28
PATHOGENESIS OF SUBACUTE CUTANEOUS LUPUS ERYTHEMATOSUS REVIEW ARTICLE Panagiotis G. Stavropoulos, MD Assistant Professor, First Department of Dermatology, School of Medicine, University of Athens, “A. Syngros” Hospital, 5 Ionos Dragoumi St, 16121 Athens, Greece. E-mail: [email protected] Andreas V. Goules, MD PhD, Research Fellow, First Department of Dermatology, School of Medicine, University of Athens, “A. Syngros” Hospital, 5 Ionos Dragoumi St, 16121 Athens, Greece. E-mail: [email protected] Georgia Avgerinou, MD Associate Professor, First Department of Dermatology, School of Medicine, University of Athens, “A. Syngros” Hospital, 5 Ionos Dragoumi St, 16121 Athens, Greece. E-mail: [email protected] Andreas D. Katsambas, MD Professor and Chairman, First Department of Dermatology, School of Medicine, University of Athens, “A. Syngros” Hospital, 5 Ionos Dragoumi St, 16121 Athens, Greece.

pathogenesis of subacute cutaneous lupus erythematosus

Embed Size (px)

Citation preview

Page 1: pathogenesis of subacute cutaneous lupus erythematosus

PATHOGENESIS OF SUBACUTE CUTANEOUS LUPUS

ERYTHEMATOSUS

REVIEW ARTICLE

Panagiotis G. Stavropoulos, MD

Assistant Professor, First Department of Dermatology, School of Medicine,

University of Athens, “A. Syngros” Hospital, 5 Ionos Dragoumi St, 16121 Athens,

Greece.

E-mail: [email protected]

Andreas V. Goules, MD

PhD, Research Fellow, First Department of Dermatology, School of Medicine,

University of Athens, “A. Syngros” Hospital, 5 Ionos Dragoumi St, 16121 Athens,

Greece.

E-mail: [email protected]

Georgia Avgerinou, MD

Associate Professor, First Department of Dermatology, School of Medicine,

University of Athens, “A. Syngros” Hospital, 5 Ionos Dragoumi St, 16121 Athens,

Greece.

E-mail: [email protected]

Andreas D. Katsambas, MD

Professor and Chairman, First Department of Dermatology, School of Medicine,

University of Athens, “A. Syngros” Hospital, 5 Ionos Dragoumi St, 16121 Athens,

Greece.

Page 2: pathogenesis of subacute cutaneous lupus erythematosus

2

E-mail: [email protected], [email protected]

Key Words: Subacute cutaneous lupus erythematosus, pathogenesis, plasmacytoid

dendritic cells, apoptosis, type I IFNs, antibody depended cell mediated cytotoxicity

Correspondence

Andreas D. Katsambas, MD

First Department of Dermatology, School of Medicine, University of Athens, “A.

Syngros” Hospital, Greece.

Ionos Dragoumi St 5

Athens 16121

Greece

Tel: 210-7210839

Fax: 210-7211122

E-mail: [email protected], [email protected],

Page 3: pathogenesis of subacute cutaneous lupus erythematosus

3

ABSTRACT

Subacute Cutaneous Lupus Erythematosus (SCLE) is a photosensitive form of lupus

specific skin lesion which is strongly associated with the presence of anti-Ro/SSA

autoantibody. The pathogenesis of SCLE includes genetic, environmental and

immunologic factors. Recent studies provide strong evidences for the involvement of

innate and cell mediated immunity, underlying the important role of plasmacytoid

dendritic cells (pDC), IFNα and antibody depended cell cytotoxicity (ADCC). In

addition, a variety of cytokines, chemokines and adhesion molecules have been found

to participate in the expansion phase of the autoimmune effector mechanisms. This

article summarizes the recent immunological findings and reviews the current

mechanisms which are implied in the development of the disease.

INTRODUCTION

Subacute Cutaneous Lupus Erythematosus (SCLE) represents a distinct lupus-

specific cutaneous lesion, intermediate between the acute lesion of malar rash and the

chronic lesions such as discoid lupus or lupus profundus which usually cause scarring.

SCLE is a photosensitive, non-fixed, non-scarring, exacerbating and remitting skin

disease which commonly occurs in sun-exposed areas and may be generalized. It may

present as a papulosquamus eruption that resembles psoriasis or as an annular lesion

that resembles erythema multiforme. The most commonly affected areas are the

shoulders, the upper back, the extensor arms and the V-area of the upper chest

Page 4: pathogenesis of subacute cutaneous lupus erythematosus

4

while the face and the neck are less commonly affected. SCLE presents in the third

or fourth decade of life and women are 3-4 times more likely to be affected than men

1. Half of the patients who exhibit SCLE meet the diagnostic criteria for SLE and

develop a less severe systemic disease although the skin lesions may be more

refractory.

Sometimes, SCLE needs to be differentiated from other clinical subtypes

of chronic cutaneous lupus erythematosus (LE) such as chronic discoid LE

(CDLE), LE tumidus (LET) and LE profundus (LEP). Interestingly, 20% of

patients with SCLE have concomitant CDLE 2. However, CDLE presents as one or

more erythematous papules and/or plaques which are more likely to appear on

the face, the external ears, the scalp, the extensor aspects of the forearms and the

trunk and have a tendency for scarring and atrophy 3, 4

. LET corresponds to

discoid, urticaria-like single or multiple plaques with a bright reddish or

violaceous smooth surface on sun exposed areas. The eruption is remarkable

photosensitive and persistent without a tendency for scarring 5.

Histopathologically, is characterized by dermal inflammation and the absence of

epidermal involvement 6. Finally, LEP is an inflammatory condition involving

the subcutaneous adipose tissue (lupus panniculitis) and clinically presents as

deep cutaneous nodules which are not painful and are symmetrically distributed

on the upper arms and the face 4.

The pathogenesis of SCLE is multifactorial and although the exact cause

remains unknown genetic, environmental and immunologic factors seem to contribute

to the development of the disease. Recent genetic data has elucidated potential

candidate genes for SCLE 7-9. Among the environmental factors, ultraviolet light

(UVL) and drugs appear to play the most important role in the pathogenesis of SCLE

and cutaneous lupus in general 10, 11

. Many studies have led to the postulate that innate

Page 5: pathogenesis of subacute cutaneous lupus erythematosus

5

and cell mediated immunity participates in the development of the cutaneous skin

lesions 12-15

. Finally, a complex network of cutaneous cytokines, chemokines and

adhesion molecules orchestrate and promote tissue injury observed in skin lesions of

SCLE 16, 17

. In summary, the etiopathogenesis of SCLE is considered to result from

three distinct stages in genetic susceptible patients: initiation, amplification and

maintenance of autoimmune response and finally induction of tissue injury.

IMMUNOPATHOLOGY-HISTOPATHOLOGY

Histological, LE-specific lesions share many common features but SCLE can

be distinguished from the other forms 18. The major characteristics are moderate

hyperkeratosis with focal disorientation and liquefactive degeneration of the basal

layer, mild to prominent atrophy and periappendageal mononuclear cell infiltrate

confined to the superficial dermis. A dermal edema can be also observed but

basement membrane thickening and follicular plugging is minimal or absent 19. The

inflammatory infiltrate of the characteristic interface dermatitis observed in SCLE

lesions, consists mainly of activated T cells and macrophages. These inflammatory

cells appear to be in close apposition to epidermal basal keratinocytes which undergo

apoptosis and cytotoxic injury 18. The interface dermatitis is characterized by a

hybrid pattern comprising cell-poor vacuolar foci alternating with zones of

lichenoid dermatitis 20. Immunohistochemical studies including staining for the

cytotoxic molecule granzyme B, revealed a significant lower expression in

lesional lymphocytes of patients with SCLE compared to patients with CDLE 21.

Furthermore, when analyzing the expression of both granzyme B and Tia1 in

skin biopsies of patients with CDLE, LET, LEP and SCLE, it was found that

Page 6: pathogenesis of subacute cutaneous lupus erythematosus

6

granzyme B and Tia1 positive cells were present in all subsets but their number

was lower in patients with SCLE 15. The above data suggest that the number of

CD8 lymphocytes is lower in SCLE lesions compared to other clinical subtypes

of cutaneous LE. Immunofluorescence studies demonstrate deposition of IgG

immunoglobulin and/or complement components in a granular pattern at the dermal-

epidermal junction, in about 60% of patients with SCLE. A dust like pattern of IgG

deposition overlying epidermal basal cells and cells below the dermal-epidermal

junction is considered by some investigators, a specific immunopathological feature

of SCLE 22.

Anti-SS-A/Ro and anti-SS-B/La autoantibodies have been strongly associated

with cutaneous lupus and SCLE 23, 24

. Approximately 90% of SCLE patients have

positive anti-Ro/SS-A antibodies 25 while a smaller percentage is being positive for

anti-La/SS-B 23. Other autoantibodies such as antinuclear antibodies (ANA), anti-

dsDNA and anti-Sm, have also been found in SCLE patients but less commonly than

anti-Ro/SS-A 2, 25

. Previous studies have focused on the close association of

antibodies against the Ro/SS-A antigen with the development of clinical symptoms 23,

26. Deposition of immunoglobulins and complement at the dermoepidermal junction

suggests a direct participation of anti-Ro/SS-A antibodies in the pathogenesis of

SCLE. Several mechanisms though have been implicated in the pathophysiology of

the disease including direct autoantibody effects, immunoglobulin deposits and cell-

mediated immunity 27.

Independently of the underlying immunological mechanisms that mediate the

tissue damage, epidermal keratinocytes and especially the basal cells seem to be the

major target of the immune system. Dermal-epidermal localization of

immunoglobulins and complement possibly results from nuclear material deriving

from apoptotic keratinocytes 28. These nuclear autoantigens can react with circulating

Page 7: pathogenesis of subacute cutaneous lupus erythematosus

7

autoantibodies and form immune complexes which precipitate and initiate tissue

injury. Besides this model which is also observed in SLE and drug induced lupus, a

direct effect of autoantibody has been also proposed as a possible mechanism for the

pathogenesis of cutaneous lupus and SCLE. It has been previously shown that the

binding of anti-Ro/SS-A antibodies to the surface of epidermal cells, is an important

inducer of antibody-dependent keratinocyte damage in photosensitive cutaneous LE

29. In addition, these autoantibodies have been found to bind to UVB-irradiated human

keratinocytes both in vivo and in vitro 30-34

. It has also been shown that binding is

dependent on UVB dose and glycation 31.

Antibody-dependent cell–mediated cytotoxicity (ADCC) and CD8+

cytotoxicity may also be involved in tissue damage. Keratinocytes can be killed both

by complement-mediated cytotoxicity lysis and ADCC 13, 14

. Gershwin et al

demonstrated a destruction of DNA-coated targets by lupus antisera and lymphocyte

effectors 35. In a recent study Furukawa et al supported that keratinocytes from

patients with SLE and SCLE showed enhanced cytotoxicity to UV radiation and to

antibody-mediated cytotoxicity 36. More specifically, keratinocytes from SCLE

patients have been found to be more susceptible to ultraviolet radiation-induced

cytotoxicity and binding of anti-Ro/SS-A and showed significant ADCC after

irradiation, incubation in their own patients’ sera and exposure to mononuclear cells

from normal individuals. Furthermore, antiRo/SS-A IgG fractions induce enhanced

cytotoxicity of irradiated keratinocytes from SCLE patients. Taking into consideration

that basal keratinocytes are relative resistant to ultraviolet radiation-induced apoptosis

while suprabasal keratinocytes undergo apoptosis in vivo after irradiation 37 it seems

possible that suprabasal keratinocytes are the major targets of ADCC in SCLE lesions

38.

Page 8: pathogenesis of subacute cutaneous lupus erythematosus

8

Tissue injury in SLE is mainly attributed to immune complex formation and

deposition 39. This concept is also confirmed in SCLE skin lesions, since

immunoglobulin and complement have been identified at the dermal-epidermal

junction, leading to complement activation, membrane attack complex formation and

cellular injury. Moreover, the presence of immunoglobulin at the dermal-epidermal

junction is not always accompanied by tissue damage. Besides, the presence of a

lymphocytic infiltrate implies a cell-mediated immunity. Recent findings suggest

ADCC and CD8+ cytotoxicity as underlying immunological mechanisms responsible

for the injury phase and the development of clinical symptoms. Taking together, we

can speculate that immune complexes and a direct autoantibody effect possibly

characterize the early phase of tissue damage. ADCC and direct T cell-mediated

cytotoxicity probably occur later enhancing the inflammatory response. It appears that

the injury phase of SCLE is a complex phenomenon, attributed to distinct

autoimmune effectors mechanisms which include both humoral and cell-mediated

immunity. The initial hypothesis of immune complex deposition has been enriched by

new findings which point out the role of cell mediated immunity and provide new

insights in our understanding the pathophysiology of the disease.

In another recent study, a quantitative immunohistochemical analysis of CD4+

T cells from patients with cutaneous lupus erythematosus including SCLE, revealed

that the number of Foxp3+ Treg (CD4+ CD25+) was significantly reduced compared

to that in lesions from patients with other inflammatory diseases 40. There was no

correlation between disease subtypes and the frequency of Foxp3+ Treg in the skin of

patients with cutaneous lupus erythematosus. Referring to the phenotype and number

of Treg as well as to their sensitivity of apoptosis, no differences were observed in

peripheral blood between SCLE patients and normal donors 40. The fact that Treg are

reduced in skin lesions but not in peripheral blood of patients with CLE, reflects a

Page 9: pathogenesis of subacute cutaneous lupus erythematosus

9

limitation of the disease to the skin while a systemic decrease could explain an

involvement of multiple organs in patients with SLE. The relative lack of Treg could

amplify the autoimmune process and enhance the inflammatory response in skin

lesions of SCLE patients. However, a more careful interpretation is needed since this

reduction could be the result of the disease process rather than a causative factor.

GENETIC CONSIDERATIONS

Multiple genes have been involved in the development of CLE and especially

SCLE. It is well established that polymorphisms of genes encoding HLA, TNFa and

complements molecules, have the strongest genetic association with SCLE. Specific

combinations of these genes may determine individual susceptibility to SCLE. It is

currently believed that a specific genetic background is necessary for peculiar

environmental factors to act and lead the immune system to autoimmunity and SCLE

lesions.

Firstly, the HLA1, B8, DR3 haplotype has been found in 25% of a cohort of

SCLE patients 41 and since then it has been widely extended

7. Subsequently, the

HLA1, B8, DR3, DQ2, DRw52 and C4 null ancestral haplotype has been proposed as

a susceptibility haplotype for SCLE, especially for patients with positive anti-Ro

autoantibodies 42. In addition, HLA-DR3 women have been associated with SCLE,

SLE and Sjögren syndrome as well as with the presence of anti-Ro autoantibodies

suggesting a crucial immunogenetic role for the antibody production. In a past study,

the presence of HLA DQ1 and DQ2 has been found to enhance the production of

autoantibodies in patients with Sjögren syndrome 43. The mechanisms, by which the

HLA genes are involved in the pathogenesis of the disease, have not been elucidated

Page 10: pathogenesis of subacute cutaneous lupus erythematosus

10

yet. It is well documented though that MHC loci influence many autoimmune

diseases and many models have been proposed. It is generally accepted that HLA

molecules are possibly involved in T cell repertoire selection and antigen presentation

leading to abnormal autoimmune responses 44.

In addition, deficiencies of C2 and C4 components of complement have been

associated with both SLE and SCLE 45, 46

. The majority of patients with C2 or C4

homozygous deficiency have anti-Ro autoantibodies implying a possible role for this

antibody in the development of SCLE 47. In a recent study a homozygous single

nucleotide polymorphism of the complement C1qA gene which encodes the A chain

of the C1q complement component, has been associated with decreased levels of C1q

and a SCLE phenotype 8. Similarly, patients with a complete deficiency of C1q, C1r

or C1s are prone to develop SLE and present with photosensitive cutaneous lesions 48.

More specifically, Pickering et al supported that C1q component of complement is

very important in the physiological clearance of apoptotic cells 49. Furthermore,

complement deficiency states may lead to impaired clearance of immune complexes

allowing subsequent deposition in tissues and injury via ligation of Fcγ receptors on

leukocytes. Thus, another physiological activity of complement is processing and

clearance of immune complexes and apoptotic cells. In this context, complement

system contributes in the resolution of immune response and prevention of

autoimmune phenomena.

Finally, a polymorphism of the TNFα gene promoter (-308A) has been found

to encode increased expression of TNFα by UV-B irradiated keratinocytes and has

been associated with SLCE lesions 8, 9

. However, the TNFα gene is located within the

HLA region and thus shares the same extended haplotype with DR3, implying a

possible linkage equilibrium. Evidence in SLE patients though, supports that each is

likely to contribute independently to disease susceptibility 50.

Page 11: pathogenesis of subacute cutaneous lupus erythematosus

11

THE EFFECT OF ULTRAVIOLET LIGHT

There is a clear relationship between ultraviolet light (UV) and

photosensitivity in patients with SLE and SCLE. Photosensitivity is included in the

diagnostic criteria of SLE while SCLE is considered to be the most photosensitive

lesion of cutaneous lupus. The UV spectrum is divided into two major segments:

UVB which represents the wavelengths between 290-320nm and UVA which consists

of wavelengths between 320-400nm. Epidermis is a major absorbent of UVB and less

than 10% penetrates to dermis. On the contrary, UVA penetrates to the dermis and

contribute in altering structural and matrix proteins. Although solar light includes both

UVB and UVA, UVB is more efficient in evoking photosensitive responses and has

been markedly involved in forms of CLE 10, 51-53

. However, UVA has been also found

to induce lupus skin lesions 11, 27, 54

.

DNA damage and apoptosis

UV light is capable of DNA damage. More specifically, UVB can cause

excitation of DNA molecules leading to formation of pyrimidine dimmers acting on a

direct way 55, 56

. On the contrary UVA seems to damage DNA indirectly via a

photosensitized reaction by modifying singlet oxygen generating purine bases 55, 56

.

Altered DNA molecules with specific modifications might possess immunogenic

properties and in combination with a possible insufficiency of repairing mechanisms

could lead the immune system to autoimmunity. Consistent with this speculation is

Page 12: pathogenesis of subacute cutaneous lupus erythematosus

12

the observation that SLE patients can develop immune responses to UV altered DNA

molecules 57.

Apoptosis is considered a programmed cell death which is characterized by

specific steps such as DNA cleavage and fragmentation, nuclear condensation, surface

blebbing, cytoplasmic contraction and finally packaging of cellular components

within membranes and formation of apoptotic bodies. During this process toxic agents

such as cytolytic enzymes are released and many self antigens can be redistributed

and presented to the immune system. In addition, enzymatic degradation can cause

alterations in these autoantigens and thereby increase their immunogenicity 58.

Normally, apoptotic debris is eliminated by phagocytes and overexposure of

autoantigens to immune system is prevented. Therefore, an increase rate of apoptosis

or a decreased rate of clearance of apoptotic cells predisposes to autoimmunity.

UV exposure is known to induce apoptosis in keratinocytes although basal

keratinocytes are more resistant than suprabasal cells 37, 59

. UVB is considered to be a

strong inducer of apoptosis and in that way enhances the exposure of autoantigens at

the surface of apoptotic cells. Carrichio et al supported that UVB dose plays a crucial

role in inflammation and autoantigens redistribution and determines the rate of

apoptosis 60. Low and intermediate doses induce non-inflammatory apoptotic

procedures while high doses result to proinflammatory necrosis. More specifically,

UVB can cause an upregulation of both Fas and FasL leading to apoptotic death via

the activation of this particular pathway 61. In cutaneous lupus lesions, the increased

rate of apoptosis results to translocation and display of autoantigens such as Ro/SSA

and La/SSB to keratinocytes cell surface. In a past study, it was demonstrated that

UVB irradiated keratinocytes from SLE patients, underwent apoptosis and formed

apoptotic blebs rich in lupus autoantigens including both Ro/SSA and La/SSB 62. This

finding has led to the suggestion that these autoantigens can be phagocytozed,

Page 13: pathogenesis of subacute cutaneous lupus erythematosus

13

processed and presented to lymphocytes and thus contributing to the beak of immune

tolerance and generation of primary immune responses to self-antigens 63.

Furthermore, Golan et al demonstrated enhanced membrane binding of antibodies to

Ro/SSA and La/SSB to cultured keratinocytes of SLE patients after UV irradiation

implying a possible translocation of these antigens to the cell surface32.

Several studies supported a disturbed clearance of apoptotic cells in SLE.

More specifically, macrophages have been proposed to exhibit impaired clearance

capacity of apoptotic cells 64-66

. Furthermore, apoptotic neutrophils from SLE patients

have been found incapable of binding the C1q component of complement, leading to

accumulation of apoptotic debris 67. Similarly, C1q appears to bind directly to surface

blebs of apoptotic human keratinocytes 68. It has been mentioned previously that

decreased C1q levels have been also associated with the SCLE phenotype 8. The

decreased rate of clearance and the subsequent accumulation of apoptotic debris

predisposes to self antigen presentation and processing which may result to

autoimmune responses.

Apoptosis, inflammation and type I INFs

Besides, apoptosis has been also associated with the development of

inflammatory skin lesions in SLE. Macrophages that have ingested apoptotic cells

seem to release anti-inflammatory cytokines such as TGFβ 69 and thus a decreased

clearance rate enhances an inflammatory profile which can induce autoimmune

procedures. It has been supported that apoptotic cells can interact with Fcγ receptors

on phagocytes and thereby activate the latter cells and promote the inflammatory

response in SLE 70. In the skin of patients with SLE, it has been shown that DNA and

RNA particles released from apoptotic cells can induce the production of IFNα by

plasmacytoid dendritic cells 71. In a past study, Farkas et al found that pDC were

Page 14: pathogenesis of subacute cutaneous lupus erythematosus

14

present at skin lesions of patients with SLE and DLE suggesting that pDC are an

important source of IFNα/β in cutaneous LE lesions 72. Activation of pDC leads to

production of INFα which induces the secretion of specific chemokines such as

CXCL9, CXCL10 and CXCL11 12. These chemokines are the major ligands of the

CXCR3 which is expressed by skin homing lymphocytes and premature pDC and

therefore contribute to the recruitment of inflammatory cells at skin lesions.

Furthermore, in a recent study it has been shown that the expression pattern of

interferon–inducible proteins including CXCL9 and CXCL10, reflects the

characteristic histological distribution of infiltrating immune cells in different CLE

subsets, including SCLE 15. More specifically, in SCLE lesions it was demonstrated

an association between these specific chemokines and the distribution of

CXCR3+CD3+ focused in epidermis and upper dermis. In another study, it has been

shown that in patients with active CLE lesions, IFN-inducible chemokines such as

IP10 and CXCL10 can lead to the recruitment of CXCR3 expressing T cells into the

skin lesions, suggesting that type I INFs can induce a Th1-bias inflammatory immune

response 73. These findings have revealed the important role of innate immunity and

IFNα and open up options for novel therapeutic approaches in CLE 74.

Cytokines and adhesion molecules

UV light is also involved in cytokine expression, vascular activation and

induction of adhesion molecules, chemokines and selectins which mediate the

migration of inflammatory cells and lymphocytes to skin lesions of CLE 16, 17

. UVB

can promote the release of proinflammatory cytokines such as TNFα and IL-1 by

keratinocytes. These cytokines can cause upregulation of ICAM on keratinocytes and

therefore account for the increased homing of leukocytes to the skin 75. Additionally,

Page 15: pathogenesis of subacute cutaneous lupus erythematosus

15

TNFα is capable of inducing translocation of Ro/SSA and La/SSB autoantigens to the

surface of keratinocytes and apoptosis via the TNFR1 76. Immunohistochemical

studies in skin biopsies of SCLE patients who were receiving treatment, demonstrated

that refractory lesional skin tissue displayed a strongly positive distribution of TNFα

particularly in epidermis while no prominent staining was seen in non lesional skin

from the same group or the control group 77. These findings suggest a potential role of

TNFα in the pathogenesis of SCLE as well as a possible therapeutic target. UVA

exposure can also promote IL-12 and IFN-γ production in the skin leading to a Th1

inflammatory response 78. Blood vessels and endothelium are also involved in SCLE

lesions. Activated endothelium expresses a variety of adhesion molecules which

mediate the recruitment and transmigration of leukocytes through vascular wall, at the

sites of skin lesions. TNFα and INF-γ are potent inducers of ICAM-1 by the

endothelium after UVB exposure 79 while VCAM-1 has been found to be highly

expressed by endothelial cells in skin lesions of lupus 80, 81

. Finally, E-selectin has

also been found to be increased in lupus photosensitive lesions 81 and after UVB

exposure 82.

Although the data mentioned above are referring to SLE and cutaneous lupus,

similar alterations in vascular endothelium and cytokines are implied in SCLE. It is

obvious that the overproduction of proinflammatory cytokines affect both the

inflammatory procedures observed in skin lesions and cutaneous endothelium.

Keratinocytes are induced to express ICAM-1 which facilitates T-cell migration to the

skin. The endothelium of cutaneus lesions is activated and expresses associated

adhesion molecules such as ICAM-1, VCAM-1 and E-selectins which contribute to

recruitment of leukocytes from the circulation to the inflammatory sites. Probably,

LFA-1 is also upregulated on the surface of leukocytes and type I INFs promote the

release of specific chemokines which further attract the inflammatory cells. This

Page 16: pathogenesis of subacute cutaneous lupus erythematosus

16

complex network of cytokines and chemokines plays an important role in

perpetuating and amplification of inflammation and therefore contribute to

propagation and maintenance of the autoimmune procedures.

OVERVIEW

SCLE is the most photosensitive form of cutaneous lupus erythematous and it

has been associated with the presence of anti-Ro/SSA autoantibodies. Data over the

past years support that the etiopathogenesis of SCLE share common features with

SLE and the other forms of CLE, although it is considered a distinct skin lesion. It is

well documented that specific genetic background is an important factor for the

development of SCLE and many candidate genes have been identified. The ancestral

haplotype HLA1, B8, DR3, DQ2, DRw52 and C4 null, deficiencies of C1q, C2 and

C4 components of complement and the TNFα -308 polymorphism have been found to

predispose to the SCLE phenotype. Although the exact mechanisms have not been

established yet, it seems possible that this genetic susceptibility interferes with T cell

repertoire selection and autoantigen presentation. The effect of UVL and especially

UVB appears to play an important role at the initiation and maintenance of

autoimmunity. UVL increases the rate of apoptosis of keratinocytes and causes

redistribution of autoantigens such as Ro/SSA and La/SSB. The apoptotic blebs arise

form the nuclear structures and harbor a variety of self antigens which are processed

by phagocytes and presented to lymphocytes, leading to primary autoimmune

responses. Impaired clearance of apoptotic cells results in accumulation of apoptotic

debris and further enhancement of autoantigen presentation to the immune system.

UVL also causes damage and alteration in DNA molecules which become more

Page 17: pathogenesis of subacute cutaneous lupus erythematosus

17

immunogenic. These mechanisms participate in the induction phase and the loss of

immune tolerance. In addition, persistent apoptosis activates phagocytes via the Fcγ

receptors and contributes to the development of an inflammatory environment at the

skin lesions. UVL induces the overproduction of proinflammatory cytokines including

IL-1 and TNFα by the skin and the expression of adhesion molecules (ICAM-1,

VCAM-1, E-selectins) by both keratinocytes and endothelial cells. Plasmacytoid cells

produce high levels of IFNα which causes secretion of specific chemokines such as

CXCL9, 10, 11 by keratinocytes and fibroblasts. This complex network of cytokines,

adhesion molecules and chemokines participate in leukocyte recruitment at the skin

and is responsible for the expansion phase through perpetuation and maintenance of

inflammation. Finally, the injury phase is mediated by immune complexes deposition,

direct autoantibody effect, direct T cell cytotoxicity and ADDC. Recent studies

provide strong evidence for the dynamic role of the latter mechanisms and suggest

further therapeutic approaches. The current immunological concepts in the

pathogenesis of SCLE are summarized in Figure 1.

These new findings have improved our understanding of the genetic,

environmental and immunologic mechanisms which are involved in the pathogenesis

of SCLE. The variety of potent molecules and cytokines implies molecular orientated

therapeutic strategies which include cytokine inhibition, direct T cell therapies and

disruption of T cell interactions with adhesion molecules. The role of innate and cell

mediated immunity has now become apparent and directs our insights into new

researching fields.

Page 18: pathogenesis of subacute cutaneous lupus erythematosus

18

REFERENCES

1 Parodi A, Caproni M, Cardinali C, et al. Clinical, histological and

immunopathological features of 58 patients with subacute cutaneous lupus

erythematosus. A review by the Italian group of immunodermatology.

Dermatology 2000;200:6-10.

2 Sontheimer R D. Subacute cutaneous lupus erythematosus: a decade's

perspective. Med Clin North Am 1989;73:1073-90.

3 Callen J P. Discoid lupus erythematosus--variants and clinical associations.

Clin Dermatol 1985;3:49-57.

4 Patel P and Werth V. Cutaneous lupus erythematosus: a review. Dermatol

Clin 2002;20:373-85, v.

5 Kuhn A, Richter-Hintz D, Oslislo C, et al. Lupus erythematosus tumidus--a

neglected subset of cutaneous Lupus erythematosus: report of 40 cases. Arch

Dermatol 2000;136:1033-41.

6 Kuhn A, Sonntag M, Ruzicka T, et al. Histopathologic findings in lupus

erythematosus tumidus: review of 80 patients. J Am Acad Dermatol

2003;48:901-8.

7 Lio D, Candore G, Colombo A, et al. A genetically determined high setting of

TNF-alpha influences immunologic parameters of HLA-B8,DR3 positive

subjects: implications for autoimmunity. Hum Immunol 2001;62:705-13.

8 Millard T P, Kondeatis E, Cox A, et al. A candidate gene analysis of three

related photosensitivity disorders: cutaneous lupus erythematosus,

polymorphic light eruption and actinic prurigo. Br J Dermatol 2001;145:229-

36.

Page 19: pathogenesis of subacute cutaneous lupus erythematosus

19

9 Werth V P, Zhang W, Dortzbach K, et al. Association of a promoter

polymorphism of tumor necrosis factor-alpha with subacute cutaneous lupus

erythematosus and distinct photoregulation of transcription. J Invest Dermatol

2000;115:726-30.

10 Kochevar I E. Action spectrum and mechanisms of UV radiation-induced

injury in lupus erythematosus. J Invest Dermatol 1985;85:140s-143s.

11 Nived O, Johansen P B and Sturfelt G. Standardized ultraviolet-A exposure

provokes skin reaction in systemic lupus erythematosus. Lupus 1993;2:247-

50.

12 Meller S, Winterberg F, Gilliet M, et al. Ultraviolet radiation-induced injury,

chemokines, and leukocyte recruitment: An amplification cycle triggering

cutaneous lupus erythematosus. Arthritis Rheum 2005;52:1504-16.

13 Norris D A and Lee L A. Antibody-dependent cellular cytotoxicity and skin

disease. J Invest Dermatol 1985;85:165s-175s.

14 Norris D A, Ryan S B, Kissinger R M, et al. Systematic comparison of

antibody-mediated mechanisms of keratinocyte lysis in vitro. J Immunol

1985;135:1073-9.

15 Wenzel J, Zahn S, Mikus S, et al. The expression pattern of interferon-

inducible proteins reflects the characteristic histological distribution of

infiltrating immune cells in different cutaneous lupus erythematosus subsets.

Br J Dermatol 2007;157:752-7.

16 Bennion S D and Norris D A. Ultraviolet light modulation of autoantigens,

epidermal cytokines and adhesion molecules as contributing factors of the

pathogenesis of cutaneous LE. Lupus 1997;6:181-92.

Page 20: pathogenesis of subacute cutaneous lupus erythematosus

20

17 Middleton M H and Norris D A. Cytokine-induced ICAM-1 expression in

human keratinocytes is highly variable in keratinocyte strains from different

donors. J Invest Dermatol 1995;104:489-96.

18 Bangert J L, Freeman R G, Sontheimer R D, et al. Subacute cutaneous lupus

erythematosus and discoid lupus erythematosus. Comparative histopathologic

findings. Arch Dermatol 1984;120:332-7.

19 Gilliam J N and Sontheimer R D. Distinctive cutaneous subsets in the

spectrum of lupus erythematosus. J Am Acad Dermatol 1981;4:471-5.

20 Crowson A N and Magro C. The cutaneous pathology of lupus erythematosus:

a review. J Cutan Pathol 2001;28:1-23.

21 Wenzel J, Uerlich M, Worrenkamper E, et al. Scarring skin lesions of discoid

lupus erythematosus are characterized by high numbers of skin-homing

cytotoxic lymphocytes associated with strong expression of the type I

interferon-induced protein MxA. Br J Dermatol 2005;153:1011-5.

22 Nieboer C, Tak-Diamand Z and Van Leeuwen-Wallau H E. Dust-like

particles: a specific direct immunofluorescence pattern in sub-acute cutaneous

lupus erythematosus. Br J Dermatol 1988;118:725-9.

23 Sontheimer R D, Maddison P J, Reichlin M, et al. Serologic and HLA

associations in subacute cutaneous lupus erythematosus, a clinical subset of

lupus erythematosus. Ann Intern Med 1982;97:664-71.

24 Sontheimer R D, Thomas J R and Gilliam J N. Subacute cutaneous lupus

erythematosus: a cutaneous marker for a distinct lupus erythematosus subset.

Arch Dermatol 1979;115:1409-15.

25 Lee L A, Roberts C M, Frank M B, et al. The autoantibody response to

Ro/SSA in cutaneous lupus erythematosus. Arch Dermatol 1994;130:1262-8.

Page 21: pathogenesis of subacute cutaneous lupus erythematosus

21

26 Weston W L. Significance and character of SS-A(Ro) and SS-B(La) antigens.

J Invest Dermatol 1985;84:85.

27 Norris D A and Lee L A. Pathogenesis of cutaneous lupus erythematosus. Clin

Dermatol 1985;3:20-35.

28 Gilliam J N. The significance of cutaneous immunoglobulin deposits in lupus

erythematosus and NZB/NZW F1 hybrid mice. J Invest Dermatol

1975;65:154-61.

29 Furukawa F, Kanauchi H and Imamura S. Susceptibility to UVB light in

cultured keratinocytes of cutaneous lupus erythematosus. Dermatology

1994;189 Suppl 1:18-23.

30 Furukawa F, Ikai K, Matsuyoshi N, et al. Relationship between heat shock

protein induction and the binding of antibodies to the extractable nuclear

antigens on cultured human keratinocytes. J Invest Dermatol 1993;101:191-5.

31 Furukawa F, Kashihara-Sawami M, Lyons M B, et al. Binding of antibodies to

the extractable nuclear antigens SS-A/Ro and SS-B/La is induced on the

surface of human keratinocytes by ultraviolet light (UVL): implications for the

pathogenesis of photosensitive cutaneous lupus. J Invest Dermatol

1990;94:77-85.

32 Golan T D, Elkon K B, Gharavi A E, et al. Enhanced membrane binding of

autoantibodies to cultured keratinocytes of systemic lupus erythematosus

patients after ultraviolet B/ultraviolet A irradiation. J Clin Invest

1992;90:1067-76.

33 Jones S K. Ultraviolet radiation (UVR) induces cell-surface Ro/SSA antigen

expression by human keratinocytes in vitro: a possible mechanism for the

UVR induction of cutaneous lupus lesions. Br J Dermatol 1992;126:546-53.

Page 22: pathogenesis of subacute cutaneous lupus erythematosus

22

34 LeFeber W P, Norris D A, Ryan S R, et al. Ultraviolet light induces binding of

antibodies to selected nuclear antigens on cultured human keratinocytes. J

Clin Invest 1984;74:1545-51.

35 Gershwin M E, Glinski W, Chused T, et al. Lymphocyte-dependent antibody-

mediated cytolysis of DNA-anti-DNA-coated target cells using human and

murine SLE effector populations. Clin Immunol Immunopathol 1977;8:280-

91.

36 Furukawa F, Itoh T, Wakita H, et al. Keratinocytes from patients with lupus

erythematosus show enhanced cytotoxicity to ultraviolet radiation and to

antibody-mediated cytotoxicity. Clin Exp Immunol 1999;118:164-70.

37 Norris D A, Middleton M H, Whang K, et al. Human keratinocytes maintain

reversible anti-apoptotic defenses in vivo and in vitro. Apoptosis 1997;2:136-

48.

38 Norris D A, Whang K, David-Bajar K, et al. The influence of ultraviolet light

on immunological cytotoxicity in the skin. Photochem Photobiol

1997;65:636-46.

39 Raz E, Ben-Bassat H, Davidi T, et al. Cross-reactions of anti-DNA

autoantibodies with cell surface proteins. Eur J Immunol 1993;23:383-90.

40 Franz B, Fritzsching B, Riehl A, et al. Low number of regulatory T cells in

skin lesions of patients with cutaneous lupus erythematosus. Arthritis Rheum

2007;56:1910-20.

41 Sontheimer R D, Stastny P and Gilliam J N. Human histocompatibility antigen

associations in subacute cutaneous lupus erythematosus. J Clin Invest

1981;67:312-6.

42 Watson R M, Talwar P, Alexander E, et al. Subacute cutaneous lupus

erythematosus-immunogenetic associations. J Autoimmun 1991;4:73-85.

Page 23: pathogenesis of subacute cutaneous lupus erythematosus

23

43 Provost T T and Watson R. Anti-Ro(SS-A) HLA-DR3-positive women: the

interrelationship between some ANA negative, SS, SCLE, and NLE mothers

and SS/LE overlap female patients. J Invest Dermatol 1993;100:14S-20S.

44 Harley J B, Reichlin M, Arnett F C, et al. Gene interaction at HLA-DQ

enhances autoantibody production in primary Sjogren's syndrome. Science

1986;232:1145-7.

45 Callen J P, Hodge S J, Kulick K B, et al. Subacute cutaneous lupus

erythematosus in multiple members of a family with C2 deficiency. Arch

Dermatol 1987;123:66-70.

46 Provost T T, Arnett F C and Reichlin M. Homozygous C2 deficiency, lupus

erythematosus, and anti-Ro (SSA) antibodies. Arthritis Rheum 1983;26:1279-

82.

47 Meyer O, Hauptmann G, Tappeiner G, et al. Genetic deficiency of C4, C2 or

C1q and lupus syndromes. Association with anti-Ro (SS-A) antibodies. Clin

Exp Immunol 1985;62:678-84.

48 Walport M J, Davies K A, Morley B J, et al. Complement deficiency and

autoimmunity. Ann N Y Acad Sci 1997;815:267-81.

49 Pickering M C, Botto M, Taylor P R, et al. Systemic lupus erythematosus,

complement deficiency, and apoptosis. Adv Immunol 2000;76:227-324.

50 Rood M J, van Krugten M V, Zanelli E, et al. TNF-308A and HLA-DR3

alleles contribute independently to susceptibility to systemic lupus

erythematosus. Arthritis Rheum 2000;43:129-34.

51 Cripps D J and Rankin J. Action spectra of lupus erythematosus and

experimental immunofluorescence. Arch Dermatol 1973;107:563-7.

52 Epstein J H, Tuffanelli D and Dubois E L. Light Sensitivity and Lupus

Erythematosus. Arch Dermatol 1965;91:483-5.

Page 24: pathogenesis of subacute cutaneous lupus erythematosus

24

53 Freeman R G, Knox J M and Owens D W. Cutaneous lesions of lupus

erythematosus induced by monochromatic light. Arch Dermatol

1969;100:677-82.

54 Lehmann P, Holzle E, Kind P, et al. Experimental reproduction of skin lesions

in lupus erythematosus by UVA and UVB radiation. J Am Acad Dermatol

1990;22:181-7.

55 Runger T M, Epe B and Moller K. Processing of directly and indirectly

ultraviolet-induced DNA damage in human cells. Recent Results Cancer Res

1995;139:31-42.

56 Sutherland B M, Hacham H, Gange R W, et al. DNA damage and repair in

human skin: pathways and questions. Basic Life Sci 1990;53:149-60.

57 Davis P, Russell A S and Percy J S. Antibodies to UV light denatured DNA in

systemic lupus erythematosus: detection by filter radioimmunoassay and

clinical correlations. J Rheumatol 1976;3:375-9.

58 Utz P J, Hottelet M, Schur P H, et al. Proteins phosphorylated during stress-

induced apoptosis are common targets for autoantibody production in patients

with systemic lupus erythematosus. J Exp Med 1997;185:843-54.

59 Daniels F, Jr., Brophy D and Lobitz W C, Jr. Histochemical responses of

human skin following ultraviolet irradiation. J Invest Dermatol 1961;37:351-

7.

60 Caricchio R, McPhie L and Cohen P L. Ultraviolet B radiation-induced cell

death: critical role of ultraviolet dose in inflammation and lupus autoantigen

redistribution. J Immunol 2003;171:5778-86.

61 Aragane Y, Kulms D, Metze D, et al. Ultraviolet light induces apoptosis via

direct activation of CD95 (Fas/APO-1) independently of its ligand CD95L. J

Cell Biol 1998;140:171-82.

Page 25: pathogenesis of subacute cutaneous lupus erythematosus

25

62 Casciola-Rosen L A, Anhalt G and Rosen A. Autoantigens targeted in

systemic lupus erythematosus are clustered in two populations of surface

structures on apoptotic keratinocytes. J Exp Med 1994;179:1317-30.

63 Albert M L, Sauter B and Bhardwaj N. Dendritic cells acquire antigen from

apoptotic cells and induce class I-restricted CTLs. Nature 1998;392:86-9.

64 Tas S W, Quartier P, Botto M, et al. Macrophages from patients with SLE and

rheumatoid arthritis have defective adhesion in vitro, while only SLE

macrophages have impaired uptake of apoptotic cells. Ann Rheum Dis

2006;65:216-21.

65 Bijl M, Reefman E, Horst G, et al. Reduced uptake of apoptotic cells by

macrophages in systemic lupus erythematosus: correlates with decreased

serum levels of complement. Ann Rheum Dis 2006;65:57-63.

66 Ren Y, Tang J, Mok M Y, et al. Increased apoptotic neutrophils and

macrophages and impaired macrophage phagocytic clearance of apoptotic

neutrophils in systemic lupus erythematosus. Arthritis Rheum 2003;48:2888-

97.

67 Donnelly S, Roake W, Brown S, et al. Impaired recognition of apoptotic

neutrophils by the C1q/calreticulin and CD91 pathway in systemic lupus

erythematosus. Arthritis Rheum 2006;54:1543-56.

68 Korb L C and Ahearn J M. C1q binds directly and specifically to surface blebs

of apoptotic human keratinocytes: complement deficiency and systemic lupus

erythematosus revisited. J Immunol 1997;158:4525-8.

69 Fadok V A, Bratton D L, Konowal A, et al. Macrophages that have ingested

apoptotic cells in vitro inhibit proinflammatory cytokine production through

autocrine/paracrine mechanisms involving TGF-beta, PGE2, and PAF. J Clin

Invest 1998;101:890-8.

Page 26: pathogenesis of subacute cutaneous lupus erythematosus

26

70 Manfredi A A, Rovere P, Galati G, et al. Apoptotic cell clearance in systemic

lupus erythematosus. I. Opsonization by antiphospholipid antibodies. Arthritis

Rheum 1998;41:205-14.

71 Lovgren T, Eloranta M L, Bave U, et al. Induction of interferon-alpha

production in plasmacytoid dendritic cells by immune complexes containing

nucleic acid released by necrotic or late apoptotic cells and lupus IgG.

Arthritis Rheum 2004;50:1861-72.

72 Farkas L, Beiske K, Lund-Johansen F, et al. Plasmacytoid dendritic cells

(natural interferon- alpha/beta-producing cells) accumulate in cutaneous lupus

erythematosus lesions. Am J Pathol 2001;159:237-43.

73 Wenzel J, Worenkamper E, Freutel S, et al. Enhanced type I interferon

signalling promotes Th1-biased inflammation in cutaneous lupus

erythematosus. J Pathol 2005;205:435-42.

74 Wenzel J and Tuting T. Identification of type I interferon-associated

inflammation in the pathogenesis of cutaneous lupus erythematosus opens up

options for novel therapeutic approaches. Exp Dermatol 2007;16:454-63.

75 Stephansson E and Ros A M. Expression of intercellular adhesion molecule-1

(ICAM-1) and OKM5 in UVA- and UVB-induced lesions in patients with

lupus erythematosus and polymorphous light eruption. Arch Dermatol Res

1993;285:328-33.

76 Dorner T, Hucko M, Mayet W J, et al. Enhanced membrane expression of the

52 kDa Ro(SS-A) and La(SS-B) antigens by human keratinocytes induced by

TNF alpha. Ann Rheum Dis 1995;54:904-9.

77 Zampieri S, Alaibac M, Iaccarino L, et al. Tumour necrosis factor alpha is

expressed in refractory skin lesions from patients with subacute cutaneous

lupus erythematosus. Ann Rheum Dis 2006;65:545-8.

Page 27: pathogenesis of subacute cutaneous lupus erythematosus

27

78 Shen J, Bao S and Reeve V E. Modulation of IL-10, IL-12, and IFN-gamma in

the epidermis of hairless mice by UVA (320-400 nm) and UVB (280-320 nm)

radiation. J Invest Dermatol 1999;113:1059-64.

79 Barker J N, Sarma V, Mitra R S, et al. Marked synergism between tumor

necrosis factor-alpha and interferon-gamma in regulation of keratinocyte-

derived adhesion molecules and chemotactic factors. J Clin Invest

1990;85:605-8.

80 Jones S M, Mathew C M, Dixey J, et al. VCAM-1 expression on endothelium

in lesions from cutaneous lupus erythematosus is increased compared with

systemic and localized scleroderma. Br J Dermatol 1996;135:678-86.

81 Nyberg F, Hasan T, Skoglund C, et al. Early events in ultraviolet light-induced

skin lesions in lupus erythematosus: expression patterns of adhesion molecules

ICAM-1, VCAM-1 and E-selectin. Acta Derm Venereol 1999;79:431-6.

82 Norris P, Poston R N, Thomas D S, et al. The expression of endothelial

leukocyte adhesion molecule-1 (ELAM-1), intercellular adhesion molecule-1

(ICAM-1), and vascular cell adhesion molecule-1 (VCAM-1) in experimental

cutaneous inflammation: a comparison of ultraviolet B erythema and delayed

hypersensitivity. J Invest Dermatol 1991;96:763-70.

Page 28: pathogenesis of subacute cutaneous lupus erythematosus

28

Genetic Susceptibility

+

UVL

Suprabasal Keratinocytes

Increased Rate of Apoptosis

Decreased Clearance of Apoptotic Debris

DNA Damage

Processing by APC/DC

Autoantigen Presentation

Initiation Phase

Primary Autoimmune Response

B autoreactive clones/anti-

Ro, anti-La

T autoreactive clones

Adhesion Molecules (ICAM-1)

Proinflammatory Cytokines (TNFα, IL-1)

Redistribution of Autoantigens (Ro, La)

Amplification Phase of Autoimmune Response

Recruitment of Leukocytes

pDC

IFNα induced Chemokines

Endothelium

Adhesion Molecules

(ICAM-1, VCAM-1, E-selectins)

Injury Phase

ICs, Direct Antibody Effect,

ADCC, CD8 cytotoxicity,

Figure 1. Current immunological concepts in the pathogenesis of SCLE.