You are on page 1of 9

Vol. 122 No.

1 July 2016

Etiology and pathogenesis of oral lichen planus: an overview


Zoya B. Kurago, DDS, PhD

Oral lichen planus is a noninfectious, chronic inflammatory condition that involves the oral mucosal stratified
squamous epithelium and the underlying lamina propria and may be accompanied by skin lesions. This overview describes
the current understanding of the immunopathologic mechanisms implicated in oral lichen planus. (Oral Surg Oral Med Oral
Pathol Oral Radiol 2016;122:72-80)

Oral lichen planus (OLP) is recognized as a noninfec- response to epithelial antigens versus a dysregulated
tious, chronic inflammatory condition that involves the response to external antigens, and (4) viral infections.
oral mucosal stratified squamous epithelium and the Hypersensitivity reactions generally align with
underlying lamina propria and may be accompanied by lichenoid mucositis, an umbrella term for OLP-like
skin lesions. The causes that initiate and/or perpetuate mucositis in response to restorative and other mate-
OLP (with or without skin lesions) are, for the most part, rials (e.g., amalgam), reactions to drugs, and so on.
unknown. The prevailing theories revolve around dys- Hypersensitivity reactions usually resolve upon
regulated T-cell-mediated disorder to exogenous triggers removal of the trigger, and do not maintain a chronic
as opposed to a dysregulated response to autologous course, as is characteristic of OLP. Drugs may also be
keratinocyte antigens (autoimmune), and definitive data implicated in exacerbations of pre-existing OLP, which
necessary to resolve this dilemma, in particular, the can create challenges in patient management.
initiating triggers and the target antigens, are currently Stress is thought to play a role in the pathogenesis of
missing. Common difficulties in the study of OLP are OLP because anxiety and depression are reportedly
related to the overlap between the features of OLP and more common in patients with OLP in comparison with
those of other oral mucosal conditions, the highly vari- normal controls,2 and OLP exacerbations correlate with
able application of diagnostic criteria, and the potential episodes of anxiety.3,4 However, a cause-and-effect
coexistence of additional non-OLP inflammatory con- relationship between stress and the onset of OLP has
ditions in the same patients. Nevertheless, the growing not been demonstrated. Moreover, studies show that
database of information about this disorder suggests acute and chronic psychological stress can induce
certain immune response patterns. The accumulating pronounced changes in innate and adaptive immune
knowledge will eventually facilitate a more reliable responses. These immune response changes are
separation of this condition from other disorders that are predominantly mediated via neuroendocrine mediators
typically included on the differential diagnosis list. from the hypothalamicepituitaryeadrenal axis and the
In contrast to OLP, cutaneous LP is typically a self- sympatheticeadrenal axis (reviewed by Kemeny and
limiting condition (except for the hypertrophic form) Schedlowski5). The implication of these studies is that
and usually resolves within 6 to 12 months,1 suggesting similar to other inflammatory conditions of various
that the underlying mechanisms in OLP and cutaneous causes, stress is more likely to play a secondary,
LP may be distinct. It is not clear yet whether the rather than a primary, role in OLP pathogenesis.
mechanisms driving isolated OLP are different from Autoimmune response to epithelial self-antigens
those driving OLP with cutaneous lesions. The focus remains a possibility. A single study of cutaneous LP
of this overview is on OLP. reported evidence in support of autoimmunity by
A number of potential OLP triggers have been pro-
posed, mainly (1) local and systemic inducers of cell-
mediated hypersensitivity, (2) stress, (3) autoimmune
Statement of Clinical Relevance
Oral lichen planus (OLP), a common inflammatory
Associate Professor, Departments of Oral Health and Diagnostic disorder of unknown cause, can result in significant
Sciences and Oral Biology, Augusta University Dental College of morbidity. As studies investigate various aspects of
Georgia; Department of Pathology, Augusta University Medical this condition, updates on the accumulating knowl-
College of Georgia; Augusta University Cancer Center, Augusta, GA,
edge are of value to clinicians who manage patients
USA.
Received for publication Aug 25, 2015; returned for revision Mar 2, with this disorder. This overview is focused on the
2016; accepted for publication Mar 9, 2016. current understanding of OLP immunopathogenesis
Ó 2016 Elsevier Inc. All rights reserved. in an effort to facilitate better management of pa-
2212-4403/$ - see front matter tients with OLP.
http://dx.doi.org/10.1016/j.oooo.2016.03.011

72
OOOO REVIEW ARTICLE
Volume 122, Number 1 Kurago 73

expanding in vitro T cells isolated from the skin lesions


of two patients, followed by testing the ability of these
T cells to kill autologous keratinocytes (cytotoxicity).6
Besides the lack of other convincing studies that
support the autoimmune mechanism, it is difficult to
reconcile autoimmunity with the typically self-limiting
course of cutaneous LP. As stated above, although the
immune response in OLP is dysregulated, none of the
reported studies to date has provided definitive evi-
dence for autoimmunity in OLP, that is, a response to
self-antigens. Current knowledge about immune system
cells and molecules participating in OLP pathogenesis
is summarized below.
Contributions of viral infections are discussed under
“OLP Associations with Microorganisms and Other
Systemic Disorders.”

OLP IMMUNOPATHOGENESIS
Investigations of immune system activities in OLP have
focused largely on erosive and reticular forms, some-
times comparing the two. Published reports include
evaluations of biopsy specimens, saliva, and blood.
Studies that have examined changes in the composition
of untreated OLP lesions over time are lacking; this is a
limitation, given the chronic waxing and waning course
of OLP. Following is a summary of published papers,
complemented by a diagram and a table of important
cells and soluble factors (Figure 1, Table I). A
Fig. 1. Cells and molecules implicated in the pathogenesis of
hypothetical model of interactions implicated in OLP
OLP. The following hypothetical model of cell-mediated
immunopathogenesis combines data from OLP studies and
pathogenesis is included in the legend to Figure 1.
known immune system functions. An unidentified trigger
initiates cell/tissue damage and the immune response by Cells
activating the resident myeloid DC (Langerhans cells and/or
T cells and NK cells. Established OLP lesions are
stromal DC). Activated DC loaded with antigen undergo
maturation, produce cytokines and chemokines, and migrate
typically found to contain T cells with alpha-beta T-cell
to regional lymph nodes to present the processed antigen to T receptors, including CD4þ (“helper”) and CD8þ (“cyto-
cells. Some mature DC remain in the mucosal lamina propria toxic”) T cells, both within the epithelium and lamina
and produce IL-12, IL-18 and TNF-alpha. The apparent lack propria infiltrates. Both cell subsets can be involved in a
of B cells and antibodies in OLP suggests the absence of type 1 immune response, where CD4þ T cells produce
soluble antigen. Mast cells in the damaged tissue release Th1 factors, and CD8þ cytotoxic T cells kill host cells
granules with inflammatory mediators. Various resident cells and may contribute type 1 soluble factors. The vast
produce chemokines CCL5, CXCL9, CXCL10, which majority of oral mucosal and peripheral blood studies
particularly attract Th1 and cytotoxic T cells. The vascular show a dominant type 1 (Th1) cell-mediated immune
endothelial cells in the lamina propria also respond by pro- response. This response likely includes cell-mediated
ducing chemerin, which attracts plasmacytoid DC and NK
cytotoxicity, as CD8þ T cells were often found at the
cells. Plasmacytoid DC interact with myeloid DC, T cells and
NK cells and secrete IFN-alpha. T cells and NK cells are
further activated by IL-12, IL18 and IFN-alpha to release IFN- stimulates parakeratosis that presents clinically as white pla-
gamma and TNF-alpha, which in turn activates DC. Some ques or striae. Erosive OLP, but not reticular OLP, is asso-
cytotoxic CD8þ T cells kill basal/parabasal keratinocytes ciated with Th17 CD4þ T cells, a source of IL-17. Whether
using perforin and granzyme, resulting in apoptotic (‘colloid’) IL-17 is an initiator or a consequence of the destruction that
bodies. TNF-alpha activates anti-microbial responses and may results in mucosal ulceration, is not known. The lack of
kill host cells. IFN-gamma stimulates the expression of MHC myeloid or lymphoid suppressor cells in OLP likely also
class II on keratinocytes, which may facilitate their antigen- contributes to the chronic and destructive inflammation. It is
dependent interaction with CD4þ T cells. How this interaction unclear what a gut barrier-associated cytokine IL-22 or TLR
affects the process is not clear. Equally unclear is what may be contributing to the OLP immunopathogenesis.
ORAL AND MAXILLOFACIAL PATHOLOGY OOOO
74 Kurago July 2016

Table I. Cells and soluble factors found in OLP OLP patients.11 Moreover, Th1 cells produce interferon-
Cells Functions in OLP
gamma (IFN-g), and IFN-g-positive cells were identified
in OLP.7 Natural killer (NK) cells also can produce
Myeloid dendritic cells (DC) Produce IL-12, TNF-a, (IL-18*)
Chemokines; interact with T cells IFN-g, and cells with NK cell markers (CD56
Plasmacytoid DC Produce IFN-a, chemokines; interact
dim
CD16þ) have been identified in OLP lesions.12 This
with T cells NK cell subset is known for cytotoxicity and the ability
Th1 T cells Produce IFN-g, TNF-a to produce TNF-a and IFN-g,13,14 but NK cell func-
Th17 T cells Produce IL-17
tions in OLP are not defined. Together, OLP-associated
Cytotoxic T cells Cytotoxicity; produce IFN-g*
NK cells Produce IFN-g*; cytotoxicity* IFN-g-positive cells could be Th1 cells and/or NK cells.
Mast cells Produce TNF-a, proteases, Another proinflammatory CD4þ T cell subset iden-
vasoactive mediators, MMPs, tified recently in OLP in high numbers is Th17,15 which
chemokine CCL5 is responsible for production of interleukin-17 (IL-17)
Soluble factors rather than IFN-g. Th17 T cells are important contrib-
Chemokines Likely effects utors to the defense against many bacterial and fungal
CCL5 Attract T cells, other leukocytes pathogens and to autoimmune conditions.16
CXCL9, CXCL10 Attract Th1 and cytotoxic T cells Not surprisingly, regulatory T cells (Treg), which are
Chemerin Attract NK cells well known for suppressing inflammation, were rare in
Cytokines Potential effects both erosive and nonerosive OLP lesions.15 As already
IL-12 Stimulate IFN-g expression (T cells, emphasized, the antigens to which either CD4þ or
NK cells) CD8þ T cells are responding in OLP are unknown.
IL-18 Complex effects depending on
Antigen-presenting cells. Professional antigen pre-
context; stimulate TNF-a and
IFN-g expression, T-cell and sentation to T cells leading to T-cell activation or
NK-cell cytotoxicity tolerance depends largely on dendritic cells (DC) and
IFN-a Stimulate MxA expression typically occurs in secondary lymphoid organs, such as
(keratinocytes, leukocytes) lymph nodes. Interestingly, studies have suggested that
IFN-g Keratinocyte apoptosis, MHC class II
DCeT-cell interactions leading to T-cell activation may
expression
TNF-a Antimicrobial activity, cell apoptosis be occurring on site within OLP lesions. The DC sub-
IL-17 (erosive OLP) Proinflammatory: antibacterial, sets characterized in OLP include mature and immature
antifungal, autoimmune conditions myeloid DC (both Langerhans cells and stromal DC)
IL-22 GI barrier maintenance and plasmacytoid DC.17 Mature DC, which are DC
Other factors Potential effects expressing all the molecules required for T-cell
MMPs Matrix degradation activation, were found mainly in the lamina propria in
Factors in saliva Potential effects clusters with each other and with T cells.17 Mature
IL-17 Marker of mucosal inflammation myeloid DC can produce the Th1 cytokine, IL-12.
Soluble TLR2 Not known However, plasmacytoid DC are the major producers
Soluble TLR4 Not known of IFN-a, another cytokine that stimulates a type 1
DC, dendritic cell; GI, gastrointestinal; IFN, interferon; IL, inter- immune response18 by inducing the expression of MxA
leukin; MHC, major histocompatibility complex; MMP, matrix met- and other genes.17,19 Consistent with IFN-a production,
alloproteinase; MxA, human myxovirus resistance protein 1; NK, epithelial and inflammatory cells in OLP lesions
natural killer; OLP, oral lichen planus; TNF, tumor necrosis factor;
TLR, toll-like receptor.
strongly expressed MxA.12,17 In the context of activated
*The stated activity has not been demonstrated in OLP but is likely mature plasmacytoid DC, IFN-a, and Th1 T cells, NK
based upon the known functions of the cells. cells are also likely to contribute to the type 1 response.
Macrophages also perform antigen presentation to
epithelial-connective tissue interface and sometimes T cells and are normal residents of the oral mucosa.
adjacent to apoptotic keratinocytes.7,8 Moreover, Their participation and contributions to OLP have not
cytotoxicity-related molecules perforin, Tia-1, and gran- been fully characterized.
zyme were all detected in the epithelium and the con- B lymphocytes, which, in addition to antibody pro-
nective tissue,9,10 which occurred much more often in duction, also perform antigen presentation, appear to
OLP than in cutaneous LP.9,10 Immune-mediated cell contribute little, if anything, in OLP,20 but a detailed
death can occur through several mechanisms. The spe- analysis of B cells in OLP has not been provided in
cific mechanism of cytotoxicity directed at keratinocytes the literature. Similarly, terminally differentiated
in OLP is still unclear. Other parameters indicating a Th1 B cells (plasma cells) and antibodies have not been
profile include the Th1 T cell regulator, transcription found to contribute to OLP.20
factor T-bet, which prevailed over Th2-related tran- Mast cells. Finally, activated mast cells have been
scription factor GATA-3 in peripheral blood T cells of described in OLP.21 Mast cells, normally associated
OOOO REVIEW ARTICLE
Volume 122, Number 1 Kurago 75

with mucosal vasculature and nerves, may contribute diagnostic criteria (e.g., lichenoid mucositis vs OLP,
several soluble inflammatory mediators (Table I)21-23 as hypersensitivities are often associated with Th2
and facilitate the migration of various inflammatory responses), the simultaneous presence of other
cells from the bloodstream. inflammatory oral non-OLP conditions (e.g., peri-
It is important to note that the characteristic odontal disease), and potential ethnic differences.
composition of the inflammatory infiltrate in OLP- In recent studies, a proinflammatory Th17 cytokine
related ulcers and erosions and in the marginal IL-17 was found to be significantly increased in the
gingiva is distorted. This change in ulcers and erosions erosive OLP lesions35 and saliva of patients with
is, at least partly, caused by surface breach and expo- erosive OLP, but not in those with reticular OLP.36
sure of the lamina propria to the oral microflora. Plaque IL-17 is produced by Th17 T cells and several other
microflora likely contributes to the immune response in cell types. IL-17 is very important for clearance of
the marginal gingiva. In these areas, other inflammatory many bacterial and fungal pathogens and leads to
cells are present, particularly neutrophils, which severe inflammation with marked tissue destruction.16
respond to microorganisms and necrosis. As a result, Based on known IL-17 activities, it may be a contrib-
the mechanisms operating in such lesions are more utor to, and/or a consequence of, surface erosions and
complex, and the morphology of such lesions can ulcerations in OLP.
compromise the accuracy of the diagnosis. Incisional Other cytokines also identified in OLP are not
biopsy samples that exclude erosions, ulcers, and necessarily affiliated with a specific inflammatory profile
marginal gingiva are better for the histopathologic (e.g., IL-6). Yet another cytokine of interest that has
diagnosis of OLP. been described recently in OLP is IL-22.37 IL-22 can be
made by a subset of T cells and several other immune
Soluble factors system cells. The main function of IL-22 in the gut is to
Soluble factors include cytokines, growth factors, che- stimulate epithelial proliferation to support and maintain
moattractants, and enzymes. Both locally and in the the gastrointestinal (GI) epithelial barrier. IL-22 also
peripheral blood of OLP patients, the identified soluble facilitates barrier defense against bacterial pathogens by
factors also indicate a type 1 immune response.7,24-27 stimulating the production of antimicrobial pep-
Cytokines. Some of the cytokines have already been tides.38,39 IL-22 is implicated in multiple conditions with
mentioned earlier (IL-12, IFN-g, IFN-a). Tumor dysregulated immune responses, including autoimmune
necrosis factor-alpha (TNF-a) is another destructive disorders.38,39 The sources and functions of IL-22 in
cytokine associated with type 1 responses, and it also OLP are not known, and further studies are required.
stimulates antimicrobial activities. Important producers As cytokines are critical factors in immune re-
of TNF-a are T cells, NK cells, DC, and macrophages. sponses, the genetic regulation of their expression is
IL-12, IFN-g, and TNF-a are significantly elevated expected to affect susceptibility to, and severity of,
over the prototypical Th2 cytokine IL-4,26,27 but inflammatory diseases, such as OLP. Consistent with
Th2-associated IL-10 and IL-13 would benefit from this expectation, multiple studies have shown associa-
additional analysis. IL-12 stimulates IFN-g production tions with certain sequence variations (functional
and activates T-cell and NK-cell cytotoxicity. In addi- polymorphisms) in the regulatory regions of cytokine
tion to activating antimicrobial defenses, which are genes implicated in OLP, particularly the Th1 cytokines
particularly helpful against intracellular microorgan- TNF-a,25,40-42 IFN-g,25,43,44 IL-12,45 and IL-18.31 A
isms, IFN-g stimulates major histocompatibility com- positive correlation between certain polymorphisms
plex (MHC) class II expression. Consistent with this and the levels of cytokine production in OLP has
effect, MHC class IIþ keratinocytes (which normally been shown for IL-18.31 The details of specific
express only MHC class I) have been described in polymorphisms vary somewhat with study populations.
OLP.28 MHC class II expression can then permit a Chemokines. Identified chemoattractants (chemo-
direct antigen-specific interaction between keratino- kines), the major function of which is to regulate cell
cytes and CD4þ T cells.29 Yet another cytokine migration, also support a Th1 profile in OLP. In
identified in OLP is IL-18,30,31 which can have contrast to myeloid DC and mast cells, as mentioned
similar effects on cytotoxicity and IFN-g production to above, a-b T cells, NK cells, and plasmacytoid DC are
those of IL-12. IL-18 may be produced by antigen not normal residents of the oral mucosa. Depending on
presenting cells and epithelial cells.32 The sources of conditions, mucosal resident myeloid DC differentially
IL-18 in OLP have not been defined. express either Th1-recruiting CXCL9, 10, and 11, or
Only a few reports have suggested a dominant Th2 Th2-recruiting chemokines.46 In OLP lesions
profile33 or a mixed Th1-Th2 profile,34 raising (including the epithelium) and in peripheral blood,
questions about representative specimens (e.g., CXCL9, CXCL10, and several general inflammation-
proximity to marginal gingiva), application of associated chemokines have been identified.47
ORAL AND MAXILLOFACIAL PATHOLOGY OOOO
76 Kurago July 2016

Moreover, CD8þ T cells in OLP were found to express TNF-a biologics have not seen wide application.55
the appropriate chemokine receptors CXCR3 and Studies comparing the effects of various treatments
CCR5 and produce their own CXCL10, possibly for erosive OLP have been reviewed recently.56
leading to self-recruitment.48 In the vast majority of
OLP cases analyzed, NK cells in OLP lesions were OLP PATHOGENESIS AND POTENTIAL FOR
mainly in the lamina propria and, in contrast to ORAL SQUAMOUS CELL CARCINOMA
T cells, expressed ChemR23, a receptor for the There are numerous publications focused on OLP as a
chemoattractant chemerin produced by the endothelial possible precursor to squamous cell carcinoma (SCC),
cells of the mucosal vessels.12 Altogether, the some of which are listed here.26,57-63 So far, this question
chemokine and chemokine receptor profiles in OLP remains unresolved for a variety of reasons, including
are consistent with a type 1 immune response. inconsistent application of diagnostic criteria, insufficient
During any inflammatory response, other factors are documentation of all relevant information, incomplete
induced as well. Among them are reactive oxygen long-term follow-up, and lack of appropriate control
species that are important for antimicrobial activities populations. These limitations most likely contribute to
but also capable of inducing DNA damage. Some are the highly variable rates of oral SCC reported in patients
enzymes that contribute to matrix degradation (matrix with OLP (0.4%e12.5%).26,57-63
metalloproteinases), which facilitates cell migration and For the purposes of this overview, the issue of
removal of debris. Some are growth factors that regu- carcinogenesis in the context of OLP is of some rele-
late the proliferation and differentiation of cells. Such vance because longstanding chronic inflammation in
factors are common in various inflammatory settings, some conditions is associated with increased risk of
including OLP. cancer. For example, chronic noninfectious inflamma-
OLP immunopathogenesis and response to treat- tory bowel diseases (IBD), that is, ulcerative colitis and
ment. Following are a few points regarding response to Crohn disease, are associated with risk of colorectal
treatment that speak to the immunopathogenesis of adenocarcinoma. This risk increases over time,
OLP. The most widely used treatment for OLP is depending on the population studied, from 1% or less at
immunosuppression with topical or systemic cortico- 10 years to much higher chances at 30 years.64
steroids. Corticosteroids are effective because they Although there is some overlap in the
inhibit the activation of DC and T cells, secretion of immunopathologic mechanisms of IBD and OLP,
Th1 cytokines,49,50 as well as stimulate Th2 cytokine there are also significant differences between them,
IL-10 that also acts to interfere with Th1 cytokine between the oral mucosa and the intestinal mucosa,
activities.51 However, not all OLP cases respond to and between the carcinoma types, so much remains to
corticosteroids. Provided that the diagnosis of OLP is be learned. One unifying feature is that certain aspects
accurate and no other complicating condition is of chronic inflammation at the mucosal surfaces
present, some patients with OLP may be resistant to stimulate healing and repair and improve cell survival
steroids. This phenomenon is also seen in other while promoting proliferation, which make cells more
noninfectious inflammatory conditions (inflammatory vulnerable to mutagenesis. Mutagenesis may be
bowel disease, rheumatoid arthritis, asthma, etc.).49,52 induced by exogenous (e.g., chemical carcinogens) or
Resistance to steroids has a genetic basis, illustrated, endogenous (e.g., reactive oxygen species) triggers.
in part, by the wide variation in lymphocyte sensitivity How T-cell-mediated keratinocyte death in OLP
to steroids even in healthy individuals.52-54 With factors into the potential scenario is not clear.
functional polymorphisms in genes that encode Cytotoxicity aside, in OLP patients, it is theoretically
inflammatory mediators being responsible for disease possible that certain genetic predispositions combined
severity, additional polymorphisms in glucocorticoid with longstanding chronic activation of key
response pathways may further affect treatment options. inflammation-related prosurvival factors make the
For example, a specific polymorphism in the regulatory multistep process of initiation and progression of
sequence of the TNF-a gene is well known for carcinogenesis easier. In other words, the triggers of
imparting steroid resistance in several systemic in- mutagenesis (whether endogenous and/or exogenous)
flammatory disorders.49 in progenitor epithelial cells under such conditions are
In cases of steroid resistance, calcineurin inhibitors less likely to kill the cells and more likely to lead to
are often useful because they target T-cell activation malignant transformation. If this, indeed, is true in
and proliferation directly. Other options include bi- OLP, then controlling inflammation should be impor-
ologics, which can target individual cytokines or their tant for the prevention of chronic inflammation-related
receptors.55 The best known target is TNF-a activity carcinogenesis. With accumulating knowledge about
(the cytokine or its receptor), although in OLP anti- how various immunoregulatory drugs affect
OOOO REVIEW ARTICLE
Volume 122, Number 1 Kurago 77

inflammation and cancer risk, it will be possible to circulating IFN-g and CXCL10,66 and there are several
make better decisions about management of patients extrahepatic complications of HCV associated with a
with chronic inflammatory conditions. type 1 immune response. In this context, OLP lesions
fit the profile, but the relationship between OLP and
OLP ASSOCIATIONS WITH other extrahepatic HCV-associated disorders is unclear.
MICROORGANISMS AND OTHER DISORDERS
Viral infections
Bacteria
A number of potential associations between specific
viral infections and OLP have been investigated, Apart from the studies on Helicobacter pylori, for which
no association with OLP has been found,65 bacterial
including cytomegalovirus, herpes simplex virus-1,
studies are in early stages. Microorganisms may be
human herpesvirus-6, Epstein-Barr virus, hepatitis B
triggers or could be responsible for sustaining or
virus, and human papillomavirus, without revealing
exacerbating the chronic course of OLP. Significant
significant correlations.65 Currently, there is convincing
differences in bacteria were identified by 16S rRNA
evidence that, at least in some geographic regions, OLP
sequencing in the saliva of patients with erosive OLP
is associated with hepatitis C virus (HCV) infection.
versus patients with reticular OLP versus normal
The HCVeOLP association is likely related to the
propensity for several other common extrahepatic controls.36 Of interest are observations that
Fusobacteria and Campylobacter were significantly
effects of HCV infection, such as cryoglobulinemia
increased, whereas Porphyromonas were decreased in
and several autoimmune disorders.66 Analysis of eight
saliva of patients with erosive OLP relative to normal
qualified studies revealed a significantly higher
controls.36 Relevant to microbial colonization and OLP
probability of circulating anti-HCV antibodies or
pathogenesis are studies of toll-like receptors (TLRs), a
HCV RNA in patients with OLP (with or without skin
subset of pattern recognition receptors for microbial
lesions), with an overall odds ratio (OR) of 5.71 (95%
molecules. TLR expression in saliva and in OLP biopsy
confidence interval [CI] 3.48e9.37).67 Similar results
were reported in Chinese studies68 (OR 5.4; 95% CI samples was reportedly different from those in normal
controls.73-77 Removal of plaque and calculus improved
3.5e8.3). Seropositivity for anti-HCV antibodies is
the management of patients with gingival OLP,65 as did
believed to represent an active infection, as 75% to 85%
the use of antimicrobial rinses (personal clinical
of seropositive individuals have persistent chronic in-
experience), suggesting that bacteria may contribute to
fections. The same systematic review67 found
OLP-associated inflammation. Together, microbial and
geographic regional differences: a strong HCVeOLP
pattern recognition receptor studies have the potential to
association in Mediterranean populations (OR 6.63;
greatly improve our understanding of OLP, its subtypes,
95% CI 4.68e9.40), in contrast to Northern European
studies (OR 2.14; 95% CI 0.59e7.69). Two studies and the relationship with other inflammatory disorders.
also supported the opposite: HCV patients are more
likely to develop OLP (OR 2.5; 95% CI 2.0e3.168 OLP AND OTHER SYSTEMIC DISORDERS
and OR 4.47; 95% CI 1.84e10.86,67 respectively). Ideally, multicenter studies with inclusion of relevant
HCVeOLP associations were also suggested in Japan controls are needed to obtain sufficient data for this type
and the United States, but not in Ethiopia or Egypt,69 of analysis. There are reports of association of OLP and
although some of the studies were quite small. There cutaneous LP with several autoimmune disorders (sys-
is still ongoing debate regarding the impact of specific temic lupus erythematosus, Sjögren syndrome, and
HCV genotypes on the likelihood and strength of the others), which lack documentation of criteria used to
OLPeHCV association. The geographic variation in diagnose OLP. Unfortunately, careful studies of asso-
the HCVeOLP association could be attributed to ciations between OLP and other noninfectious systemic
population genetics, environmental factors, or even conditions that uniformly apply consistent diagnostic
HCV genotype, and so far, answers to these questions criteria for OLP using both clinical and histopathologic
have not been found.67,69 evaluations are rare. Without this approach, other ul-
The mechanisms of OLP development in patients with cerative and red/white oral mucosal lesions can
HCV are not clear. No effect of HCV genotype or viral complicate the results. Moreover, many studies do not
load on OLP prevalence was identified,70,71 and detec- separate OLP from cutaneous LP.
tion of HCV in OLP lesions is controversial. Moreover, With that said, several studies suggest a possible
HCV lacks tropism for cells other than hepatocytes, as it association between OLP and thyroid disease. Among
requires liver-specific miR122 for replication.72 The thyroid disorders, Hashimoto thyroiditis (HT)78 and
immune response to HCV infection is mainly type 1, hypothyroidism79 were identified. According to the
that is, dominated by IFNs, TNF-a, and cytotoxicity report of 15 cases in Italy,78 HT in OLP patients was
mediated by CD8þ T cells, with high levels of significantly more common (w14%) than HT-related
ORAL AND MAXILLOFACIAL PATHOLOGY OOOO
78 Kurago July 2016

hypothyroidism in the general population (w1%). profile appears to be another contributing factor. Patient
Further, HT preceded the onset of reticular OLP in 93% responses to treatment with corticosteroids and calci-
of the study group. The diagnosis of OLP in all patients neurin inhibitors further support the role of T cells and
was based on defined clinical and histopathologic Th1 factors in OLP immunopathogenesis. Based on
criteria.80 Another study79 of 152 OLP cases diagnosed available data summarized above, destruction of
using the same criteria,80 patients with hypothyroidism keratinocytes in OLP could occur as a result of auto-
were approximately twofold more likely to present with reactivity or as a bystander effect. There is a signifi-
OLP compared with individuals without cant association between OLP and HCV infection in
hypothyroidism. The reticular form of OLP was the some geographic regions. The role of oral microflora in
most common type. There was no evidence in this OLP pathogenesis remains to be determined. How the
study that thyroxine treatment had an impact on the potential association between OLP and thyroid disor-
oral lesions. The mechanisms that could explain the ders may factor into the immunopathogenesis of OLP is
described associations are not known. not clear. To further advance our understanding of this
Evidence for other OLP associations is difficult to disorder, there is a need for more studies that use well-
confirm for reasons stated earlier. This difficulty is documented OLP cases diagnosed with consistent
illustrated in reports that describe a possible link between clinical and pathologic criteria and include appropriate
OLP and Good syndrome. Good syndrome is a rare controls.
condition consisting of thymoma with an adult-onset
immunodeficiency, which is characterized by hypo- The author thanks Dr. Y.-S. Lisa Cheng, DDS, PhD, for her
valuable input.
gammaglobulinemia, low or absent B cells, and variable
defects in cell-mediated immunity that involves CD4þ
T cells.81 Thymoma without immunodeficiency is
associated with several autoimmune conditions, REFERENCES
including myasthenia gravis, pure red cell aplasia, 1. Gorouhi F, Davari P, Fazel N. Cutaneous and mucosal lichen planus: a
systemic lupus erythematosus, polymyositis.82 Because comprehensive review of clinical subtypes, risk factors, diagnosis,
of humoral and cell-mediated immunodeficiency, and prognosis. Scientific World Journal. 2014;2014:742826.
patients with Good syndrome also have a variety of viral, 2. Rojo-Moreno JL, Bagan JV, Rojo-Moreno J, Donat JS,
Milian MA, Jimenez Y. Psychologic factors and oral lichen pla-
bacterial, and fungal infections of the upper respiratory, nus. A psychometric evaluation of 100 cases. Oral Surg Oral Med
gastrointestinal, and urinary tracts, as well as infections Oral Pathol Oral Radiol Endod. 1998;86:687-691.
of the skin, joints, bone, and the central nervous sys- 3. McCartan BE. Psychological factors associated with oral lichen
tem.82 Most reported cases of OLP associated with Good planus. J Oral Pathol Med. 1995;24:273-275.
4. Eisen D. The clinical features, malignant potential, and systemic
syndrome83-85 are based on loose application of clinical
associations of oral lichen planus: a study of 723 patients. J Am
features only. In view of known coexistence of multiple Acad Dermatol. 2002;46:207-214.
infections and autoimmune disorders in patients with 5. Kemeny ME, Schedlowski M. Understanding the interaction be-
Good syndrome, oral mucositis from causes other than tween psychosocial stress and immune-related diseases: a step-
OLP is a distinct possibility. Maehara et al. described wise progression. Brain Behav Immun. 2007;21:1009-1018.
two cases of oral mucositis in the context of Good 6. Sugerman PB, Satterwhite K, Bigby M. Autocytotoxic T-cell
clones in lichen planus. Br J Dermatol. 2000;142:449-456.
syndrome and apparently found differences in several 7. Khan A, Farah CS, Savage NW, Walsh LJ, Harbrow DJ,
immunologic parameters of the oral mucosal lesions Sugerman PB. Th1 cytokines in oral lichen planus. J Oral Pathol
relative to those in seven OLP samples.86 An important Med. 2003;32:77-83.
limitation of this study is that the clinical and 8. Sugerman PB, Savage NW, Zhou X, Walsh LJ, Bigby M. Oral
histopathologic criteria used for diagnosis were not lichen planus. Clin Dermatol. 2000;18:533-539.
9. Santoro A, Majorana A, Bardellini E, Festa S, Sapelli P,
provided. Moreover, the photomicrographs depicting Facchetti F. NF-kappaB expression in oral and cutaneous lichen
examples of immunohistochemistry are of very low planus. J Pathol. 2003;201:466-472.
resolution. Altogether, the link between OLP and Good 10. Lage D, Pimentel VN, Soares TC, Souza EM, Metze K, Cintra ML.
syndrome is not supported by the available evidence. Perforin and granzyme B expression in oral and cutaneous lichen
planusda comparative study. J Cutan Pathol. 2011;38:973-978.
11. Lu R, Zhou G, Du G, Xu X, Yang J, Hu J. Expression of T-bet
CONCLUSIONS and GATA-3 in peripheral blood mononuclear cells of patients
In summary, mucosal damage in erosive and reticular with oral lichen planus. Arch Oral Biol. 2011;56:499-505.
OLP is largely caused by a type 1 immune response that 12. Parolini S, Santoro A, Marcenaro E, et al. The role of chemerin in
involves myeloid and plasmacytoid DC, CD4þ and the colocalization of NK and dendritic cell subsets into inflamed
CD8þ T cells, NK cells, and mast cells, without tissues. Blood. 2007;109:3625-3632.
13. Bjorkstrom NK, Riese P, Heuts F, et al. Expression patterns of
apparent contributions by B lymphocytes, plasma cells, NKG2 A, KIR, and CD57 define a process of CD56 dim NK-cell
or antibodies. The dominant cytokines and chemokines differentiation uncoupled from NK-cell education. Blood.
are consistent with a Th1 profile. In erosive OLP, Th17 2010;116:3853-3864.
OOOO REVIEW ARTICLE
Volume 122, Number 1 Kurago 79

14. Moretta L. Dissecting CD56 dim human NK cells. Blood. 36. Wang K, Miao T, Lu W, et al. Analysis of oral microbial com-
2010;116:3689-3691. munity and Th17-associated cytokines in saliva of patients with
15. Vered M, Furth E, Shalev Y, Dayan D. Inflammatory cells of oral lichen planus. Microbiol Immunol. 2015;59:105-113.
immunosuppressive phenotypes in oral lichen planus have a 37. Shen Z, Gao X, Ma L, Zhou Z, Shen X, Liu W. Expression of
proinflammatory pattern of expression and are associated with Foxp3 and interleukin-17 in lichen planus lesions with emphasis
clinical parameters. Clin Oral Investig. 2013;17:1365-1373. on difference in oral and cutaneous variants. Arch Dermatol Res.
16. Korn T, Bettelli E, Oukka M, Kuchroo VK. IL-17 and Th17 cells. 2014;306:441-446.
Annu Rev Immunol. 2009;27:485-517. 38. Parks OB, Pociask DA, Hodzic Z, Kolls JK, Good M. Interleukin-
17. Santoro A, Majorana A, Roversi L, et al. Recruitment of dendritic 22 signaling in the regulation of intestinal health and disease.
cells in oral lichen planus. J Pathol. 2005;205:426-434. Front Cell Dev Biol. 2015;3:85.
18. Hervas-Stubbs S, Perez-Gracia JL, Rouzaut A, Sanmamed MF, 39. Fujita H. The role of IL-22 and Th22 cells in human skin diseases.
Le Bon A, Melero I. Direct effects of type I interferons on cells of J Dermatol Sci. 2013;72:3-8.
the immune system. Clin Cancer Res. 2011;17:2619-2627. 40. Carrozzo M, Dametto E, Fasano ME, et al. Cytokine gene poly-
19. Staeheli P. Interferon-induced proteins and the antiviral state. Adv morphisms in hepatitis C virus-related oral lichen planus. Exp
Virus Res. 1990;38:147-200. Dermatol. 2007;16:730-736.
20. Sugerman PB, Savage NW, Walsh LJ, et al. The pathogenesis of 41. Xavier GM, de Sa AR, Guimaraes AL, da Silva TA, Gomez RS.
oral lichen planus. Crit Rev Oral Biol Med. 2002;13:350-365. Investigation of functional gene polymorphisms interleukin-1
21. Zhao ZZ, Sugerman PB, Zhou XJ, Walsh LJ, Savage NW. Mast beta, interleukin-6, interleukin-10 and tumor necrosis factor in
cell degranulation and the role of T cell RANTES in oral lichen individuals with oral lichen planus. J Oral Pathol Med. 2007;36:
planus. Oral Dis. 2001;7:246-251. 476-481.
22. Zhou XJ, Sugerman PB, Savage NW, Walsh LJ. Matrix metal- 42. Kimkong I, Hirankarn N, Nakkuntod J, Kitkumthorn N. Tumour
loproteinases and their inhibitors in oral lichen planus. J Cutan necrosis factor-alpha gene polymorphisms and susceptibility to
Pathol. 2001;28:72-82. oral lichen planus. Oral Dis. 2011;17:206-209.
23. Zhao ZZ, Savage NW, Sugerman PB, Walsh LJ. Mast cell/T cell in- 43. Kimkong I, Nakkuntod J, Sodsai P, Hirankarn N, Kitkumthorn N.
teractions in oral lichen planus. J Oral Pathol Med. 2002;31:189-195. Association of interferon-gamma gene polymorphisms with sus-
24. Simark Mattsson C, Jontell M, Bergenholtz G, Heyden M, ceptibility to oral lichen planus in the Thai population. Arch Oral
Dahlgren UI. Distribution of interferon-gamma mRNA-positive cells Biol. 2012;57:491-494.
in oral lichen planus lesions. J Oral Pathol Med. 1998;27:483-488. 44. Bai J, Lin M, Zeng X, et al. Association of polymorphisms in the
25. Carrozzo M, Uboldi de Capei M, Dametto E, et al. Tumor necrosis human IFN-gamma and IL-4 gene with oral lichen planus: a study in
factor-alpha and interferon-gamma polymorphisms contribute to sus- an ethnic Chinese cohort. J Interferon Cytokine Res. 2008;28:351-358.
ceptibility to oral lichen planus. J Invest Dermatol. 2004;122:87-94. 45. Jiang C, Yao H, Cui B, Zhou Y, Wang Y, Tang G. Association of
26. Lodi G, Scully C, Carrozzo M, Griffiths M, Sugerman PB, interleukin 12 A gene polymorphisms with oral lichen planus in
Thongprasom K. Current controversies in oral lichen planus: Chinese population. J Oral Pathol Med. 2015;44:602-606.
report of an international consensus meeting. Part 2. Clinical 46. Lebre MC, Burwell T, Vieira PL, et al. Differential expression of
management and malignant transformation. Oral Surg Oral Med inflammatory chemokines by Th1- and Th2-cell promoting den-
Oral Pathol Oral Radiol Endod. 2005;100:164-178. dritic cells: a role for different mature dendritic cell populations in
27. Wang Y, Zhou J, Fu S, Wang C, Zhou B. A study of association attracting appropriate effector cells to peripheral sites of inflam-
between oral lichen planus and immune balance of Th1/Th2 cells. mation. Immunol Cell Biol. 2005;83:525-535.
Inflammation. 2015;38:1874-1879. 47. Hu JY, Zhang J, Cui JL, et al. Increasing CCL5/CCR5 on CD4þ
28. Farthing PM, Cruchley AT. Expression of MHC class II antigens T cells in peripheral blood of oral lichen planus. Cytokine.
(HLA DR, DP and DQ) by keratinocytes in oral lichen planus. 2013;62:141-145.
J Oral Pathol Med. 1989;18:305-309. 48. Iijima W, Ohtani H, Nakayama T, et al. Infiltrating CD8þ T cells
29. Strange P, Skov L, Baadsgaard O. Interferon gamma-treated in oral lichen planus predominantly express CCR5 and CXCR3
keratinocytes activate T cells in the presence of superantigens: and carry respective chemokine ligands RANTES/CCL5 and IP-
involvement of major histocompatibility complex class II mole- 10/CXCL10 in their cytolytic granules: a potential self-recruiting
cules. J Invest Dermatol. 1994;102:150-154. mechanism. Am J Pathol. 2003;163:261-268.
30. Zhang Y, Liu W, Zhang S, et al. Salivary and serum interleukin- 49. De Iudicibus S, Franca R, Martelossi S, Ventura A, Decorti G.
18 in patients with oral lichen planus: a study in an ethnic Chinese Molecular mechanism of glucocorticoid resistance in inflamma-
population. Inflammation. 2012;35:399-404. tory bowel disease. World J Gastroenterol. 2011;17:1095-1108.
31. Bai J, Zhang Y, Lin M, et al. Interleukin-18 gene polymorphisms 50. Li CC, Munitic I, Mittelstadt PR, Castro E, Ashwell JD. Sup-
and haplotypes in patients with oral lichen planus: a study in an pression of dendritic cell-derived IL-12 by endogenous gluco-
ethnic Chinese cohort. Tissue Antigens. 2007;70:390-397. corticoids is protective in LPS-induced sepsis. PLoS Biol.
32. Gracie JA, Robertson SE, McInnes IB. Interleukin-18. J Leukoc 2015;13:e1002269.
Biol. 2003;73:213-224. 51. Galon J, Franchimont D, Hiroi N, et al. Gene profiling reveals
33. Pekiner FN, Demirel GY, Borahan MO, Ozbayrak S. Cytokine unknown enhancing and suppressive actions of glucocorticoids
profiles in serum of patients with oral lichen planus. Cytokine. on immune cells. FASEB J. 2002;16:61-71.
2012;60:701-706. 52. Nicolaides NC, Charmandari E, Chrousos GP, Kino T. Recent
34. Simark-Mattsson C, Bergenholtz G, Jontell M, et al. Distribution advances in the molecular mechanisms determining tissue sensi-
of interleukin-2, -4, -10, tumour necrosis factor-alpha and trans- tivity to glucocorticoids: novel mutations, circadian rhythm and
forming growth factor-beta mRNAs in oral lichen planus. Arch ligand-induced repression of the human glucocorticoid receptor.
Oral Biol. 1999;44:499-507. BMC Endocr Disord. 2014;14:71.
35. Piccinni MP, Lombardelli L, Logiodice F, et al. Potential path- 53. Hearing SD, Norman M, Smyth C, Foy C, Dayan CM. Wide
ogenetic role of Th17, Th0, and Th2 cells in erosive and reticular variation in lymphocyte steroid sensitivity among healthy human
oral lichen planus. Oral Dis. 2014;20:212-218. volunteers. J Clin Endocrinol Metab. 1999;84:4149-4154.
ORAL AND MAXILLOFACIAL PATHOLOGY OOOO
80 Kurago July 2016

54. Creed TJ, Lee RW, Newcomb PV, di Mambro AJ, Raju M, 74. Zunt SL, Burton LV, Goldblatt LI, Dobbins EE, Srinivasan M.
Dayan CM. The effects of cytokines on suppression of lymphocyte Soluble forms of toll-like receptor 4 are present in human saliva
proliferation by dexamethasone. J Immunol. 2009;183:164-171. and modulate tumour necrosis factor-alpha secretion by
55. O’Neill ID, Scully C. Biologics in oral medicine: ulcerative dis- macrophage-like cells. Clin Exp Immunol. 2009;156:285-293.
orders. Oral Dis. 2013;19:37-45. 75. Ohno S, Tateishi Y, Tatemoto Y, Morishita K, Sasabe E,
56. Cheng S, Kirtschig G, Cooper S, Thornhill M, Leonardi-Bee J, Yamamoto T. Enhanced expression of toll-like receptor 2 in
Murphy R. Interventions for erosive lichen planus affecting lesional tissues and peripheral blood monocytes of patients with
mucosal sites. Cochrane Database Syst Rev 2012;(2):CD008092. oral lichen planus. J Dermatol. 2011;38:335-344.
57. van der Meij EH, Schepman KP, Smeele LE, van der Wal JE, 76. Janardhanam SB, Prakasam S, Swaminathan VT, Kodumudi KN,
Bezemer PD, van der Waal I. A review of the recent literature Zunt SL, Srinivasan M. Differential expression of TLR-2 and
regarding malignant transformation of oral lichen planus. Oral Surg TLR-4 in the epithelial cells in oral lichen planus. Arch Oral Biol.
Oral Med Oral Pathol Oral Radiol Endod. 1999;88:307-310. 2012;57:495-502.
58. Mattsson U, Jontell M, Holmstrup P. Oral lichen planus and 77. Kho HS, Chang JY, Kim YY, Kim Y. MUC1 and Toll-like re-
malignant transformation: is a recall of patients justified? Crit Rev ceptor-2 expression in burning mouth syndrome and oral lichen
Oral Biol Med. 2002;13:390-396. planus. Arch Oral Biol. 2013;58:837-842.
59. Larsson A, Warfvinge G. Malignant transformation of oral lichen 78. Lo Muzio L, Santarelli A, Campisi G, Lacaita M, Favia G. Possible
planus. Oral Oncol. 2003;39:630-631. link between Hashimoto’s thyroiditis and oral lichen planus: a novel
60. Gonzalez-Moles MA, Scully C, Gil-Montoya JA. Oral lichen association found. Clin Oral Investig. 2013;17:333-336.
planus: controversies surrounding malignant transformation. Oral 79. Siponen M, Huuskonen L, Laara E, Salo T. Association of oral
Dis. 2008;14:229-243. lichen planus with thyroid disease in a Finnish population: a
61. Carbone M, Arduino PG, Carrozzo M, et al. Course of oral lichen retrospective case-control study. Oral Surg Oral Med Oral Pathol
planus: a retrospective study of 808 northern Italian patients. Oral Oral Radiol Endod. 2010;110:319-324.
Dis. 2009;15:235-243. 80. van der Meij EH, van der Waal I. Lack of clinicopathologic
62. Georgakopoulou EA, Achtari MD, Achtaris M, Foukas PG, correlation in the diagnosis of oral lichen planus based on the
Kotsinas A. Oral lichen planus as a preneoplastic inflammatory presently available diagnostic criteria and suggestions for modi-
model. J Biomed Biotechnol. 2012;2012:759626. fications. J Oral Pathol Med. 2003;32:507-512.
63. Fitzpatrick SG, Hirsch SA, Gordon SC. The malignant trans- 81. Kelesidis T, Yang O. Good’s syndrome remains a mystery after
formation of oral lichen planus and oral lichenoid lesions: a 55 years: a systematic review of the scientific evidence. Clin
systematic review. J Am Dent Assoc. 2014;145:45-56. Immunol. 2010;135:347-363.
64. Yashiro M. Ulcerative colitis-associated colorectal cancer. World 82. Bernard C, Frih H, Pasquet F, et al. Thymoma associated with
J Gastroenterol. 2014;20:16389-16397. autoimmune diseases: 85 cases and literature review. Autoimmun
65. Lodi G, Scully C, Carrozzo M, Griffiths M, Sugerman PB, Rev. 2016;15:82-92.
Thongprasom K. Current controversies in oral lichen planus: 83. Arnold SJ, Hodgson T, Misbah SA, Patel SY, Cooper SM,
report of an international consensus meeting. Part 1. Viral in- Venning VA. Three difficult cases: the challenge of autoimmu-
fections and etiopathogenesis. Oral Surg Oral Med Oral Pathol nity, immunodeficiency and recurrent infections in patients with
Oral Radiol Endod. 2005;100:40-51. Good syndrome. Br J Dermatol. 2015;172:774-777.
66. Calvaruso V, Craxi A. Immunological alterations in hepatitis C 84. Macdonald JB, Mangold AR, Connolly SM. Good syndrome and
virus infection. World J Gastroenterol. 2013;19:8916-8923. lichen planus: case and review. J Eur Acad Dermatol Venereol.
67. Lodi G, Pellicano R, Carrozzo M. Hepatitis C virus infection and 2014;28:1828-1830.
lichen planus: a systematic review with meta-analysis. Oral Dis. 85. Malphettes M, Gerard L, Galicier L, et al. Good syndrome: an
2010;16:601-612. adult-onset immunodeficiency remarkable for its high incidence
68. Shengyuan L, Songpo Y, Wen W, Wenjing T, Haitao Z, Binyou W. of invasive infections and autoimmune complications. Clin Infect
Hepatitis C virus and lichen planus: a reciprocal association deter- Dis. 2015;61:e13-e19.
mined by a meta-analysis. Arch Dermatol. 2009;145:1040-1047. 86. Maehara T, Moriyama M, Kawano S, et al. Cytokine profiles
69. Carrozzo M, Brancatello F, Dametto E, et al. Hepatitis C virus- contribute to understanding the pathogenic difference between
associated oral lichen planus: is the geographical heterogeneity Good syndrome and oral lichen planus: two case reports and
related to HLA-DR6? J Oral Pathol Med. 2005;34:204-208. literature review. Medicine (Baltimore). 2015;94:e704.
70. Nagao Y, Sata M, Itoh K, Tanikawa K, Kameyama T. Quanti-
tative analysis of HCV RNA and genotype in patients with
chronic hepatitis C accompanied by oral lichen planus. Eur J Clin
Reprint requests:
Invest. 1996;26:495-498.
71. Farhi D, Dupin N. Pathophysiology, etiologic factors, and clinical Zoya B. Kurago, DDS, PhD
management of oral lichen planus, part I: Facts and controversies. Dental College of Georgia
Clin Dermatol. 2010;28:100-108. Medical College of Georgia
72. Luna JM, Scheel TK, Danino T, et al. Hepatitis C virus RNA Georgia Cancer Center
functionally sequesters miR-122. Cell. 2015;160:1099-1110. 1430 JW Gilbert Dr.
73. Srinivasan M, Kodumudi KN, Zunt SL. Soluble CD14 and toll-like Augusta, GA 30912
receptor-2 are potential salivary biomarkers for oral lichen planus USA
and burning mouth syndrome. Clin Immunol. 2008;126:31-37. ZKURAGO@gru.edu; zkurago@augusta.edu

You might also like