Schenke in 2006

17
Host responses in maintaining periodontal health and determining periodontal disease H ARVEY A. S CHENKEIN Inflammation and destruction of periodontal tissues are largely considered to result from the response of a susceptible host to a microbial biofilm containing gram-negative bacterial pathogens. Many of the bac- teria that can contribute to periodontitis have been identified [206]; characteristically, their presence is strongly associated with destructive periodontal lesions and they display virulence factors associated with the pathology of soft and hard tissue destruction. Most of the bacteria that share these characteristics are not present in healthy periodontal sites or are present in lower concentrations than in diseased sites. The manner in which destructive periodontal lesions are initiated by bacteria in previously healthy sites is not entirely clear, but likely mechanisms for progressive destruction of previously compromised periodontal sites have been extensively described. As individuals are not equally susceptible to the destructive effects of periodontal infections, it is clear that variability in host responses among individuals contributes significantly to the expression of these diseases in the population [233]. This likely holds true both for the protective host mechanisms that prevent periodontitis or impede its progression and for the destructive mechanisms that initiate lesions and pro- mote progressive disease. The intent of this review is to outline some of the host response mechanisms that might contribute both to maintenance of periodontal health and to initiation and progression of disease. Host characteristics that contribute to prevention of periodontitis Protective host cells – polymorphonuclear leukocytes Histologic examination of gingivitis or periodontitis lesions reveals that the polymorphonuclear leukocyte appears to play a key role in maintenance of perio- dontal health [89, 145]. These cells line the junctional epithelium in large numbers and appear to attempt to wall off the underlying tissues from the bacterial biofilm. Their appearance is the result of the pre- sence or generation of chemotactic factors in the gingival sulcus and underlying tissues [93, 219]. Plaque bacteria are replete with chemotactic factors, they are further capable of generating chemotactic factors from plasma via activation of the comple- ment, coagulation, fibrinolytic, and kinin systems or from surrounding cells following interactions with cellular receptors [192, 193]. The paramount importance of polymorphonuclear leukocytes in the protective response against patho- gens of the plaque biofilm is underscored by the prevalence and severity of periodontitis in patients with syndromes characterized by polymorphonuclear leukocyte dysfunction. Patients with diseases such as leukocyte adhesion deficiency, chronic neutropenia, and cyclic neutropenia suffer from frequent and severe extraoral infections and are frequently afflic- ted with severe and early onset forms of periodontitis [4, 37, 239]. Molecular defects in polymorphonuclear leukocytes with a variety of functional consequences can be shown to result in accelerated periodontitis. For example, leukocyte adhesion deficiency, charac- terized by total lack of, or decreased expression of, the beta chain of CD18 or alternatively a defect in selectin ligand expression, results in a decreased ability of polymorphonuclear leukocytes to recognize inflamed endothelium. The failure of polymorpho- nuclear leukocytes to transmigrate the endothelium results in a greatly attenuated inflammatory response to bacterial challenges and restricts the protective response against periodontal pathogens [138]. A second example is the mutation of the gene encoding cathepsin C that results in the Papillon–Lefe `vre 77 Periodontology 2000, Vol. 40, 2006, 77–93 Printed in the UK. All rights reserved ȑ 2006 The Author. Journal compilation ȑ 2006 Blackwell Munksgaard PERIODONTOLOGY 2000

description

tt

Transcript of Schenke in 2006

Page 1: Schenke in 2006

Host responses in maintainingperiodontal health anddetermining periodontal disease

HARVEY A. SCHENKEIN

Inflammation and destruction of periodontal tissues

are largely considered to result from the response of a

susceptible host to a microbial biofilm containing

gram-negative bacterial pathogens. Many of the bac-

teria that can contribute to periodontitis have been

identified [206]; characteristically, their presence is

strongly associated with destructive periodontal

lesions and they display virulence factors associated

with the pathology of soft and hard tissue destruction.

Most of the bacteria that share these characteristics

are not present in healthy periodontal sites or are

present in lower concentrations than in diseased sites.

The manner in which destructive periodontal lesions

are initiated by bacteria in previously healthy sites is

not entirely clear, but likely mechanisms for

progressive destruction of previously compromised

periodontal sites have been extensively described.

As individuals are not equally susceptible to the

destructive effects of periodontal infections, it is clear

that variability in host responses among individuals

contributes significantly to the expression of these

diseases in the population [233]. This likely holds true

both for the protective host mechanisms that prevent

periodontitis or impede its progression and for the

destructive mechanisms that initiate lesions and pro-

mote progressive disease. The intent of this review is to

outline some of the host response mechanisms that

might contribute both to maintenance of periodontal

health and to initiation and progression of disease.

Host characteristics that contributeto prevention of periodontitis

Protective host cells –polymorphonuclear leukocytes

Histologic examination of gingivitis or periodontitis

lesions reveals that the polymorphonuclear leukocyte

appears to play a key role in maintenance of perio-

dontal health [89, 145]. These cells line the junctional

epithelium in large numbers and appear to attempt

to wall off the underlying tissues from the bacterial

biofilm. Their appearance is the result of the pre-

sence or generation of chemotactic factors in the

gingival sulcus and underlying tissues [93, 219].

Plaque bacteria are replete with chemotactic factors,

they are further capable of generating chemotactic

factors from plasma via activation of the comple-

ment, coagulation, fibrinolytic, and kinin systems or

from surrounding cells following interactions with

cellular receptors [192, 193].

The paramount importance of polymorphonuclear

leukocytes in the protective response against patho-

gens of the plaque biofilm is underscored by the

prevalence and severity of periodontitis in patients

with syndromes characterized by polymorphonuclear

leukocyte dysfunction. Patients with diseases such as

leukocyte adhesion deficiency, chronic neutropenia,

and cyclic neutropenia suffer from frequent and

severe extraoral infections and are frequently afflic-

ted with severe and early onset forms of periodontitis

[4, 37, 239]. Molecular defects in polymorphonuclear

leukocytes with a variety of functional consequences

can be shown to result in accelerated periodontitis.

For example, leukocyte adhesion deficiency, charac-

terized by total lack of, or decreased expression of,

the beta chain of CD18 or alternatively a defect in

selectin ligand expression, results in a decreased

ability of polymorphonuclear leukocytes to recognize

inflamed endothelium. The failure of polymorpho-

nuclear leukocytes to transmigrate the endothelium

results in a greatly attenuated inflammatory response

to bacterial challenges and restricts the protective

response against periodontal pathogens [138]. A

second example is the mutation of the gene encoding

cathepsin C that results in the Papillon–Lefevre

77

Periodontology 2000, Vol. 40, 2006, 77–93

Printed in the UK. All rights reserved

� 2006 The Author.

Journal compilation � 2006 Blackwell Munksgaard

PERIODONTOLOGY 2000

Page 2: Schenke in 2006

syndrome [85–87, 227]. Cathepsin C is a lysosomal

protease found in polymorphonuclear leukocytes and

macrophages that removes dipeptides from the

amino terminus of proteins and is likely important in

the activation of other pro-enzymes. Papillon–Lefevre

syndrome is also characterized by early and aggres-

sive periodontitis and characteristic palmar and

plantar hyperkeratosis. It is notable that the patho-

genic mechanisms responsible for the periodontal

syndromes observed in the leukocyte adhesion defi-

ciency and Papillon–Lefevre syndromes are not

known, but the associations with defects in leukocyte

function are clear. An additional condition of interest

in this regard is Morbus–Kostmann syndrome, an

autosomal recessive disease characterized by a lack

of the polymorphonuclear leukocyte antibacterial

cathelicidin LL-37. Patients with this disease have

been reported to have severe periodontitis and

members of one affected family have been found to

have an overgrowth of Actinobacillus actinomyce-

temcomitans in the oral flora [175].

Additional evidence of the importance of poly-

morphonuclear leukocytes for protection against

periodontitis is provided by the observation that up

to 80% of patients with localized aggressive perio-

dontitis have a measurable decrease in chemotactic

function when compared to age- and race-matched

periodontally healthy individuals [195, 232, 234, 235].

Symptoms in such patients differ significantly from

those in patients with syndromic defects in poly-

morphonuclear leukocyte function. Patients with

localized aggressive periodontitis and chemotactic

dysfunction are systemically healthy, early expression

of periodontitis being the only clinical manifestation

of the defect. The chemotactic dysfunction is subtle

in that cells appear to migrate at about 50% or less of

the velocity of normal cells. Interestingly, not all

patients with clinical signs of localized aggressive

periodontitis demonstrate a reproducible defect in

chemotactic function, raising the question of whether

localized aggressive periodontitis is truly a manifes-

tation of the polymorphonuclear leukocyte defect,

whether the association is fortuitous but unrelated, or

whether there are multiple etiologic subforms of

localized aggressive periodontitis only some of which

are related to chemotactic dysfunction.

Protective host responses – antibodies

Periodontitis patients characteristically produce sys-

temic and local antibodies reactive with a variety of

periodontal bacterial pathogens [1, 25, 61, 62, 222,

244]. It is apparent that these antibodies do not

generally appear in significant titers until after peri-

odontal destruction has already occurred; clinically

periodontal healthy subjects only rarely produce high

levels of such antibodies [1, 150, 170]. This and the

observation that such antibodies persist long after

treatment of periodontal infections raises the ques-

tion of whether antibodies produced during the

course of disease are functional or are merely serving

as markers of previous infection.

A series of studies of antibody responses and

specificity in aggressive periodontitis patients was

performed to address the issue of functionality of

antibodies reactive with the periodontal patho-

gens A. actinomycetemcomitans and Porphyromonas

gingivalis. These studies were based upon the initial

observations of Ranney and coworkers [83, 182] that

the presence or absence of high levels of such anti-

bodies were directly associated with disease severity

as indicated by loss of attachment. Those authors

observed that aggressive periodontitis patients who

were seropositive for both A. actinomycetemcomitans

and P. gingivalis displayed significantly fewer sites

with severe attachment loss (sites with more than

5 mm attachment loss) than did patients who

were seronegative for both organisms. Patients sero-

positive for one or the other bacteria displayed

intermediate percentages of periodontal sites with

attachment loss. The concept that antibody function

could be related to clinical status in aggressive perio-

dontitis was reinforced by studies demonstrating that

periodontal therapy resulting in decreased bacterial

load could induce production of antibodies to plaque

microorganisms in patients with low serum antibody

levels and decreased titer but increase antibody

avidity in patients who had high levels of specific

antibody [30]. These associations can be interpreted to

mean that the antibodies, likely raised during the

course of disease or due to therapy, diminished the

progression of periodontitis in these patients by

modifying or reducing components of the bacterial

microflora. However, in several other studies, effects

of periodontal therapy on the titers and avidities of

bacteria-specific antibodies were extremely variable

[9, 38, 47, 63, 99, 149, 151, 190, 230, 238].

To better examine the effects of antibody titer on

clinical status, studies have been performed that

assess antibody responses to specific bacterial viru-

lence factors. Several of such studies verified that

strong antibody responses against specific bacterial

antigens, such as the lipopolysaccharide or leuko-

toxin of A. actinomycetemcomitans and the lipo-

polysaccharide or hemagglutinin of P. gingivalis,

were associated with less severe disease in patients

78

Schenkein

Page 3: Schenke in 2006

with aggressive or chronic periodontitis [24, 26–28].

These antibodies were identified to be predominantly

of the IgG2 subclass. Other studies also identified the

predominant subclass response to pathogens such as

P. gingivalis to be IgG2 but failed to show that high

levels of these antibodies were associated decreased

clinical severity [243]. Despite the lack of clarity

regarding the impact of disease-induced antibody on

periodontal clinical status, several investigators have

developed vaccines based upon immunization with

whole bacteria or agents related to specific virulence

factors that appear to be effective in reducing meas-

ures of disease in animal models [19, 50, 65, 76, 78,

157, 166, 178, 185, 249]. This approach, which has

mainly targeted P. gingivalis antigens such as cap-

sular polysaccharide and the gingipain proteases,

sheds some light on the pathogenic mechanisms of

disease in these models of periodontitis.

The protective role of local antibody production, as

detected by elevated concentrations of gingival

crevice fluid antibodies, is unclear. Studies by Tew

et al. [222] demonstrated the local production of

antibody reactive with A. actinomycetemcomitans

and P. gingivalis but failed to note significant asso-

ciations between such antibody and clinical meas-

ures of disease. Other studies did demonstrate that

colonization of specific bacteria such as A. actino-

mycetemcomitans was lower in sites containing

locally produced antibodies [62] or that therapy

decreased local antibody production [47], but the

impact of this �protective� response on clinical disease

was not definitively established.

Protection of the periodontium by theepithelial barrier

The oral epithelium, particularly the junctional and

sulcular epithelia, represents a dynamic physical and

chemical barrier against the pathologic properties of

the microbial biofilm that exists in its vicinity. The

characteristics of the junctional epithelium in health

and disease have been recently described in an

excellent review by Bosshardt & Lang [21].

Fundamentally, healthy epithelial tissues that are

challenged by bacteria react to this challenge by

mobilizing their own antimicrobial mechanisms and

by permitting cells of the innate and adaptive

immune systems to access the pathogens. Histologi-

cally, tissues in health comprise not only epithelial

cells but also cellular sentinels such as dendritic cells

[35, 54, 108–110] and polymorphonuclear leukocytes

[197], as well as other leukocytes including lympho-

cytes and mononuclear phagocytes [196].

Cytokines that reflect innate responses of the host

to the microbial biofilm have been shown to be

expressed by cells of the junctional epithelium,

including chemotactic cytokines interleukin (IL)-8

[225, 226] and cytokine-induced neutrophil chemo-

attractant-2 [146], as well as IL-1 and tumor necrosis

factor-a [147]. Furthermore, cells of the junctional

epithelium and their resident leukocytes contribute

to host defense by expression of antibacterial pep-

tides including calprotectin [154], a- and b-defensins[34, 130, 237] andcathelicidin LL-37 (recently reviewed

by Dale [46]). Recent data demonstrate that perio-

dontal pathogens induce production of b-defensinsand IL-8 by polymorphonuclear leukocytes [237], and

that defensins are up-regulated in tissues of patients

with chronic periodontitis [130]. However, healthy

tissues appear to have high concentrations of defen-

sins indirectly supporting their role in homeostasis of

the gingival sulcus in health [20, 131].

Destructive host responses toperiodontal microorganisms –innate immunity

It is remarkably difficult to separate and individually

account for the relative impact of innate and

acquired host responses in the pathology of perio-

dontitis lesions. The histopathology of periodontitis

illustrates that most stages of the disease comprise

elements of both types of responses and of both

acute and chronic inflammation. Furthermore, cel-

lular and biochemical constituents of both categories

of response elements interact to determine the

balance of responses in such complex lesions. It is

therefore more useful to organize a discussion of

destructive host responses as a continuum of

responses of various cell types that participate in

pathologic reactions at different stages of disease and

at different locations within the lesion.

Polymorphonuclear leukocytes

Though polymorphonuclear leukocytes appear to

maintain a defensive posture in the periodontal

lesion, it is well known that these cells also can be

major players in immunopathology [16, 241]. Early

studies in animals illustrated that induction of

Arthus-type reactions in the gingiva could lead to

periodontal inflammation and attachment loss [181,

183]. However, the apparent lack of readily demon-

strable antigen–antibody complexes in periodontal

tissues argue that this phenomenon is not likely to be

79

Host responses in periodontal health and disease

Page 4: Schenke in 2006

operant in human disease [192]. Nevertheless, the

presence of high concentrations of periodontitis-

specific antibodies in the serum and gingival crevice

fluid, the abundance of opsonized bacteria in the

subgingival region, and evidence for the interaction

of bacteria with phagocytes in the periodontal lesion

argue that polymorphonuclear leukocytes are likely

to contribute to pathology in periodontitis [8, 11, 113,

145].

Polymorphonuclear leukocytes synthesize and

release a number of substances, such as lysosomal

enzymes including tissue-destructive proteases [18,

125, 135, 172, 173, 213, 242] that are associated with

the breakdown of periodontal tissues. Interactions of

polymorphonuclear leukocytes with bacteria have

been shown to damage a variety of crucial cell types,

including fibroblasts, endothelial cells, and kera-

tinocytes [241]. Though polymorphonuclear leuko-

cytes constitutively represent a first line of protection

against periodontal pathogens, their interaction with

overwhelming masses of biofilm lead to the release of

constitutive enzymes into the surrounding tissues as

well as the synthesis and secretion of proinflamma-

tory molecules such as arachidonic acid metabolites.

The presence of tissue-damaging enzymes, bone-

resorbing lipids, and other inflammatory mediators

that are released upon encounter with bacterial

pathogens is likely to contribute to the inflammatory

response and attachment loss observed in perio-

dontitis. It has been proposed that much of the

periodontal tissue destruction observed in aggressive

periodontitis is the result of hyperactivity or priming

of polymorphonuclear leukocytes in these patients

[112, 113, 200, 236], resulting in elevated production

of inflammatory and bone-resorbing lipids.

Macrophages

A currently accepted paradigm, explaining how the

predominantly gram-negative infection associated

with periodontitis lesions is translated into destruc-

tion of bone and connective tissue, revolves around

mononuclear phagocytes. In sites of chronic inflam-

mation, bacteria participate in attracting leukocytes

into the periodontal tissues either by directly

expressing chemoattractant peptides or by stimula-

ting the production and secretion of chemoattractant

cytokines and inflammatory lipids from host cells.

This leads to accumulation of leukocytes in the host

tissue [8, 93, 94]. Bacterial lipopolysaccharide can

subsequently interact with macrophage or dendritic

cell receptors, including CD14 and Toll-like recep-

tors, to stimulate production of inflammatory

cytokines and other mediators. This model of tissue

destruction focuses on the production of IL-1 as a key

mediator of periodontal tissue destruction due to the

association of this cytokine with stimulation of

collagenolytic and bone-destructive processes [217].

Studies in model systems have indicated that inhi-

bition or interruption of these pathways results in

inhibition of bacterial-mediated periodontal tissue

destruction [7, 52, 53, 81, 156, 179, 180]. This model of

pathogenesis has the particular appeal of explaining

as well some of the differences in susceptibility to

periodontitis observed in the population, due to the

presence of genetic polymorphisms in the IL-1 genes

that appear to influence the severity of periodontitis

(see below).

The interaction of lipopolysaccharide with macro-

phages also stimulates production of prostanoids, in

particular prostaglandin E2, which is notably found at

high concentrations in gingival crevice fluid from

sites undergoing periodontal breakdown. Prosta-

glandin E2 is a bone-resorbing inflammatory lipid,

and experimental data indicate that this pathway is

likely to be important in human periodontal bone

loss [79, 91, 159, 160, 161, 198, 251]. Studies per-

formed both in animal models and in human clinical

trials have demonstrated that production of prosta-

glandin E2 and loss of alveolar bone due to perio-

dontitis are substantially attenuated following local

or systemic use of nonsteroidal anti-inflammatory

drugs [92, 100, 162, 171, 245].

It is important to note that cells other than macro-

phages that are present in periodontal lesions (such

as fibroblasts) also produce inflammatory cytokines,

lipid mediators, and matrix metalloproteinases and

are likely to participate in the accumulation of these

molecules [59, 60, 153, 155, 164, 186, 209, 224].

Destructive host responses toperiodontal microorganisms –acquired immunity

Although current thinking ascribes a major role for

innate immune mechanisms in periodontal tissue

destruction, the histopathology of the destructive

periodontal lesion and systemic effects of periodontal

bacteria on immune factors such as antibodies and

cytokines implicates acquired immunity as playing a

role in the disease process.

The classic studies of Page and Schroeder des-

cribed a continuum of histologic observations that

indicated that development of destructive periodon-

titis entails a progression of cell types [167, 168]. Early

80

Schenkein

Page 5: Schenke in 2006

lesions of chronic periodontitis are characterized by a

cellular infiltrate comprising mainly macrophages

and T lymphocytes. More advanced lesions demon-

strating connective tissue loss and bone resorption

contain large numbers of B lymphocytes and plasma

cells. It has been proposed that the histologic com-

position, cytokine profile, and nature and specificity

of the local immune response in periodontitis lesions

reflects the presence and function of subclasses of

regulatory T cells induced by the microbial biofilm

[221, 248]. The histologic picture of the early lesion,

clinical gingivitis, is most consistent with a Th1

response [199]. Such a lesion is classically developed

in response to intracellular pathogens. On the other

hand, the histology of the advanced lesion is more

consistent with a Th2 response, which is typically

mounted to fight extracellular pathogens. The

pathogen itself may dictate which type of response is

generated through its interaction with accessory cells

and resulting production of cytokines characteristic

of the response [51, 106, 114]. For example, the Th1

response is usually characterized by IL-12 produc-

tion, which in turn induces interferon-c production,

leading to macrophage activation, increased phago-

cytic activity, and protective immunity. The Th2 re-

sponse, on the other hand, entails production of

alternative cytokines such as IL-4, IL-10, and IL-13,

leading to antibody production. An exception to this,

and one which is relevant to immune responses to

some oral microorganisms such as A. actinomyce-

temcomitans, is that some pathogens with high levels

of surface carbohydrates induce high levels of anti-

bodies of the IgG2 subclass that are dependent upon

Th1 rather than Th2 cytokines for their production [2,

117].

During an immune response either the Th1 or the

Th2 cytokine profile will usually dominate, indicating

polarization of the response. The nature of this

response in periodontitis has been examined with

conflicting results. Investigators have examined gin-

gival tissues for expression of cytokine mRNA or

observed the interaction of periodontal pathogens

with leukocytes. Results have frequently demonstra-

ted a mixture of Th1 and Th2 cytokines or a pre-

dominance of one type over another [69–73, 163, 199,

218, 220, 221, 248]. Those data would favor the

hypothesis that chronic periodontitis lesions are

more associated with the Th2 response. Results

demonstrating a mixture of Th1 and Th2 cytokines

are not surprising in view of the numerous bacterial

species that may interact with the immune system in

a periodontitis lesion. Furthermore, tissues from

lesions displaying differing histologic stages of

disease, different states of disease activity, or differing

forms of periodontitis would likely be heterogeneous

with respect to cytokine profiles.

Aggressive periodontitis andTh1-dependent antibody production

One of the interesting aspects of the biology of

aggressive periodontitis is that patients with localized

disease have higher serum concentrations of IgG2

protein than do other age- and race-matched perio-

dontal groups and healthy controls [129]. In addition,

localized aggressive periodontitis patients also dis-

play very high serum concentrations of specific IgG2

antibodies reactive with carbohydrate antigens of the

periodontal pathogen A. actinomycetemcomitans [26]

and IgG2 plays a predominant role in the antibody

response to P. gingivalis [27, 243]. The increase in

serum concentration of IgG2 (> 1 mg ⁄ ml) found in

localized aggressive periodontitis patients is far too

great to be accounted for only by the antibody

reactive with A. actinomycetemcomitans, which

appears to average 70–80 lg ⁄ ml in seropositive

patients. The presence of elevated Th1-dependant

specific antibody and total IgG2 has led us to spe-

culate that the immune response of localized

aggressive periodontitis patients may be polarized

towards Th1-dependent responses [215]. Serum IgG2

levels appear to be under genetic control, possible

explaining the predominance of Th1 responses in

localized aggressive periodontitis patients [56].

Examination of peripheral blood leukocytes from

localized aggressive periodontitis patients reveals

that adherent monocytes and soluble factors secreted

by these cells appear to direct increased IgG2 pro-

duction observed in cell culture. In addition to being

controlled by Th1 cytokines such as IL-12, IL-18, and

interferon-c, IgG2 production is also influenced by

the concentrations of the inflammatory lipids

prostaglandin E2 and platelet-activating factor [2,

102, 103, 117, 215]. Monocytes from localized aggres-

sive periodontitis patients produce lower levels of

platelet-activating factor-acetylhydrolase, a platelet-

activating factor-degrading enzyme, which likely

accounts for the higher concentrations of platelet-

activating factor. Monocytes from these patients have

a tendency to differentiate into dendritic cells, which

produce lower levels of platelet-activating factor-

acetylhydrolase than macrophages [3, 14, 215]. A

series of studies exploring the mechanism by which

platelet-activating factor influences IgG2 production

have led to the observation that platelet-activating

factor enhances prostaglandin E2, IL-12, and IL-18

81

Host responses in periodontal health and disease

Page 6: Schenke in 2006

production by dendritic cells as well as interferon-cproduction by natural killer cells. All of these media-

tors contribute to elevated Th1 responses. In addition,

the periodontal pathogen A. actinomycetemcomitans

interacts with dendritic cells and natural killer cells to

rapidly produce interferon-c and also induces high

levels of Th1 cytokines such as IL-12.

The persistence of Th1 responses can cause tissue

damage as the cytokines and inflammatory mediators

associated with this pathway can induce harmful

molecules including nitric oxide (NO), reactive oxy-

gen intermediates, IL-1, interferon-c, and tumor

necrosis factor [106]. These factors can synergize to

induce pathology in sites of chronic inflammation

and thus may contribute to the uniquely rapid and

severe tissue destruction seen in localized aggressive

periodontitis. Thus, Th1 polarization in localized

aggressive periodontitis leads to high levels of pro-

tective IgG2 antibody, large amounts of probably

irrelevant IgG2, and cell-mediated immune mecha-

nisms of chronic inflammation.

Acquired immunity and bone resorption

An emerging area of research demonstrates rela-

tionships between the immune system, oral bacterial

pathogens, and alveolar bone loss. Key components

of this system of bone resorption and remodeling are

tumor necrosis factor receptor superfamily proteins

including RANKL (receptor activator of nuclear factor

kappaB ligand), RANK, and osteoprotegerin. RANK,

the receptor for RANKL, is found on osteoclast pre-

cursor cells. RANKL is expressed on osteoblasts and

can be regulated by IL-1, IL-6, IL-11, IL-17, tumor

necrosis factor-a, and prostaglandin E2 as well as by

various hormones. Bone loss and remodeling are

controlled both by the balance between RANK and

RANKL and by the RANKL decoy receptor osteopro-

tegerin [97, 98, 211, 223]. The interplay between this

system and the immune system is demonstrated by

the observation that activated T cells express RANKL

and that bone resorption is a consequence of T-cell

activation and up-regulation of RANKL. In inflam-

matory diseases such as arthritis and in infectious

diseases characterized by bone resorption it is

thought that RANKL plays a key role in inducing

osteoclasts and bone resorption. In conditions char-

acterized by both T-cell activation and inflammatory

cytokine production, bone resorption is further

enhanced by the effects of such cytokines on RANKL

expression in other cell types [223].

Links between RANK ⁄ RANKL and periodontal

diseases have been established via examination of

gingival tissues and gingival crevice fluid as well as in

animal model systems. Gingival tissues have been

shown to express both RANKL and osteoprotegerin

mRNA with high frequency, and RANKL mRNA levels

have been shown to be highest in tissues from sites

with advanced disease, whereas osteoprotegerin

levels have been found to be very low in such sites

[127, 128]. Notably, RANKL mRNA appears to be ex-

pressed by lymphocytes and macrophages in the

gingiva. Both RANKL and osteoprotegerin have also

been quantitated in gingival crevice fluid and it has

been noted that the ratio of RANKL to osteoproteg-

erin is greater in fluids from diseased sites than from

healthy sites [148].

In vitro and animal studies have shown that both

A. actinomycetemcomitans and P. gingivalis can up-

regulate RANKL [17, 31, 107, 121, 132]. In studies

using NOD ⁄ SCID mice reconstituted with human

mononuclear cells from periodontitis patients it

was shown that A. actinomycetemcomitans activates

CD4+ T cells and up-regulates RANKL, leading to

alveolar bone resorption. Likewise, P. gingivalis also

up-regulates RANKL on mononuclear cells, with

resulting bone loss.

Bacterial factors promoting evasion ofprotective host factors

Periodontal pathogens, like other microbial patho-

gens, have evolved mechanisms that both promote

disease and enhance their own survival. The creation

and ⁄ or maintenance of a biological niche suitable

for biofilm formation and survival must occur in a

manner that protects the colonizing organisms

against the host responses meant to control their

numbers. The ability of pathogenic periodontal

microorganisms to flourish appears to entail more

subtle mechanisms than simple overwhelming of the

immune system by sheer number. The pathogenic

species P. gingivalis and A. actinomycetemcomitans

exemplify this principle. Both organisms have a

common attribute of being able to invade structural

cells of the periodontium, thereby theoretically

�hiding� from elements of the immune system [32, 33,

67, 126, 187, 188]. However, they additionally have

interesting mechanisms for perturbing host response

mechanisms.

P. gingivalis

As described above, P. gingivalis can contribute to

periodontal inflammation and destruction through

the interaction of its lipopolysaccharide with

inflammatory cells as well as with cells of the

82

Schenkein

Page 7: Schenke in 2006

immune system that respond to its antigenicity.

These interactions can result in the production of

proinflammatory cytokines (IL-1b, tumor necrosis

factor-a, IL-6 and IL-8) as well as anti-inflammatory

cytokines (IL-1ra, IL-4) and promote antibody and

cell-mediated protective immune responses [12, 69,

70, 115, 116, 209, 250]. However, this species also

produces potent proteases, termed gingipains, that

utilize host proteins as nutritional substrates and that

are thought to play a significant role in the patho-

genesis of periodontitis [44, 74, 101, 152, 158, 174].

Studies have demonstrated that such proteases

readily degrade host-protective molecules such as

complement proteins and immunoglobulins, thereby

evading the opsonic activities of these molecules [45,

191] and generating proinflammatory molecules such

as complement anaphylatoxins [246]. Furthermore,

these proteases can degrade cytokines and their

receptors such as tumor necrosis factor-a, IL-6, andIL-8 [13, 29, 139, 144, 165] and cellular receptors such

as that for complement (C) 5a [104, 105] and cell

adhesion molecules on endothelial cells [202]. The

functional sequelae of the interactions of these bac-

terial proteases with host proteins would depend

upon the constituents of the local site at any given

time but evasion of the host antibacterial inflamma-

tory response would be a likely outcome.

Interestingly, the lipopolysaccharide of P. gingiva-

lis is unusual in both its structure and its interaction

with Toll-like receptors on leukocytes and other cells

[10, 43, 184]. Unlike Escherichia coli lipopolysaccha-

ride, which interacts mainly with Toll-like receptor-4,

P. gingivalis lipopolysaccharide interacts signifi-

cantly with Toll-like receptor-2. Recent studies

indicate that P. gingivalis lipopolysaccharide appears

to be antagonistic to Toll-like receptor-4 and com-

petes with lipopolysaccharide from other species

for Toll-like receptor-4 [36, 48, 49]. It has been

hypothesized that this property could represent a

mechanism of evasion of the host innate immune

system whereby recognition of bacterial pathogens

normally recognized by Toll-like receptor-4 could be

blocked.

A. actinomycetemcomitans

Several of the properties of A. actinomycetemcomi-

tans related to evasion of host responsiveness are

directed at phagocytes. A notable property of A.

actinomycetemcomitans is its toxicity to host leuko-

cytes such as neutrophils and monocytes via pro-

duction of a leukotoxin [229]. This toxin is well

characterized and strains of A. actinomycetemcomi-

tans producing high levels of the toxin are associated

epidemiologically with risk for localized aggressive

periodontitis [57, 58, 90]. This ability to kill phago-

cytes imparts obvious survival advantages to the

organism. Additionally, strains of A. actinomycetem-

comitans also produce immunosuppressive factors

that suppress specific and nonspecific immune re-

sponses as well as fibroblast proliferation [75, 123,

124, 177, 203–205, 208, 214]. These properties plus its

capacity for invasion of epithelial and endothelial

cells provide substantial means for evasion of host

protective responses.

Host characteristics thatpredispose to periodontal infection

Contribution of genes to host responsesin periodontal infection

The host response to infection depends upon both

the nature of the pathogen and its virulence charac-

teristics and is significantly influenced by genetic

factors. Genetic susceptibility to most infections is a

function of the interaction of multiple genes and

environmental factors with the pathogen [39, 96].

This multifactorial etiologic and pathogenic model

appears to apply to susceptibility to periodontal

pathogens [88, 140].

Data from studies of twins and families indicate

that genes influence periodontal disease risk and

pathogenesis. Studies of chronic periodontitis in

twins indicate that there is greater concordance of

periodontal measures in monozygotic twins than in

dizygotic twins and that environmental factors do not

account for this difference [40, 141, 142]. It is esti-

mated that about 50% of the variance in disease

expression in the population is due to genetic influ-

ences. Furthermore, family studies of patients with

aggressive periodontitis verify that there is a genetic

contribution to this form of disease as well [23, 133].

The challenge for investigators is to identify the genes

responsible and to understand how they interact with

each other and with the patient’s environment to

influence the host’s response to the microflora.

Since the pathogenesis of periodontitis largely

involves host responses to periodontal microorgan-

isms, it follows that it is likely that genetic variation in

the expression of these responses or in factors that

determine the characteristics of the microflora

should influence disease expression. Genetic vari-

ation influencing the host response to environmental

or systemic risk factors for periodontitis could also

influence the impact of such factors on disease.

83

Host responses in periodontal health and disease

Page 8: Schenke in 2006

Several genetic polymorphisms impacting the host

response have been examined for variants that could

be associated with periodontitis. As mentioned

above, a primary example of this relates to genetic

control of the secretion of IL-1 by macrophages. It

has been observed that polymorphic genes regulating

the production of IL-1 in response to bacterial lipo-

polysaccharide may impact on susceptibility to

severe periodontitis [122]. Such genetic variants of

the IL-1 A and IL-1B genes predispose macrophages

to produce varying amounts of IL-1 in response to a

given stimulus, and a higher proportion of patients

genetically programmed to produce high levels of

IL-1 appear to demonstrate increased suscepti-

bility to increased disease severity. Interestingly, the

association observed between IL-1 polymorphic

genes and aggressive periodontitis is with the alleles

that would promote lower levels of IL-1, or otherwise

no association at all has been detected [55, 240].

Several studies have examined the role of IL-1 poly-

morphisms in periodontal risk in a variety of popu-

lations as well as in smoking and nonsmoking subsets

of patients with variable results [6, 42, 68, 80, 82,

134, 136, 137, 189]. These relationships appear to be

population-dependent, and also may indicate a dif-

ference in pathogenesis of chronic and aggressive

periodontitis that could relate to the role of IL-1 in

antibody production. Although it is likely that genetic

variants in the IL-1 genes are likely to be among

many other such genes that contribute to genetic risk

for periodontitis, the biology of IL)1 and the associ-

ation of high innate levels of IL-1 and severe disease

provide a compelling argument that this is an

important pathway from innate immunity to perio-

dontal destruction.

Polymorphisms in the gene for a second related

proinflammatory cytokine tumor necrosis factor-ahave also been examined, some studies demon-

strating associations with periodontitis severity and

other failing to demonstrate such a relationship [41,

64, 66, 118, 201, 207, 228]. Several investigators have

examined polymorphisms in the gene coding for the

anti-inflammatory cytokine IL)10, searching for

associations with chronic or aggressive periodonti-

tis. For the most part, these studies have not

supported a role for the tested polymorphisms in

periodontal pathogenesis [77, 95, 118, 247]. This is

also true for studies examining IL-4 polymorphisms

[111, 143].

Studies of polymorphisms that influence the

function of leukocyte Fc receptors have shown

interesting associations with disease expression and

severity. Such polymorphisms influence the affinity

of the interaction of Fc receptors with immuno-

globulins, thereby affecting phagocytic capacity and

consequent antibacterial efficacy. FccRIIIb (CD16)

gene polymorphisms, which influence the affinity of

IgG1 and IgG3 interactions with FcRIII, have been

shown to be associated with periodontitis severity in

Japanese populations. The polymorphic alleles cod-

ing for lower affinity receptors are more commonly

present in patients with more severe and more rap-

idly progressing disease [119, 120, 212].

Smoking

Modification of the host response by an environmental

factor: Studies of risk factors for periodontitis clearly

implicate smoking as a potent risk factor for perio-

dontal disease. Although the precise biological

mechanism behind this relationship are not entirely

clear, physiologic effects of smoking on vasocon-

striction, revascularization of wounds, collagen and

collagenase production, oxygen transport, and

wound healing are almost certain to be important in

the effects on the periodontium. Additionally, several

studies have demonstrated that host responses may

be significantly altered in smokers and by cigarette

smoke.

We have examined the influence of smoking on the

expression of aggressive periodontitis and noted that

generalized aggressive periodontitis patients who

smoked had greater extent and severity of disease

than did patients who did not smoke [194]. As we had

previously found associations between elevated

antibody levels against the pathogens P. gingivalis

and A. actinomycetemcomitans and decreased sever-

ity of disease, we studied how smoking might influ-

ence antibody levels. Because racial groups differ in

serum immunoglobulin levels we analyzed these data

for race-matched groups of subjects. Interestingly, we

noted that IgG2, a predominant subclass of antibody

reactive with periodontal pathogens, was substan-

tially lower in the serum of race-matched generalized

aggressive periodontitis patients who smoked than in

those who did not smoke, consistent with the clinical

findings in this group [176]. This was not the case for

other IgG subclasses nor was it true for patients with

localized aggressive periodontitis or chronic perio-

dontitis. When we examined specific serum anti-

body concentrations to A. actinomycetemcomitans in

generalized aggressive periodontitis patients we

observed much lower concentrations of antibody in

serum from smokers than in serum from nonsmokers

[216]. Although the mechanism for specific suppres-

sion of serum IgG2 and antibody response in this

84

Schenkein

Page 9: Schenke in 2006

group of patients with severe disease is not clear,

these observations demonstrate that a risk factor for

clinical expression of disease may act by modifying

the host immune response and thereby influencing

disease expression [15].

Smoking has also been shown to influence aspects

of the inflammatory response. For example, several

studies demonstrate an impairment of polymorpho-

nuclear leukocyte function by tobacco and its by-

products, including chemotaxis and phagocytosis by

oral and peripheral polymorphonuclear leukocytes,

oxidative burst, and superoxide and hydrogen per-

oxide production [231]. Studies have also demon-

strated that tobacco components can stimulate the

production of proinflammatory cytokines such as

IL-1, IL-6, IL-8, and tumor necrosis factor-a, as well

as transforming growth factor-b, thereby promoting

increased fibrosis, bone resorption, and decreased

bone formation [5, 22, 84, 169, 210].

Conclusion

In attempting to synthesize the large amount of data

on host responses in periodontal diseases one must

consider several fundamental issues that complicate

our understanding of this topic. First, there are mul-

tiple subforms of periodontitis that likely entail dif-

fering combinations of pathogenic mechanisms of

tissue destruction. In addition, each periodontal site

comprises a complex microflora that may interact

with, direct, and modify the host response in a dif-

ferent way. Furthermore, each patient with perio-

dontitis is exposed to different nonmicrobial envi-

ronmental factors that can modify the host response

to the etiologic agents of their periodontal condition.

Finally, each individual possesses amostly immutable

innate level of host susceptibility to periodontal

infections which permits disease expression in a

manner modifiable by the microflora and environ-

ment. It is clear thatmultiplemodels of host response-

based pathogenesis are likely necessary to describe

the way in which periodontal infections can lead to

destruction of hard and soft tissues. Likewise, host-

based mechanisms of maintenance of periodontal

health would vary amongst individuals depending

upon the susceptibility factors outlined above.

Acknowledgment

Supported in part by grantDE 13102 from theNational

Institute of Dental and Craniofacial Research.

References

1. Albandar JM, DeNardin AM, Adesanya MR, Diehl SR, Winn

DM. Associations between serum antibody levels to

periodontal pathogens and early-onset periodontitis.

J Periodontol 2001: 72: 1463–1469.

2. Al-Darmaki S, Knightshead K, Ishihara Y, Best A,

Schenkein HA, Tew JG, Barbour SE. Delineation of the role

of platelet-activating factor in the immunoglobulin G2

antibody response. Clin Diagn Lab Immunol 2004: 11:

720–728.

3. Al-Darmaki S, Schenkein HA, Tew JG, Barbour SE.

Differential expression of platelet-activating factor

acetylhydrolase in macrophages and monocyte-derived

dendritic cells. J Immunol 2003: 170: 167–173.

4. Anderson DC, Schmalsteig FC, Finegold MJ, Hughes BJ,

Rothlein R, Miller LJ, Kohl S, Tosi MF, Jacobs RL, Waldrop

TC, Goldman AS, Shearer WT, Springer TA. The severe and

moderate phenotypes of heritable Mac-1, LFA-1 defici-

ency. Their quantitative definitions and relation to leu-

kocyte dysfunction and clinical features. J Infect Dis 1985:

152: 668–689.

5. Andreou V, D’Addario M, Zohar R, Sukhu B, Casper RF,

Ellen RP, Tenenbaum HC. Inhibition of osteogenesis

in vitro by a cigarette smoke-associated hydrocarbon

combined with Porphyromonas gingivalis lipopolysac-

charide: reversal by resveratrol. J Periodontol 2004: 75:

939–948.

6. Armitage GC, Wu Y, Wang HY, Sorrell J, di Giovine FS, Duff

GW. Low prevalence of a periodontitis-associated inter-

leukin-1 composite genotype in individuals of Chinese

heritage. J Periodontol 2000: 71: 164–171.

7. Assuma R, Oates T, Cochran D, Amar S, Graves DT. IL-1

and TNF antagonists inhibit the inflammatory response

and bone loss in experimental periodontitis. J Immunol

1998: 160: 403–409.

8. Attstrom R, Egelberg J. Emigration of blood neutrophils

and monocytes into the gingival crevice. J Periodontal Res

1970: 5: 48–50.

9. Aukhil I, Lopatin DE, Syed SA, Morrison EC, Kowalski CJ.

The effects of periodontal therapy on serum antibody

(IgG) levels to plaque microorganisms. J Clin Periodontol

1988: 15: 544–550.

10. Bainbridge BW, Darveau RP. Porphyromonas gingivalis

lipopolysaccharide: an unusual pattern recognition

receptor ligand for the innate host defense system. Acta

Odontol Scand 2001: 59: 131–138.

11. Baker P, Wilson M. Opsonic IgG antibody against Actino-

bacillus actinomycetemcomitans in localized juvenile per-

iodontitis. Oral Microbiol Immunol 1989: 4: 98–105.

12. Baker PJ, Dixon M, Evans RT, Dufour L, Johnson E,

Roopenian DC. CD4(+) T cells and the proinflammatory

cytokines gamma interferon and interleukin-6 contribute

to alveolar bone loss in mice. Infect Immun 1999: 67:

2804–2809.

13. Banbula A, Bugno M, Kuster A, Heinrich PC, Travis J,

Potempa J. Rapid and efficient inactivation of IL-6 gingi-

pains, lysine- and arginine-specific proteinases from

Porphyromonas gingivalis. Biochem Biophys Res Commun

1999: 261: 598–602.

14. Barbour SE, Ishihara Y, Fakher M, Al-Darmaki S, Caven

TH, Shelburne CP, Best AM, Schenkein HA, Tew JG.

85

Host responses in periodontal health and disease

Page 10: Schenke in 2006

Monocyte differentiation in localized juvenile periodonti-

tis is skewed toward the dendritic cell phenotype. Infect

Immun 2002: 70: 2780–2786.

15. Barbour SE, Nakashima K, Zhang JB, Tangada S, Hahn CL,

Schenkein HA, Tew JG. Tobacco and smoking: environ-

mental factors that modify the host response (immune

system) and have an impact on periodontal health. Crit

Rev Oral Biol Med 1997: 8: 437–460.

16. Baumann H, Gauldie J. The acute-phase response.

Immunol Today 1994: 15: 74–80.

17. Belibasakis GN, Johansson A, Wang Y, Chen C, Lagergard

T, Kalfas S, Lerner UH. Cytokine responses of human

gingival fibroblasts to Actinobacillus actinomycetemcomi-

tans cytolethal distending toxin. Cytokine 2005: 30: 56–63.

18. Benedek-Spat E, Di Felice R, Andersen E, Cimasoni G.

In vitro release of elastase from human blood and gingival

crevicular neutrophils. Arch Oral Biol 1991: 36: 507–510.

19. Bird PS, Gemmell E, Polak B, Paton RG, Sosroseno W,

Seymour GJ. Protective immunity to Porphyromonas

gingivalis infection in a murine model. J Periodontol 1995:

66: 351–362.

20. Bissell J, Joly S, Johnson GK, Organ CC, Dawson D,

McCray PB Jr, Guthmiller JM. Expression of beta-defen-

sins in gingival health and in periodontal disease. J Oral

Pathol Med 2004: 33: 278–285.

21. Bosshardt DD, Lang NP. The junctional epithelium: from

health to disease. J Dent Res 2005: 84: 9–20.

22. Bostrom L, Linder LE, Bergstrom J. Smoking and cervic-

ular fluid levels of IL-6 and TNF-alpha in periodontal

disease. J Clin Periodontol 1999: 26: 352–357.

23. Boughman JA, Charon JA, Suzuki JB. Biological and

genetical aspects of early onset periodontitis. J Periodontol

1988: 7: 249–257.

24. Califano JV, Gunsolley JC, Nakashima K, Schenkein HA,

Wilson ME, Tew JG. Influence of anti-Actinobacillus

actinomycetemcomitans Y4 (serotype b) lipopoly-

saccharide on severity of generalized early-onset perio-

dontitis. Infect Immun 1996: 64: 3908–3910.

25. Califano JV, Gunsolley JC, Schenkein HA, Tew JG. A

comparison of IgG antibody reactive with Bacteroides

forsythus and Porphyromonas gingivalis in adult and

early-onset periodontitis. J Periodontol 1997: 68: 734–738.

26. Califano JV, Pace BE, Gunsolley JC, Schenkein HA, Lally

ET, Tew JG. Antibody reactive with Actinobacillus actin-

omycetemcomitans leukotoxin in early-onset periodontitis

patients. Oral Microbiol Immunol 1997: 12: 20–26.

27. Califano JV, Schifferle RE, Gunsolley JC, Best AM,

Schenkein HA, Tew JG. Antibody reactive with Porphyro-

monas gingivalis serotypes K1–6 in adult and generalized

early-onset periodontitis. J Periodontol 1999: 70: 730–735.

28. Califano JV, Chou D, Lewis JP, Rogers JD, Best AM,

Schenkein HA. Antibody reactive with Porphyromonas

gingivalis hemagglutinin in chronic and generalized

aggressive periodontitis. J Periodontal Res 2004: 39:

263–268.

29. Calkins CC, Platt K, Potempa J, Travis J. Inactivation of

tumor necrosis factor-alpha by proteinases (gingipains)

from the periodontal pathogen, Porphyromonas gingivalis.

Implications of immune evasion. J Biol Chem 1998: 273:

6611–6614.

30. Chen HA, Johnson BD, Sims TJ, Darveau RP, Moncla BJ,

Whitney CW, Engel D, Page RC. Humoral immune

responses to Porphyromonas gingivalis before and fol-

lowing therapy in rapidly progressive periodontitis

patients. J Periodontol 1991: 62: 781–791.

31. Choi BK, Moon SY, Cha JH, Kim KW, Yoo YJ. Prostaglandin

E (2) is a main mediator in receptor activator of nuclear

factor-kappaB ligand-dependent osteoclastogenesis in-

duced by Porphyromonas gingivalis. Treponema denticola,

and Treponema socranskii. J Periodontol 2005: 76: 813–820.

32. Christersson LA, Albini B, Zambon JJ, Wikesjo UM, Genco

RJ. Tissue localization of Actinobacillus actinomycetem-

comitans in human periodontitis. I. Light, immunofluo-

rescence and electron microscopic studies. J Periodontol

1987: 58: 529–539.

33. Christersson LA. Actinobacillus actinomycetemcomitans

and localized juvenile periodontitis. Clinical, microbio-

logic and histologic studies. Swed Dent J Suppl 1993: 90:

1–46.

34. Chung WO, Hansen SR, Rao D, Dale BA. Protease-activa-

ted receptor signaling increases epithelial antimicrobial

peptide expression. J Immunol 2004: 173: 5165–5170.

35. Cirrincione C, Pimpinelli N, Orlando L, Romagnoli P.

Lamina propria dendritic cells express activation markers

and contact lymphocytes in chronic periodontitis.

J Periodontol 2002: 73: 45–52.

36. Coats SR, Reife RA, Bainbridge BW, Pham TT, Darveau RP.

Porphyromonas gingivalis lipopolysaccharide antagonizes

Escherichia coli lipopolysaccharide at toll-like receptor 4

in human endothelial cells. Infect Immun 2003: 71: 6799–

6807.

37. Cohen EW, Morris AL. Periodontal manifestations of cyclic

neutropenia. J Periodontol 1961: 32: 159–168.

38. Colombo AP, Sakellari D, Haffajee AD, Tanner A, Cugini

MA, Socransky SS. Serum antibodies reacting with sub-

gingival species in refractory periodontitis subjects. J Clin

Periodontol 1998: 25: 596–604.

39. Cooke GS, Hill AV. Genetics of susceptibility to human

infectious disease. Nat Rev Genet 2001: 2: 967–977.

40. Corey LA, Nance WE, Hofstede P, Schenkein HA. Self-

reported periodontal disease in a Virginia twin population.

J Periodontol 1993: 64: 1205–1208.

41. Craandijk J, van Krugten MV, Verweij CL, van der Velden

U, Loos BG. Tumor necrosis factor-alpha gene polymor-

phisms in relation to periodontitis. J Clin Periodontol

2002: 29: 28–34.

42. Cullinan MP, Westerman B, Hamlet SM, Palmer JE, Faddy

MJ, Lang NP, Seymour GJ. A longitudinal study of inter-

leukin-1 gene polymorphisms and periodontal disease in a

general adult population. J Clin Periodontol 2001: 28:

1137–1144.

43. Cunningham MD, Bajorath J, Somerville JE, Darveau RP.

Escherichia coli and Porphyromonas gingivalis lipopoly-

saccharide interactions with CD14: implications for mye-

loid and nonmyeloid cell activation. Clin Infect Dis 1999:

28: 497–504.

44. Curtis MA, Aduse-Opoku J, Rangarajan M. Cysteine pro-

teases of Porphyromonas gingivalis. Crit Rev Oral Biol Med

2001: 12: 192–216.

45. Cutler C, Arnold R, Schenkein H. Inhibition of C3 and IgG

proteolysis enhances phagocytosis of Porphyromonas

gingivalis. J Immunol 1993: 151: 7016–7029.

46. Dale BA. Periodontal epithelium: a newly recognized role

in health and disease. Periodontol 2000 2002: 30: 70–78.

86

Schenkein

Page 11: Schenke in 2006

47. Darby IB, Mooney J, Kinane DF. Changes in subgingival

microflora and humoral immune response following per-

iodontal therapy. J Clin Periodontol 2001: 28: 796–805.

48. Darveau RP, Arbabi S, Garcia I, Bainbridge B, Maier RV.

Porphyromonas gingivalis lipopolysaccharide is both

agonist and antagonist for p38 mitogen-activated protein

kinase activation. Infect Immun 2002: 70: 1867–1873.

49. Darveau RP, Pham TT, Lemley K, Reife RA, Bainbridge BW,

Coats SR, Howald WN, Way SS, Hajjar AM. Porphyro-

monas gingivalis lipopolysaccharide contains multiple

lipid A species that functionally interact with both toll-like

receptors 2 and 4. Infect Immun 2004: 72: 5041–5051.

50. DeCarlo AA, Huang Y, Collyer CA, Langley DB, Katz J.

Feasibility of an HA2 domain-based periodontitis vaccine.

Infect Immun 2003: 71: 562–566.

51. de Jong EC, Smits HH, Kapsenberg ML. Dendritic cell-

mediated T cell polarization. Springer Semin Immunopa-

thol 2005: 26: 289–307.

52. Delima AJ, Karatzas S, Amar S, Graves DT. Inflammation

and tissue loss caused by periodontal pathogens is reduced

by interleukin-1 antagonists. J Infect Dis 2002: 186: 511–516.

53. Delima AJ, Oates T, Assuma R, Schwartz Z, Cochran D,

Amar S, Graves DT. Soluble antagonists to interleukin-1

(IL-1) and tumor necrosis factor (TNF) inhibits loss of

tissue attachment in experimental periodontitis. J Clin

Periodontol 2001: 28: 233–240.

54. Dereka XE, Tosios KI, Chrysomali E, Angelopoulou E.

Factor XIIIa+ dendritic cells and S-100 protein+ Langerh-

ans’ cells in adult periodontitis. J Periodontal Res 2004: 39:

447–452.

55. Diehl SR, Wang Y, Brooks CN, Burmeister JA, Califano JV,

Wang S, Schenkein HA. Linkage disequilibrium of inter-

leukin-1 genetic polymorphisms with early-onset perio-

dontitis. J Periodontol 1999: 70: 418–430.

56. Diehl SR, Wu T, Burmeister JA, Califano JV, Brooks CN,

Tew JG, Schenkein HA. Evidence of a substantial genetic

basis for IgG2 levels in families with aggressive perio-

dontitis. J Dent Res 2003: 82: 708–712.

57. DiRienzo J, McKay T. Identification and characterization

of genetic cluster groups of Actinobacillus actinomyce-

temcomitans isolated from the human oral cavity. J Clin

Microbiol 1994: 32: 75–81.

58. DiRienzo JM, Slots J, Sixou M, Sol MA, Harmon R, McKay

TL. Specific genetic variants of Actinobacillus actin-

omycetemcomitans correlate with disease and health in a

regional population of families with localized juvenile

periodontitis. Infect Immun 1994: 62: 3058–3065.

59. Domeij H, Yucel-Lindberg T, Modeer T. Signal pathways

involved in the production ofMMP-1 andMMP-3 in human

gingival fibroblasts. Eur J Oral Sci 2002: 110: 302–306.

60. Dongari-Bagtzoglou AI, Ebersole JL. Production of

inflammatory mediators and cytokines by human gingival

fibroblasts following bacterial challenge. J Periodontal Res

1996: 31: 90–98.

61. Ebersole J. Systemic humoral immune responses in perio-

dontal disease. Crit Rev Oral Biol Med 1990: 1: 283–331.

62. Ebersole JL, Cappelli D. Gingival crevicular fluid antibody

to Actinobacillus actinomycetemcomitans in periodontal

disease. Oral Microbiol Immunol 1994: 9: 335–344.

63. Ebersole JL, Taubman MA, Smith DJ, Haffajee AD. Effect

of subgingival scaling on systemic antibody responses to

oral microorganisms. Infect Immun 1985: 48: 534–539.

64. Endo M, Tai H, Tabeta K, Kobayashi T, Yamazaki K, Yoshie

H. Analysis of single nucleotide polymorphisms in the

5¢-flanking region of tumor necrosis factor-alpha gene in

Japanese patients with early-onset periodontitis. J Perio-

dontol 2001: 72: 1554–1559.

65. Evans RT, Klausen B, Sojar HT, Bedi GS, Sfintescu C,

Ramamurthy NS, Golub LM, Genco RJ. Immunization

with Porphyromonas (Bacteroides) gingivalis fimbriae

protects against periodontal destruction. Infect Immun

1992: 60: 2926–2935.

66. Fassmann A, Holla LI, Buckova D, Vasku A, Znojil V, Vanek

J. Polymorphisms in the +252 (A ⁄ G) lymphotoxin-alpha

and the )308 (A ⁄ G) tumor necrosis factor-alpha genes

and susceptibility to chronic periodontitis in a Czech

population. J Periodontal Res 2003: 38: 394–399.

67. Fives-Taylor P, Meyer D, Mintz K. Characteristics of

Actinobacillus actinomycetemcomitans invasion of and

adhesion to cultured epithelial cells. Adv Dent Res 1995: 9:

55–62.

68. Galbraith GM, Hendley TM, Sanders JJ, Palesch Y, Pandey

JP. Polymorphic cytokine genotypes as markers of disease

severity in adult periodontitis. J Clin Periodontol 1999: 26:

705–709.

69. Gemmell E, Carter CL, Bird PS, Seymour GJ. Genetic

dependence of the specific T-cell cytokine response to

Porphyromonas gingivalis in mice. J Periodontol 2002: 73:

591–596.

70. Gemmell E, Kjeldsen M, Yamazaki K, Nakajima T, Aldred

MJ, Seymour GJ. Cytokine profiles of Porphyromonas

gingivalis-reactive T lymphocyte line and clones derived

from P. gingivalis-infected subjects. Oral Dis 1995: 1: 139–

146.

71. Gemmell E, Marshall RI, Seymour GJ. Cytokines and

prostaglandins in immune homeostasis and tissue

destruction in periodontal disease. Periodontol 2000 1997:

14: 112–143.

72. Gemmell E, Seymour GJ. Cytokine profiles of cells

extracted from humans with periodontal diseases. J Dent

Res 1998: 77: 16–26.

73. Gemmell E, Yamazaki K, Seymour GJ. Destructive perio-

dontitis lesions are determined by the nature of the

lymphocytic response. Crit Rev Oral Biol Med 2002: 13:

17–34.

74. Genco CA, Potempa J, Mikolajczyk-Pawlinska J, Travis J.

Role of gingipains R in the pathogenesis of Porphyro-

monas gingivalis-mediated periodontal disease. Clin Infect

Dis 1999: 28: 456–465.

75. Ghoneum M, Saglie R, Brown J, Regala C. Actinobacillus

actinomycetemcomitans suppresses rat natural killer cell

activity in vivo. Acta Otolaryngol 2004: 124: 621–627.

76. Gibson FC 3rd, Genco CA. Prevention of Porphyromonas

gingivalis-induced oral bone loss following immunization

with gingipain R1. Infect Immun 2001: 69: 7959–7963.

77. Gonzales JR, Michel J, Diete A, Herrmann JM, Bodeker RH,

Meyle J. Analysis of genetic polymorphisms at the inter-

leukin-10 loci in aggressive and chronic periodontitis.

J Clin Periodontol 2002: 29: 816–822.

78. Gonzalez D, Tzianabos AO, Genco CA, Gibson FC 3rd.

Immunization with Porphyromonas gingivalis capsular

polysaccharide prevents P. gingivalis-elicited oral bone

loss in a murine model. Infect Immun 2003: 71: 2283–

2287.

87

Host responses in periodontal health and disease

Page 12: Schenke in 2006

79. Goodson J, Dewhirst F, Brunetti A. Prostaglandin E2 levels

and human periodontal disease. Prostaglandins 1974: 6:

81–85.

80. Gore EA, Sanders JJ, Pandey JP, Palesch Y, Galbraith GM.

Interleukin-1beta+3953 allele 2: association with disease

status in adult periodontitis. J Clin Periodontol 1998: 25:

781–785.

81. Graves DT, Delima AJ, Assuma R, Amar S, Oates T,

Cochran D. Interleukin-1 and tumor necrosis factor

antagonists inhibit the progression of inflammatory cell

infiltration toward alveolar bone in experimental perio-

dontitis. J Periodontol 1998: 69: 1419–1425.

82. Greenstein G, Hart TC. A critical assessment of interleu-

kin-1 (IL-1) genotyping when used in a genetic suscepti-

bility test for severe chronic periodontitis. J Periodontol

2002: 73: 231–247.

83. Gunsolley JC, Burmeister JA, Tew JG, Best AM, Ranney

RR. Relationship of serum antibody to attachment level

patterns in young adults with juvenile periodontitis or

generalized severe periodontitis. J Periodontol 1987: 58:

314–320.

84. Gustafsson A, Asman B, Bergstrom K. Cigarette smoking

as an aggravating factor in inflammatory tissue-destruc-

tive diseases. Increase in tumor necrosis Factor-alpha

priming of peripheral neutrophils measured as genera-

tion of oxygen radicals. Int J Clin Lab Res 2000: 30: 187–

190.

85. Hart TC, Hart PS, Bowden DW, Michalec MD, Callison SA,

Walker SJ, Zhang Y, Firatli E. Mutations of the cathepsin C

gene are responsible for Papillon–Lefevre syndrome. J Med

Genet 1999: 36: 881–887.

86. Hart TC, Hart PS, Michalec MD, Zhang Y, Firatli E, Van

Dyke TE, Stabholz A, Zlotogorski A, Shapira L, Soskolne

WA. Haim–Munk syndrome and Papillon–Lefevre syn-

drome are allelic mutations in cathepsin C. J Med Genet

2000: 37: 88–94.

87. Hart TC, Hart PS, Michalec MD, Zhang Y, Marazita ML,

Cooper M, Yassin OM, Nusier M, Walker S. Localisation of

a gene for prepubertal periodontitis to chromosome 11q14

and identification of a cathepsin C gene mutation. J Med

Genet 2000: 37: 95–101.

88. Hart TC, Kornman KS. Genetic factors in the pathogenesis

of periodontitis. Periodontol 2000 1997: 14: 202–215.

89. Hart TE, Shapira L, Van Dyke TE. Neutrophil defects as

risk factors for periodontal diseases. J Periodontol 1994:

65: 521–529.

90. Haubek D, Poulsen K, Asikainen S, Kilian M. Evidence for

absence in northern Europe of especially virulent clonal

types of Actinobacillus actinomycetemcomitans. J Clin

Microbiol 1995: 33: 395–401.

91. Heasman P, Collins J, Offenbacher S. Changes in crevic-

ular fluid levels of interleukin-1 beta, leukotriene B4,

prostaglandin E2, thromboxane B2 and tumour necrosis

factor alpha in experimental gingivitis in humans. J Perio-

dontal Res 1993: 28: 241–247.

92. Heasman P, Seymour R. The effect of a systemically-

administered non-steroidal anti-inflammatory drug

(flurbiprofen) on experimental gingivitis in humans. J Clin

Periodontol 1989: 16: 551–556.

93. Hellden L, Ericson T, Lindhe J. Neutrophil chemotactic

substances in different fractions of soluble dental plaque

material. Scand J Dent Res 1973: 81: 276–284.

94. Hellden L, Lindhe J. Enhanced emigration of crevicular

leukocytes mediated by factors in human dental plaque.

Scand J Dent Res 1973: 81: 123–129.

95. Hennig BJ, Parkhill JM, Chapple IL, Heasman PA, Taylor JJ.

Dinucleotide repeat polymorphism in the interleukin-10

gene promoter (IL-10.G) and genetic susceptibility to

early-onset periodontal disease. Genes Immun 2000: 1:

402–404.

96. Hill AV. The genomics and genetics of human infectious

disease susceptibility. Annu Rev Genomics Hum Genet

2001: 2: 373–400.

97. Hofbauer LC, Heufelder AE. Role of receptor activator of

nuclear factor-kappaB ligand and osteoprotegerin in bone

cell biology. J Mol Med 2001: 79: 243–253.

98. Horowitz MC, Xi Y, Wilson K, Kacena MA. Control of

osteoclastogenesis and bone resorption by members of

the TNF family of receptors and ligands. Cytokine Growth

Factor Rev 2001: 12: 9–18.

99. Hosaka Y, Saito A, Nakagawa T, Seida K, Yamada S, Okuda

K. Effect of initial therapy on dynamics of immunogloblin

G levels to some periodontopathic bacteria in serum and

gingival crevicular fluid. Bull Tokyo Dent Coll 1994: 35:

207–216.

100. Howell T, Williams R. Nonsteroidal antiinflammatory

drugs as inhibitors of periodontal disease progression. Crit

Rev Oral Biol Med 1993: 4: 177–196.

101. Imamura T. The role of gingipains in the pathogenesis of

periodontal disease. J Periodontol 2003: 74: 111–118.

102. Ishihara Y, Zhang JB, Fakher M, Best AM, Schenkein HA,

Barbour SE, Tew JG. Non-redundant roles for interleukin-

1 alpha and interleukin-1 beta in regulating human IgG2.

J Periodontol 2001: 72: 1332–1339.

103. Ishihara Y, Zhang JB, Quinn SM, Schenkein HA, Best AM,

Barbour SE, Tew JG. Regulation of immunoglobulin G2

production by prostaglandin E (2) and platelet-activating

factor. Infect Immun 2000: 68: 1563–1568.

104. Jagels MA, Ember JA, Travis J, Potempa J, Pike R, Hugli

TE. Cleavage of the human C5A receptor by proteinases

derived from Porphyromonas gingivalis: cleavage of leu-

kocyte C5a receptor. Adv Exp Med Biol 1996: 389:

155–164.

105. Jagels MA, Travis J, Potempa J, Pike R, Hugli TE. Proteo-

lytic inactivation of the leukocyte C5a receptor by pro-

teinases derived from Porphyromonas gingivalis. Infect

Immun 1996: 64: 1984–1991.

106. Jankovic D, Liu Z, Gause WC. Th1- and Th2-cell com-

mitment during infectious disease: asymmetry in diver-

gent pathways. Trends Immunol 2001: 22: 450–457.

107. Jiang Y, Mehta CK, Hsu TY, Alsulaimani FF. Bacteria

induce osteoclastogenesis via an osteoblast-independent

pathway. Infect Immun 2002: 70: 3143–3148.

108. Jotwani R, Cutler CW. Multiple dendritic cell (DC) sub-

populations in human gingiva and association of mature

DCs with CD4+ T-cells in situ. J Dent Res 2003: 82:

736–741.

109. Jotwani R, Palucka AK, Al-Quotub M, Nouri-Shirazi M,

Kim J, Bell D, Banchereau J, Cutler CW. Mature dendritic

cells infiltrate the T cell-rich region of oral mucosa in

chronic periodontitis: in situ, in vivo, and in vitro studies.

J Immunol 2001: 167: 4693–4700.

110. Juhl M, Stoltze K, Reibel J. Distribution of Langerhans cells

in clinically healthy-human gingival epithelium with

88

Schenkein

Page 13: Schenke in 2006

special emphasis on junctional epithelium. Scand J Dent

Res 1988: 96: 199–208.

111. Kang BY, Choi YK, Choi WH, Kim KT, Choi SS, Kim K, Ha

NJ. Two polymorphisms of interleukin-4 gene in Korean

adult periodontitis. Arch Pharm Res 2003: 26: 482–486.

112. Kantarci A, Oyaizu K, Van Dyke TE. Neutrophil-mediated

tissue injury in periodontal disease pathogenesis: findings

from localized aggressive periodontitis. J Periodontol 2003:

74: 66–75.

113. Kantarci A, Van Dyke TE. Neutrophil-mediated host re-

sponse to Porphyromonas gingivalis. J Int Acad Period-

ontol 2002: 4: 119–125.

114. Kapsenberg ML. Dendritic-cell control of pathogen-driven

T-cell polarization. Nat Rev Immunol 2003: 3: 984–993.

115. Katz J, Black KP, Michalek SM. Host responses to recom-

binant hemagglutinin B of Porphyromonas gingivalis in an

experimental rat model. Infect Immun 1999: 67: 4352–4359.

116. Kesavalu L, Chandrasekar B, Ebersole JL. In vivo induction

of proinflammatory cytokines in mouse tissue by Por-

phyromonas gingivalis and Actinobacillus actinomyce-

temcomitans. Oral Microbiol Immunol 2002: 17: 177–180.

117. Kikuchi T, Hahn CL, Tanaka S, Barbour SE, Schenkein HA,

Tew JG. Dendritic cells stimulated with Actinobacillus

actinomycetemcomitans elicit rapid gamma interferon

responses by natural killer cells. Infect Immun 2004: 72:

5089–5096.

118. Kinane DF, Hodge P, Eskdale J, Ellis R, Gallagher G. Ana-

lysis of genetic polymorphisms at the interleukin-10 and

tumour necrosis factor loci in early-onset periodontitis.

J Periodontal Res 1999: 34: 379–386.

119. Kobayashi T, Sugita N, van der Pol WL, Nunokawa Y,

Westerdaal NA, Yamamoto K, van de Winkel JG, Yoshie H.

The Fcgamma receptor genotype as a risk factor for gen-

eralized early-onset periodontitis in Japanese patients.

J Periodontol 2000: 71: 1425–1432.

120. Kobayashi T, Yamamoto K, Sugita N, van der Pol WL,

Yasuda K, Kaneko S, van de Winkel JG, Yoshie H. The Fc

gamma receptor genotype as a severity factor for chronic

periodontitis in Japanese patients. J Periodontol 2001: 72:

1324–1331.

121. Kobayashi-Sakamoto M, Hirose K, Isogai E, Chiba I.

NF-kappaB-dependent induction of osteoprotegerin by

Porphyromonas gingivalis in endothelial cells. Biochem

Biophys Res Commun 2004: 315: 107–112.

122. Kornman KS, di Giovine FS. Genetic variations in cytokine

expression: a risk factor for severity of adult periodontitis.

Ann Periodontol 1998: 3: 327–338.

123. Kurita-Ochiai T, Ochiai K, Ikeda T. Immunosuppressive

effect induced by Actinobacillus actinomycetemcomitans:

effect on immunoglobulin production and lymphokine

synthesis. Oral Microbiol Immunol 1992: 7: 338–343.

124. Kurita-Ochiai T, Ochiai K, Saito N, Ikeda T. Adoptive

transfer of suppressor T cells induced by Actinobacillus

actinomycetemcomitans regulates immune response.

J Periodontal Res 1994: 29: 1–8.

125. Lah TT, Babnik J, Schiffmann E, Turk V, Skaleric U.

Cysteine proteinases and inhibitors in inflammation:

their role in periodontal disease. J Periodontol 1993: 64:

485–491.

126. Lamont R, Chan A, Belton C, Izutsu K, Vasel D, Weinberg

A. Porphyromonas gingivalis invasion of gingival epithelial

cells. Infect Immun 1995: 63: 3878–3885.

127. Liu D, Xu JK, Figliomeni L, Huang L, Pavlos NJ, Rogers M,

Tan A, Price P, Zheng MH. Expression of RANKL and OPG

mRNA in periodontal disease: possible involvement in

bone destruction. Int J Mol Med 2003: 11: 17–21.

128. Lossdorfer S, Gotz W, Jager A. Immunohistochemical

localization of receptor activator of nuclear factor kappaB

(RANK) and its ligand (RANKL) in human deciduous teeth.

Calcif Tissue Int 2002: 71: 45–52.

129. Lu H, Wang M, Gunsolley JC, Schenkein HA, Tew JG.

Serum immunoglobulin G subclass concentrations in

periodontally healthy and diseased individuals. Infect

Immun 1994: 62: 1677–1682.

130. Lu Q, Jin L, Darveau RP, Samaranayake LP. Expression of

human beta-defensins-1 and -2 peptides in unresolved

chronic periodontitis. J Periodontal Res 2004: 39: 221–227.

131. Lundy FT, Orr DF, Shaw C, Lamey PJ, Linden GJ. Detec-

tion of individual human neutrophil alpha-defensins

(human neutrophil peptides 1, 2 and 3) in unfractionated

gingival crevicular fluid – a MALDI-MS approach. Mol

Immunol 2005: 42: 575–579.

132. Mahamed DA, Marleau A, Alnaeeli M, Singh B, Zhang X,

Penninger JM, Teng YT. G(–) anaerobes-reactive CD4+

T-cells trigger RANKL-mediated enhanced alveolar bone

loss in diabetic NOD mice. Diabetes 2005: 54: 1477–1486.

133. Marazita ML, Burmeister JA, Gunsolley JC, Koertge TE,

Lake K, Schenkein HA. Evidence for autosomal dominant

inheritance and race-specific heterogeneity in early-onset

periodontitis. J Periodontol 1994: 65: 623–630.

134. Mark LL, Haffajee AD, Socransky SS, Kent RL Jr, Guerrero

D, Kornman K, Newman M, Stashenko P. Effect of the

interleukin-1 genotype on monocyte IL-1beta expression

in subjects with adult periodontitis. J Periodontal Res 2000:

35: 172–177.

135. McCulloch CA. Host enzymes in gingival crevicular fluid

as diagnostic indicators of periodontitis. J Clin Periodontol

1994: 21: 497–506.

136. Meisel P, Siegemund A, Dombrowa S, Sawaf H, Fanghae-

nel J, Kocher T. Smoking and polymorphisms of the

interleukin-1 gene cluster (IL-1alpha, IL-1beta, and

IL-1RN) in patients with periodontal disease. J Periodontol

2002: 73: 27–32.

137. Meisel P, Siegemund A, Grimm R, Herrmann FH, John U,

Schwahn C, Kocher T. The interleukin-1 polymorphism,

smoking, and the risk of periodontal disease in the pop-

ulation-based SHIP study. J Dent Res 2003: 82: 189–193.

138. Meyle J. Leukocyte adhesion deficiency and prepubertal

periodontitis. Periodontol 2000 1994: 6: 26–36.

139. Mezyk-Kopec R, Bzowska M, Potempa J, Jura N, Sroka A,

Black RA, Bereta J. Inactivation of membrane tumor nec-

rosis factor alpha by gingipains from Porphyromonas

gingivalis. Infect Immun 2005: 73: 1506–1514.

140. Michalowicz BS. Genetic and inheritance considerations

in periodontal disease. Curr Opin Periodontol 1993: 11–17.

141. Michalowicz BS. Genetic risk factors for the periodontal

diseases. Compendium 1994: 15: 1036, 1038, 1040.

142. Michalowicz BS, Diehl SR, Gunsolley JC, Sparks BS, Brooks

CN, Koertge TE, Califano JV, Burmeister JA, Schenkein HA.

Evidence of a substantial genetic basis for risk of adult

periodontitis. J Periodontol 2000: 71: 1699–1707.

143. Michel J, Gonzales JR, Wunderlich D, Diete A, Herrmann

JM, Meyle J. Interleukin-4 polymorphisms in early onset

periodontitis. J Clin Periodontol 2001: 28: 483–488.

89

Host responses in periodontal health and disease

Page 14: Schenke in 2006

144. Mikolajczyk-Pawlinska J, Travis J, Potempa J. Modulation

of interleukin-8 activity by gingipains from Porphyro-

monas gingivalis: implications for pathogenicity of perio-

dontal disease. FEBS Lett 1998: 440: 282–286.

145. Miyasaki K. The neutrophil: mechanisms of controlling

periodontal bacteria. J Periodontol 1991: 62: 761–774.

146. Miyauchi M, Kitagawa S, Hiraoka M, Saito A, Sato S, Kudo

Y, Ogawa I, Takata T. Immunolocalization of CXC chem-

okine and recruitment of polymorphonuclear leukocytes

in the rat molar periodontal tissue after topical application

of lipopolysaccharide. Histochem Cell Biol 2004: 121:

291–297.

147. Miyauchi M, Sato S, Kitagawa S, Hiraoka M, Kudo Y,

Ogawa I, Zhao M, Takata T. Cytokine expression in rat

molar gingival periodontal tissues after topical application

of lipopolysaccharide. Histochem Cell Biol 2001: 116:

57–62.

148. Mogi M, Otogoto J, Ota N, Togari A. Differential expression

of RANKL and osteoprotegerin in gingival crevicular

fluid of patients with periodontitis. J Dent Res 2004: 83:

166–169.

149. Mouton C, Desclauriers M, Allard H, Bouchard M. Serum

antibodies to Bacteroides gingivalis in periodontitis: a

longitudinal study. J Periodontal Res 1987: 22: 426–430.

150. Mouton C, Hammond PG, Slots J, Genco RJ. Serum anti-

bodies to oral Bacteroides asaccharolyticus (Bacteroides

gingivalis): relationship to age and periondontal disease.

Infect Immun 1981: 31: 182–192.

151. Murray PA, Burstein DA, Winkler JR. Antibodies to

Bacteroides gingivalis in patientswith treatedanduntreated

periodontal disease. J Periodontol 1989: 60: 96–103.

152. Nakayama K. Molecular genetics of Porphyromonas gin-

givalis: gingipains and other virulence factors. Curr

Protein Pept Sci 2003: 4: 389–395.

153. Nichols FC, Levinbook H, Shnaydman M, Goldschmidt J.

Prostaglandin E2 secretion from gingival fibroblasts trea-

ted with interleukin-1beta: effects of lipid extracts from

Porphyromonas gingivalis or calculus. J Periodontal Res

2001: 36: 142–152.

154. Nisapakultorn K, Ross KF, Herzberg MC. Calprotectin

expression in vitro by oral epithelial cells confers resist-

ance to infection by Porphyromonas gingivalis. Infect

Immun 2001: 69: 4242–4247.

155. Nishikawa M, Yamaguchi Y, Yoshitake K, Saeki Y. Effects of

TNFalpha and prostaglandin E2 on the expression of

MMPs in human periodontal ligament fibroblasts. J Perio-

dontal Res 2002: 37: 167–176.

156. Oates TW, Graves DT, Cochran DL. Clinical, radiographic

and biochemical assessment of IL-1 ⁄ TNF-alpha antag-

onist inhibition of bone loss in experimental periodontitis.

J Clin Periodontol 2002: 29: 137–143.

157. O’Brien-Simpson NM, Paolini RA, Reynolds EC. RgpA-Kgp

peptide-based immunogens provide protection against

Porphyromonas gingivalis challenge in a murine lesion

model. Infect Immun 2000: 68: 4055–4063.

158. O’Brien-Simpson NM, Veith PD, Dashper SG, Reynolds

EC. Porphyromonas gingivalis gingipains: the molecular

teeth of a microbial vampire. Curr Protein Pept Sci 2003: 4:

409–426.

159. Offenbacher S, Farr D, Goodson J. Measurement of pros-

taglandin E in crevicular fluid. J Clin Periodontol 1981: 8:

359–367.

160. Offenbacher S, Heasman P, Collins J. Modulation of host

PGE2 secretion as a determinant of periodontal disease

expression. J Periodontol 1993: 64: 432–444.

161. Offenbacher S, Odle BM, Gray RC, Van Dyke TE. Crevic-

ular fluid prostaglandin E levels as a measure of the per-

iodontal disease status of adult and juvenile periodontitis

patients. J Periodontal Res 1984: 19: 1–13.

162. Offenbacher S, Williams R, Jeffcoat M, Howell T, Odle B,

Smith M, Hall C, Johnson H, Goldhaber P. Effects of

NSAIDs on beagle crevicular cyclooxygenase metabolites

and periodontal bone loss. J Periodontal Res 1992: 27:

207–213.

163. Okada H, Murakami S. Cytokine expression in periodontal

health and disease. Crit Rev Oral Biol Med 1998: 9:

248–266.

164. Okamatsu Y, Kobayashi M, Nishihara T, Hasegawa K.

Interleukin-1 alpha produced in human gingival fibro-

blasts induces several activities related to the progression

of periodontitis by direct contact. J Periodontal Res 1996:

31: 355–364.

165. Oleksy A, Banbula A, Bugno M, Travis J, Potempa J. Pro-

teolysis of interleukin-6 receptor (IL-6R) by Porphyro-

monas gingivalis cysteine proteinases (gingipains) inhibits

interleukin-6-mediated cell activation. Microb Pathog

2002: 32: 173–181.

166. Page RC. Vaccination and periodontitis: myth or reality.

J Int Acad Periodontol 2000: 2: 31–43.

167. Page RC, Schroeder HE. Pathogenesis of inflammatory

periodontal disease. Lab Invest 1976: 33: 235–249.

168. Page RC, Schroeder H. Periodontitis in man and other

animals. A comparative review. Basel: S. Karger, 1982.

169. Palmer RM, Wilson RF, Hasan AS, Scott DA. Mechanisms

of action of environmental factors – tobacco smoking.

J Clin Periodontol 2005: 32 (Suppl. 6): 180–195.

170. Papapanou PN, Neiderud AM, Disick E, Lalla E, Miller GC,

Dahlen G. Longitudinal stability of serum immunoglob-

ulin G responses to periodontal bacteria. J Clin Period-

ontol 2004: 31: 985–990.

171. Paquette D. Potential role of nonsteroidal anti-inflam-

matory drugs in the treatment of periodontitis. Compen-

dium 1992: 13: 1174–1179.

172. Pippin DJ. Increased intracellular levels of beta-glucu-

ronidase in polymorphonuclear leucocytes from humans

with rapidly progressive periodontitis. Arch Oral Biol 1990:

35: 325–328.

173. Pippin DJ, Cobb CM, Feil P. Increased intracellular levels

of lysosomal beta-glucuronidase in peripheral blood

PMNs from humans with rapidly progressive periodonti-

tis. J Periodontal Res 1995: 30: 42–50.

174. Potempa J, Sroka A, Imamura T, Travis J. Gingipains, the

major cysteine proteinases and virulence factors of Por-

phyromonas gingivalis: structure, function and assembly

of multidomain protein complexes. Curr Protein Pept Sci

2003: 4: 397–407.

175. Putsep K, Carlsson G, Boman HG, Andersson M. Defici-

ency of antibacterial peptides in patients with morbus

Kostmann: an observation study. The Lancet 2002: 360:

1144–1149.

176. Quinn SM, Zhang JB, Gunsolley JC, Schenkein JG,

Schenkein HA, Tew JG. Influence of smoking and race on

immunoglobulin G subclass concentrations in early-onset

periodontitis patients. Infect Immun 1996: 64: 2500–2505.

90

Schenkein

Page 15: Schenke in 2006

177. Rabie G, Lally ET, Shenker BJ. Immunosuppressive prop-

erties of Actinobacillus actinomycetemcomitans leuko-

toxin. Infect Immun 1988: 56: 122–127.

178. Rajapakse PS, O’Brien-Simpson NM, Slakeski N, Hoff-

mann B, Reynolds EC. Immunization with the RgpA-Kgp

proteinase-adhesin complexes of Porphyromonas gingi-

valis protects against periodontal bone loss in the rat

periodontitis model. Infect Immun 2002: 70: 2480–2486.

179. Ramamurthy NS, Rifkin BR, Greenwald RA, Xu JW, Liu Y,

Turner G, Golub LM, Vernillo AT. Inhibition of matrix

metalloproteinase-mediated periodontal bone loss in rats:

a comparison of 6 chemically modified tetracyclines.

J Periodontol 2002: 73: 726–734.

180. Ramamurthy NS, Xu JW, Bird J, Baxter A, Bhogal R, Wills R,

Watson B, Owen D, Wolff M, Greenwald RA. Inhibition of

alveolar bone loss by matrix metalloproteinase inhibitors

in experimental periodontal disease. J Periodontal Res

2002: 37: 1–7.

181. Ranney RR. Specific antibody in gingiva and submandib-

ular nodes of monkeys with allergic periodontal disease.

J Periodontal Res 1970: 5: 1–7.

182. Ranney RR, Yanni NR, Burmeister JA, Tew JG. Relationship

between attachment loss and precipitating serum anti-

body to Actinobacillus actinomycetemcomitans in adoles-

cents and young adults having severe periodontal

destruction. J Periodontol 1982: 53: 1–7.

183. Ranney RR, Zander HA. Allergic periodontal disease in

sensitized squirrel monkeys. J Periodontol 1970: 41: 12–21.

184. Reife RA, Shapiro RA, Bamber BA, Berry KK, Mick GE,

Darveau RP. Porphyromonas gingivalis lipopolysaccharide

is poorly recognized by molecular components of innate

host defense in a mouse model of early inflammation.

Infect Immun 1995: 63: 4686–4694.

185. Ross BC, Czajkowski L, Hocking D, Margetts M, Webb E,

Rothel L, Patterson M, Agius C, Camuglia S, Reynolds E,

Littlejohn T, Gaeta B, Ng A, Kuczek ES, Mattick JS, Gearing

D, Barr IG. Identification of vaccine candidate antigens

from a genomic analysis of Porphyromonas gingivalis.

Vaccine 2001: 19: 4135–4142.

186. Ruwanpura SM, Noguchi K, Ishikawa I. Prostaglandin E2

regulates interleukin-1beta-induced matrix metalloprote-

inase-3 production in human gingival fibroblasts. J Dent

Res 2004: 83: 260–265.

187. Saglie FR. Scanning electron microscope and intragingival

microorganisms in periodontal diseases. Scanning Microsc

1988: 2: 1535–1540.

188. Saglie FR, Pertuiset J, Rezende MT, Nestor M, Marfany A,

Cheng J. In situ correlative immuno-identification of

mononuclear infiltrates and invasive bacteria in diseased

gingiva. J Periodontol 1988: 59: 688–696.

189. Sakellari D, Koukoudetsos S, Arsenakis M, Konstantinidis

A. Prevalence of IL-1A and IL-1B polymorphisms in a

Greek population. J Clin Periodontol 2003: 30: 35–41.

190. Sandholm L, Tolo K. Serum antibody levels to 4 perio-

dontal pathogens remain unaltered after mechanical

therapy of juvenile periodontitis. J Clin Periodontol 1986:

13: 646–650.

191. Schenkein H. Failure of Bacteroides gingivalis W83 to

accumulate bound C3 following opsonization with serum.

J Periodontal Res 1989: 24: 20–27.

192. Schenkein HA. The role of complement in periodontal

diseases. Crit Rev Oral Biol Med 1991: 2: 65–81.

193. Schenkein HA, Berry CR. Activation of complement by

Treponema denticola. J Dent Res 1991: 70: 107–110.

194. Schenkein HA, Gunsolley JC, Koertge TE, Schenkein JG,

Tew JG. Smoking and its effects on early-onset periodon-

titis. J Am Dent Assoc 1995: 126: 1107–1113.

195. Schenkein HA, Van Dyke TE. Early-onset periodontitis

systemic aspects of etiology and pathogenesis. Periodontol

2000 1994: 6: 7–25.

196. Schroeder HE. Transmigration and infiltration of leuko-

cytes in human junctional epithelium. Helv Odontol Acta

1973: 17: 6–18.

197. Schroeder HE, Listgarten MA. The gingival tissues: The

architecture of peridontal protection. Periodontol 2000

1997: 13: 91–120.

198. Sengupta S, Fine J, Wu-Wang C, Gordon J, Murty V,

Slomiany A, Slomiany B. The relationship of prostaglan-

dins to cAMP, IgG, IgM and alpha-2-macroglobulin in

gingival crevicular fluid in chronic adult periodontitis.

Arch Oral Biol 1990: 35: 593–596.

199. Seymour GJ, Gemmell E. Cytokines in periodontal disease:

where to from here? Acta Odontol Scand 2001: 59:

167–173.

200. Shapira L, Borinski R, Sela MN, Soskolne A. Superoxide

formation and chemiluminescence of peripheral poly-

morphonuclear leukocytes in rapidly progressive perio-

dontitis patients. J Clin Periodontol 1991: 18: 44–48.

201. Shapira L, Stabholz A, Rieckmann P, Kruse N. Genetic

polymorphism of the tumor necrosis factor (TNF)-alpha

promoter region in families with localized early-onset

periodontitis. J Periodontal Res 2001: 36: 183–186.

202. Sheets SM, Potempa J, Travis J, Casiano CA, Fletcher HM.

Gingipains from Porphyromonas gingivalis W83 induce

cell adhesion molecule cleavage and apoptosis in endot-

helial cells. Infect Immun 2005: 73: 1543–1552.

203. Shenker BJ, Kushner ME, Tsai CC. Inhibition of fibroblast

proliferation by Actinobacillus actinomycetemcomitans.

Infect Immun 1982: 38: 986–992.

204. Shenker BJ, McArthur WP, Tsai CC. Immune suppression

induced by Actinobacillus actinomycetemcomitans. I.

Effects on human peripheral blood lymphocyte responses

to mitogens and antigens. J Immunol 1982: 128: 148–154.

205. Shenker B, Vitale L, Welham D. Immune suppression

induced by Actinobacillus actinomycetemcomitans: effects

on immunoglobulin production by human B cells. Infect

Immun 1990: 58: 3856–3862.

206. Socransky SS, Haffajee AD. Periodontal microbial ecology.

Periodontol 2000 2005: 38: 135–187.

207. Soga Y, Nishimura F, Ohyama H, Maeda H, Takashiba S,

Murayama Y. Tumor necrosis factor-alpha gene (TNF-

alpha) -1031 ⁄ -863-857 single-nucleotide polymorphisms

(SNPs) are associated with severe adult periodontitis in

Japanese. J Clin Periodontol 2003: 30: 524–531.

208. Sosroseno W, Herminajeng E. The role of macrophages in

the induction of murine immune response to Actinoba-

cillus actinomycetemcomitans. J Med Microbiol 2002: 51:

581–588.

209. Steffen MJ, Holt SC, Ebersole JL. Porphyromonas gingivalis

induction of mediator and cytokine secretion by human

gingival fibroblasts. Oral Microbiol Immunol 2000: 15:

172–180.

210. Stein SH, Green BE, Scarbecz M. Augmented transforming

growth factor-beta1 in gingival crevicular fluid of smokers

91

Host responses in periodontal health and disease

Page 16: Schenke in 2006

with chronic periodontitis. J Periodontol 2004: 75:

1619–1626.

211. Suda T, Takahashi N, Udagawa N, Jimi E, Gillespie MT,

Martin TJ. Modulation of osteoclast differentiation and

function by the new members of the tumor necrosis factor

receptor and ligand families. Endocr Rev 1999: 20:

345–357.

212. Sugita N, Kobayashi T, Ando Y, Yoshihara A, Yamamoto K,

van de Winkel JG, Miyazaki H, Yoshie H. Increased fre-

quency of FcgammaRIIIb-NA1 allele in periodontitis-

resistant subjects in an elderly Japanese population. J Dent

Res 2001: 80: 914–918.

213. Taichman NS, Tsai CC, Baehni PC, Stoller N, McArthur

WP. Interaction of inflammatory cells and oral micro-

organisms. IV. In vitro release of lysosomal constituents

from polymorphonuclear leukocytes exposed to supra-

gingival and subgingival bacterial plaque. Infect Immun

1977: 16: 1013–1023.

214. Tan KS, Song KP, Ong G. Cytolethal distending toxin of

Actinobacillus actinomycetemcomitans. Occurrence and

association with periodontal disease. J Periodontal Res

2002: 37: 268–272.

215. Tanaka S, Barbour SE, Best AM, Schenkein HA, Tew JG.

Prostaglandin E2-mediated regulation of immunoglobulin

G2 via interferon gamma. J Periodontol 2003: 74: 771–779.

216. Tangada SD, Califano JV, Nakashima K, Quinn SM, Zhang

JB, Gunsolley JC, Schenkein HA, Tew JG. The effect of

smoking on serum IgG2 reactive with Actinobacillus

actinomycetemcomitans in early-onset periodontitis

patients. J Periodontol 1997: 68: 842–850.

217. Tatakis D. Interleukin-1 and bone metabolism: a review.

J Periodontol 1993: 64: 416–431.

218. Taubman MA, Kawai T. Involvement of T-lymphocytes in

periodontal disease and in direct and indirect induction of

bone resorption. Crit Rev Oral Biol Med 2001: 12: 125–135.

219. Tempel TR, Snyderman R, Jordan HV, Mergenhagen SE.

Factors from saliva and oral bacteria, chemotactic for

polymorphonuclear leukocytes: Their possible role in

gingival inflammation. J Periodontol 1970: 41: 71–80.

220. Teng YT. Mixed periodontal Th1-Th2 cytokine profile in

Actinobacillus actinomycetemcomitans-specific osteopro-

tegerin ligand (or RANK-L)-mediated alveolar bone

destruction in vivo. Infect Immun 2002: 70: 5269–5273.

221. Teng YT. The role of acquired immunity and periodontal

disease progression. Crit Rev Oral Biol Med 2003: 14: 237–

252.

222. Tew J, Marshall D, Burmeister J, Ranney R. Relationship

between gingival crevicular fluid and serum antibody

titers in young adults with generalized and localized

periodontitis. Infect Immun 1985: 49: 487–493.

223. Theill LE, Boyle WJ, Penninger JM. RANK-L and RANK: T

cells, bone loss, and mammalian evolution. Annu Rev

Immunol 2002: 20: 795–823.

224. Tipton DA, Flynn JC, Stein SH, Dabbous M. Cyclooxy-

genase-2 inhibitors decrease interleukin-1beta-stimulated

prostaglandin E2 and IL-6 production by human gingival

fibroblasts. J Periodontol 2003: 74: 1754–1763.

225. Tonetti MS, Gerber L, Lang NP. Vascular adhesion mole-

cules and initial development of inflammation in clinically

healthy-human keratinized mucosa around teeth and

osseointegrated implants. J Periodontal Res 1994: 29: 386–

392.

226. Tonetti MS, Imboden MA, Lang NP. Neutrophil migration

into the gingival sulcus is associated with transepithelial

gradients of interleukin-8 and ICAM-1. J Periodontol 1998:

69: 1139–1147.

227. Toomes C, James J, Wood AJ, Wu CL, McCormick D, Lench

N, Hewitt C, Moynihan L, Roberts E, Woods CG, Markham

A, Wong M, Widmer R, Ghaffar KA, Pemberton M, Hussein

IR, Temtamy SA, Davies R, Read AP, Sloan P, Dixon MJ,

Thakker NS. Loss-of-function mutations in the cathepsin

C gene result in periodontal disease and palmoplantar

keratosis. Nat Genet 1999: 23: 421–424.

228. Trevilatto PC, Tramontina VA, Machado MA, Goncalves

RB, Sallum AW, Line SR. Clinical, genetic and microbio-

logical findings in a Brazilian family with aggressive

periodontitis. J Clin Periodontol 2002: 29: 233–239.

229. Tsai C, McArthur W, Baehni P, Hammond B, Taichman N.

Extraction and partial characterization of a leukotoxin

from a plaque-derived Gram-negative microorganism.

Infect Immun 1979: 25: 427–439.

230. Unsal BT, Ozcan G, Balos K, Gun H. The effects of perio-

dontal therapy on serum antibody levels to Actinobacillus

actinomycetemcomitans and Porphyromonas gingivalis

(part II). J Marmara Univ Dent Fac 1996: 2: 474–478.

231. van der Vaart H, Postma DS, Timens W, ten Hacken NH.

Acute effects of cigarette smoke on inflammation and

oxidative stress: a review. Thorax 2004: 59: 713–721.

232. Van Dyke TE, Schweinebraten M, Cianciola LJ, Offenba-

cher S, Genco RJ. Neutrophil chemotaxis in families with

localized juvenile periodontitis. J Periodontal Res 1985: 20:

503–514.

233. Van Dyke TE, Sheilesh D. Risk factors for periodontitis.

J Int Acad Periodontol 2005: 7: 3–7.

234. Van Dyke TE, Vaikuntam J. Neutrophil function and dys-

function in periodontal disease. Curr Opin Periodontol

1994: 1: 19–27.

235. Van Dyke TE, Warbington M, Gardner M, Offenbacher S.

Neutrophil surface protein markers as indicators of

defective chemotaxis in LJP. J Periodontol 1990: 61:

180–184.

236. Van Dyke TE, Zinney W, Winkel K, Taufiq A, Offenba-

cher S, Arnold RR. Neutrophil function in localized

juvenile periodontitis – phagocytosis, superoxide pro-

duction and specific granule release. J Periodontol 1986:

57: 703–708.

237. Vankeerberghen A, Nuytten H, Dierickx K, Quirynen M,

Cassiman JJ, Cuppens H. Differential induction of human

Beta-defensin expression by periodontal commensals and

pathogens in periodontal pocket epithelial cells. J Period-

ontol 2005: 76: 1293–1303.

238. Vincent JW, Falkler WA Jr, Cornett WC, Suzuki JB. Effect of

periodontal therapy on specific antibody responses to

suspected periodontopathogens. J Clin Periodontol 1987:

14: 412–417.

239. Waldrop TC, Anderson DC, Hallmon WW, Schmalstieg FC,

Jacobs RL. Periodontal manifestations of the heritable

Mac-1, LFA)1, deficiency syndrome. J Periodontol 1987:

58: 400–416.

240. Walker SJ, Van Dyke TE, Rich S, Kornman KS, di Giovine

FS, Hart TC. Genetic polymorphisms of the IL-1alpha and

IL-1beta genes in African-American LJP patients and an

African-American control population. J Periodontol 2000:

71: 723–728.

92

Schenkein

Page 17: Schenke in 2006

241. Weiss SJ. Tissue destruction by neutrophils. N Engl J Med

1989: 320: 365–376.

242. White RR, Montgomery EH. Exocytosis of polymorpho-

nuclear leukocyte lysosomal contents induced by dental

plaque. Infect Immun 1977: 16: 934–937.

243. Whitney C, Ant J, Moncla B, Johnson B, Page RC, Engel D.

Serum immunoglobulin G antibody to Porphyromonas

gingivalis in rapidly progressive periodontitis: titer, avid-

ity, and subclass distribution. Infect Immun 1992: 60:

2194–2200.

244. Williams BL, Ebersole JL, Spektor MD, Page RC. Assess-

ment of serum antibody patterns and analysis of subgin-

gival microflora of members of a family with a high

prevalence of early-onset periodontitis. Infect Immun

1985: 49: 742–750.

245. Williams R, Jeffcoat M, Howell T, Rolla A, Stubbs D, Teoh

K, Reddy M, Goldhaber P. Altering the progression of

human alveolar bone loss with the non-steroidal anti-

inflammatory drug flurbiprofen. J Periodontol 1989: 60:

485–490.

246. Wingrove JA, DiScipio RG, Chen Z, Potempa J, Travis J,

Hugli TE. Activation of complement components C3 and

C5 by a cysteine proteinase (gingipain-1) from Porphyro-

monas (Bacteroides) gingivalis. J Biol Chem 1992: 267:

18902–18907.

247. Yamazaki K, Tabeta K, Nakajima T, Ohsawa Y, Ueki K, Itoh

H, Yoshie H. Interleukin-10 gene promoter polymorphism

in Japanese patients with adult and early-onset perio-

dontitis. J Clin Periodontol 2001: 28: 828–832.

248. Yamazaki K, Yoshie H, Seymour GJ. T cell regulation of the

immune response to infection in periodontal diseases.

Histol Histopathol 2003: 18: 889–896.

249. Yonezawa H, Ishihara K, Okuda K. Arg-gingipain a DNA

vaccine induces protective immunity against infection by

Porphyromonas gingivalis in a murine model. Infect

Immun 2001: 69: 2858–2864.

250. Zhou Q, Desta T, Fenton M, Graves DT, Amar S. Cytokine

profiling of macrophages exposed to Porphyromonas

gingivalis, its lipopolysaccharide, or its FimA protein.

Infect Immun 2005: 73: 935–943.

251. Zhou J, Zou S, Zhao W, Zhao Y. Prostaglandin E2 level in

gingival crevicular fluid and its relation to the periodontal

pocket depth in patients with periodontitis. Chin Med Sci J

1994: 9: 52–55.

93

Host responses in periodontal health and disease