cutaneous immune response during ageing – macrophage activation and cytokine secretion. Tregs
evidence for reduced immune surveillance have been shown to inhibit TNF-a secretion and steer
Previous studies indicated that old subjects have reduced macrophages towards and anti-inflammatory functional
ability to mount cutaneous DTH reactions after chal- profile [51].
lenge with recall antigens and that this occurs in both
humans and rodents [17–19,61]. This included decreased Conclusion
erythema and induration at the site of antigenic challenge In older humans, there is increased incidence of certain
as well as decreased infiltration of T cells as assessed by cutaneous malignancies and infections that point to

Current Opinion in Immunology 2011, 23:525–531 www.sciencedirect.com


Skin and old age Vukmanovic-Stejic et al. 529

defective immunity in the skin. Immune ageing is a 11. Agius E, Lacy KE, Vukmanovic-Stejic M, Jagger AL,
 Papageorgiou AP, Hall S, Reed JR, Curnow SJ, Fuentes-
multifactorial process and in the skin there is evidence Duculan J, Buckley CD et al.: Decreased TNF-{alpha}
that various cells exhibit defects during ageing. The synthesis by macrophages restricts cutaneous
immunosurveillance by memory CD4+ T cells during aging.
DTH reaction, a memory T cell response in the skin is J Exp Med 2009, 206:1929-1940.
also defective in older humans. However, the defect may A study investigating the basis for age-associated reductions in DTH
responses in old individuals. Age-associated decreased T cell infiltration
not be in the memory T cells themselves instead, con- was observed, secondary to reduced activation of the endothelium. Age-
ditioning of the skin environment at the site of the associated reductions in TNF-a production in cutaneous macrophages
and increase in cutaneous Treg cells are reported as possible causes.
antigenic challenge is defective, resulting in suboptimal
endothelial activation that prevents circulating memory T 12. Reed JR, Vukmanovic-Stejic M, Fletcher JM, Soares MV, Cook JE,
Orteu CH, Jackson SE, Birch KE, Foster GR, Salmon M et al.:
cells from homing to the appropriate location in the skin. Telomere erosion in memory T cells induced by telomerase
This decreased cutaneous response after antigenic chal- inhibition at the site of antigenic challenge in vivo. J Exp Med
2004, 199:1433-1443.
lenge may be related to suppression of either the acquired
or innate arms of the immune response by Tregs that are 13. Vukmanovic-Stejic M, Agius E, Booth N, Dunne PJ, Lacy KE,
Reed JR, Sobande TO, Kissane S, Salmon M, Rustin MH,
found at significantly higher numbers in the skin of older Akbar AN: The kinetics of CD4Foxp3 T cell accumulation
humans. These finding raise the question of whether during a human cutaneous antigen-specific memory response
in vivo. J Clin Invest 2008, 118:3639-3650.
modulating Treg activity in the skin may be a way to
boost cutaneous immunosurveillance of older humans 14. Kenney RT, Rangdaeng S, Scollard DM: Skin blister
immunocytology. A new method to quantify cellular kinetics in
that may reduce the risk of developing malignancy or vivo. J Immunol Methods 1987, 97:101-110.
infections in these individuals.
15. Wakim LM, Waithman J, van Rooijen N, Heath WR, Carbone FR:
 Dendritic cell-induced memory T cell activation in
Acknowledgements nonlymphoid tissues. Science 2008, 319:198-202.
Authors would like to acknowledge support of the MRC UK (grant award A study providing important insight into the priming and execution of
G0901102 to MV-S), BBSRC, British Skin Foundation and Dermatrust (to antiviral immunity to a local viral infection. Using a model of herpes
simplex virus reactivation to examine the stimulation of tissue-resident
AA) and National Institutes of Health (grant awards AG20719 and
T cells during secondary challenge. The results revealed that memory
AG03319, National Institute on Ageing to JN-Ž). CD8+ T cell responses can be initiated within peripheral tissues through a
tripartite interaction that includes CD4+ T cells and recruited dendritic
References and recommended reading cells. These findings support the existence of a response mechanism in
extra-lymphoid tissues that can act to control localised infection.
Papers of particular interest, published within the annual period of
review, have been highlighted as: 16. Gebhardt T, Wakim LM, Eidsmo L, Reading PC, Heath WR,
 Carbone FR: Memory T cells in nonlymphoid tissue that
 of special interest provide enhanced local immunity during infection with herpes
 of outstanding interest simplex virus. Nat Immunol 2009, 10:524-530.
Related to reference above. This study reports that memory CTLs in the
skin remain in the tissue and maximise protective immunity to subsequent
1. Laube S: Skin infections and ageing. Ageing Res Rev 2004, challenge with HSV-1. Using GFP expressing mice and serial transplan-
3:69-89. tation of latently infected ganglia, the authors carry out an elegant set of
experiments to demonstrate that HSV-specific cells reside in the skin near
2. Lasithiotakis K, Leiter U, Meier F, Eigentler T, Metzler G, the primary site of HSV-1 infection. These tissue-resident memory T cells
Moehrle M, Breuninger H, Garbe C: Age and gender are do not readily enter circulation once they establish residency in a given
significant independent predictors of survival in primary tissue and can proliferate locally after secondary viral challenge.
cutaneous melanoma. Cancer 2008, 112:1795-1804.
17. Vissinga C, Nagelkerken L, Zijlstra J, Hertogh-Huijbregts A,
3. Desai A, Krathen R, Orengo I, Medrano EE: The age of skin Boersma W, Rozing J: A decreased functional capacity of CD4+
cancers. Sci Aging Knowl Environ 2006, 2006:e13. T cells underlies the impaired DTH reactivity in old mice.
4. Turk JL: Delayed Hypersensitivity. edn 3. Amsterdam: Elsevier; Mech Ageing Dev 1990, 53:127-139.
1980.
18. Toichi E, Hanada K, Hosokawa T, Higuchi K, Hosokawa M,
5. Platt JL, Grant BW, Eddy AA, Michael AF: Immune cell Imamura S, Hosono M: Age-related decline in humoral
populations in cutaneous delayed-type hypersensitivity. J Exp immunity caused by the selective loss of TH cells and decline
Med 1983, 158:1227-1242. in cellular immunity caused by the impaired migration of
inflammatory cells without a loss of TDTH cells in SAMP1
6. Jacysyn JF, Abrahamsohn IA, Macedo MS: Modulation of mice. Mech Ageing Dev 1997, 99:199-217.
delayed-type hypersensitivity during the time course of
immune response to a protein antigen. Immunology 2001, 19. Castle SC, Norman DC, Perls TT, Chang MP, Yoshikawa TT,
102:373-379. Makinodan T: Analysis of cutaneous delayed-type
hypersensitivity reaction and T cell proliferative response
7. Poulter LW, Seymour GJ, Duke O, Janossy G, Panayi G: in elderly nursing home patients: an approach to
Immunohistological analysis of delayed-type hypersensitivity identifying immunodeficient patients. Gerontology 1990,
in man. Cell Immunol 1982, 74:358-369. 36:217-229.
8. Boyman O, Conrad C, Tonel G, Gilliet M, Nestle FO: The 20. Iwasaki A, Medzhitov R: Toll-like receptor control of the
pathogenic role of tissue-resident immune cells in psoriasis. adaptive immune responses. Nat Immunol 2004, 5:987-995.
Trends Immunol 2007, 28:51-57.
21. Matzinger P: An innate sense of danger. Ann NY Acad Sci 2002,
9. Tomura M, Honda T, Tanizaki H, Otsuka A, Egawa G, Tokura Y, 961:341-342.
Waldmann H, Hori S, Cyster JG, Watanabe T et al.: Activated
regulatory T cells are the major T cell type emigrating from the 22. Shaw AC, Panda A, Joshi SR, Qian F, Allore HG, Montgomery RR:
skin during a cutaneous immune response in mice. J Clin Invest Dysregulation of human Toll-like receptor function in aging.
2010, 120:883-893. Ageing Res Rev 2010, 10:346-353.
10. Kupper TS, Fuhlbrigge RC: Immune surveillance in the skin: 23. Panda A, Qian F, Mohanty S, van Duin D, Newman FK, Zhang L,
mechanisms and clinical consequences. Nat Rev Immunol Chen S, Towle V, Belshe RB, Fikrig E et al.: Age-associated
2004, 4:211-222. decrease in TLR function in primary human dendritic cells

www.sciencedirect.com Current Opinion in Immunology 2011, 23:525–531


530 Immune Senescence

predicts influenza vaccine response. J Immunol 2010, 43. Clark RA, Chong BF, Mirchandani N, Yamanaka K, Murphy GF,
184:2518-2527. Dowgiert RK, Kupper TS: A novel method for the isolation of
skin resident T cells from normal and diseased human skin.
24. Shaw AC, Joshi S, Greenwood H, Panda A, Lord JM: Aging of the J Invest Dermatol 2006, 126:1059-1070.
innate immune system. Curr Opin Immunol 2010, 22:507-513.
44. Akbar AN, Henson SM: Are senescence and exhaustion
25. Mahbub S, Brubaker AL, Kovacs EJ: Aging of the innate immune  intertwined or unrelated processes that compromise
system: an update. Curr Immunol Rev 2011, 7:104-115. immunity? Nat Rev Immunol 2011, 11:289-295.
A review article untangling the relationship between senescence and
26. Kovacs EJ, Palmer JL, Fortin CF, Fulop T Jr, Goldstein DR,
exhaustion and their role in age-associated decrease in immune function.
Linton PJ: Aging and innate immunity in the mouse: impact
of intrinsic and extrinsic factors. Trends Immunol 2009, 45. Day CL, Kaufmann DE, Kiepiela P, Brown JA, Moodley ES,
30:319-324. Reddy S, Mackey EW, Miller JD, Leslie AJ, DePierres C et al.: PD-1
expression on HIV-specific T cells is associated with T-cell
27. Grolleau-Julius A, Harning EK, Abernathy LM, Yung RL: Impaired
exhaustion and disease progression. Nature 2006,
dendritic cell function in aging leads to defective antitumor
443:350-354.
immunity. Cancer Res 2008, 68:6341-6349.
46. Blackburn SD, Shin H, Haining WN, Zou T, Workman CJ, Polley A,
28. Agrawal A, Agrawal S, Cao JN, Su H, Osann K, Gupta S: Altered
Betts MR, Freeman GJ, Vignali DA, Wherry EJ: Coregulation of
innate immune functioning of dendritic cells in elderly
CD8+ T cell exhaustion by multiple inhibitory receptors during
humans: a role of phosphoinositide 3-kinase-signaling
chronic viral infection. Nat Immunol 2009, 10:29-37.
pathway. J Immunol 2007, 178:6912-6922.
47. Sharpe AH, Wherry EJ, Ahmed R, Freeman GJ: The function of
29. Conrad C, Meller S, Gilliet M: Plasmacytoid dendritic cells in the
programmed cell death 1 and its ligands in regulating
skin: to sense or not to sense nucleic acids. Semin Immunol
autoimmunity and infection. Nat Immunol 2007, 8:239-245.
2009, 21:101-109.
48. Akbar AN, Vukmanovic-Stejic M: Telomerase in T lymphocytes:
30. Lai Y, Di Nardo A, Nakatsuji T, Leichtle A, Yang Y, Cogen AL,
use it and lose it? J Immunol 2007, 178:6689-6694.
Wu ZR, Hooper LV, Schmidt RR, von Aulock S et al.: Commensal
bacteria regulate Toll-like receptor 3-dependent inflammation 49. Effros RB, Dagarag M, Spaulding C, Man J: The role of CD8+
after skin injury. Nat Med 2009, 15:1377-1382. T-cell replicative senescence in human aging. Immunol Rev
2005, 205:147-157.
31. Gregorio J, Meller S, Conrad C, Di Nardo A, Homey B, Lauerma A,
Arai N, Gallo RL, Digiovanni J, Gilliet M: Plasmacytoid dendritic 50. Clark RA, Chong B, Mirchandani N, Brinster NK, Yamanaka K,
cells sense skin injury and promote wound healing through Dowgiert RK, Kupper TS: The vast majority of CLA+ T cells are
type I interferons. J Exp Med 2010, 207:2921-2930. resident in normal skin. J Immunol 2006, 176:4431-4439.
32. Shodell M, Siegal FP: Circulating, interferon-producing 51. Tiemessen MM, Jagger AL, Evans HG, van Herwijnen MJ, John S,
plasmacytoid dendritic cells decline during human ageing. Taams LS: CD4+CD25+Foxp3+ regulatory T cells induce
Scand J Immunol 2002, 56:518-521. alternative activation of human monocytes/macrophages.
Proc Natl Acad Sci USA 2007, 104:19446-19451.
33. Jing Y, Shaheen E, Drake RR, Chen N, Gravenstein S, Deng Y:
Aging is associated with a numerical and functional decline in 52. Schwarz A, Schwarz T: UVR-induced regulatory T cells switch
plasmacytoid dendritic cells, whereas myeloid dendritic cells antigen-presenting cells from a stimulatory to a regulatory
are relatively unaltered in human peripheral blood. Hum phenotype. J Invest Dermatol 2010, 130:1914-1921.
Immunol 2009, 70:777-784.
53. Richards H, Williams A, Jones E, Hindley J, Godkin A, Simon AK,
34. Fujita H, Nograles KE, Kikuchi T, Gonzalez J, Carucci JA, Gallimore A: Novel role of regulatory T cells in limiting early
Krueger JG: Human Langerhans cells induce distinct IL-22- neutrophil responses in skin. Immunology 2010, 131:583-592.
producing CD4+ T cells lacking IL-17 production. Proc Natl
Acad Sci USA 2009, 106:21795-21800. 54. Lages CS, Suffia I, Velilla PA, Huang B, Warshaw G, Hildeman DA,
Belkaid Y, Chougnet C: Functional regulatory T cells
35. de Jong A, Pena-Cruz V, Cheng TY, Clark RA, Van Rhijn I, accumulate in aged hosts and promote chronic infectious
Moody DB: CD1a-autoreactive T cells are a normal component disease reactivation. J Immunol 2008, 181:1835-1848.
of the human alphabeta T cell repertoire. Nat Immunol 2010,
11:1102-1109. 55. Kaporis HG, Guttman-Yassky E, Lowes MA, Haider AS, Fuentes-
Duculan J, Darabi K, Whynot-Ertelt J, Khatcherian A, Cardinale I,
36. Bobr A, Olvera-Gomez I, Igyarto BZ, Haley KM, Hogquist KA, Novitskaya I et al.: Human basal cell carcinoma is associated
Kaplan DH: Acute ablation of Langerhans cells enhances skin with Foxp3+ T cells in a Th2 dominant microenvironment.
immune responses. J Immunol 2010, 185:4724-4728. J Invest Dermatol 2007, 127:2391-2398.
37. Schwarz A, Noordegraaf M, Maeda A, Torii K, Clausen BE, 56. Ahmadzadeh M, Felipe-Silva A, Heemskerk B, Powell DJ Jr,
Schwarz T: Langerhans cells are required for UVR-induced Wunderlich JR, Merino MJ, Rosenberg SA: FOXP3 expression
immunosuppression. J Invest Dermatol 2010, 130:1419-1427. accurately defines the population of intratumoral regulatory T
cells that selectively accumulate in metastatic melanoma
38. Schwarz A, Maeda A, Schwarz T: Alteration of the migratory
lesions. Blood 2008, 112:4953-4960.
behavior of UV-induced regulatory T cells by tissue-specific
dendritic cells. J Immunol 2007, 178:877-886. 57. Clark RA, Huang SJ, Murphy GF, Mollet IG, Hijnen D,
 Muthukuru M, Schanbacher CF, Edwards V, Miller DM, Kim JE
39. Igyarto BZ, Jenison MC, Dudda JC, Roers A, Müller W, Koni PA,
et al.: Human squamous cell carcinomas evade the immune
Campbell DJ, Shlomchik MJ, Kaplan DH: Langerhans cells
response by down-regulation of vascular E-selectin and
suppress contact hypersensitivity responses via cognate CD4
recruitment of regulatory T cells. J Exp Med 2008,
interaction and Langerhans cell-derived IL-10. J Immunol 2009,
205:2221-2234.
183:5085-5093.
Reports decreased expression of adhesion molecule E-selectin on the
40. Cumberbatch M, Dearman RJ, Kimber I: Influence of ageing on blood vessels in SCC and a reduced proportion of skin homing CLA+ T
Langerhans cell migration in mice: identification of a putative cells compared to normal skin; Furthermore, SCCs are heavily infiltrated
deficiency of epidermal interleukin-1beta. Immunology 2002, by FOXP3+ T reg cells recruited from blood (approx 50% of T cells).
105:466-477. Showed that topical treatment with imiquimod (TLR7 agonist) increased
expression of E-selectin and reduced percentage and suppressive func-
41. Bhushan M, Cumberbatch M, Dearman RJ, Kimber I, Griffiths CE: tion of Tregs in SCC.
Exogenous interleukin-1beta restores impaired Langerhans
cell migration in aged skin. Br J Dermatol 2004, 150:1217-1218. 58. Klages K, Mayer CT, Lahl K, Loddenkemper C, Teng MW,
 Ngiow SF, Smyth MJ, Hamann A, Huehn J, Sparwasser T:
42. Schaerli P, Ebert L, Willimann K, Blaser A, Roos RS, Loetscher P, Selective depletion of Foxp3+ regulatory T cells improves
Moser B: A skin-selective homing mechanism for human effective therapeutic vaccination against established
immune surveillance T cells. J Exp Med 2004, 199:1265-1275. melanoma. Cancer Res 2010, 70:7788-7799.

Current Opinion in Immunology 2011, 23:525–531 www.sciencedirect.com


Skin and old age Vukmanovic-Stejic et al. 531

A study using transgenic DEREG (depletion of regulatory T cells) mice, with nursing home and hospitalized senior citizens. J Allergy
which express a diphtheria toxin receptor under control of the Foxp3 Clin Immunol 1988, 81:836-843.
locus, to allow selective depletion of Foxp3+ Tregs even during ongoing
immune responses. Foxp3+ Treg depletion induced partial regression of 62. Moesgaard F, Lykkegaard NM, Norgaard LP, Christophersen S,
established ovalbumin (OVA)-expressing B16 melanoma, which was Mosbech H: Cell-mediated immunity assessed by skin testing
associated with an increased intratumoral accumulation of activated (Multitest). I. Normal values in healthy Danish adults. Allergy
CD8+ cytotoxic T cells. The antitumor effect was significantly enhanced 1987, 42:591-596.
when Treg depletion was combined with vaccination against OVA.
63. McHale JF, Harari OA, Marshall D, Haskard DO: Vascular
59. Simon AK, Jones E, Richards H, Wright K, Betts G, Godkin A, endothelial cell expression of ICAM-1 and VCAM-1 at
Screaton G, Gallimore A: Regulatory T cells inhibit Fas ligand- the onset of eliciting contact hypersensitivity in mice:
induced innate and adaptive tumour immunity. Eur J Immunol evidence for a dominant role of TNF-alpha. J Immunol 1999,
2007, 37:758-767. 162:1648-1655.
60. Gregg R, Smith CM, Clark FJ, Dunnion D, Khan N, Chakraverty R, 64. Radovits BJ, Kievit W, Laan RF: Tumour necrosis factor-alpha
Nayak L, Moss PA: The number of human peripheral blood  antagonists in the management of rheumatoid arthritis in the
CD4+ CD25high regulatory T cells increases with age. elderly: a review of their efficacy and safety. Drugs Aging 2009,
Clin Exp Immunol 2005, 140:540-546. 26:647-664.
A review of currently available evidence relating to efficacy and safety of
61. Marrie TJ, Johnson S, Durant H: Cell-mediated immunity of anti-TNF therapy in elderly RA patients (over 65 years old).
healthy adult Nova Scotians in various age groups compared

www.sciencedirect.com Current Opinion in Immunology 2011, 23:525–531

You might also like