You are on page 1of 45

1353

Send Orders for Reprints to reprints@benthamscience.net

Current Topics in Medicinal Chemistry, 2020, 20, 1353-1397


REVIEW ARTICLE
ISSN: 1568-0266
eISSN: 1873-4294

Periodontal Pathogens and Neuropsychiatric Health Impact


Factor:
3.442

The international
journal for
in-depth
reviews on
Current Topics in
Medicinal
Chemistry

BENTHAM
SCIENCE

Abhishek Wadhawan1,2, Mark A. Reynolds3, Hina Makkar1, Alison J. Scott4, Eileen Potocki5,
Current Topics in Medicinal Chemistry

Andrew J. Hoisington6, Lisa A. Brenner7,8,9, Aline Dagdag1, Christopher A. Lowry7,9,8,10,11,


Yogesh Dwivedi12 and Teodor T. Postolache1,8,9,13,*

1
Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, USA;
2
Department of Psychiatry, Saint Elizabeths Hospital, Washington, D.C. 20032, USA; 3Department of Advanced Oral
Sciences & Therapeutics, University of Maryland School of Dentistry, Baltimore 21201, USA; 4Department of Microbial
Pathogenesis, University of Maryland School of Dentistry, Baltimore, USA; 5VA Maryland Healthcare System, Balti-
more VA Medical Center, Baltimore, USA; 6Air Force Institute of Technology, Wright-Patterson Air Force Base, USA;
7
Departments of Psychiatry, Neurology, and Physical Medicine & Rehabilitation, University of Colorado Anschutz
Medical Campus, Aurora, USA; 8Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC),
Veterans Integrated Service Network (VISN) 19, Aurora, USA; 9Military and Veteran Microbiome: Consortium for Re-
search and Education (MVM-CoRE), Aurora, USA; 10Department of Integrative Physiology, Center for Neuroscience
and Center for Microbial Exploration, University of Colorado Boulder, Boulder, USA; 11Rocky Mountain Mental Illness
Research Education and Clinical Center (MIRECC), Rocky Mountain Regional Veterans Affairs Medical Center
(RMRVAMC), Aurora, USA; 12Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Bir-
mingham, Alabama, USA; 13Mental Illness Research, Education and Clinical Center (MIRECC), Veterans Integrated
Service Network (VISN) 5, VA Capitol Health Care Network, Baltimore, USA

Abstract: Increasing evidence incriminates low-grade inflammation in cardiovascular, metabolic dis-


eases, and neuropsychiatric clinical conditions, all important causes of morbidity and mortality. One of
the upstream and modifiable precipitants and perpetrators of inflammation is chronic periodontitis, a
polymicrobial infection with Porphyromonas gingivalis (P. gingivalis) playing a central role in the dis-
ARTICLE HISTORY ease pathogenesis. We review the association between P. gingivalis and cardiovascular, metabolic, and
neuropsychiatric illness, and the molecular mechanisms potentially implicated in immune upregulation
Received: September 01, 2019 as well as downregulation induced by the pathogen. In addition to inflammation, translocation of the
Revised: December 04, 2019
Accepted: December 04, 2019 pathogens to the coronary and peripheral arteries, including brain vasculature, and gut and liver vascula-
DOI: ture has important pathophysiological consequences. Distant effects via translocation rely on virulence
10.2174/1568026620666200110161105
factors of P. gingivalis such as gingipains, on its synergistic interactions with other pathogens, and on its
capability to manipulate the immune system via several mechanisms, including its capacity to induce
production of immune-downregulating micro-RNAs. Possible targets for intervention and drug devel-
opment to manage distal consequences of infection with P. gingivalis are also reviewed.

Keywords: Chronic periodontitis, Porphyromonas gingivalis, Dementia, Cardiovascular disease, Metabolic syndrome, Suicidal
behavior, Mood disorders, micro-RNAs.

1. INTRODUCTION of periodontal diseases. Gingivitis is characterized by gingi-


val inflammation without the destruction of the supporting
1.1. Periodontal Diseases periodontal ligament and bone. Gingivitis can generally be
Periodontal diseases are infections of the supporting tis- resolved by effective oral hygiene [1-8], although other risk
sue(s) of the teeth. Oral bacterial biofilms are the primary factors may also lead to inflammation of the gums (reviewed
etiology of periodontal diseases, which are often chronic in in [9]), which can then advance to periodontitis, if not treated
nature. Gingivitis and periodontitis are the two major types appropriately [10]. In periodontitis, the inflammatory condi-
tion results in the breakdown of the periodontal ligament and
*Address correspondence to this author at 685 W. Baltimore Street, Suite # resorption of alveolar bone, resulting in loss of tooth support
930, Baltimore, MD 21201, USA; Tel: +1-(301) 996-9040; [3]. This destructive process, which results in apical migra-
E-mail: tpostola@som.umaryland.edu tion of the gingival epithelium, is characterized clinically by

1873-4294/20 $65.00+.00 © 2020 Bentham Science Publishers


1354 Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 Wadhawan et al.

formation of periodontal pockets. With advancing destruc- 2012 indicated that 46% of the adult population of the
tion of the periodontal tissues, an increase in tooth mobility United States (≥ 30 years) consisting of approximately 65
and masticatory impairment occurs. Tooth loss can eventu- million adults, had periodontitis, and 8.9% among those had
ally occur if periodontitis is not treated [11]. Based on a severe periodontitis [14]. Additionally, 44.7% of the adults
combination of clinical measures, i.e., clinical Attachment aged ≥ 30 years had periodontitis between the years 2011 to
Loss (CAL) and Periodontal Probing Depth (PPD), used to 2012 [14]. Prevalence of periodontitis has been reported to
diagnose and characterize periodontal disease, a frequently be higher in older adults (≥ 65 years old), with almost two-
used composite case definition of periodontitis was intro- thirds of that subpopulation being affected throughout the
duced by the American Academy of Periodontology (AAP) United States [15], likely due to lack of access to dental care,
and the Centers for Disease Control and Prevention (CDC). in addition to other factors [16].
“Severe periodontitis” is defined as “≥ 1 interproximal site
with PPD ≥ 5 mm and ≥ 2 interproximal sites with CAL ≥ 6 1.2. Associations of Periodontal Disease with Cardiovas-
mm (not on the same tooth).” “Moderate periodontitis” is cular, Metabolic and Neuropsychiatric Disorders
defined as “≥ 2 interproximal sites with PPD ≥ 5 mm (not on
same tooth), or ≥ 2 interproximal sites with CAL ≥ 4 mm Increasing evidence has demonstrated that periodontitis
(not on same tooth)” “Mild periodontitis” is defined as “≥ 2 is associated with conditions that pose a significant risk of
interproximal sites with CAL ≥ 3 mm, and one site with PPD morbidity and mortality (Fig. 1), such as cardiovascular dis-
≥ 5 mm or ≥ 2 interproximal sites with PPD ≥ 4 mm (not on eases (e.g., heart attack, coronary artery disease, and stroke)
the same tooth)” “No periodontitis” is defined as “No evi- and type 2 diabetes mellitus [17, 18]. Moreover, periodontal
dence of mild, moderate, or severe periodontitis” [12]. disease and systemic conditions, such as cardiovascular dis-
ease, often share common risk factors, including obesity,
Periodontitis is a major public health issue and its preva- smoking, and aging [19]. Accumulating data indicate that
lence is high in the adult population of the United States patients with other systemic diseases, such as Acquired Im-
[13]. Estimates of the prevalence of periodontitis across munodeficiency Syndrome (AIDS), Down’s syndrome, and
populations vary substantially because no sets of thresholds leukemia, are at increased risk of developing periodontitis
have been consistently used in epidemiological studies. De- [20, 21]. Periodontitis appears to be a risk indicator or factor
spite these shortcomings, based on the definitions provided for major chronic systemic conditions and health-related
by AAP and CDC, the data obtained from the National outcomes, including in pregnant women [22].
Health and Nutrition Examination Survey (NHANES) 2009-

Barrier Systemic
Periodontium breach Circulation spread Distal sites

Atherosclerosis Alzheimer’s
Viable
Ginglval disease
bacteria
Tooth epithelium
enamel
Periodontal Stroke
pocket

Obesity Pulmonary
Leukocytes infections
Bacterial
Bacteria products
LPS Rheumatoid Inflammatory
arthritis bowel
disease
Acute-
Alveolar phase Colon
Pro-inflammatory response
bone cancer
mediators

IL-17 TNF-a Diabetes


Vasculature
Junctional IL-1b IL-6 Pregnancy
epithelium Osteoclast complications

Fig. (1). Periodontitis and systemic diseases. Ulceration of the gingival epithelium initiates periodontitis, which is followed by influx of im-
mune cells and bacterial invasion that eventually leads to destruction of the supporting alveolar bone and inflammatory damage of the perio-
dontal tissues. These processes mediated by chronic inflammation allow leakage of host inflammatory factors, pathogenic oral bacteria, and
bacterial products into the systemic circulation, from where they are translocated to distal tissue sites and organs. When periodontally-
derived products reach the bloodstream, a multitude of systemic diseases are potentially adversely affected by them, either indirectly via
amplification of the systemic inflammatory response, or directly in situ. IL: interleukin; TNF-α: tumor necrosis factor-alpha; LPS: lipopoly-
saccharide. (Reproduced from: Konkel JE, O’Boyle C and Krishnan S (2019) Distal Consequences of Oral Inflammation. Front. Immunol.
10:1403. doi: 10.3389/fimmu.2019.01403.
Periodontal Pathogens and Neuropsychiatric Health Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 1355

Previous research has reported an association between tiple tooth loss as significant risk factors for dementia [82].
periodontitis and cardiovascular diseases, including ischemic However, when compared to healthy individuals, under-
heart disease [23], cerebrovascular disease [24], atheroscle- standably demented patients have been shown to exhibit sig-
rotic cardiovascular disease [25], and peripheral arterial dis- nificantly poorer measures of periodontal health [83]. Simi-
ease [26, 27]. Specifically, several systematic reviews, co- larly, a prospective cohort study from Japan reported an in-
hort, cross-sectional, and case-control studies have also dem- crease in the risk of all-cause dementia and AD with tooth
onstrated a statistically significant relationship between pe- loss [84]. Specifically, an increase in multivariable-adjusted
riodontitis and myocardial infarction [28-41]; however, this hazard ratio of all-cause dementia was reported with decreas-
association has not been observed in all studies [42-48]. ing number of remaining teeth (p for trend = .04), after ad-
Many candidate mechanisms have been postulated to explain justing for potential confounders [84]. Another prospective
the positive association between periodontal and cardiovas- cohort study also confirmed an increased risk of developing
cular diseases. An individual may be at a higher risk for de- dementia in patients with complete tooth loss [hazard ratio
veloping both periodontal disease and atherosclerosis in the (HR) = 1.10, 95% CI = 1.04-1.16] and those not undergoing
presence of an underlying inflammatory response trait [49]. periodontal treatment [HR = 1.14, 95% CI = 1.04-1.24],
Once periodontal disease occurs, it may serve to initiate and when compared with individuals who received dental pro-
exacerbate atherogenesis and thromboembolic events by phylaxis, following adjustment for confounders [85]. Addi-
providing a biological burden of inflammatory molecular tionally, as compared to AD patients without periodontitis,
mediators [thromboxane A2, interleukin (IL)-1 beta, prosta- AD patients with periodontitis have been reported to have a
glandin E2 and tumor necrosis factor-alpha (TNF-α)] and sixfold increase in cognitive decline over a period of 6-
immune triggers such as endotoxin [49]. Additionally, the months in a prospective observational study [86].
presence of bacteria in the bloodstream has been associated
Patients with AD have been reported to have altered cy-
with elevation of biomarkers for inflammation [50, 51]. Oral
tokine profiles, microglial activation, inflammasome activa-
bacteria can pass into the bloodstream due to disruptions in tion, and complement activation, features that are consistent
the integrity of gingival sulcular/pocket epithelium (barrier),
with infectious pathologies [87, 88]. Interest in identifying a
which is characteristic of periodontitis [52, 53]. Periodontitis
potential infectious cause of AD has grown with the recent
has been reported to be associated with ischemic stroke as
characterization of amyloid-β (Aβ) as an antimicrobial pep-
well. Based on five case-control studies (pooled Relative
tide [89-91]. Porphyromas gingivalis (P. gingivalis) infec-
Risk [RR] = 3.04, 95% CI: 1.10–8.43) [54-58] and three co-
tion, as well as periodontitis, have been reported to be risk
hort studies (pooled RR = 2.52, 95% CI: 1.77–3.58) [49, 59, factors for the development of AD, Aβ plaques, and demen-
60], a statistically significant association was reported in a
tia [92-96]. Data from a study on Apoe−/− mice indicated
meta-analytical study [61].
that brain infection and activation of the complement path-
A relationship between periodontitis and metabolic dis- way occurred only upon oral infection with P. gingivalis,
eases has also been reported. As shown by several studies, which was not seen upon infection of these mice with other
chronic periodontitis has been associated with obesity [62- two oral bacteria used in this study [97]. As compared to
64], and this relationship even exists in adolescents [65]. control mice, infection of the oral cavity with P. gingivalis
Cross-sectional evidence exists for a negative correlation resulted in alveolar bone loss, impaired cognitive function,
between periodontitis and good physical fitness after adjust- and increased deposition of AD-like plaques in transgenic
ing for a number of confounders, including higher fasting mice overexpressing mutated hAPP-J20 [98]. Detection of P.
plasma glucose, old age, and smoking habits [66]. Prospec- gingivalis lipopolysaccharide (LPS) in the postmortem hu-
tive studies also indicate an increased risk of periodontitis man brain tissue of AD patients [99] supports the hypothesis
being associated with a sedentary lifestyle, as well as a linear that infection of the brain by P. gingivalis may affect the
and inverse association between periodontitis and sustained pathophysiology of AD [100]. More recently, Dominy et al.
physical activity [67-69]. When compared with non- identified P. gingivalis DNA in the cerebrospinal fluid (CSF)
diabetics, diabetic patients have been reported to have higher of living subjects with probable AD and the postmortem
plaque deposits and poorer oral hygiene [70]. Bad periodon- brain tissue of AD patients, but not in patients with other
tal health has been reported in individuals with metabolic neurodegenerative diseases. These results suggest a certain
syndrome as compared to healthy controls [71-72]. Moreo- pathophysiological specificity and that DNA of P. gingivalis
ver, clinical measures of periodontitis have been reported to could be used as a potential diagnostic marker in AD [101].
have a relationship with various components of metabolic Moreover, evidence from a recent animal study points to-
syndrome in a dose-dependent manner [73]. Specifically, an wards neuroinflammatory mechanisms potentially mediating
association has been exhibited between periodontitis and the associations between ischemic stroke and periodontitis
components of metabolic syndrome, including hypertension, [102], which perhaps, could also have a role in the patho-
obesity, dyslipidemia, and hyperglycemia [74-79]. physiology of vascular dementia.
Periodontal disease has been reported to be positively as- Psychiatric patients have also been shown to have an ele-
sociated with dementia as well [80]; however, the data are vated risk of periodontal disease. In a study of institutional-
inconsistent. Although one systematic review reported a ized psychiatric patients, the majority had periodontal bleed-
positive association between periodontal disease and Alz- ing on probing and periodontal pockets; moreover, older
heimer’s disease (AD) [Odds ratio (OR): 1.69, 95% confi- patients were more likely to be edentulous (i.e., lacking
dence intervals (CI): 1.21-2.35] [81], another review failed to teeth) [103]. Moderately deep pockets have been reported in
provide conclusive evidence for either periodontitis or mul- about 59% of psychiatric outpatients [104] and 15-28% of
1356 Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 Wadhawan et al.

psychiatric inpatients [103, 105, 106]. A meta-analysis re- tal disease with cardiovascular, metabolic and psychiatric
ported that patients with severe mental illness had a signifi- disorders, with inflammation being proposed as the mediator
cantly more likelihood of losing all their teeth, as compared for these associations [9]. Moreover, comorbidities exist
to the general community (OR: 2.8, 95% CI: 1.7-4.6) [107]. between metabolic and psychiatric disorders/symptoms
More recently, patients with schizophrenia have been re- [130] that may be explained by genes of the prolactin-
ported to have a higher evidence of periodontal disease, pathway and the shared inherited predisposition to psycho-
higher unmet dental prosthetic needs, and greater dental car- neuro-endocrine dysfunction [131].
ies [108]. Moreover, the severity of schizophrenia and ex-
trapyramidal side-effects were associated with poor oral 2. PERIODONTAL PATHOGENS
health [108]. Likewise, in a separate study, poorer oral hy-
giene and periodontal health, as evidenced by plaque index, The oral cavity is continuously bathed in saliva and char-
gingival index, and probing pocket depths, were reported in acterized by a pH close to neutrality and in a narrow range of
patients who had a diagnosis of schizophrenia for a duration temperature (34 to 36 ⁰C) creating a distinct habitat for a
of ≥ 11 years (p < 0.001) [109]. In a case-control study, bi- variety of microorganisms [132]. In addition to bacteria, the
polar disorder was associated with an increased risk for pe- oral cavity harbors other types of microflora as well, includ-
riodontitis (OR = 2.13; 95% CI: 1.39-3.27), and periodontitis ing protozoa, fungi, viruses, and mycoplasma [133]. While
was also strongly associated with the depressive phase of there are over 700 species of bacteria that can colonize the
bipolar disorder (OR = 28.94; 95% CI: 4.44-177.27; oral cavity [134], only a relatively small number of bacterial
p<0.001) and the total bacterial load (OR = 1.91; 95% CI: species are closely associated with periodontitis [135]. Bac-
1.0-1.99; p < 0.001) [110]. Similar associations were found teria in the oral cavity are classified broadly in two ways: a)
in a separate study, which reported higher caries prevalence, according to Gram-staining-Gram-positive and Gram-
poorer periodontal health, and poorer oral hygiene in bipolar negative; and b) according to their oxygen requirements-
patients as compared to healthy controls [111]. A recent aerobic or anaerobic. The anaerobic bacteria are further sub-
cross-sectional study indicated that depression was signifi- divided into facultative and obligate anaerobic subsets (Ta-
cantly associated with alcohol use (OR = 1.47; 95% CI: 1.38, ble 1). Nevertheless, the oral cavity has a stable community
1.57) and periodontal disease during pregnancy (OR = 1.76; of microbes that coexist symbiotically, despite the presence
95% CI: 1.73, 1.80), and that depression during pregnancy of such diversity in the oral microbiome. Hence, periodontal
was positively associated with having a preterm delivery disease may result from microbial dysbiosis, leading to the
(OR = 1.18; 95% CI: 1.15, 1.21 and having low birth weight emergence of pathogenic microorganisms. In fact, a shift in
(OR = 1.21; 95% CI: 1.17, 1.21], with depression possibly the predominant species of microbes in the gingival sulcus to
serving as a mediator in the association between adverse Gram-negative, proteolytic, anaerobic and chemoorganotro-
birth outcomes and periodontal disease [112]. A prospective phic organisms from an initial preponderance of Gram-
cohort study also revealed a positive association between positive, fermentative and facultative microorganisms has
depressive symptoms and moderate/severe periodontitis been associated with damage to the periodontal tissues [136].
[RR: 1.18], as well as higher risk of periodontitis (RR: 1.19)
[113]. Moreover, in the Old Order Amish, tooth loss has Seminal work by Socransky and colleagues utilized
been associated with cardinal symptoms of depression, such whole genomic DNA probes and checkerboard DNA–DNA
as anhedonia and dysphoria/hopelessness. These effects were hybridization to characterize periodontal microbial commu-
reported to be mediated by inflammation [114]. nities based on a color-coded system that reflected associated
disease severity, community ordination, and cluster analysis
The associations between psychiatric disorders and [137]. These investigators reported that a total of five major
periodontal disease and its manifestations have been related complexes were observed consistently, using their analytical
to poor compliance of dental treatments, negative outlook methods, which were initially color-coded as red, orange,
towards the physician, dental phobia, poor oral hygiene, green, yellow, and purple complexes [137]. Amongst these
trouble accessing health care services, lack of self-care, poor groups, the foremost complex was the “red complex” that
nutrition, and medication side-effects (dry mouth) in patients consisted of three species, i.e., Tannerella forsythia (T. for-
with schizophrenia, depression, and anxiety disorders [115- sythia), Treponema denticola (T. denticola) and P. gin-
124]. Negative thought patterns may be precipitated or wors- givalis, and their detection was associated with diseased
ened in patients with periodontitis due to the social stigma sites, as well as with each other [137]. Whereas orange com-
associated with halitosis, as well as their altered appearance plex [consisting of Parvimonas micra (P. micra), Prevotella
resulting from pathologic tooth migration, tooth loss, or both intermedia (P. intermedia), Fusobacterium nucleatum (F.
[125, 126]. In contrast to bipolar disorder patients experienc- nucleatum), Eubacterium nodatum (E. nodatum), Prevotella
ing depressive and mixed states when self-neglect may be nigrescens (P. nigrescens), and Campylobacter rectus (C.
the most distinctive feature requiring motivating and educa- rectus)] is also correlated with diseased periodontal tissues,
tional efforts, those patients experiencing mania may have yellow, green, and purple complexes have been associated
mucosal or gingival lacerations or dental abrasions resulting
with healthy periodontal status [137, 138].
from overenthusiastic flossing and tooth brushing [118], re-
quiring supervision of dental hygiene. In subjects with psy- Additionally, these complexes likely reflect early to late
chiatric illnesses, co-morbid substance use disorders (psy- colonizers in developing dental biofilms as part of microbial
chostimulants, alcohol, tobacco) may also increase the risk succession [139]. Members of the purple (e.g., Veillonella
of oral health problems [127-129]. An extensive review by parvula and Actinomyces odontolyticus), yellow (some
Makkar et al. lists the bidirectional associations of periodon- Streptococcus species, e.g., Streptococcus mitis) and green
Periodontal Pathogens and Neuropsychiatric Health Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 1357

Table 1. More frequently isolated microorganisms from human oral cavity (Reproduced from How KY, Song KP, and Chan KG
(2016) Porphyromonas gingivalis: An Overview of Periodontopathic Pathogen below the Gum Line. Front. Microbiol. 7:53.
doi: 10.3389/fmicb.2016.00053).

Microbial group Microbial genus/species

Streptococcus (S. gordonii, S. mitis, S. auralis, S. salivarius)

Staphylococcus (S. aureus, S. epidermidis)

Enterococcus (E. faecalis)

Lactobacillus (L. casei, L. fermentum)


Aerobic or facultative
Corynebacterium (C. matruchotii)

Gram-positive Actinomyces (A. naeslundii, A. israelli, A. viscosus)

Arachnia (A. propionica)

Rothia (R. dentocariosa)

Bacillus (B. cereus)

Obligate anaerobes Propionibacterium (P. acnes)

Peptostreptococcus (P. micros, P. anaerobius)

Campylobacter (C. rectus, C. concisus, C. gracilis)


Aerobic or facultative
Actinobacillus (A. actinomycetemcomitans)

Gram-negative Fusobacterium (F. nucleatum)

Obligate anaerobes Porphyromonas (P. gingivalis)

Prevotella (P. melaninogenica, P. oralis, P. intermedia)

[e.g., Aggregatibacter actinomycetemcomitans (A. actinomy- 85% of the sites with periodontal destruction [141, 142].
cetemcomitans), Eikenella corrodens, Capnocytophaga spu- Also, in healthy sites, P. gingivalis, is found rarely or in low
tigena, Capnocytophaga ochracea, and Campylobacter con- counts. It forms black-pigmented colonies on blood agar
cisus] complexes constitute most of the early colonizing mi- plates after 6 to 10 days due to heme accumulation, and iron
crobial species. Generally, after establishment of the early is an absolute requirement for the growth of this bacterium
colonizers, the orange complex bacteria (i.e., F. nucleatum, [143]. Within the human oral cavity, the major habitat of P.
P. micra, P. intermedia, P. nigrescens, E. nodatum, and C. gingivalis is the subgingival pocket where availability of
rectus) appear, which are putative periodontal pathogens. All sugar is low. Hence, the survival of this bacterium in the
three members of the red complex (i.e., P. gingivalis, T. for- deep periodontal pocket is dependent upon the fermentation
sythia and T. denticola) are also on the list of putative perio- of amino acids for energy production [144]. As P. gingivalis
dontal pathogens and are considered as the climax commu- is an obligate anaerobe, it often attaches to the primary colo-
nity [137]. Follow-up studies placed early colonizers, includ- nizers of dental plaque, including P. intermedia and Strepto-
ing Actinomyces species, in a sixth group, i.e., the blue com- coccus gordonii, and therefore, it is referred to as the secon-
plex [138, 140]. Notably, putative periodontal pathogens are dary colonizer of dental plaque. In advanced periodontal
present in all groups of colonizers, i.e., early to late [137, lesions, it has been demonstrated that P. gingivalis associates
138, 140]. These bacteria could contribute significantly to with T. denticola and T. forsythia to establish the red com-
the initiation and progression of periodontal infections under plex [145]. Evidence from immunological studies also indi-
appropriate conditions. Despite the presence of other oral cates that patients diagnosed with adult periodontitis have
pathogens of interest, we have narrowed our discussion in higher serum levels of antibodies to P. gingivalis [146-149].
this review to P. gingivalis, because of the recent emergence
Based on their capacity to develop abscesses in experi-
of data that has implicated this bacterium in AD [97-101]
mental animal models, strains of P. gingivalis have been
and due to the ongoing development of drugs against this
divided into two types: invasive and non-invasive. Under
pathogen, as described in later parts of this review.
both in vivo and in vitro conditions, the invasive strain of P.
2.1. Porphyromonas gingivalis gingivalis has been reported to demonstrate greater patho-
genicity than the non-invasive strain [150, 151]. The process
The bacterial species that is highly implicated with both
of periodontal tissue destruction is complex, and its induc-
chronic and aggressive periodontitis is the Gram-negative,
tion and progression depend on the release of bacterial sub-
asaccharolytic, P. gingivalis, which can be found in almost
1358 Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 Wadhawan et al.

stances, host inflammatory response, and plaque accumula- proteolysis [158]. While gingipain K has one sub-type, i.e.,
tion. Constituents or metabolites of a pathogen that cause Kgp; gingipain R has two sub-types, RgpA and RgpB. The
damage to the host and are essential in various stages of the extracellular matrix components, such as collagen, immuno-
pathogen’s life cycle are referred to as virulence factors. P. globulins, complement factors, cytokines and the integrin–
gingivalis possesses virulence factors that can penetrate the fibronectin-binding are degraded by gingipain R [156, 159].
gingivae and cause tissue destruction directly or indirectly by Additionally, maturation and processing of the major fim-
induction of inflammation; these virulence factors include briae (FimA) are also carried out by this enzyme [160].
LPS, fimbriae, capsule, gingipains or cysteine proteinases, Available evidence suggests a direct role of P. gingivalis
outer membrane proteins, outer membrane vesicles, and proteases in the colonization and destruction of supporting
haemagglutinins [152-155]. Changes in the external envi- periodontal tissues [161, 162]. Furthermore, high resistance
ronment of the periodontopathogen often determine the ex- to host defense mechanisms is conferred to the microorgan-
pression of virulence factors. If these virulence factors are isms by these enzymes. P. gingivalis proteases also disrupt
activated, induction of host immune responses via cytokine inflammatory response of the host, activate the matrix metal-
production, inhibition of host protective mechanisms, bone loproteinases in the host, inactivate inhibitors of plasma pro-
resorption, as well as rapid and significant destruction of teinases, and cleave the receptors on the surface of host cells
periodontal tissues can occur in hosts. A paucity of informa- [159, 163]. Within biofilms consisting of mixed-species in-
tion is available on the mechanisms and the expression of cluding P. gingivalis, these enzymes mediate detachment
these virulence determinants during various stages of perio- and reduction of A. actinomycetemcomitans aggregation
dontal disease. Below, we summarize the virulence factors [164], as well as reduction in the numbers of T. forsythia
(Fig. 2) possessed by P. gingivalis: [165]. Furthermore, gingipains contribute to inhibition of
blood coagulation and, consequently, increased bleeding by
degrading fibrinogen and host heme proteins, hence promot-
ing bacterial growth by enhancing the availability of hemin
[166]. Since erythrocytes are found in abundance in perio-
Capsule
dontal pockets, it is not surprising to find a high proliferation
of P. gingivalis within them. Gingipains also have a role in
OM and Gingipains
LPS
the breakdown of complement factors (e.g., C4 and C3), T-
cell receptors (e.g., CD8 and CD4), and antibacterial pep-
tides (e.g., neutrophil-derived α -defensins) [165, 167]. P.
Fimbriae
gingivalis and its gingipains have been reported to proac-
tively manipulate host molecules and prevent bacterial clear-
ance by interfering with the crosstalk between toll-like re-
ceptor (TLR) and C5a receptor signaling [168]. Evidence
from in vitro studies indicates that inflammatory mediators
from various host cells are also regulated by gingipains, such
as soluble triggering receptor expressed on myeloid cells-1
Fig. (2). Virulence factors of Porphyromonas gingivalis. [169], protease-activated receptor-2 [170], and various inter-
leukins (IL-1α, IL-1β, IL-18) [171]. However, it remains
unclear whether periodontitis progresses due to the concur-
2.1.1. Gingipains rent effects of gingipains on the abovementioned inflamma-
tory mediators. Toxicity of P. gingivalis has been reported to
A virulence factor that allows most P. gingivalis strains be mediated by gingipains within the epithelial cells, endo-
to colonize the oral cavity is their ability to secrete toxic me- thelial cells and fibroblasts [172-174]. Furthermore, gingi-
tabolites together with numerous lipolytic, proteolytic and pains are implicated as narrow-spectrum virulence targets
hydrolytic enzymes. Some of the enzymes are transported because eradication of P. gingivalis rarely occurs on treat-
from the outer membrane into outer membrane vesicles dur- ment with broad-spectrum antibiotics and may result in resis-
ing bacterial growth, while others are found within the perip- tance [175-179]. P. gingivalis virulence can be reduced by
lasmic space. Usually, these bacterial enzymes are present using short peptide analogs that block the proteolytic activity
adjacent to the host cells. Of these enzymes, the progression of gingipains [180]. In a recent study, it was reported that as
of periodontal disease mainly involves proteolytic enzymes, compared to the non-AD control individuals, AD brains had
including peptidases, as well as caseinolytic-, trysin- and significantly higher gingipain immunoreactivity [101]. These
thiol-proteinases [156, 157]. Generally, P. gingivalis pro- researchers performed further testing in vivo of the potent,
duces proteases that belong to two distinct families: a) serine selective, brain-penetrant, small-molecule gingipain inhibi-
proteinase family, and b) cysteine proteinase family (also tors developed by them and reported that a disease-
known as “trypsin-like” enzyme) [144]. Cleavage of poly- modifying effect in AD could be achieved by small-molecule
peptides at the C-terminal after the lysine or arginine residue inhibition of gingipains [101].
is carried out by the “trypsin-like” enzymes, which are usu-
2.1.2. Lipopolysaccharide
ally referred to as gingipains. Depending on whether gingi-
pains cleave after the lysine or arginine residue, they are LPS composes the outer membrane of Gram-negative
called gingipain K or R, respectively. Of the total extracellu- bacteria and is an important factor for innate immune recog-
lar proteolytic activity that occurs at the sites infected by P. nition by TLR 4 [181, 182]. Three structurally distinct re-
gingivalis, these enzymes responsible for about 85% of the gions characterize LPS – O-antigen polysaccharide, core
Periodontal Pathogens and Neuropsychiatric Health Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 1359

oligosaccharide, and lipid A – with structural diversity de- 9 was seen in exposed mice versus mice that received saline
creasing proximally [183]. LPS serves as an essential immu- [209]. Additionally, LPS Pg also stimulates the expression of
nomodulatory tool for P. gingivalis, with modifications to thrombospondin-1 [210] and plasminogen activator inhibitor
the canonical structure resulting in altered innate recognition type I (PAI-1) [211], both of which also play a role in the
and adaptive functions [184]. Lipid A, commonly known as cardiovascular disease pathophysiology [212, 213].
endotoxin, is the predominantly hydrophobic moiety of LPS
2.1.3. Outer Membrane Proteins
that forms the outer leaflet of the asymmetric outer mem-
brane bilayer [185]. TLR4 recognition of lipid A is based on P. gingivalis consists of two cell membranes, the inner
the positioning, length, and quantity of acyl chains along membrane (IM) and the outer membrane (OM) as part of its
with the presence and modification of terminal phosphates cell envelope, which are separated by the periplasm that con-
on the diglucosamine framework [186, 187]. The most po- tains an additional layer of peptidoglycan. Within the asym-
tent TLR4 agonists are the hexaacylated bis-phosphorylated metrical bilayer of OM, the outer and inner leaflets consist of
enteric lipid A structures, but subtle modifications to this LPS and phospholipids, respectively. The cell membrane
structure can lead to a range of potency from partial or weak offers protection and acts as a selective barrier permitting
agonists to full antagonists [188]. P gingivalis LPS (LPSPg), exchange of several materials via the porin proteins in the
specifically lipid A, is unique in structure and activity, and OM [214]. There are two categories of OM proteins, one that
both are important considerations to understand innate in- consists of integral proteins containing membrane-spanning
flammation stemming from infection [184]. The heteroge- regions, and the other that consists of proteins anchored to
nous lipid A structures of LPSPg range from tetra- and penta- the OM by an N-terminal lipid tail [215]. The proteins in the
acylated forms with mono- and bis-phosphorylation; how- OM are considered to mediate the interaction among perio-
ever, none elicits robust TLR4 signaling [189, 190]. Struc- dontal microflora, which in turn, is postulated to be associ-
tures of the tetra-acylated forms are condition dependent, ated with the formation and maintenance of periodontal
including non-phopshorylated (TLR4 silent) and monophos- biofilms [215]. Additionally, data suggest that when acti-
phorylated forms (TLR4 antagonist) [191, 192]. Whereas vated with P. gingivalis OM proteins, T-helper cells of ag-
most Gram-negative organisms found in the body (commen- gressive periodontitis patients, as compared with T-helper
sals and pathogens) have lipid A structures with acyl chain cells of healthy controls, produced higher levels of proin-
lengths of even carbon numbers, lipid A of P. gingivalis has flammatory cytokines such as IL-1β and IL-6 [216]. A major
branched odd carbon length primary acyl chains that drasti- OM protein (75-kDa) from P. gingivalis has been reported to
cally reduce recognition by the MD2/TLR4 signaling com- stimulate production of IL-1 in mouse peritoneal macro-
plex [184]. Importantly, TLR4 is the only recognized recep- phages, as well as activation of polyclonal B-cells [217].
tor for LPSPg, a point that has been clarified after numerous Another OM protein (40-kDa) from P. gingivalis has been
early reports of TLR2 activity; this was later attributed to reported to act as an aggregation factor and the aggregation
lipoprotein contamination in LPSPg biological extracts [184, activity of P. gingivalis cells for A. viscosus (an early colo-
193-198]. Exclusivity of TLR4 activation by LPSPg, though nizer of tooth surfaces) is inhibited by the antibody against
it is a weak agonist, has been definitively demonstrated using this protein [218]. Yet another P. gingivalis OM protein
both ultra-purified biological and synthetic lipid A prepara- named LptO (PG0027) is considered to be essential to pro-
tions, thoroughly discussed elsewhere [194, 199-202]. Struc- vide attachment to host cells, as it is also required for the O-
tural modification of P. gingivalis lipid A is influenced by deacylation of LPS of this bacterium that further confers this
environmental (niche) and host factors, notably hemin (oxi- property to P. gingivalis [219]. Moreover, production of ac-
dized heme) [203, 204]. Hemin levels initiate a lipid A modi- tive gingipains involves the role of another OM protein re-
fication cascade that switches between weakly agonistic to ferred to as PG534 [220]. In fact, upon comparison with the
antagonistic structures, directly manipulating the host innate wild-type strain of P. gingivalis, the strain with defective
immune response in turn [203, 205]. PG534 protein has been reported to have reduced activities
of gingipains R (RgpA and RgpB) and gingipain K (Kgp)
P. gingivalis can subvert detection by the innate host sur-
[220].
veillance via LPS hypoacylation and phosphate cleavage,
which possibly interferes with the distribution of leukocytes 2.1.4. Fimbriae
in the area surrounding the bacterial colonies. Additionally,
Bacterial cells can have surface appendages extending
activation of neutrophils, eosinophils, and basophils is af-
from their outer membrane, referred to as fimbriae, which
fected due to a reduction in the ability of gingival epithelial
are characteristically thin and proteinaceous. Most P. gin-
cells to secrete IL-8 (a chemokine) after being stimulated
givalis strains harbor these structures, which are about 3-25
with LPSPg, a phenomenon that is referred to as chemokine
μm long [221, 222]. On the cell surface of P. gingivalis, two
paralysis [206, 207]. Thus, P. gingivalis may become resis-
tant to damage mediated by the oxidative burst from the po- different types of fimbriae are expressed: a) major or long
fimbriae (consists of a subunit protein named FimA or fim-
lymorphonuclear neutrophils. Within the periodontal liga-
brillin that is encoded by the fimA gene), and b) Mfa1 or
ment stem cells that participate in periodontal tissue regen-
minor or short fimbriae (consists of a subunit Mfa protein
eration, mineralization and osteoblastic differentiation are
that is encoded by the mfa1 gene). Despite differing in their
also inhibited by LPSPg [208]. It has been demonstrated that
amino acid composition and being antigenically distinct,
in mice, cardiac dysfunction occurs after being exposed to a
low dose of LPSPg, which was mediated by the induction of these two types of fimbriae play a role in the development of
inflammatory reactions associated with periodontal disease
an inflammatory response in the left ventricle [209]. Fur-
[223]. For instance, macrophage-mediated production of IL-
thermore, a significant increase in matrix metalloproteinase-
1α, IL-1β, IL-6 and TNF-α has been reported to be induced
1360 Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 Wadhawan et al.

by minor fimbriae [224]. Additionally, fimbriae play an es- tion amongst these microbes is required to cause the destruc-
sential role in binding to and subsequent invasion of P. gin- tive events involved in periodontal disease progression,
givalis into the host cells [225], as well as in adherence to which cannot be precipitated by only one of these microbial
several molecules and oral substrates, including the oral epi- species in isolation [132, 135, 230].
thelium and commensal bacteria (streptococci and Actinomy- Hajishengallis and Lamont have proposed “a polymicro-
ces viscosus) and the extracellular matrix proteins [226,
bial synergy and dysbiosis (PSD) model” of periodontal dis-
227]. Specifically, due to their ability to attach to cellular
ease [231]. According to this model, even though diverse
α5β1-integrin, type II fimbriae lead to rearrangement of the
microbiota colonize the gingival crevicular space, hetero-
actin cytoskeleton and facilitate the phagocytosis of bacteria
typic communities are assembled only between compatible
by the host dendritic cells and phagocytes [223]. The host
microorganisms [231]. Despite the fact that these heterotypic
cellular function is eventually impaired by the virulence fac- communities can produce toxic products such as proteases
tors of these intracellular bacteria [228]. Moreover, progres-
and promote inflammation, the host controls their over-
sion of atherosclerosis may involve P. gingivalis fimbriae.
growth and overt pathogenicity. Moreover, there can be a
Reduction in regulatory T cells (Tregs) has been reported in
person-to-person and site-to-site variation of the microbial
atherosclerotic patients with P. gingivalis infection as com-
constituents of these communities over time. The virulence
pared with healthy, non-atherosclerotic patients, and P. gin-
of the entire community can be elevated after colonization by
givalis FimA genotype II has been reported to be the domi- keystone pathogens (e.g., P. gingivalis) and their interactive
nant type associated with reduced Treg population in patients
communication with other pathogens (e.g., Streptococcus
with atherosclerosis [229].
mitis). Furthermore, tissue homeostasis is disrupted, and de-
struction of periodontal tissues occurs upon impairment of
2.2. Interactions between Periodontal Pathogens the host immune surveillance and an increase in the number
Whether periodontal tissue breakdown would occur or of dysbiotic communities [231].
not is determined by the balance between host protection and In agreement with this model, evidence from animal
bacterial aggression, factors that are further influenced by the studies suggests that tissue damage during periodontitis can
complex interactions between bacterial flora and the host be significantly enhanced by co-infection with P. gingivalis,
defense mechanisms [152]. Several experimental studies T. denticola, and F. nucleatum, when compared to infection
have indicated that Gram-negative rods, including P. gin- with these species in isolation [232-234] (Fig. 3). More re-
givalis, F. nucleatum, P. intermedia, P. nigrescens, A. cently, Deng et al. [235] reported a synergistic interaction
actinomycetemcomitans and T. forsythia (previously desig- between these three microbes that enhances their pathogenic-
nated Bacteroides forsythus), are the primary etiological ity, a finding that was based on their in vivo transcriptional
agents of periodontal diseases. However, a concerted interac-

P. gingivalis
Pr
ote min
a
oly o a
ity
bil

sis cid
Mo

fre
s
e

T. denticola F. nucleatum
Hemolysis free iron
from heme

Free amino acids


Fig. (3). Synergistic pathogenicity of Treponema denticola (T. denticola), Porphyromas gingivalis (P. gingivalis), Fusobacterium nucleatum
(F. nucleatum) in chronic periodontitis, as demonstrated by an interaction model. Free amino acids, which could be contributed by other
members of the periodontal microbiota, are indicated by the dashed line. (Adapted from Deng Z-L, Sztajer H, Jarek M, Bhuju S and Wagner-
Döbler I (2018) Worlds Apart – Transcriptome Profiles of Key Oral Microbes in the Periodontal Pocket Compared to Single Laboratory
Culture Reflect Synergistic Interactions. Front. Microbiol. 9:124. doi: 10.3389/fmicb.2018.00124).
Periodontal Pathogens and Neuropsychiatric Health Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 1361

profiles. Two-way synergistic interactions, where each spe- in regulating the production of proinflammatory cytokines
cies gives and takes, were proposed in this study via a sche- [250, 251].
matic interaction model [235]. Enhanced adhesion in the
To understand periodontal disease and its association
periodontal niche was indicated by the upregulation of many
with coronary artery disease, P. gingivalis serves as a model
surface proteins in vivo [236], and the authors reported that pathogen and has been researched more extensively at the
mobility of P. gingivalis was enhanced by attaching to T.
molecular level. It expresses cysteine proteases (Arg-specific
denticola via upregulation of hemagglutinin and fimbriae
gingipains), which release biologically active C5a by cleav-
[235]. Since P. gingivalis has strong proteolytic activities, a
ing C5. This process occurs independent of the complement
bacterial species like F. nucleatum that exhibits no or weak
cascade activation and an interaction between the TLR2 and
intrinsic proteolytic activity, would benefit from coexisting
C5a receptor (C5aR, also known as CD88) can be induced
with P. gingivalis [237]. Deng and colleagues also reported by P. gingivalis [228].
that F. nucleatum up-regulates peptide transporters and uses
the proteolytic activity of other species to its advantage, par- IL-6 has been identified as an independent risk factor for
ticularly of P. gingivalis and T. denticola that have an in- rupture of atherosclerotic plaque and coronary artery disease
creased cysteine protease activity [235]. Shared labor be- [252]. This cytokine also has a well-documented role in
tween periodontal pathogens has been observed in terms of chronic periodontitis. IL-6 mediates periodontal tissue de-
iron as well, which is an important pathogenic element struction and is elevated in periodontal tissues after being
[238]. Whereas T. denticola has been reported to have down- stimulated by other cytokines (TNF-α and IL-1β) or by LPS.
regulation in hemolysins, F. nucleatum has been shown to A recent study reported that as compared to healthy subjects,
engage in lysing erythrocytes by upregulating hemolysins a significantly higher level of IL-6 was observed in patients
[235]. Thus, the hemolytic activity of F. nucleatum profits P. with chronic periodontitis [253], a finding that was consis-
gingivalis and T. denticola, both of which can uptake free tent with a previous study [254].
iron or bind heme to acquire iron for themselves [235]. Another recognized risk factor for acute coronary syn-
drome is increased white blood cell count [255, 256], which
3. IMMUNE RESPONSE TO PERIDONTAL PATHO- is increased in infections, including chronic periodontitis
GENS [253]. However, a shared risk factor for chronic periodontitis
Various innate immune mechanisms operate via some as well as acute coronary syndrome is smoking, which can
acute-phase reactants in periodontitis (mainly a chronic con- also increase the white blood cell count [257]. Besides, as
dition), which is indicative that systemic inflammation exists compared to non-smokers with periodontitis, smokers with
in periodontitis [239, 240]. The acute-phase reactants pos- periodontitis have been reported to have significantly higher
sess proinflammatory properties such as neutralizing invad- white blood cell counts [258].
ing pathogens, activating complement factors, and regenerat- Heat-shock proteins (HSPs) protect human cells in the
ing and repairing different tissues. Plasminogen activator state of stress, for instance, during inflammation, and can
inhibitor-1 (PAI-1) and C-reactive protein (CRP) are the result in the production of HSP-specific antibodies as well as
acute-phase reactants that have received the most focus in T-cells by interacting with the innate and adaptive immune
research. A number of meta-analyses have noted that in- responses [259]. In the same way, the antigens that are ex-
flammation is the chief mediator of the association between pressed by the bacteria under stress could simulate/mimic
periodontal and cardiovascular disease [241-246]. human HSPs, which could further stimulate the production
Inflammation plays a central role in both periodontitis of antibodies and autoreactive T-cells against human cells
and coronary artery disease. Indeed, acute coronary syn- [260]. Additionally, evidence suggests that increased levels
drome has been linked to periodontitis, with the recognition of these HSPs occur in patients with periodontitis, which
of immunological burden as an important mediator between could stimulate atherosclerosis by promoting inflammation
these conditions [34, 247]. Although no causal relationship [261]. Several studies have stated that periodontal pathogens,
has been identified between chronic periodontitis and a like P. gingivalis, express HSPs that have been involved in
higher risk for cardiovascular diseases, an association be- atherosclerosis [262-264]. Furthermore, the presence of
tween them has been confirmed in several studies, even after cross-reactivity between periodontal bacterial HSPs and en-
adjusting for known confounding variables, including obe- dothelial cell-HSPs has been reported by numerous studies
sity, smoking and diabetes mellitus [29]. An inflammatory [265-268], which further suggests that HSP-induced immune
reaction and periodontal bone loss has been reported to be responses might promote atherosclerosis.
induced after implantation of P. gingivalis in an animal study One of the markers for activated cell-mediated immunity
[248]. A critical role in osteoclast development and innate includes neopterin [269]. The production of neopterin results
immunity against invading pathogens is played by tumor from the interaction between T-helper (Th)1 lymphocytes
necrosis factor receptor-associated factor 6 (TRAF6) [249]. and phagocytic cells, which include granulocytes, macro-
It has been shown that in the IL‐1 receptor/TLR phages, monocytes and dendritic cells [269-271]. Interferon-
(IL‐1R/TLR) signaling pathway that mediates inflammation, gamma (IFN-γ) is the main stimulating factor leading to the
TRAF6 expression is increased by P. gingivalis and bacterial formation of neopterin [270]. On the other hand, increased
pathogen‐associated molecular patterns (PAMPs), which neopterin levels can also result from additional proinflamma-
eventually activates the secretion of proinflammatory cyto- tory markers like LPS and TNF-α [272]. Also, neopterin
kines in human periodontal ligament fibroblasts (HPDLFs); concentration in the bodily fluids marks the immune-related
thereby suggesting that TRAF6 might have an important role oxidative stress [273]. Additionally, there exists an associa-
1362 Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 Wadhawan et al.

tion between neopterin concentration and reactive oxygen NAs inhibit gene expression, upregulation of TNF-α by miR-
species (ROS), which in turn is stimulated by IFN-γ [274]. 155 could be an indirect regulation via several other inter-
Ozmeriç et al. [275] reported that a significantly higher total mediates. Thus, being pleotropic in nature, miR-155 demon-
amount of neopterin in gingival crevicular fluid (GCF) was strates both inhibiting and enhancing properties to achieve a
present in individuals with aggressive periodontitis com- specific goal. On the other hand, miR-155 can be upregu-
pared to that of healthy controls. They further reported that lated by various TLR ligands. For instance, in the early
systemically healthy individuals with aggressive periodonti- phase of TLR4 signaling, miR-155 was found to be upregu-
tis had significantly higher salivary neopterin levels than the lated [301]. Conversely, miR-146 expression can dysregulate
systemically and periodontally healthy individuals [275]. In a set of TLRs with an immediate effect on cytokine produc-
another study, salivary neopterin levels increased signifi- tion. This innate immune effect functions as a negative feed-
cantly with the number of affected teeth in periodontitis pa- back loop. miR-146 in knock-out mice show widespread
tients, with higher salivary neopterin levels reported in sub- immune suppressive ability, primarily via IL-1 receptor-
jects with ≥ 20 diseased teeth when compared to subjects associated kinase-1 (IRAK1) and TRAF6 [302]. It is upregu-
with < 20 teeth affected by periodontitis [276]. A positive lated in the monocytes in response to bacterial components
association has been reported between CAL and GCF neop- (e.g., LPS) and inflammatory cytokines [303]. In turn, miR-
terin concentrations in a different study, with lowest GCF 146a leads to downregulation of the adaptor proteins that
neopterin concentrations seen in the healthy group of indi- operate downstream of cytokine receptors and TLRs, includ-
viduals (mean = 1.36 nmol/l) and the highest concentrations ing TRAF6 and IRAK1 [303]. Consequently, this abolishes
seen in individuals with moderate to severe periodontitis the activation of a key transcription factor, nuclear factor
(mean = 51 nmol/l) [277]. kappa-light-chain-enhancer of activated B cells (NF-κB),
which has a role in the transcription of several genes in-
3.1. Role of Micro-RNAs volved in inflammation, including adhesion molecules, pros-
MicroRNAs (miRs) belong to a family of 19-24 nucleo- taglandins, chemokines, TNF-α, IL-1β, IL-6, and IL-8 [304].
tide-long RNAs that are post-transcriptional regulators of Eventually, miR-146a leads to inhibition of the inflammatory
gene expression (they do so by destabilizing the mRNA response in keratinocytes, macrophages, and monocytes
translation) and have important physiological functions [278, [305, 306].
279]. Since the discovery of miRNAs in 1993 [280], they Wang et al. have shown the inability of miR19a-3p in
have emerged as a critical player in almost all key cellular suppressing TNF-α expression in the brain of suicide sub-
processes, including development of diseases [281-283]. jects [293]. It appears that RNA binding protein human anti-
Several studies have shown that miRs have a role in inflam- gen R (HuR) competitively inhibits miR-19a-3p binding to
mation and immune response [284-287]. The role of miR- TNF-α 3’UTR region, thus blocking the action of this
NAs has been well established in various psychiatric ill- miRNA in regulating TNF-α expression [293]. Another anti-
nesses, such as major depression, schizophrenia, and bipolar inflammatory miRNA is miR-124, which prevents activation
disorder [288-293]. More recently, the role of miRNAs has of microglia, and hence, has a protective role in neurodegen-
been studied in various immune functions [294]. In the CNS, erative diseases and CNS. Elevated levels of miR-124 can
selective activation of microglia and astrocyte are critical in negatively regulate TLR signaling by targeting TLR6 and
triggering cascades of events associated with neuroinflam- TNF-α. Part of the adaptive immune response can also be
mation [295]. Interestingly, miRNAs modulate both innate targeted by miR-124. Data show that miR-124 can induce T
and adaptive immunity in the CNS [296-298]. Accumulating cell activity with a marked increase in effector response.
reports suggest that a set of miRNAs can generate proin- This increased activity was found to be associated with
flammatory response by inhibiting translation of many anti- upregulated expression of IL-2, IFN-γ, and TNF-α [307].
inflammatory cytokines. On the contrary, an anti- Role of miR-181 family members, including miR-181a,
inflammatory response can be triggered via a set of miRNAs miR-181b, miR-181c, and miR-181d, have been implicated
by altering the expression and functions of proinflammatory in immune cell homeostasis with proinflammatory response.
modulators [299]. Both clinical and preclinical studies sug- Both vesicular and neurological changes have been associ-
gest a significant involvement of miR-155, miR-124, miR- ated with miR-181 expression causing altered expression of
146a, miR-21, miR-27b, miR-223 and let-7 in this process TNF-α, IL-6, IL-1β, and IL-8 in the proinflammatory arm.
[300]. Depending on the types of inflammatory molecules The overproduction of these proinflammatory cytokines also
released by microglia at their different maturation states, the resulted in an elevated level of anti-inflammatory cytokine
target response of different miRNAs or different sets of IL-10 [308]. Some other prominent miRNAs with immuno-
miRNAs can be unique. M1 microglia, which represent the modulatory roles include miR-223, miR-34, and miR-21.
proinflammatory state, produce inflammatory mediators such miR-223 has anti-inflammatory properties in peripheral im-
as proinflammatory cytokines (IL-6, IL-1β and TNF-α), ROS mune cells. In T cells, miR-223 was upregulated in patients
and matrix metalloproteinase-9. Whereas, microglia in their with rheumatoid arthritis and miR-223 impaired activation of
anti-inflammatory state or M2, release anti-inflammatory a protective IGF-1/IL-10 axis. Exacerbation of traumatic
cytokines including IL-10, transforming growth factor beta brain injury mediated via inflammation and apoptosis has
(TGF-β), IL-4, and IL-13 [298]. This dynamic is signifi- been linked with increased expression of miR-34. miR-21 is
cantly modified by participating miRNAs. upregulated in activated immune cells, including neutrophils,
The role of miR-155 has been well-documented in sev- dendritic cells, monocytes/macrophages, and T cells. miR-21
eral inflammatory disorders. Increased level of miR-155 has also affects differentiation of other immune cells, including
been associated with increased level of TNF-α. Since miR- T cells and dendritic cells. Dendritic cells, that are deficient
Periodontal Pathogens and Neuropsychiatric Health Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 1363

in miR-21, express higher IL-12 level. On the other hand, fibroblasts, miR-146a regulated the proinflammatory cyto-
miR-21 deficiency enhances Th1 and Th17 cell responses. kine expression by negatively regulating the inflammatory
Let-7 miRNAs modulate inflammation. Increasing let-7 ex- response via NF-κB and IRAK1, post-stimulation with LPS
pression in macrophages promotes differentiation likely by of P. gingivalis [7, 8].
reducing expression of the transcription factor
Human studies have also reported similar observations.
CCAAT/enhancer-binding protein alpha (CEBP-α) [295,
As compared to healthy controls, individuals with chronic
298-299, 302].
periodontitis have been reported to have an upregulation of
Several studies suggest that inflammatory response asso- miR-146a, suggesting that this miR may facilitate the sur-
ciated with diabetes mellitus may be critical factor in depres- vival of the periodontopathic bacteria [4]. These findings
sion development in diabetes mellitus patients and that corroborate the hypothesis that bacterial components of
miRNAs may be playing crucial role in such development. plaque (e.g., LPS) in aggressive periodontitis, might increase
Interestingly, diabetes mellitus, when associated with sys- the miR-146a via stimulation of TLRs, which, via a negative
temic inflammation (whether it is related to autoimmune or feedback loop, may subsequently control the level of proin-
metabolic disease), is associated with differential miRNA flammatory cytokines [5]. Ghotloo et al. reported that, as
expression. For example, the autoimmune process in type 1 compared to healthy controls, individuals with aggressive
diabetes mellitus has been linked to the islet-specific miR- periodontitis had significantly increased levels of miR-146a
326 [309], as well as other miRNAs known to be involved in (p<0.001), and the severity of the disease was directly asso-
islet cell function (miR-24, 26, 127-3p, 148, 182, 184, 195, ciated with the increases in these levels [5]. Moreover, a re-
375, and 493) [281]. Alterations in expression of miRNAs duction in the levels of proinflammatory cytokines was ac-
have also been linked to system metabolic inflammatory companied with an overexpression of miR-146a [5]. Given
disease and diabetes mellitus complications (e.g., miR-16, that inflammation plays a central role in the pathophysiology
21, 29, 93, 98, 133, 192, 200a, 200b, 200c, 216, 320, 429, of periodontitis, the unexpected finding of reduced expres-
and 503) [281, 283]. It has recently been shown that tran- sion of major proinflammatory cytokines (IL-6, TNF-α and
scriptional regulation of miR-146b is involved in TNF-α and IL-1β) in this study suggests that either a nonimmunologic-
IL-6 induced inflammatory response in obesity [310]. dependent mechanism operates in the progression of perio-
Recently, extracellular vesicular miRNAs have been dontal disease, and/or other inflammatory mediators are in-
studied for their role in regulating distal targets besides their volved in the disease process [5]. However, in a study by
autocrine role in the CNS. Depending on the pathogenicity, Bagavad Gita et al., in response to the negative regulatory
the microvesicles can be uploaded with a selective set of role of miR-146a, the expected downregulation of its poten-
miRNAs to deliver distantly in peripheral circulation. For tial targets (TNF-α and IL-1β) was not seen in individuals
example, let-7b can travel in vesicle and modulates inflam- with chronic periodontitis and acute coronary syndrome, and
matory response by selectively binding and masking TLR7 an upregulation of these cytokines (TNF-α, IL-6 and IL-1β)
[311]. Let-7, miR-124 and miR-21 have been found in was reported instead [253]. Alternatively, many genes are
exosomes, which are released from neurons. These miRNAs regulated by miR-146a, including downregulation of TGF-β
regulate microglia functions [298]. Altogether, it appears and epidermal growth factor receptor [355-357]; hence, it is
that miRNAs are actively involved in regulating immune possible that the periodontal tissue repair and regeneration
response associated with various disorders. This also opens that is stimulated by both TGF-β and epidermal growth fac-
the possibility to use miRNAs for diagnostic and therapeutic tor receptor are disrupted by miR-146a [355, 357].
purposes.
Many factors, including miRs, strictly and finely regulate
As reviewed earlier in the manuscript, P. gingivalis has the balance of TLR4 signaling pathways [358, 359]. Asso-
several virulence factors. Production of a variety of miRs is ciations have been reported between response to antidepres-
stimulated by P. gingivalis, including immunoinflammatory sants and miR-146a from whole blood [360, 361]. In a recent
miR-146a that orchestrates the innate host immune response study by Hung et al., associations between major depressive
[4, 312], miR-203 production that inhibits SOCS3 and disorder and the expression of intracellular miRs that regu-
SOCS6, and miR-105 that suppresses TLR2 production late TLR4 signaling in monocytes and peripheral blood
[153]. MiR-146a has been reported to have a critical regula- mononuclear cells (PBMCs) were investigated [6]. Authors
tory role in periodontal disease [313, 314]. In an in vitro reported that miR-146a levels in PBMCs were significantly
study, Tang et al. reported that HPDLFs had a significantly lower before treatment with antidepressants in individuals
increased expression of miR-146a after being stimulated with a diagnosis of major depressive disorder than in healthy
with P. gingivalis and its LPS [315]. Additionally, signifi- controls, and after antidepressant treatment, miR-146a levels
cant suppression of TRAF6 expression, p38 phosphorylation, increased significantly [6]. A similar pattern was observed
and of IL-6, IL-8, and IL-1β secretion was reported with for miR-146a in monocytes. Moreover, a negative associa-
overexpression of miR-146a [315]. Moreover, P. gingivalis- tion was found between 17-item Hamilton Depression Rat-
stimulated HPDLFs, but not P. gingivalis-LPS stimulated
ing Scale scores and the levels of miR-146a [6]. Together,
HPDLFs, had direct binding of miR-146a to the 3’-UTR of
these results suggest that an aberrant expression of intracel-
TRAF6 [315]. Phosphorylation of p38 could be inhibited by
lular regulatory miRs that regulate TLR4 signaling may play
the suppression of TRAF6. Lastly, P. gingivalis-induced
a role in those with a diagnosis of major depressive disorder
miR-146a upregulation in HPDLFs was significantly abol-
and that treatment with antidepressants can ameliorate these
ished by inhibition of p38 and TRAF6 [315]. Similarly, pre-
impairments [6].
vious studies had also reported that in HPDLFs and human
1364 Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 Wadhawan et al.

Another miR of interest is miR-21 which is expressed [292, 376-379]. Dwivedi (2018) recently presented a thor-
universally in mammalian organs, including the spleen, ough review of the role of miRs in suicidal behavior and
heart, colon, and the small intestine [362]. Significant atten- depression and commented that miRs that were particularly
tion has been given to the roles of miR-21 in cardiovascular involved in stress response, synaptic plasticity, and neu-
biology and disease [363]. Specifically, miR-21 is greatly rotrophic activity were the ones that were highly associated
expressed by the major types of cardiovascular cells that with depressive symptoms [380]. Specifically, miR-21 has
include cardiac fibroblast [364], vascular smooth muscle cell also been reported to have a role in schizophrenia, alcohol-
[365], endothelial cell [366] and cardiomyocyte [367]. Re- ism, and major depressive disorder [334]. Miguel-Hidalgo et
cently, miR-21 was reported to be overexpressed in patients al. reported that, as compared to non-psychiatric controls,
having a combination of heart failure and diabetes mellitus, human subjects with depression and alcoholism had signifi-
in patients with a combination of coronary artery disease and cantly low levels of miR-21 in the white matter adjacent to
diabetes mellitus, as well as in patients having diabetes mel- the orbitofrontal cortex [334]. Another study was done by
litus alone [368]. When compared to that of healthy control performing miR microarray analysis in PBMCs obtained
subjects, the expression of miR-21 in these groups of pa- from healthy controls and from patients with schizophrenia,
tients was elevated 2.21, 1.79 and 1.30 times, respectively who were not on antipsychotic medication [381]. It was ob-
[368]. The authors further reported that in asymptomatic served that after antipsychotic treatment, serum miR-21 ex-
type-2 diabetes mellitus patients, miR-21 expression could pression decreased in patients with schizophrenia. Moreover,
be useful in predicting the onset of heart failure [368]. An- there was a negative correlation observed between improve-
other recent study reported that in elderly patients with acute ment of positive, general psychopathology and aggressive-
myocardial infarction, serum levels of miR-21 were posi- ness symptoms of schizophrenia and the change in miR-21
tively correlated with both serum levels of cardiac troponin I expression [381]. In a different study by Beech et al., it was
(r = 0.5128, p = 0.009) and serum levels of creatine kinase reported that, following psychological stress, expression of
MB isoenzyme (r = 0.3683, p = 0.0229) [329]. The authors miR-21 was upregulated in non-smoking heavy alcohol
also reported that the upregulation of miR-21 expression was drinkers but not in non-smoking moderate drinkers [382].
present in the serum of elderly patients with acute myocar- Even though the differences were not statistically significant
dial infarction, which further, via activation of the c-Jun N- between the two groups, the expression levels of miR-21
terminal kinases/p38/caspase-3 signaling pathway, inhibited were correlated [miR-21; R2 = 0.57] with the amount of al-
the TNF-α-induced apoptosis in human cardiac myocytes cohol drunk in the heavy drinkers group, but not in the mod-
[329]. Similarly, other studies have also reported a higher erate drinkers group [382]. Thus, alteration in miRs, espe-
miR-21 expression in human hearts from patients with heart cially the elevation of miR-21 expression, could be a part of
failure, when compared with that of healthy controls [369, the shared molecular inflammatory mechanisms that link the
370]. pathophysiology of periodontal, cardiovascular, and psychi-
atric disorders. In addition, other miRs that are implicated in
Upregulation of miR-21 also has been reported in re-
periodontal and cardiovascular disease are listed in Table 2.
sponse to stimulation by periodontal pathogen [330]. Moreo-
ver, miR-21 has also been demonstrated to be involved in the
development of gingival mesenchymal stem cells and perio- 3.2. Kynurenine Pathway
dontal ligament cells [371-374]. P. gingivalis virulence fac- The kynurenine pathway is a critical pathway involved in
tors signal via TLRs, inducing inflammation by activating immunoregulation, by limiting very intense or prolonged
the NF-κB pathway, which further leads to miR-21 upregula- inflammation, and in the containment of infection, by depriv-
tion [198, 375]. LPSPg has also been reported to induce ing microorganisms of critical supply of tryptophan. Moreo-
upregulation of miR-21 [330-332], thereby suggesting that ver, the kynurenine pathway is a central pathway connecting
periodontal pathogen-induced inflammation is closely asso- inflammation with brain function and behavior, in principal
ciated with miR-21. Furthermore, during pathogenesis of contributing to both adaptive and maladaptive reactions to
periodontitis, NF-κB was predicted to be regulated by miR- local and systemic infections and low-grade inflammation.
21 and other miRs [330, 332]. More recently, Zhou et al.
reported a greater expression of miR-21 in LPSPg-challenged One of the essential amino acids, tryptophan, is found in
cells, ligated mice, as well as in periodontitis patients. The many foods that are rich in protein [383]. It is transported
authors also concluded that the absence of miR-21 increased across the blood-brain barrier by a transporter for large neu-
NF-κB activation, proinflammatory cytokine production, and tral amino acids. Two pathways are involved in the metabo-
programmed cell death protein 4 (PDCD4) expression [333]. lism of this essential amino acid in the brain. The less preva-
Additionally, miR-21 down-regulated LPSPg-induced in- lent metabolic pathway involves tryptophan acting as the
flammation [333]. Together, these results point towards precursor for the neurotransmitter, serotonin, both centrally
miR-21's negative regulatory functions on proinflammatory and peripherally [384-385]. On the other hand, almost 90%
responses induced by the periodontal pathogens, and protec- of tryptophan metabolism occurs via synthesis of kynurenine
tion offered by this miR in terms of progression of periodon- [386] and the rate-limiting enzymes of this pathway include
titis, which was shown in an in vivo periodontitis animal tryptophan-2, 3-dioxygenase and indoleamine-2, 3-
model [333]. dioxygenase (IDO). Expression of IDO can be induced by
inflammation in the brain, which further increases the syn-
Psychiatric disorders, including schizophrenia, alcohol- thesis of downstream metabolites of this pathway, such as
ism, and depression, have been reported to have miRNA quinolinic acid (QA) and kynurenic acid (KYNA) [387].
alterations in particular brain regions, reflecting the patho- Whereas quinolinic acid produces free radicals and is an
physiological roles of these non-coding type of mRNAs
Periodontal Pathogens and Neuropsychiatric Health Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 1365

Table 2. Micro-RNAs involved in periodontitis, cardiovascular diseases, and depression.

Micro-RNA Upregulation (↑) or Upregulation (↑) or Upregulation (↑) or Reported function References
(miR) downregulation (↓) in downregulation (↓) in downregulation (↓) in
periodontitis cardiovascular disease depression

miR-16 ↑ ↑ ↓ Facilitates cell-cycle [316-322]


arrest and apoptosis,
activated by p53

miR-19a ↑ ↑ ↓ Targets TNF-α, protects [313, 323-326]


cardiomyocytes by
inhibiting apoptosis or
activating autophagy,
negatively regulates
neurogenin 1 to inhibit
the differentiation of
neural stem cells into
neurons

miR-21 ↑ ↑ ↓ Promotes the post- [327-334]


translational degradation
of a transporter that
regulates cholesterol
efflux (ABCG1) in
macrophages and en-
hances macrophage
apoptosis; increased
expression of Col-1, α-
SMA and F-actin, pro-
motes the effects of
TGF-β1-induced activa-
tion of cardiac fibro-
blasts; absence of miR-
21 increased NF-κB
activation, proinflamma-
tory cytokine produc-
tion, and programmed
cell death protein 4
(PDCD4) expression

miR-29a ↑ ↑ - Mediates osteoblast [335-338]


differentiation; targets
IL-12p19 and p40 and
downregulates IL-23;
inhibits both intrinsic
and extrinsic apoptosis;
induces angiogenesis in
hypoxia, hypoxamir

miR-30e ↓ ↓ ↑ Inhibiting cytotoxicity [339-342]


of natural killer cells

miR-146a ↑ ↑ ↓ Regulates the proin- [5, 6, 315, 343, 344]


flammatory cytokine
expression by negatively
regulating the inflamma-
tory response via NF-κB
and IRAK1; regulate
TLR4 signaling
(Table 2) contd….
1366 Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 Wadhawan et al.

Micro-RNA Upregulation (↑) or Upregulation (↑) or Upregulation (↑) or Reported function References
(miR) downregulation (↓) in downregulation (↓) in downregulation (↓) in
periodontitis cardiovascular disease depression

miR-155 ↑ ↑ ↑ Activation by nuclear [344-347]


factor-kappaB, fos/jun,
targeting nuclear factor-
kappaB signaling; medi-
ating type 1 interferon
response to infection;
critical for dendritic cell
function and maturation;
inducing activation of
natural killer cells; es-
sential for T-cell devel-
opment and T-helper 17
response; inhibits DNA
damage-induced apopto-
sis

miR-210 ↓ ↑ - Impairs immune- [314, 343, 348]


suppressive functions of
regulatory T-cells by
targeting forkhead box
P3; hypoxamir, controls
host responses in hy-
poxia by induction of
autophagy and targeting
of hypoxia-inducible
factor-1a (pro-survival)

miR-223 ↑ ↑ ↑ Modulates differentia- [313, 314, 349-352]


tion and activation of
myeloid cells, enhances
granulopoiesis, inhibits
macrophage differentia-
tion, controls excessive
innate immune re-
sponses

miR-486 ↑ ↑ - Leads to stronger activa- [314, 337, 353, 354]


tion of NF-κB by dis-
rupting NF-κB negative-
feedback loops

agonist at (N-methyl-D-aspartate receptor (NMDA) recep- to limit the formation of QA [389]. Together with activation
tors that eventually results in neurotoxicity, KYNA antago- of IDO, metabolites of kynurenine pathway - including tryp-
nizes glutamate to confer neuroprotection and acts as a free tophan, kynurenine, KYNA, and others are involved in the
radical scavenger. QA is primarily produced by the brain pathophysiology of psychiatric disorders and suicidal behav-
microglia and the peripheral macrophages invading the ior [390]. Individuals with both treatment-resistant depres-
brain. Thus, activation of the kynurenine pathway may ex- sion and suicidal behavior reportedly have kynurenine path-
plain the association between inflammation in the brain and way dysregulation, which is associated with neuroinflamma-
degeneration of neurons [387]. Another neurotoxic metabo- tion [391]. It has been hypothesized that NMDA receptors
lite of the kynurenine pathway of tryptophan metabolism and alteration in glutamate neurotransmission may mediate
includes 3-hydroxykynurenine (3-HK), which like QA acts the pathogenesis of depression [392-396].
as an NMDA receptor agonist and generates oxidative free
Increasing evidence points towards an association be-
radicals. 3-HK is a product of enzymatic action by
tween dysregulation of the kynurenine pathway as a conse-
kynurenine-3-monooxygenase on kynurenine and it is con-
quence of inflammation, which eventually leads to an imbal-
verted into QA after a few more enzymatic steps [388]. Also,
ance of neuroactive metabolites in suicidal patients [397,
a neuroprotective metabolite of kynurenine pathway called
398]. Sublette et al. [399] analyzed group differences in
picolinic acid (PIC) is produced competitively by the amino- KYN levels between three groups of individuals, i.e., pa-
β-carboxymuconate-semialdehyde-decarboxylase (ACMSD)
tients with major depressive disorder with a history of sui-
Periodontal Pathogens and Neuropsychiatric Health Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 1367

cide attempt, patients with major depressive disorder without man gingival tissues [418]. As compared to healthy gingiva,
history of suicide attempt, and healthy volunteers. The chronic periodontitis has been reported to have an upregula-
authors reported that, as compared to the patients with major tion in the expression of this enzyme, which may occur, in
depressive disorder without a history of suicide attempt, pa- part, due to the inflammatory cytokines- and bacterial prod-
tients with major depressive disorder with a history of sui- ucts-induced activation of human gingival fibroblasts [418].
cide attempt had a higher level of kynurenine in their plasma LPS treatment of primary human periodontal ligament cells
(t = 2.105, df = 58, p = 0.040) [399]. Dysfunction of gluta- has been reported to induce IDO mRNA expression as well,
mate receptors may also be implicated in the pathophysiol- in a dose-dependent manner, peaking at 8 hours post-
ogy of suicide, as evidenced by a post-mortem study assess- treatment. Moreover, LPS treatment also increased the pro-
ing the differences in glutamate receptors in the frontal corti- duction of kynurenine after 72 hours of treatment [419].
ces of human suicide victims versus those of controls who Obese individuals may have activation of IDO, which is
died of sudden death due to causes other than suicide [394].
suggested by an increased kynurenine:tryptophan ratio and
Additionally, as compared to healthy controls, suicide at-
lower levels of tryptophan in their adipose tissue [420, 421].
tempters have been reported to have a decreased PIC/QA
Kynurenine pathway metabolites may also mediate the asso-
ratio and reduced PIC levels in both plasma and CSF [400].
ciations between metabolic and psychiatric disorders [401].
Alterations in the kynurenine pathway have also been re- For instance, in a study of individuals with bipolar disorder,
ported in bipolar disorder [401-404] and schizophrenia, and serum kynurenine and the kynurenine:tryptophan ratio were
it is hypothesized that KYNA may play a major role in the reported to be positively correlated with high levels of car-
pathophysiology of these disorders [405], as it is a naturally bohydrate craving [422]. Authors also reported that serum
occurring astrocyte-derived NMDA and alpha-7 nicotinic kynurenine:tryptophan ratio was increased in patients with
acetylcholine (a7nACh) receptor antagonist [406, 407]. bipolar disorder, who were also overweight/obese [422].
Higher levels of KYNA have been reported in the postmor- Moreover, increased tryptophan breakdown in individuals
tem prefrontal cortex (2.9 pmol/mg protein vs. 1.9 pmol/mg with cardiovascular disease often results in increased serum
protein in controls) [408] and the CSF (approximately 1.7 kynurenine:tryptophan ratios and are also linked to greater
nM vs. 1.0 nM in healthy volunteers) [409] of patients with risk for fatal outcomes of cardiovascular pathologies [423].
schizophrenia. These findings were confirmed in later stud- Cardiovascular disease mortality has been reported to be
ies that demonstrated increased KYNA levels in both drug- associated with tryptophan catabolites, including kynurenine
naïve [410] and drug-treated [411, 412] schizophrenia pa- and 3-HK [424]. Macrophage-rich cores of human advanced
tients. More recently, a meta-analysis on 13 studies revealed atherosclerotic plaques have been reported to have an in-
that specifically within the central nervous system, KYNA creased expression of IDO [425]. Furthermore, in both gen-
levels were moderately increased in patients with schizo- ders, more advanced atherosclerosis has been positively as-
phrenia, when compared to healthy controls [standardized sociated with IDO activity in the blood [426]. In patients
mean differences (SMDs) = 0.66, 95% CI = 0.25-1.06, p = with suspected stable angina pectoris, increased risk of acute
.001] [413]. Alterations in glutamatergic and cholinergic myocardial infarction can be predicted by elevated levels of
signaling, and indirectly, in dopaminergic signaling, may be plasma kynurenines, including, among others, KYNA and 3-
caused by increased concentration of KYNA, which may HK [427]. Post-stroke patients have also been reported to
eventually lead to schizophrenia symptoms [405]. have higher concentration of several kynurenine metabolites
(such as KYNA) and activation of IDO [428], factors that
Moreover, disturbances in the kynurenine pathway have
have also been associated with development of post-stroke
also been reported in AD [414], cardiovascular disorders,
and metabolic disorders. As compared to cognitively cognitive impairment and increased mortality [429]. Thus,
kynurenine pathway metabolites are involved in neuropsy-
screened controls, older patients with histopathologically
chiatric, cardiovascular, and metabolic conditions, and might
confirmed AD were reported to have an association between
have a role in periodontal disease and the pathogens contrib-
poor cognition and elevated QA levels in the plasma [414].
uting to the local and systemic progression of infection, in-
Furthermore, peripheral monocytes from patients with AD
cluding P. gingivalis.
have been reported to have increased expression of the exci-
totoxic metabolite of the kynurenine pathway of tryptophan
metabolism, i.e., QA [415]. 4. INFLAMMATION AND MENTAL HEALTH

To the best of our knowledge, there are no studies avail- It has been reported that activation of microglial cells as
able that have directly examined the levels of kynurenine part of a chronic inflammatory process in the brain (neuroin-
metabolites in periodontal disease. However, Buczko et al. flammation) can be stimulated by periodontitis, which is
[416] reported that, in comparison with healthy volunteers well-known to induce systemic inflammation as well [9,
and patients with hypertension or diabetes alone, patients 430-432]. In fact, several neurodegenerative disorders, in-
with both diabetes and hypertension had significantly higher cluding AD, amyotrophic lateral sclerosis, multiple sclerosis
concentrations of kynurenine and KYNA in their saliva, and Parkinson’s disease, have been reported to have neuroin-
which could eventually contribute to the local and distant flammation as one of their major features [433-435]. Psychi-
effects of periodontal pathogens, downregulation of antimi- atric disorders have received attention over the past 20 years,
crobial immune pressure, and thus facilitation of transloca- in terms of the possible role that neuroinflammation might
tion. Additionally, the onset and development of periodontal play in their pathogenesis. Specifically, the brains of major
disease may involve the kynurenine derivatives in the human depression [436, 437] and schizophrenia [438-440] patients
saliva [417]. IDO expression has also been confirmed in hu- have been reported to have activation of the microglia, as
evidenced by studies utilizing positron emission tomography
1368 Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 Wadhawan et al.

and immunohistochemistry. Hence, neuroinflammation croglia [460]. Elevation of excitotoxic kynurenine metabolite
likely has a significant role in the overall neuropsychiatric QA may lead to neurodegenerative changes that could be
health of an individual. linked to the reduction of astrocytes [461]. Neuronal repair
and neuroregeneration are also suppressed by chronic stress.
Psychological stress is defined as the emotional and
physiological sequelae experienced by individuals faced with A decrease in the synthesis of brain-derived neurotrophic
factor (BDNF) has been associated with a stress-induced
adverse life events that exceed perceived coping skills. Both
increase in proinflammatory cytokines, and chronic admini-
physiologic and emotional reactions are evoked by stress,
stration of antidepressant medications can reverse these al-
although modifiable, which serves as an important risk factor
terations [462]. Depending upon their concentrations, gluco-
for both physical and mental illnesses. Stressful life events
corticoids may have either pro- or anti-inflammatory effects.
are robustly, and presumably causally, associated with major
depressive disorder [441, 442]. Both molecular and cellular Studies have demonstrated that, while higher concentrations
of cortisol equivalent to those seen following a major physi-
abnormalities are thought to play a role in the neurobiology
cal stress increase the inflammatory response triggered by
of stress and depression, and these abnormalities also have
LPS, basal cortisol concentrations exert an anti-
interactions with environmental and genetic determinants
inflammatory effect [463]. Activation of the NF-κB pathway
[443]. The role of psychological stress as a potential cofactor
and mitogen-activated protein kinase signaling, which are
in the pathogenesis of infectious disease is exemplified by an
association between dose-dependent stress and an increased essential for the synthesis of proinflammatory cytokines and
chemokines, results in this proinflammatory effect [464].
risk of acute infectious respiratory illness [444]. The immune
Depression is also accompanied by neurodegeneration, par-
system may be impacted directly by stress. For instance,
ticularly in the hippocampus, frontal cortex, and amygdala,
after adjustment for age, gender, and cigarette smoking,
and the frequency and severity of depressive episodes are
Genco et al. reported a higher risk of more severe periodon-
positively associated with the amount of neurodegeneration
tal CAL (OR = 2.24) and alveolar bone loss (OR = 1.91)
among individuals with financial strain and an inadequate [465, 466]. These neurotoxic effects are mediated by the
proinflammatory cytokines, hypercortisolemia, and the syn-
coping style, as opposed to those with low levels of financial
thesis of QA, an NMDA agonist, together with xanthurenic
strain within the same coping group [445]. Data also support
acid and other intermediates via the kynurenine pathway,
a higher risk of infection and immune reaction in the care-
which leads to the production of ROS. The kynurenine
givers of those with chronic illnesses [446-448]. Further-
pathway is further activated via the induction of IDO by the
more, clinical wound healing may be altered by stress [449],
with reports of slow wound healing in individuals with proinflammatory cytokines (particularly IFN-γ) in response
to infections [467-468]. Moreover, infectious agents, includ-
higher psychological stress levels that were independent of
ing Toxoplasma gondii, have also been associated with
the duration of stressors [449-451]. Stress has also been re-
symptoms of depression [469, 470], suggesting the possible
ported to impede oral mucosal wound healing [452]. An
role of periodontal pathogens, especially P. gingivalis, as
overview of the mechanisms that link chronic stress, perio-
well as periodontal disease/tooth loss, in depression as well
dontal disease, and depression can be found in an extensive
review by Warren et al. [115]. [114, 471].
Major psychiatric disorders, including depression [472-
A part of the adaptive mechanism in the ‘fight or flight’
475], schizophrenia [476-479], and bipolar disorder [480-
response is the stress-induced hypercortisolemia. However,
483], have been reported to manifest a proinflammatory
the stress response can have detrimental effects on the brain
state. Neuroinflammation has been reported to be involved in
and the peripheral organs if it is prolonged and dissociated
from the initiating stimulus. For instance, hypercortisolemia, the pathophysiology of AD as well [484, 485]. Moreover,
onset of AD may be delayed, and its progression may be
which indicates the hyperactivity of the hypothalamic-
slowed by conventional non-steroidal anti-inflammatory
pituitary-adrenal axis, is present in about 80% of severely
drugs (NSAIDs) [486, 487]. Increased risk for all-cause de-
depressed patients but in only about 50% of depressed pa-
mentia has been reported in subjects treated for chronic in-
tients overall [453]. Counterintuitively, major depression is
flammatory disorders, including inflammatory arthritis (OR:
frequently associated with an increase in proinflammatory
cytokines, chemokines, and prostaglandins, despite the pres- 1.33, 95% CI: 1.06-1.63), bullous skin diseases (OR: 1.55,
95% CI: 1.11-2.18), systemic vasculitis (OR: 1.64, 95% CI:
ence of increased glucocorticoids that are usually associated
1.24-2.18) and Crohn's disease (OR: 2.08, 95% CI: 1.16-
with a reduction in proinflammatory mediators [454, 455].
3.74) [488]. The authors also reported that a reduced risk
Although glucocorticoids suppress expression of inflamma-
for dementia across most conditions, especially for systemic
tion after an acute stress response, glucocorticoid receptor
autoimmune disorders (OR: 0.41, 95% CI: 0.18-0.95), was
desensitization after chronic stress ultimately leads to a
proinflammatory state [456]. Following chronic stress, mi- associated with combined glucocorticoids and NSAIDs ther-
apy [488].
croglia change from the resting to the active state in several
regions of the brain [457]. Depressed patients may also evi- Neuroinflammation has been reported to play a role in
dence these changes [458]. Indirect evidence in support of suicidal behavior [489]. Meta-analytical evidence exists for
stress-induced microglial activation in depression is provided the presence of immune activation in the blood and CSF of
by clinical studies reporting enhancement of peripheral suicide attempters as well as in the postmortem brain sam-
macrophage activity following chronic social stress [459]. ples of individuals who died by suicide [490-492]. Victims
Furthermore, reduction in the oligodendroglia and astrocytes who died by suicide have been demonstrated to have pro-
in certain regions of the brain, such as the sub-genual pre- nounced microgliosis [458], as well as mRNA transcripts of
frontal cortex and amygdala, accompany the changes in mi- inflammatory cytokines in their orbitofrontal cortex [493].
Periodontal Pathogens and Neuropsychiatric Health Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 1369

Exacerbation of aggression, an endophenotype associated cyte count. In addition, a biological cluster containing these
with suicidal behavior [494], can be precipitated, at least two factors together with increased levels of IL-6, lympho-
partially, by the effects of individual proinflammatory cyto- cytes and monocytes, significantly impacted suicide risk
kines [495-497]. At both the mRNA and protein levels, in- [527]. An independent and negative association was uncov-
creased levels of proinflammatory cytokines like IL-1β, IL- ered between the cytokine IL-8 and higher risk for suicide.
6, and TNF-α, have been reported in the anterior prefrontal Moreover, even after adjusting for confounders, these results
cortex of teenagers who died by suicide [498]. The signifi- remained significant [527]. IL-6 is a proinflammatory cyto-
cantly elevated levels of IL-6 in the CSF of suicide attempt- kine and has been previously associated with suicidality as
ers further supports the role of neuroinflammation in suicidal well, and may be a part of the immunobiological network
behavior [499]. Other inflammatory triggers, such as trau- that plays a role in increasing the risk for suicide [499, 507,
matic brain injury, have also been implicated in suicidal be- 528, 529]. Many cells of the immune system, including
havior [500]. monocytes and granulocytes of the innate immune system,
can secrete IL-6, which is both a regulatory and a proin-
Peripheral inflammatory changes are also associated with
flammatory cytokine [530-532]. Moreover, this cytokine can
what we consider to be primary depression [501-504]. Both
also be secreted in response to exercise and by adipose tissue
central and peripheral innate immune systems have been
[533-536].
linked to depression [505]. In fact, more data are surfacing
that confirm the presence of pronounced inflammatory TLRs mediate the recognition of PAMPs from endoge-
changes in both the CSF and blood of patients with suicidal nous and infectious pathogens, and hence, play an important
ideations and behavior [490, 499, 506, 507]. In the past, only role in early innate immune response. Once the pathogens
a limited number of inflammatory factors have been studied are recognized, production of large amounts of inflammatory
in association with suicide risk and depressive symptoms cytokines is induced, such as IL-1β, TNF-α and IL-6, via
[499, 508, 509]. However, a complex interaction exists activation of NF-κB and other transcription factors [537].
among the inflammatory factors with specific inflammatory Protein expressions of TLR2, TLR3, TLR4, TLR6 and
factors and groups of inflammatory factors yet to be defined TLR10, and mRNA expression of TLR2 and TLR3 have
etiologically in suicidal and depressive symptoms. been recently reported to be significantly increased in the
postmortem brain of depressed and suicide subjects [538].
Neopterin has also been implicated in psychiatric disor-
Additionally, peripheral blood mRNA levels of TLR3,
ders, including bipolar disorder [510], schizophrenia [511,
512], and major depression [513]. As compared to those of TLR4, TLR5, and TLR7 in depressed patients have been
reported to be significantly increased as well [539].
healthy controls, significantly higher plasma neopterin levels
[475, 514], as well as urinary neopterin levels [515, 516]
have been reported in individuals with depression. In fact, 5. OTHER MECHANISMS POTENTIALLY CON-
neopterin can serve as an independent predictor for the de- TRIBUTING TO CARDIOVASCULAR AND NEURO-
velopment of major depression 6-months post-acute PSYCHIATRIC DISORDERS
ischemic stroke [517]. Moreover, as compared to healthy 5.1. Translocation of Bacteria
controls, antipsychotic-naive patients with schizophrenia had
significantly higher serum neopterin levels before treatment. While frequently found in individuals with periodontal
Neopterin levels declined significantly within this cohort of infections, P. gingivalis is also found in a significant propor-
schizophrenic patients after three months of treatment with tion of individuals without any evidence of local periodontal
antipsychotic medications. [518]. conditions [540]. This suggests that translocation of this bac-
terium into other organs and tissues may contribute to its
Elevated levels of CRP have also been reported to be as- presence in distal sites. Besides dental procedures, common
sociated with an increased risk for suicidal ideation, with and less traumatic activities including chewing, brushing,
suicidal ideation being significantly more prevalent in the and flossing, can also lead to transient bacteremia of P. gin-
highest compared with the lowest serum CRP quartile (OR: givalis [53], which results in translocation of this bacterium
1.79; 95% CI: 1.11–2.89), even after adjustment for multiple to several distant tissues, such as liver [541], coronary arter-
confounders [519]. Many studies have reported that patients ies [542], and placenta [543], thereby contributing to the
with depression have higher levels of IL-1β, IL-6, TNF-α development of diabetes mellitus (by influencing hepatic
and CRP [498, 520-522]. Studies have reported a correlation glycogenesis), atherosclerosis, and pre-term delivery, respec-
between increased white blood cell counts and suicidality tively (Figs. 1 and 4). In fact, P. gingivalis arterial coloniza-
(ideation and behavior) in depressive patients [523]. Moreo- tion was recently reported in 100% of patients with cardio-
ver, increased white blood cell counts have been associated vascular disease [544].
with impulsivity as well [524], which in turn, has been
linked to suicide [525]. For example, Batty et al. evaluated Periodontopathic bacteria can gain access to the systemic
over 300,000 women and reported that in about 1,000 who circulation due to breakdown of tissues in the oral cavity, a
eventually died by suicide, suicide-related mortality was characteristic of periodontitis [52, 53]. Animal models of
predicted by higher levels of white blood cell counts [526]. oral infection with P. gingivalis have described stimulation
of inflammatory reactions in distal sites, such as atheroma-
In a recent study by Keaton et al., a distinct inflammatory tous plaques [545-547]. Other common oral pathogens in-
profile in women patients who were at an increased risk for cluding Streptococcus sanguis, Chlamydia pneumoniae, and
suicide was reported [527]. It was found that the strongest Streptococcus mutans (S. mutans) have been isolated in hu-
influence on suicide risk was conferred by the increased lev- man atherosclerotic plaques as well [548-551]. Several spe-
els of white blood cell count and polymorphonuclear leuko- cies of oral bacteria have also been detected in human
1370 Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 Wadhawan et al.

Mfa1

Dendritic Cell
Internalization Membrane

Escape to ?
cytosol?

Vacuole

Fig. (4). Dendritic cell entry can be manipulated by P. gingivalis using its accessory proteins and minor fimbriae Mfa1 that interact with host
cell α5β1-integrin, an adhesion molecule. (Reproduced from How KY, Song KP and Chan KG (2016) Porphyromonas gingivalis: An Over-
view of Periodontopathic Pathogen below the Gum Line. Front. Microbiol. 7:53. doi: 10.3389/fmicb.2016.00053).

cardiac valve samples, including S. mutans, P. intermedia, P. mune responses may promote inflammatory reactions driven
gingivalis, and T. denticola [552]. Hence, systemic as well as by gut microbiota [561, 562], ultimately resulting in insulin
local systemic inflammatory reactions could be promoted by resistance [563]. Commensal bacteria that exist under
the periodontal bacteria residing within the atheromas and physiological conditions in the gut prevent pathogens from
the cardiac valves, or by the proinflammatory components invading tissues and organs, mediate food digestion, and
produced by these bacteria [553]. Moreover, murine models strengthen the immune system. However, when detrimental
have reported induction of platelet aggregation by P. gin- bacteria become predominant resulting in dysbiosis, the gut
givalis and S. mutans inoculation, presumably leading to epithelial wall may be damaged by noxious agents, including
formation of thrombi and accelerating the process of athero- bacterial metabolites and toxins, which could result in im-
genic plaque formation [554, 555]. pairment of various tissue and organs after being absorbed
into the systemic circulation via the disrupted gut epithelium
Yet, treatment for periodontitis may in itself be a trigger
[564]. In fact, P. gingivalis has been reported to be associ-
for a transient rise in systemic inflammation [556]. In fact,
ated with non-alcoholic steatohepatitis (NASH) in mice, was
periodontal treatment has been associated with a temporary
found to aggravate NASH via promoting inflammation, and
increase in systemic pro-thrombotic/inflammatory mediators
was present in injured liver as well [565]. Moreover, massive
lasting for at least one week [556], as well as with endothe-
lial dysfunction 24 hours post-treatment [557]. These asso- invasions of the gut by oral bacterial species have been im-
plicated in the major changes observed in gut microbiota of
ciations can be explained by the trauma, and bacteremia fol-
the patients with liver cirrhosis [566].
lowing periodontal treatment [558]. However, the eventual
reduction in inflammation after treatment for periodontitis is Periodontopathic bacteria may also have effects on insu-
lasting and prolonged, which outweighs the transient initial lin action [567] and glycogenesis in liver [541], and could
rise in inflammation post treatment. For example, in a self- lead to systemic inflammation and metabolic alterations by
controlled case-series study, it was reported that after exclud- inducing a change in gut microbiome [568]. In particular,
ing subjects with hypertension, diabetes and coronary artery increased systemic inflammation and insulin resistance have
disease, as well as the individuals with prescriptions for sali- been reported in mice after P. gingivalis was administered
cylate or antiplatelet drugs before dental treatment, the inci- via oral route to them [568]. In another study by this group
dence rate ratio for rate of vascular events in the first 4 of researchers, it was reported that upon comparison with
weeks after invasive dental treatment was significantly sham-inoculated mice, C57BL/6 mice that received P. gin-
higher (1.50; 95% CI: 1.09-2.06) [559]. On follow-up of givalis (strain W83) orally had increased serum endotoxin
these individuals, authors further found that within 6 months levels and significantly altered gut microbiota [569]. Down-
of receiving the invasive dental treatment, the increased rate regulation of the expression of occludin and tjp-1, genes that
of vascular events normalized to baseline values [559]. play a role in intestinal permeability, was also observed
[569]. These findings coincided with the dissemination of
Metabolic diseases are characterized by chronic low-
grade inflammation [560] in which innate and adaptive im- enterobacteria to the liver, and the alterations in gut microbi-
Periodontal Pathogens and Neuropsychiatric Health Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 1371

ota occurred prior to the changes in systemic inflammation 5.2. Neuroinflammation


[569]. Hence, periodontal pathogens may induce alterations
Proinflammatory cytokines that are produced peripher-
in the gut microbiota [153, 568, 570] and altered nutrition
ally from systemic inflammation, which in turn is generated
associated with periodontal disease [571] may affect the gut
by periodontitis, may lead to neuroinflammation even with-
microbiota as well [572]. Psychiatric symptomatology can
also be affected by changes induced in the gut microbiome out the periodontal pathogens or virulence factors directly
coming in contact with the neural tissues. Potentially, these
[573], through the gut-brain connection. This is evident from
neuroinflammatory effects of the peripherally produced
a number of clinical studies [574], imaging studies [575],
proinflammatory cytokines can be mediated through three
and animal [576-582] studies. Additionally, microbes in the
pathways (cellular, neural and humoral). Periodontal patho-
gut may have immuno-modulatory effects on the brain [583-
gens may communicate with microglia in the brain at the
585], and have implications for mental illness. For example,
more recently, Painold et al. [586] performed a cross- meningeal interface. Meninges function as a physical barrier
between the bloodstream and the CSF and cover the paren-
sectional study by performing 16S rRNA gene sequencing of
chymal surface of the brain. Receptors for LPS of P. gin-
stool samples in 10 healthy controls and 32 individuals with
givalis, such as TLR 2 and 4, are also expressed on the men-
bipolar disorder. Authors reported that microbial alpha di-
ingeal cells. P. gingivalis LPS circulating in the bloodstream
versity was negatively correlated with bipolar disorder ill-
can lead to activation of the meningeal cells, which eventu-
ness duration (R = −0.408, p = 0.021) and that bacterial
clades in patients with bipolar disorder were associated with ally form proinflammatory cytokines inside the neural tis-
sues lined by these cells [595, 596]. Thus, neuroinflamma-
several highly relevant variables, including metabolic syn-
tion may ultimately ensue via microglial activation resulting
drome, inflammatory status, depressive symptoms, trypto-
from the proinflammatory cytokines that were originally
phan levels, serum lipids, and oxidative stress [586] (Fig. 5).
produced by the meningeal cells. Also, systemic inflamma-
After establishing infection in the mouth, P. gingivalis tion manifested by a greater number of monocytes circulat-
may spread via several routes and eventually can access the ing in the bloodstream together with activated endothelial
brain. For example, it may: a) infect and spread via cranial cells is involved in the cellular pathway [597]. Endothelial
nerves [i.e., olfactory [587] or trigeminal] to the brain; b) cells that express receptors for IL-1 and TNF-α can be acti-
infect the monocytes that may then undergo recruitment to vated by these proinflammatory cytokines in the bloodstream
the brain [588, 589]; and/or c) directly infect and damage the [598]. Upon activation, these endothelial cells interact with
endothelial cells that protect the blood-brain barrier [172]. If the macrophages located in the perivascular space [598].
it gains entry into the brain, P. gingivalis may slowly spread Eventually, activation of the microglia occurs due to the in-
along anatomically connected pathways from neuron to neu- teraction between these perivascular macrophages and the
ron over many years, a phenomenon that has been described microglia. Besides producing proinflammatory cytokines,
for vascular cell-to-cell transmission of this bacterium as upon activation, microglia release chemokines (including
well [590]. Moreover, neuron-to-neuron spread, a pattern monocyte chemoattractant protein-1) and proteases as well.
that mimics the course of an infection, has been suggested In fact, recruitment of monocytes in the brain regions that
for tau pathology as well [591]. Dominy et al. [101] pro- are implicated in behavioral control, such as basal ganglia,
posed that transneuronal spread of P. gingivalis may be re- motor cortex and hippocampus, is likely mediated by mono-
sponsible for the tau pathology seen in AD brains. Gingi- cyte chemoattractant protein-1 [599]. Neural pathway in-
pains of this bacterium activate the human proteases operat- volves the direct activation of the vagus nerve or other pri-
ing on tau, and since tau is a target of gingipain proteolysis, mary afferent nerves by the cytokines in the systemic circu-
they can directly damage the tau themselves. Blood-brain lation. The nucleus of tractus solitarius serves as the primary
barrier can be damaged by bacterial LPS via aberrant activa- projection of the neural pathway and receives the incoming
tion of matrix metalloproteinase, thereby increasing its per- signal, which is then relayed onto hypothalamic nuclei in the
meability [592], and eventually, allowing the penetration of brain that act as the secondary projection of this pathway
brain by the periodontal bacteria in the blood stream. Detec- [600]. Although, peripheral production of proinflammatory
tion of LPS derived from P. gingivalis has been reported in cytokines induced by LPS is not affected by subdiaphrag-
AD patients’ brains [99] and DNA of this periodontal patho- matic vagotomy, it does lead to the impeding of the sickness
gen has also been found in the brain of mice that were in- behavior induced by LPS in rats [601]. Circumventricular
fected with P. gingivalis orally via quantitative polymerase organs and choroid plexus that are devoid of the blood-brain
chain reaction [101]. Additionally, areas of the brain which barrier are implicated in the humoral pathway. Through vol-
are deficient of a contiguous blood-brain barrier, such as the ume diffusion via these permeable areas, the proinflamma-
choroid plexuses and circumventricular organs, could allow tory cytokines circulating in the bloodstream can gain access
the periodontal pathogens that are circulating in the blood- to the brain tissue and may modify the functioning of the
stream to gain access into the brain. brain by inducing signaling events downstream [597].
Thus, periodontal pathogens or their virulence factors 6. UPCOMING CONCEPTS AND POSSIBLE INTER-
could infiltrate the brain through any of the pathways (as VENTIONAL STRATEGIES
described above) and may eventually lead to microglial acti-
vation, as evidenced by the reports that have implicated LPS Dominy et al. reported multiple lines of evidence sug-
of P. gingivalis or the P. gingivalis bacterium itself in in gesting a causal role of P. gingivalis in AD [101]. For exam-
vitro production of proinflammatory cytokines (IL-6, IL-1 ple: 1) both P. gingivalis RgpB and Kgp gingipain loads in
and TNF-α) via activation of microglial cells [593, 594]. human brains were correlated with AD diagnosis, ubiquitin
load, and tau load; 2) co-localization of RgpB was observed
1372 Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 Wadhawan et al.

Depression
Dementia
Myocardial Coronary Carotid Schizophrenia
Endothelial Bipolar disorder
infarction artery artery
cells Suicidal behavior
Atherosclerosis domain domain
Stroke

Translocation

Liver Gut
P. gingivalis

Micro-RNAs
(miR-21,
miR-146a)

Inflammation

Metabolic dysregulation
Gut dysbiosis
Diabetes
Gut permeability
Dyslipidemia
Immune dysregulation
Metabolic syndrome

Fig. (5). Schematic showing possible mechanisms by which periodontal infection with Porphyromas gingivalis (P. gingivalis) may result in
neuropsychiatric and cardiovascular disorders. Infection with P. gingivalis is associated with an increase in inflammation that helps in sup-
pressing this pathogen. However, anti-inflammatory micro-RNAs (miRs), i.e., miR-21 and miR-146a are induced by P. gingivalis infection
that inhibit inflammation, thereby facilitating the survival of this pathogen in the host and giving it an opportunity to translocate to distant
sites via blood vessels. Then, P. gingivalis localizes in the endothelial cells of the carotid and coronary artery domains, and gains access to
vital organs. This advance of P. gingivalis could further lead to multiple neuropsychiatric and cardiovascular disorders. Additionally, when
present in the gut, it may induce dysbiosis of host gut microbiome that leads to immune dysregulation and an increase in inflammation.

with human hippocampal astrocytes, neurons, and pathology TNF-α levels in mice brain tissue with established P. gin-
in AD (i.e., intraneuronal Aβ and tau tangles); 3) P. gin- givalis infection [101].
givalis 16S rRNA gene, hmuY genes, and Kgp gingipain Based on the above detailed commentary in our manu-
were all detected in the cerebral cortex of AD brains; 4) P.
script so far, it becomes essential to consider future direc-
gingivalis DNA was identified in the CSF of individuals
tions for developing novel treatments for reducing the load
clinically diagnosed with probable AD; 5) observation of
of periodontal pathogens and their associated systemic and
fragmentation of tau by gingipains; 6) P. gingivalis gingi-
psychological manifestations. Dominy et al. [101] have
pains were found to have toxic effects on neurons in vitro
shown that twice-daily oral administration of COR271, a
and these effects could be reversed by administering small- Kgp inhibitor capable of bypassing the blood brain barrier,
molecule gingipain inhibitors in mouse models; 7) brain in-
results in significantly reduced brain bacterial load at 5
fection with P. gingivalis and induction of Aβ1-42 occurred in
weeks in P. gingivalis-infected BALB/c mice as compared to
mice infected orally with this bacterium; 8) antibacterial ef-
control mice. Even though prevention of the increasing colo-
fects of Aβ1-42 on P. gingivalis were uncovered; 9) effective
nization of the brain by P. gingivalis between day 35 and day
treatment of P. gingivalis brain infection and prevention of
70 was achieved by the broad-spectrum antibiotic moxiflox-
loss of hippocampal glutamic acid decarboxylase-67+ in- acin, it was not effective in decreasing the load of P. gin-
terneurons in vivo were achieved by oral administration of a
givalis below the baseline levels seen (i.e., 5-weeks prior)
Kgp inhibitor; and 10) dose-dependent efficacy of COR388
[101]. Besides, efficacy was not improved with combina-
(a selective irreversible small-molecule inhibitor of Kgp)
tions of COR271 with moxifloxacin or COR286 versus that
treatment in reducing P. gingivalis load, host A β1-42, and
achieved with COR271 administration in isolation. Another
highly potent and selective irreversible small-molecule in-
Periodontal Pathogens and Neuropsychiatric Health Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 1373

hibitor of Kgp is COR388 which has significant CNS pene- Kgp inhibitor may result in the prevention or slowing of fur-
tration and superior oral pharmacokinetic properties. ther neurodegeneration and accumulation of pathology in
Dominy et al. [101] reported that this compound was effec- AD patients, in the reduction in brain infection with P. gin-
tive in treating an in vivo brain infection with P. gingivalis in givalis, and in the possible modification of the neuropsy-
a mouse model. Dose-dependent efficacy to treat P. gin- chiatric and systemic manifestations of P. gingivalis infec-
givalis brain infection that had already been established was tion in the brain, cardiovascular system, and metabolic or-
reported for twice daily dosing with COR388. For instance, a gans which may positively impact the overall health of its
3 mg/kg dosage of COR388 showed some decrease in the human host.
load of brain P. gingivalis, but there was no drop in the lev-
Notable too is that, activity of P. gingivalis can also be
els of TNF-α or Aβ1-42 in the brain. On the other hand, in
inhibited by several plant-derived compounds. Murakami et
comparison with infected animals treated with vehicle, al. performed a study on anti-inflammatory activity of res-
higher doses of 10 mg/kg and 30 mg/kg of this compound
veratrol, quercetin, and their related compounds (4-
reduced P. gingivalis load, TNF-α, and Aβ1-42 levels in the
allylphenol, catechin, epicatechin and orcinol) [605, 606].
brain tissue of infected mice. In fact, COR388 is currently
The authors reported inhibitory effects of these natural com-
under clinical trials (GAIN Trial) [602].
pounds on the activity of fimbriae of P. gingivalis, although
Additionally, neuroprotective effects of the small- more research is needed on the various strains of this perio-
molecule gingipain inhibitors have been demonstrated in dontal pathogen, which may have differing pathogenicity
mice. After a single administration of gingipain inhibitors by and virulence. Inhibition of the proinflammatory effects of P.
combining COR286 (given subcutaneously) and COR271 gingivalis LPS by several natural compounds has also been
(given by oral gavage), or both vehicles in eight-week-old reported. For instance, a reduction in IL-8 and IL-6 expres-
BALB/c mice, stereotactic injection was performed 1.5 sion has been demonstrated in human gingival fibroblasts
hours later with a combination of RgpB and Kgp into the stimulated by P. gingivalis LPS by using alpha-mangostin
hippocampus of these mice [101]. After 7 days, neurodegen- [607]. The roles of α-tocopherol in mitigating the harmful
eration was assessed in the brains of these mice. It was re- effects of LPS have been reported to be mediated by elevat-
ported that, as compared to those of the saline-injected mice, ing β-defensins (an antimicrobial peptide), stimulating the
significantly greater numbers of degenerating neurons were migration and growth of gingival fibroblasts, and by decreas-
present in the mice that were injected with gingipains [101]. ing the inflammatory cytokines [608]. P. gingivalis LPS-
Moreover, the pre-treatment with gingipain inhibitors such induced inflammatory response in human gingival fibro-
as COR286 and COR271, blocked the neurodegeneration blasts has been reported to be inhibited by tormentic acid,
[101], indicating the need for more research on these agents which can be a possible therapeutic agent for periodontal
to be developed into potential novel treatments for infection disease as it possesses anti-inflammatory effects by inhibit-
with P. gingivalis and prophylactic prevention or reduction ing IL-6 and IL-8 production [609]. P. gingivalis LPS has
of the subsequent burden of AD. also been reported to be inhibited by grape seed proantho-
Dominy et al. [92] identified gingipain antigens in the cyanidin extracts [610]. Downregulation of two protease
genes that are associated with inactivation of host defense
brains of individuals with both diagnosed and probable AD.
mechanisms and tissue destruction, i.e., RgpA and Kgp, can
This finding suggests that poor dental care following the
be achieved by rhein, an anthraquinone from rhubarb roots
onset of dementia does not result in the brain infection with
that exhibits antibacterial synergistic effects with other poly-
P. gingivalis; rather, infection seems to be primary and oc-
phenols [611]. Additionally, marked inhibition of the prote-
curs at a much earlier age, thereby explaining why AD pa-
thology can appear in middle-aged individuals prior to a de- inase activities of P. gingivalis gingipains occurs in a dose-
dependent manner via theaflavins, the main polyphenols in
cline in cognitive functioning [603]. Reasonably, P. gin-
black tea [612]. Moreover, a synthetic dual protease inhibitor
givalis strains may exhibit differences in virulence. Studies
of Rgp and Kgp proteases, named KYT-41, is known to pos-
are necessary to identify and characterize different strains
sess anti-inflammatory and anti-bacterial properties against
[603]. Also, more research needs to be done to determine
P. gingivalis and could be promising in treating and prevent-
which strains present in the CSF and in the brain. Moreover,
it needs to be ruled out whether other Porphyromonas spe- ing periodontitis [613]. Studies on diminishing the effects of
OM proteins of P. gingivalis are also available. P. gingivalis
cies possessing gingipains such as Porphyromonas gulae,
adhesion and invasion have been shown to be reduced by
which inhabits the oral cavity of pets including dogs, can be
about 50% with natural plant extracts that interact with bac-
transmitted to their owners’ oral cavity, and could result in
terial OM proteins, such as polyphenols of Myrothamnus
an infection of their brain [604].
flabellifolia [614]. Cytoprotective effects relating to cytokine
It has been demonstrated that protection against in vitro secretion also accompany these anti-adhesive effects [614].
P. gingivalis-induced cell death can be achieved by gingipain Although, it is important to note that testing of this plant
inhibitors but not by broad-spectrum antibiotics [101]. Also, extract was not done on other virulence factors of P. gin-
to help achieve in vivo eradication of P. gingivalis from the givalis and was only carried out on OM proteins.
brain, a high-dose subcutaneous broad-spectrum antibiotic
was less efficacious than an orally administered Kgp inhibi- 6.1. Caution
tor [101]. Resistance is not developed by P. gingivalis to the
Kgp inhibitor COR388, but it develops rapidly to moxiflox- The capability of P. gingivalis of translocation and ma-
acin (a broad-spectrum antibiotic) [101]. Together, these nipulation of the immune system locally and systemically
findings indicate that treatment with a selective and potent may lead to different interpretations of the microbe having a
causal effect on a number of periodontal and distant patholo-
1374 Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 Wadhawan et al.

gies, such as AD. Yet, the causality may be spurious, and the insomnia) of which inflammation is a compelling mechanis-
criterion of “necessary and sufficient” is likely to not be met tic hypothesis. Inflammation has been implicated in mood
by P. gingivalis infection. disorders, anxiety disorders, schizophrenia and cognitive
disorders such as dementia, and behavioral abnormalities
It is possible that unmeasured confounders or upstream
factors (such as genetic and epigenetic) may contribute to a such as suicidal behavior and aggression. As a proximate
modifiable cause of inflammation of demonstrated clinical
spurious association. The presence of the pathogen in the
relevance, P. gingivalis infection stands apart, both in terms
brain or arterial wall could be an epiphenomenon and could
of robustness of its pathophysiological effects, as well as in
be, non-consequential. Furthermore, and even more impor-
the availability of multiple preventative interventions known
tantly, strong enthusiasm for periodontal treatment in the
to affect chronic periodontitis prospectively (flossing, rins-
clinical domain may overlook the fact that the dental proce-
dures, and especially procedures targeting periodontal dis- ing, tooth brushing, dental checkups, and routine preventa-
tive periodontal interventions). Presumably, therefore, teach-
ease, are acutely linked to an increase in cardiovascular
ing oral hygiene to patients and mental health providers, as
events, and only longer term with improvement. Thus, if one
well as increasing access to preventative dental care and
would conceptualize that acute inflammation secondary to P.
treatment, coupled with motivation to attend dental appoint-
gingivalis infection and/or induction of bacteremia may con-
ments, should have a positive effect in reducing the intensity
tribute to exacerbation of mood disorders and suicidal behav-
ior (previously associated with inflammation-elevating and frequency of exacerbation of mood and anxiety disor-
ders, and in reducing the slope of deterioration in cognitive
events), one would have to consider, theoretically and clini-
disorders and schizophrenia, as well as in lowering the
cally, the exacerbation of mental illness and mortality after
prevalence of metabolic disease, cardiovascular symptoms,
periodontal procedures and their potential longer-term bene-
and mortality in patients with mental illness.
fits.
Presuming that the connection between P. gingivalis
CONCLUSION, FUTURE DIRECTIONS AND RE- translocation, immune activation, mental symptoms and be-
SEARCH IMPLICATIONS havioral dysregulation will be confirmed by future studies,
one particularly relevant aspect is the temporality of the phe-
P. gingivalis is a highly successful and intriguing patho- nomena. The initial consequence of periodontal treatment is
gen, in part because of its multiple unique capabilities. First, increased translocation and immune activation, and thus, an
to manipulate the immune system and hijacking phagocytes initial worsening of symptoms, functioning, and cardiovas-
for traveling to distant sites, as well as for evading detection cular and suicide mortality is expected. This might be met
by other immune cells. Second, to simultaneously lead to a with the recommendation of increased efforts for mental
proinflammatory state, and at the same time, to induce pro- health professional visits scheduled during the intervals of
duction of miRNAs that have immunomodulatory effects. increased risk, and with prophylactic visits and pharmacol-
The activation of these miRNAs may induce escape from ogical and psychotherapeutic approaches.
immune surveillance and immune pressure, resulting in the
capability of P. gingivalis to migrate to other distant sites. Large population studies need to test the effect of perio-
Third, the impressive capability of the microbe to localize in dontal intervention on mental health and mental health mor-
the wall of blood vessels, and also in the atheroma lesions tality, including cardiovascular mortality in patients with
and thrombi. Fourth, although naturally a microbe tends to mental illness. The intervention could be analyzed through
compete with other pathogens, P. gingivalis does not show marginal structural modeling, reducing the risk of bias, and
evidence of competition between its serotypes and with other potentially, by contributing to a model of a “quasi-clinical
bacteria, and it appears that it mostly provides synergistic trial” nested in a population study.
interactions with a number of other pathogens. The capacity One way to start the investigation of P. gingivalis con-
of P. gingivalis to build a biofilm is a formidable defense tributing to mood disorders and increased suicide risk factors
mechanism, and its biofilm includes other pathogens con- is to investigate the role of translocated P. gingivalis in ani-
tributing to their co-evading of the immune system. Finally, mal models of anxiety-like and depressive-like behaviors, as
when translocated to the arterial wall, P. gingivalis can in- well as decision making and aggression modeling, testing
duce local downregulation of inflammation employing cer- various psychotropic interventions, and in particular, gingi-
tain miRNAs as well as elevation of anti-inflammatory cyto- pain-targeting pharmacological agents, followed by human
kines such as IL-10 and downregulation of proinflammatory experiments and clinical trials.
cytokines. Yet, when the microorganism dies, release of its
LPS induces inflammation, locally and systemically, elevat- The capability to identify translocated P. gingivalis using
ing proinflammatory cytokine production. The immune ma- positron-emission tomography imaging, correspondence
nipulation and, especially, the anti-inflammatory induction between oral microbiome identification of P. gingivalis,
by P. gingivalis are factors that could be studied for potential identification of serotypes with particularly high pathogenic-
large applications in metabolic disease, cancer, autoimmune ity in terms of effects at a distance (via immune activation or
disease, and neuropsychiatric illness. translocation) are important efforts. A special focus is repre-
sented by the pregnant woman, when identifying and ad-
Mental health symptoms contribute to periodontal dis- dressing infection with P. gingivalis, as periodontal inflam-
ease (either through self-neglect, or other factors leading to mation may contribute to placental health, fetal outcome, and
reduced resilience of the mucosa; e.g., medications, reduced peripartum depression in the mother. Moreover, immuno-
dental hygiene, eating night snacks), but could also be a con- suppression tends to occur early in the phase of pregnancy
sequence of them through a number of factors (such as pain, (to favor immunotolerance of the fetus) [615], thereby giving
Periodontal Pathogens and Neuropsychiatric Health Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 1375

more opportunities to oral pathogens to infect, disseminate IDO = Indoleamine-2,3-


and manifest diverse symptomatology. However, immune Dioxygenase
activation may occur in the later phases of pregnancy and
IFN = Interferon-γ
contribute to depressive symptomatology [616, 617]. Hence,
it becomes essential to clarify the risks vs. benefits of perio- IL = Interleukin
dontal interventions in pregnancy, demonstrate windows of IRAK1 = Interleukin-1 Receptor-
vulnerability for negative effects of translocation on placen- Associated Kinase-1
tal circulation and fetal outcomes, and windows of opportu-
nity when periodontal interventions are expected to result in IM = Inner Membrane
strong benefits. Finally, studies on family aggregation (with KYNA = Kynurenic Acid
differential effects of heritability and household, such as
possible in the Old Order Amish of Lancaster, Pennsylvania) LPS = Lipopolysaccharide
– may compare potential infectious transmission between miR = Micro-RNA
spouses and between siblings and parents and offspring. This
may change the way we think about prevention of putative NASH = Non-Alcoholic Steatohepa-
infectious causes of AD, and by extension, brain involve- titis
ment in neuropsychiatric illness, and may have a consider- NF-κB = Nuclear Factor Kappa-
able prophylactic impact. Light-Chain-Enhancer Of
Activated B Cells
LIST OF ABBREVIATIONS NHANES = National Health and Nutri-
3-HK = 3-Hydroxykynurenine tion Examination Survey
a7nACh receptor = Alpha7 Nicotinic Acetyl- NMDA = N-Methyl-D-Aspartate
choline Receptor NSAIDs = Non-Steroidal Anti-
AAP = American Academy of Pe- Inflammatory Drugs
riodontology OM = Outer Membrane
A. actinomycetemcomitans = Aggregatibacter actinomy- OR = Odds Ratio
cetemcomitans
PAI-1 = Plasminogen Activator In-
ACMSD = Amino-β- hibitor-1
Carboxymuconate-
Semialdehyde- PAMPs = Pathogen‐Associated Mo-
Decarboxylase lecular Patterns
AD = Alzheimer’s Disease PBMCs = Peripheral Blood Mononu-
clear Cells
BDNF = Brain-Derived Neurotrophic
Factor PDCD4 = Programmed Cell Death
Protein 4
CAL = Clinical Attachment Loss
P. gingivalis = Porphyromas gingivalis
CEBPA = CCAAT/Enhancer-Binding
Protein Alpha PIC = Picolinic Acid
CDC = Centers for Disease Control P. intermedia = Prevotella intermedia
and Prevention PPD = Periodontal Probing Depth
CI = Confidence Intervals PSD = Polymicrobial Synergy and
CRP = C-reactive Protein Dysbiosis
CSF = Cerebrospinal Fluid QA = Quinolinic Acid
F. nucleatum = Fusobacterium nucleatum ROS = Reactive Oxygen Species
GCF = Gingival Crevicular Fluid RR = Relative Risk
hAPP-J20 = Human Amyloid Precursor T. forsythia = Tannerella forsythia
Protein T. denticola = Treponema denticola
HPDLFs = Human Periodontal Liga- TGF-β = Transforming Growth Fac-
ment Fibroblasts tor Beta
HR = Hazard Ratio TNF-α = Tumor Necrosis Factor-
HSPs = Heat-Shock Proteins Alpha
HuR = Human Antigen R TLR = Toll-Like Receptor
1376 Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 Wadhawan et al.

TRAF6 = Tumor Necrosis Factor Re- http://dx.doi.org/10.1902/annals.1999.4.1.1 PMID: 10863370


ceptor-Associated Factor 6 [4] Motedayyen, H.; Ghotloo, S.; Saffari, M.; Sattari, M.; Amid, R.
Evaluation of microRNA-146a and its targets in gingival tissues of
CONSENT FOR PUBLICATION patients with chronic periodontitis. J. Periodontol., 2015, 86(12),
1380-1385.
Not applicable. http://dx.doi.org/10.1902/jop.2015.150319 PMID: 26313020
[5] Ghotloo, S.; Motedayyen, H.; Amani, D.; Saffari, M.; Sattari, M.
Assessment of microRNA-146a in generalized aggressive
FUNDING periodontitis and its association with disease severity. J. Periodontal
The project was supported by intramural funds from the Res., 2019, 54(1), 27-32.
http://dx.doi.org/10.1111/jre.12538 PMID: 30328616
University of Maryland Baltimore, MD. Dr. Dwivedi’s con- [6] Hung, Y.Y.; Wu, M.K.; Tsai, M.C.; Huang, Y.L.; Kang, H.Y.
tribution was supported by intramural funding from the De- Aberrant expression of intracellular let-7E, MIR-146a, and MIR-
partment of Psychiatry and Behavioral Neurobiology/ Uni- 155 correlates with severity of depression in patients with major
versity of Alabama at Birmingham, Birmingham, AL and, by depressive disorder and is ameliorated after antidepressant
the grant number R01MH082802 from the National Institute treatment. Cells, 2019, 8(7), E647.
http://dx.doi.org/10.3390/cells8070647 PMID: 31252530
of Mental Health (NIMH / National Institutes of Health [7] Jiang, S.Y.; Xue, D.; Xie, Y.F.; Zhu, D.W.; Dong, Y.Y.; Wei, C.C.;
(NIH), Bethesda, MD. Rocky Mountain MIRECC for Sui- Deng, J.Y. The negative feedback regulation of microRNA-146a in
cide Prevention, Aurora, CO and Military and Veteran Mi- human periodontal ligament cells after Porphyromonas gingivalis
crobiome Consortium for Research and Education (MVM- lipopolysaccharide stimulation. Inflamm. Res., 2015, 64(6), 441-
CoRE), Aurora, CO supported Drs. Brenner, Postolache, 451.
http://dx.doi.org/10.1007/s00011-015-0824-y PMID: 25948157
Lowry and Hoisington. The Department of Veterans Affairs [8] Xie, Y.F.; Shu, R.; Jiang, S.Y.; Liu, D.L.; Ni, J.; Zhang, X.L.
Office of Research and Development (VA-ORD) RR&D MicroRNA-146 inhibits proinflammatory cytokine secretion
Small Projects in Rehabilitation Research (SPiRE: grant through IL-1 receptor-associated kinase 1 in human gingival
number 1 I21 RX002232-01) supported in part Drs. Brenner, fibroblasts. J. Inflamm. (Lond.), 2013, 10(1), 20.
Lowry, Hoisington and Postolache. The Capitol MIRECC, http://dx.doi.org/10.1186/1476-9255-10-20 PMID: 23680172
[9] Makkar, H.; Reynolds, M.A.; Wadhawan, A.; Dagdag, A.;
VISN 5, Baltimore, MD, supported, in part, the input of Drs. Merchant, A.T.; Postolache, T.T. Periodontal, metabolic, and
Potocki and Postolache. The NIMH/ NIH, Bethesda, MD cardiovascular disease: Exploring the role of inflammation and
(grant number 1R21MH116263), and the Department of the mental health. Pteridines, 2018, 29(1), 124-163.
Navy, Office of Naval Research Multidisciplinary University http://dx.doi.org/10.1515/pteridines-2018-0013 PMID: 30705520
Research Initiative (MURI) Award (grant number N00014- [10] Mariotti, A. Dental plaque-induced gingival diseases. Ann.
Periodontol., 1999, 4(1), 7-19.
15-1-2809) partially supported the writing contribution of http://dx.doi.org/10.1902/annals.1999.4.1.7 PMID: 10863371
Dr. Lowry. The DC Department of Behavioral Health, [11] Oliver, R.C.; Brown, L.J.; Löe, H. Periodontal diseases in the
Washington, DC, supported, in part, contributions of Drs. United States population. J. Periodontol., 1998, 69(2), 269-278.
Wadhawan and Postolache. No funding agency was involved http://dx.doi.org/10.1902/jop.1998.69.2.269 PMID: 9526927
in article conception, writing, and finalizing the manuscript. [12] Eke, P.I.; Page, R.C.; Wei, L.; Thornton-Evans, G.; Genco, R.J.
Update of the case definitions for population-based surveillance of
The views expressed here belong to the authors, and do not
periodontitis. J. Periodontol., 2012, 83(12), 1449-1454.
necessarily represent the views of National Institutes of http://dx.doi.org/10.1902/jop.2012.110664 PMID: 22420873
Health, Veterans Health Administration, the Department of [13] Eke, P.I.; Dye, B.A.; Wei, L.; Thornton-Evans, G.O.; Genco, R.J.
Defense, or the DC Department of Behavioral Health. CDC Periodontal Disease Surveillance workgroup: James Beck
(University of North Carolina, Chapel Hill, USA); Gordon Douglass
(Past President, American Academy of Periodontology); Roy Page
CONFLICT OF INTEREST (University of Washington). Prevalence of periodontitis in adults in
The authors declare no conflict of interest, financial or the United States: 2009 and 2010. J. Dent. Res., 2012, 91(10), 914-
920.
otherwise. http://dx.doi.org/10.1177/0022034512457373 PMID: 22935673
[14] Eke, P.I.; Dye, B.A.; Wei, L.; Slade, G.D.; Thornton-Evans, G.O.;
ACKNOWLEDGEMENTS Borgnakke, W.S.; Taylor, G.W.; Page, R.C.; Beck, J.D.; Genco, R.J.
Update on prevalence of periodontitis in adults in the United States:
Support for the writing of this manuscript was provided NHANES 2009 to 2012. J. Periodontol., 2015, 86(5), 611-622.
by the Rocky Mountain MIRECC, Aurora, CO and the Mili- http://dx.doi.org/10.1902/jop.2015.140520 PMID: 25688694
tary and Veteran Microbiome: Consortium for Research and [15] Eke, P.I.; Wei, L.; Borgnakke, W.S.; Thornton-Evans, G.; Zhang,
X.; Lu, H.; McGuire, L.C.; Genco, R.J. Periodontitis prevalence in
Education (MVM-CoRE), Aurora, CO, and in part, by Tun- adults ≥ 65 years of age, in the USA. Periodontol. 2000, 2016,
ing, Inc, Silver Spring, MD, the CS-MAP Washington, DC, 72(1), 76-95.
and by the DC Department of Behavioral Health. The views, http://dx.doi.org/10.1111/prd.12145 PMID: 27501492
opinions and findings contained in this article belong to the [16] Bersell, C.H. Access to Oral Health Care: A national crisis and call
authors. for reform. J. Dent. Hyg., 2017, 91(1), 6-14.
PMID: 29118145
[17] Saremi, A.; Nelson, R.G.; Tulloch-Reid, M.; Hanson, R.L.; Sievers,
REFERENCES M.L.; Taylor, G.W.; Shlossman, M.; Bennett, P.H.; Genco, R.;
Knowler, W.C. Periodontal disease and mortality in type 2 diabetes.
[1] Page, R.C.; Schroeder, H.E. Periodontitis in man and other animals.
Diabetes Care, 2005, 28(1), 27-32.
A comparative review. S. karger: Basel, 1982.
http://dx.doi.org/10.2337/diacare.28.1.27 PMID: 15616229
[2] Lang, N.P.; Lindhe, J. Clinical Periodontology and Implant
[18] Friedewald, V.E.; Kornman, K.S.; Beck, J.D.; Genco, R.; Goldfine,
Dentistry. John Wiley & Sons: New York, 2, 2015.
A.; Libby, P.; Offenbacher, S.; Ridker, P.M.; Van Dyke, T.E.;
[3] Armitage, G.C. Development of a classification system for
Roberts, W.C. The American Journal of Cardiology and Journal of
periodontal diseases and conditions. Ann. Periodontol., 1999, 4(1),
Periodontology editors’ consensus: periodontitis and atherosclerotic
1-6.
cardiovascular disease. J. Periodontol., 2009, 80(7), 1021-1032.
Periodontal Pathogens and Neuropsychiatric Health Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 1377

http://dx.doi.org/10.1902/jop.2009.097001 PMID: 19563277 [34] Renvert, S.; Ohlsson, O.; Pettersson, T.; Persson, G.R. Periodontitis:
[19] Genco, R.J.; Borgnakke, W.S. Risk factors for periodontal disease. a future risk of acute coronary syndrome? A follow-up study over 3
Periodontol. 2000, 2013, 62(1), 59-94. years. J. Periodontol., 2010, 81(7), 992-1000.
http://dx.doi.org/10.1111/j.1600-0757.2012.00457.x PMID: http://dx.doi.org/10.1902/jop.2010.090105 PMID: 20350154
23574464 [35] Holmlund, A.; Hedin, M.; Pussinen, P.J.; Lerner, U.H.; Lind, L.
[20] Mehta, A. Risk factors associated with periodontal diseases and Porphyromonas gingivalis (Pg) a possible link between impaired
their clinical considerations. Int. J. Contemp. Dent. Med. Rev., 2015, oral health and acute myocardial infarction. Int. J. Cardiol., 2011,
2015, 1-5. 148(2), 148-153.
[21] Komatsu, T. Oxidative stress and periodontal disease in Down http://dx.doi.org/10.1016/j.ijcard.2009.10.034 PMID: 19913930
Syndrome In Studies on Periodontal Disease; Springer: Berlin, [36] Khosravi Samani, M.; Jalali, F.; Seyyed Ahadi, S.M.; Hoseini, S.R.;
2014, pp. 211-223. Dabbagh Sattari, F. The relationship between acute myocardial
http://dx.doi.org/10.1007/978-1-4614-9557-4_15 infarction and periodontitis. Caspian J. Intern. Med., 2013, 4(2),
[22] Ali, T.B.; Abidin, K.Z. Relationship of periodontal disease to pre- 667-671.
term low birth weight infants in a selected population--a prospective PMID: 24009957
study. Community Dent. Health, 2012, 29(1), 100-105. [37] Li, P.; He, L.; Sha, Y.; Luan, Q. Periodontal status of patients with
PMID: 22482259 post-acute myocardial infarction. Beijing da xue xue bao Yi xue ban,
[23] Leng, W.D.; Zeng, X.T.; Kwong, J.S.; Hua, X.P. Periodontal 2013, 45 (1), 22-26.
disease and risk of coronary heart disease: An updated meta- [38] Kodovazenitis, G.; Pitsavos, C.; Papadimitriou, L.; Vrotsos, I.A.;
analysis of prospective cohort studies. Int. J. Cardiol., 2015, 201, Stefanadis, C.; Madianos, P.N. Association between periodontitis
469-472. and acute myocardial infarction: a case-control study of a
http://dx.doi.org/10.1016/j.ijcard.2015.07.087 PMID: 26313869 nondiabetic population. J. Periodontal Res., 2014, 49(2), 246-252.
[24] Sfyroeras, G.S.; Roussas, N.; Saleptsis, V.G.; Argyriou, C.; http://dx.doi.org/10.1111/jre.12101 PMID: 23713486
Giannoukas, A.D. Association between periodontal disease and [39] Rydén, L.; Buhlin, K.; Ekstrand, E.; de Faire, U.; Gustafsson, A.;
stroke. J. Vasc. Surg., 2012, 55(4), 1178-1184. Holmer, J.; Kjellström, B.; Lindahl, B.; Norhammar, A.; Nygren, Å.
http://dx.doi.org/10.1016/j.jvs.2011.10.008 PMID: 22244863 Periodontitis increases the risk of a first myocardial infarction: a
[25] Mattila, K.J.; Valle, M.S.; Nieminen, M.S.; Valtonen, V.V.; report from the PAROKRANK study. Circulation, 2016, 133(6),
Hietaniemi, K.L. Dental infections and coronary atherosclerosis. 576-583.
Atherosclerosis, 1993, 103(2), 205-211. [40] Yu, Y.H.; Chasman, D.I.; Buring, J.E.; Rose, L.; Ridker, P.M.
http://dx.doi.org/10.1016/0021-9150(93)90263-T PMID: 8292096 Cardiovascular risks associated with incident and prevalent
[26] Soto-Barreras, U.; Olvera-Rubio, J.O.; Loyola-Rodriguez, J.P.; periodontal disease. J. Clin. Periodontol., 2015, 42(1), 21-28.
Reyes-Macias, J.F.; Martinez-Martinez, R.E.; Patiño-Marin, N.; http://dx.doi.org/10.1111/jcpe.12335 PMID: 25385537
Martinez-Castañon, G.A.; Aradillas-Garcia, C.; Little, J.W. [41] Holmlund, A.; Holm, G.; Lind, L. Severity of periodontal disease
Peripheral arterial disease associated with caries and periodontal and number of remaining teeth are related to the prevalence of
disease. J. Periodontol., 2013, 84(4), 486-494. myocardial infarction and hypertension in a study based on 4,254
http://dx.doi.org/10.1902/jop.2012.120051 PMID: 22680302 subjects. J. Periodontol., 2006, 77(7), 1173-1178.
[27] Chen, Y.W.; Umeda, M.; Nagasawa, T.; Takeuchi, Y.; Huang, Y.; http://dx.doi.org/10.1902/jop.2006.050233 PMID: 16805679
Inoue, Y.; Iwai, T.; Izumi, Y.; Ishikawa, I. Periodontitis may [42] Bazile, A.; Bissada, N.F.; Nair, R.; Siegel, B.P. Periodontal
increase the risk of peripheral arterial disease. Eur. J. Vasc. assessment of patients undergoing angioplasty for treatment of
Endovasc. Surg., 2008, 35(2), 153-158. coronary artery disease. J. Periodontol., 2002, 73(6), 631-636.
http://dx.doi.org/10.1016/j.ejvs.2007.08.016 PMID: 17964192 http://dx.doi.org/10.1902/jop.2002.73.6.631 PMID: 12083536
[28] Lockhart, P.B.; Bolger, A.F.; Papapanou, P.N.; Osinbowale, O.; [43] Buhlin, K.; Gustafsson, A.; Håkansson, J.; Klinge, B. Oral health
Trevisan, M.; Levison, M.E.; Taubert, K.A.; Newburger, J.W.; and cardiovascular disease in Sweden. J. Clin. Periodontol., 2002,
Gornik, H.L.; Gewitz, M.H.; Wilson, W.R.; Smith, S.C., Jr; 29(3), 254-259.
Baddour, L.M. Periodontal disease and atherosclerotic vascular http://dx.doi.org/10.1034/j.1600-051x.2002.290312.x PMID:
disease: does the evidence support an independent association?: a 11940146
scientific statement from the American Heart Association. [44] Parkar, S.M.; Modi, G.N.; Jani, J. Periodontitis as risk factor for
Circulation, 2012, 125(20), 2520-2544. acute myocardial infarction: A case control study. Heart Views,
http://dx.doi.org/10.1161/CIR.0b013e31825719f3 PMID: 22514251 2013, 14(1), 5-11.
[29] Humphrey, L.L.; Fu, R.; Buckley, D.I.; Freeman, M.; Helfand, M. http://dx.doi.org/10.4103/1995-705X.107113 PMID: 23580918
Periodontal disease and coronary heart disease incidence: a [45] Joshipura, K.J.; Rimm, E.B.; Douglass, C.W.; Trichopoulos, D.;
systematic review and meta-analysis. J. Gen. Intern. Med., 2008, Ascherio, A.; Willett, W.C. Poor oral health and coronary heart
23(12), 2079-2086. disease. J. Dent. Res., 1996, 75(9), 1631-1636.
http://dx.doi.org/10.1007/s11606-008-0787-6 PMID: 18807098 http://dx.doi.org/10.1177/00220345960750090301 PMID: 8952614
[30] Rutger Persson, G.; Ohlsson, O.; Pettersson, T.; Renvert, S. Chronic [46] Howell, T.H.; Ridker, P.M.; Ajani, U.A.; Hennekens, C.H.;
periodontitis, a significant relationship with acute myocardial Christen, W.G. Periodontal disease and risk of subsequent
infarction. Eur. Heart J., 2003, 24(23), 2108-2115. cardiovascular disease in U.S. male physicians. J. Am. Coll.
http://dx.doi.org/10.1016/j.ehj.2003.10.007 PMID: 14643271 Cardiol., 2001, 37(2), 445-450.
[31] Cueto, A.; Mesa, F.; Bravo, M.; Ocaña-Riola, R. Periodontitis as http://dx.doi.org/10.1016/S0735-1097(00)01130-X PMID:
risk factor for acute myocardial infarction. A case control study of 11216961
Spanish adults. J. Periodontal Res., 2005, 40(1), 36-42. [47] Dorn, J.M.; Genco, R.J.; Grossi, S.G.; Falkner, K.L.; Hovey, K.M.;
http://dx.doi.org/10.1111/j.1600-0765.2004.00766.x PMID: Iacoviello, L.; Trevisan, M. Periodontal disease and recurrent
15613077 cardiovascular events in survivors of myocardial infarction (MI):
[32] Andriankaja, O.M.; Genco, R.J.; Dorn, J.; Dmochowski, J.; Hovey, the Western New York Acute MI Study. J. Periodontol., 2010,
K.; Falkner, K.L.; Scannapieco, F.; Trevisan, M. The use of 81(4), 502-511.
different measurements and definitions of periodontal disease in the http://dx.doi.org/10.1902/jop.2009.090499 PMID: 20367093
study of the association between periodontal disease and risk of [48] Willershausen, I.; Weyer, V.; Peter, M.; Weichert, C.; Kasaj, A.;
myocardial infarction. J. Periodontol., 2006, 77(6), 1067-1073. Münzel, T.; Willershausen, B. Association between chronic
http://dx.doi.org/10.1902/jop.2006.050276 PMID: 16734583 periodontal and apical inflammation and acute myocardial
[33] Andriankaja, O.M.; Genco, R.J.; Dorn, J.; Dmochowski, J.; Hovey, infarction. Odontology, 2014, 102(2), 297-302.
K.; Falkner, K.L.; Trevisan, M. Periodontal disease and risk of http://dx.doi.org/10.1007/s10266-013-0112-7 PMID: 23604464
myocardial infarction: the role of gender and smoking. Eur. J. [49] Beck, J.; Garcia, R.; Heiss, G.; Vokonas, P.S.; Offenbacher, S.
Epidemiol., 2007, 22(10), 699-705. Periodontal disease and cardiovascular disease. J. Periodontol.,
http://dx.doi.org/10.1007/s10654-007-9166-6 PMID: 17828467 1996, 67(10)(Suppl.), 1123-1137.
http://dx.doi.org/10.1902/jop.1996.67.10.1123
1378 Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 Wadhawan et al.

[50] Rintala, E.M.; Aittoniemi, J.; Laine, S.; Nevalainen, T.J.; [65] Modeer, T.; Blomberg, C.; Wondimu, B.; Lindberg, T. Y.; Marcus,
Nikoskelainen, J. Early identification of bacteremia by biochemical C. Association between obesity and periodontal risk indicators in
markers of systemic inflammation. Scand. J. Clin. Lab. Invest., adolescents. Int. J. Pediatr. Obes, 2011, 6 (2-2), e264-e270.
2001, 61(7), 523-530. http://dx.doi.org/10.3109/17477166.2010.495779
http://dx.doi.org/10.1080/003655101753218283 PMID: 11763410 [66] Wakai, K.; Kawamura, T.; Umemura, O.; Hara, Y.; Machida, J.;
[51] Pierrakos, C.; Vincent, J-L. Sepsis biomarkers: a review. Crit. Care, Anno, T.; Ichihara, Y.; Mizuno, Y.; Tamakoshi, A.; Lin, Y.;
2010, 14(1), R15. Nakayama, T.; Ohno, Y. Associations of medical status and
http://dx.doi.org/10.1186/cc8872 PMID: 20144219 physical fitness with periodontal disease. J. Clin. Periodontol.,
[52] Sconyers, J.R.; Crawford, J.J.; Moriarty, J.D. Relationship of 1999, 26(10), 664-672.
bacteremia to toothbrushing in patients with periodontitis. J. Am. http://dx.doi.org/10.1034/j.1600-051X.1999.261006.x PMID:
Dent. Assoc., 1973, 87(3), 616-622. 10522778
http://dx.doi.org/10.14219/jada.archive.1973.0453 PMID: 4516507 [67] Merchant, A.T.; Pitiphat, W.; Rimm, E.B.; Joshipura, K. Increased
[53] Forner, L.; Larsen, T.; Kilian, M.; Holmstrup, P. Incidence of physical activity decreases periodontitis risk in men. Eur. J.
bacteremia after chewing, tooth brushing and scaling in individuals Epidemiol., 2003, 18(9), 891-898.
with periodontal inflammation. J. Clin. Periodontol., 2006, 33(6), http://dx.doi.org/10.1023/A:1025622815579 PMID: 14561049
401-407. [68] Gortmaker, S.L.; Dietz, W.H., Jr; Cheung, L.W. Inactivity, diet, and
http://dx.doi.org/10.1111/j.1600-051X.2006.00924.x PMID: the fattening of America. J. Am. Diet. Assoc., 1990, 90(9), 1247-
16677328 1252, 1255.
[54] Lafon, A.; Tala, S.; Ahossi, V.; Perrin, D.; Giroud, M.; Béjot, Y. PMID: 2398216
Association between periodontal disease and non-fatal ischemic [69] Ching, P.L.; Willett, W.C.; Rimm, E.B.; Colditz, G.A.; Gortmaker,
stroke: a case-control study. Acta Odontol. Scand., 2014, 72(8), S.L.; Stampfer, M.J. Activity level and risk of overweight in male
687-693. health professionals. Am. J. Public Health, 1996, 86(1), 25-30.
http://dx.doi.org/10.3109/00016357.2014.898089 PMID: 24720864 http://dx.doi.org/10.2105/AJPH.86.1.25 PMID: 8561237
[55] Sim, S.J.; Kim, H.D.; Moon, J.Y.; Zavras, A.I.; Zdanowicz, J.; Jang, [70] Khader, Y.S.; Dauod, A.S.; El-Qaderi, S.S.; Alkafajei, A.; Batayha,
S.J.; Jin, B.H.; Bae, K.H.; Paik, D.I.; Douglass, C.W. Periodontitis W.Q. Periodontal status of diabetics compared with nondiabetics: a
and the risk for non-fatal stroke in Korean adults. J. Periodontol., meta-analysis. J. Diabetes Complications, 2006, 20(1), 59-68.
2008, 79(9), 1652-1658. http://dx.doi.org/10.1016/j.jdiacomp.2005.05.006 PMID: 16389170
http://dx.doi.org/10.1902/jop.2008.080015 PMID: 18771365 [71] D’Aiuto, F.; Sabbah, W.; Netuveli, G.; Donos, N.; Hingorani, A.D.;
[56] Pradeep, A.R.; Hadge, P.; Arjun Raju, P.; Shetty, S.R.; Shareef, K.; Deanfield, J.; Tsakos, G. Association of the metabolic syndrome
Guruprasad, C.N. Periodontitis as a risk factor for cerebrovascular with severe periodontitis in a large U.S. population-based survey. J.
accident: a case-control study in the Indian population. J. Clin. Endocrinol. Metab., 2008, 93(10), 3989-3994.
Periodontal Res., 2010, 45(2), 223-228. http://dx.doi.org/10.1210/jc.2007-2522 PMID: 18682518
http://dx.doi.org/10.1111/j.1600-0765.2009.01220.x PMID: [72] Khader, Y.; Khassawneh, B.; Obeidat, B.; Hammad, M.; El-Salem,
19778330 K.; Bawadi, H.; Al-akour, N. Periodontal status of patients with
[57] Dörfer, C.E.; Becher, H.; Ziegler, C.M.; Kaiser, C.; Lutz, R.; Jörss, metabolic syndrome compared to those without metabolic
D.; Lichy, C.; Buggle, F.; Bültmann, S.; Preusch, M.; Grau, A.J. syndrome. J. Periodontol., 2008, 79(11), 2048-2053.
The association of gingivitis and periodontitis with ischemic stroke. http://dx.doi.org/10.1902/jop.2008.080022 PMID: 18980512
J. Clin. Periodontol., 2004, 31(5), 396-401. [73] Shimazaki, Y.; Saito, T.; Yonemoto, K.; Kiyohara, Y.; Iida, M.;
http://dx.doi.org/10.1111/j.1600-051x.2004.00579.x PMID: Yamashita, Y. Relationship of metabolic syndrome to periodontal
15086623 disease in Japanese women: the Hisayama Study. J. Dent. Res.,
[58] Grau, A.J.; Becher, H.; Ziegler, C.M.; Lichy, C.; Buggle, F.; Kaiser, 2007, 86(3), 271-275.
C.; Lutz, R.; Bültmann, S.; Preusch, M.; Dörfer, C.E. Periodontal http://dx.doi.org/10.1177/154405910708600314 PMID: 17314261
disease as a risk factor for ischemic stroke. Stroke, 2004, 35(2), 496- [74] Cutler, C.W.; Shinedling, E.A.; Nunn, M.; Jotwani, R.; Kim, B.O.;
501. Nares, S.; Iacopino, A.M. Association between periodontitis and
http://dx.doi.org/10.1161/01.STR.0000110789.20526.9D PMID: hyperlipidemia: cause or effect? J. Periodontol., 1999, 70(12),
14707235 1429-1434.
[59] Jimenez, M.; Krall, E.A.; Garcia, R.I.; Vokonas, P.S.; Dietrich, T. http://dx.doi.org/10.1902/jop.1999.70.12.1429 PMID: 10632517
Periodontitis and incidence of cerebrovascular disease in men. Ann. [75] Nishimura, F.; Murayama, Y. Periodontal inflammation and insulin
Neurol., 2009, 66(4), 505-512. resistance--lessons from obesity. J. Dent. Res., 2001, 80(8), 1690-
http://dx.doi.org/10.1002/ana.21742 PMID: 19847898 1694.
[60] Wu, T.; Trevisan, M.; Genco, R.J.; Dorn, J.P.; Falkner, K.L.; http://dx.doi.org/10.1177/00220345010800080201 PMID:
Sempos, C.T. Periodontal disease and risk of cerebrovascular 11669476
disease: the first national health and nutrition examination survey [76] Al-Zahrani, M.S.; Borawski, E.A.; Bissada, N.F. Periodontitis and
and its follow-up study. Arch. Intern. Med., 2000, 160(18), 2749- three health-enhancing behaviors: maintaining normal weight,
2755. engaging in recommended level of exercise, and consuming a high-
http://dx.doi.org/10.1001/archinte.160.18.2749 PMID: 11025784 quality diet. J. Periodontol., 2005, 76(8), 1362-1366.
[61] Leira, Y.; Seoane, J.; Blanco, M.; Rodríguez-Yáñez, M.; http://dx.doi.org/10.1902/jop.2005.76.8.1362 PMID: 16101370
Takkouche, B.; Blanco, J.; Castillo, J. Association between [77] Chaffee, B.W.; Weston, S.J. Association between chronic
periodontitis and ischemic stroke: a systematic review and meta- periodontal disease and obesity: a systematic review and meta-
analysis. Eur. J. Epidemiol., 2017, 32(1), 43-53. analysis. J. Periodontol., 2010, 81(12), 1708-1724.
http://dx.doi.org/10.1007/s10654-016-0170-6 PMID: 27300352 http://dx.doi.org/10.1902/jop.2010.100321 PMID: 20722533
[62] Suvan, J.; D’Aiuto, F.; Moles, D.R.; Petrie, A.; Donos, N. [78] Marchetti, E.; Monaco, A.; Procaccini, L.; Mummolo, S.; Gatto, R.;
Association between overweight/obesity and periodontitis in adults. Tetè, S.; Baldini, A.; Tecco, S.; Marzo, G. Periodontal disease: the
A systematic review. Obes. Rev., 2011, 12(5), e381-e404. influence of metabolic syndrome. Nutr. Metab. (Lond.), 2012, 9(1),
http://dx.doi.org/10.1111/j.1467-789X.2010.00808.x PMID: 88.
21348914 http://dx.doi.org/10.1186/1743-7075-9-88 PMID: 23009606
[63] Saito, T.; Shimazaki, Y.; Sakamoto, M. Obesity and periodontitis. [79] Desvarieux, M.; Demmer, R.T.; Jacobs, D.R., Jr; Rundek, T.;
N. Engl. J. Med., 1998, 339(7), 482-483. Boden-Albala, B.; Sacco, R.L.; Papapanou, P.N. Periodontal
http://dx.doi.org/10.1056/NEJM199808133390717 PMID: 9705695 bacteria and hypertension: the oral infections and vascular disease
[64] Saito, T.; Shimazaki, Y.; Koga, T.; Tsuzuki, M.; Ohshima, A. epidemiology study (INVEST). J. Hypertens., 2010, 28(7), 1413-
Relationship between upper body obesity and periodontitis. J. Dent. 1421.
Res., 2001, 80(7), 1631-1636. http://dx.doi.org/10.1097/HJH.0b013e328338cd36 PMID:
http://dx.doi.org/10.1177/00220345010800070701 PMID: 20453665
11597023
Periodontal Pathogens and Neuropsychiatric Health Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 1379

[80] Pazos, P.; Leira, Y.; Domínguez, C.; Pías-Peleteiro, J.M.; Blanco, http://dx.doi.org/10.1016/j.neurobiolaging.2014.10.038 PMID:
J.; Aldrey, J.M. Association between periodontal disease and 25491073
dementia: A literature review. Neurologia, 2018, 33(9), 602-613. [94] Noble, J.M.; Borrell, L.N.; Papapanou, P.N.; Elkind, M.S.;
[English Edition]. Scarmeas, N.; Wright, C.B. Periodontitis is associated with
http://dx.doi.org/10.1016/j.nrl.2016.07.013 PMID: 27780615 cognitive impairment among older adults: analysis of NHANES-III.
[81] Leira, Y.; Domínguez, C.; Seoane, J.; Seoane-Romero, J.; Pías- J. Neurol. Neurosurg. Psychiatry, 2009, 80(11), 1206-1211.
Peleteiro, J.M.; Takkouche, B.; Blanco, J.; Aldrey, J.M. Is http://dx.doi.org/10.1136/jnnp.2009.174029 PMID: 19419981
periodontal disease associated with Alzheimer’s disease? A [95] Kaye, E.K.; Valencia, A.; Baba, N.; Spiro, A., III; Dietrich, T.;
systematic review with meta-analysis. Neuroepidemiology, 2017, Garcia, R.I. Tooth loss and periodontal disease predict poor
48(1-2), 21-31. cognitive function in older men. J. Am. Geriatr. Soc., 2010, 58(4),
http://dx.doi.org/10.1159/000458411 PMID: 28219071 713-718.
[82] Tonsekar, P.P.; Jiang, S.S.; Yue, G. Periodontal disease, tooth loss http://dx.doi.org/10.1111/j.1532-5415.2010.02788.x PMID:
and dementia: Is there a link? A systematic review. Gerodontology, 20398152
2017, 34(2), 151-163. [96] Gatz, M.; Mortimer, J.A.; Fratiglioni, L.; Johansson, B.; Berg, S.;
http://dx.doi.org/10.1111/ger.12261 PMID: 28168759 Reynolds, C.A.; Pedersen, N.L. Potentially modifiable risk factors
[83] Maldonado, A.; Laugisch, O.; Bürgin, W.; Sculean, A.; Eick, S. for dementia in identical twins. Alzheimers Dement., 2006, 2(2),
Clinical periodontal variables in patients with and without 110-117.
dementia-a systematic review and meta-analysis. Clin. Oral http://dx.doi.org/10.1016/j.jalz.2006.01.002 PMID: 19595867
Investig., 2018, 22(7), 2463-2474. [97] Poole, S.; Singhrao, S.K.; Chukkapalli, S.; Rivera, M.; Velsko, I.;
http://dx.doi.org/10.1007/s00784-018-2523-x PMID: 29934798 Kesavalu, L.; Crean, S. Active invasion of Porphyromonas
[84] Takeuchi, K.; Ohara, T.; Furuta, M.; Takeshita, T.; Shibata, Y.; gingivalis and infection-induced complement activation in ApoE-/-
Hata, J.; Yoshida, D.; Yamashita, Y.; Ninomiya, T. Tooth loss and mice brains. J. Alzheimers Dis., 2015, 43(1), 67-80.
risk of dementia in the community: the Hisayama study. J. Am. http://dx.doi.org/10.3233/JAD-140315 PMID: 25061055
Geriatr. Soc., 2017, 65(5), e95-e100. [98] Ishida, N.; Ishihara, Y.; Ishida, K.; Tada, H.; Funaki-Kato, Y.;
http://dx.doi.org/10.1111/jgs.14791 PMID: 28272750 Hagiwara, M.; Ferdous, T.; Abdullah, M.; Mitani, A.; Michikawa,
[85] Lee, Y.L.; Hu, H.Y.; Huang, L.Y.; Chou, P.; Chu, D. Periodontal M.; Matsushita, K. Periodontitis induced by bacterial infection
disease associated with higher risk of dementia: population-based exacerbates features of Alzheimer’s disease in transgenic mice. NPJ
cohort study in Taiwan. J. Am. Geriatr. Soc., 2017, 65(9), 1975- Aging Mech. Dis., 2017, 3, 15.
1980. http://dx.doi.org/10.1038/s41514-017-0015-x PMID: 29134111
http://dx.doi.org/10.1111/jgs.14944 PMID: 28598507 [99] Poole, S.; Singhrao, S.K.; Kesavalu, L.; Curtis, M.A.; Crean, S.
[86] Ide, M.; Harris, M.; Stevens, A.; Sussams, R.; Hopkins, V.; Determining the presence of periodontopathic virulence factors in
Culliford, D.; Fuller, J.; Ibbett, P.; Raybould, R.; Thomas, R.; short-term postmortem Alzheimer’s disease brain tissue. J.
Puenter, U.; Teeling, J.; Perry, V.H.; Holmes, C. Periodontitis and Alzheimers Dis., 2013, 36(4), 665-677.
cognitive decline in alzheimer’s disease. PLoS One, 2016, 11(3), http://dx.doi.org/10.3233/JAD-121918 PMID: 23666172
e0151081. [100] Singhrao, S.K.; Harding, A.; Poole, S.; Kesavalu, L.; Crean, S.
http://dx.doi.org/10.1371/journal.pone.0151081 PMID: 26963387 Porphyromonas gingivalis periodontal infection and its putative
[87] Kaushal, V.; Dye, R.; Pakavathkumar, P.; Foveau, B.; Flores, J.; links with alzheimer’s disease. Mediators Inflamm., 2015,
Hyman, B.; Ghetti, B.; Koller, B.H.; LeBlanc, A.C. Neuronal 2015137357
NLRP1 inflammasome activation of Caspase-1 coordinately http://dx.doi.org/10.1155/2015/137357 PMID: 26063967
regulates inflammatory interleukin-1-beta production and axonal [101] Dominy, S.S.; Lynch, C.; Ermini, F.; Benedyk, M.; Marczyk, A.;
degeneration-associated Caspase-6 activation. Cell Death Differ., Konradi, A.; Nguyen, M.; Haditsch, U.; Raha, D.; Griffin, C.;
2015, 22(10), 1676-1686. Holsinger, L.J.; Arastu-Kapur, S.; Kaba, S.; Lee, A.; Ryder, M.I.;
http://dx.doi.org/10.1038/cdd.2015.16 PMID: 25744023 Potempa, B.; Mydel, P.; Hellvard, A.; Adamowicz, K.; Hasturk, H.;
[88] Wyss-Coray, T.; Rogers, J. Inflammation in Alzheimer disease-a Walker, G.D.; Reynolds, E.C.; Faull, R.L.M.; Curtis, M.A.;
brief review of the basic science and clinical literature. Cold Spring Dragunow, M.; Potempa, J. Porphyromonas gingivalis in
Harb. Perspect. Med., 2012, 2(1), a006346. Alzheimer’s disease brains: Evidence for disease causation and
http://dx.doi.org/10.1101/cshperspect.a006346 PMID: 22315714 treatment with small-molecule inhibitors. Sci. Adv., 2019, 5(1),
[89] Spitzer, P.; Condic, M.; Herrmann, M.; Oberstein, T.J.; Scharin- eaau3333.
Mehlmann, M.; Gilbert, D.F.; Friedrich, O.; Grömer, T.; Kornhuber, http://dx.doi.org/10.1126/sciadv.aau3333 PMID: 30746447
J.; Lang, R.; Maler, J.M. Amyloidogenic amyloid-β-peptide variants [102] Chi, L.; Cheng, X.; He, X.; Sun, J.; Liang, F.; Pei, Z.; Teng, W.
induce microbial agglutination and exert antimicrobial activity. Sci. Increased cortical infarction and neuroinflammation in ischemic
Rep., 2016, 6, 32228. stroke mice with experimental periodontitis. Neuroreport, 2019,
http://dx.doi.org/10.1038/srep32228 PMID: 27624303 30(6), 428-433.
[90] Soscia, S.J.; Kirby, J.E.; Washicosky, K.J.; Tucker, S.M.; Ingelsson, http://dx.doi.org/10.1097/WNR.0000000000001220 PMID:
M.; Hyman, B.; Burton, M.A.; Goldstein, L.E.; Duong, S.; Tanzi, 30829959
R.E.; Moir, R.D. The Alzheimer’s disease-associated amyloid beta- [103] Angelillo, I.F.; Nobile, C.G.; Pavia, M.; De Fazio, P.; Puca, M.;
protein is an antimicrobial peptide. PLoS One, 2010, 5(3), e9505. Amati, A. Dental health and treatment needs in institutionalized
http://dx.doi.org/10.1371/journal.pone.0009505 PMID: 20209079 psychiatric patients in Italy. Community Dent. Oral Epidemiol.,
[91] Kumar, D.K.; Choi, S.H.; Washicosky, K.J.; Eimer, W.A.; Tucker, 1995, 23(6), 360-364.
S.; Ghofrani, J.; Lefkowitz, A.; McColl, G.; Goldstein, L.E.; Tanzi, http://dx.doi.org/10.1111/j.1600-0528.1995.tb00263.x PMID:
R.E.; Moir, R.D. Amyloid-β peptide protects against microbial 8681519
infection in mouse and worm models of Alzheimer’s disease. Sci. [104] Burchell, A.; Fernbacher, S.; Lewis, R.; Neil, A. “Dental as
Transl. Med., 2016, 8(340), 340ra72. Anything” inner south community health service dental outreach to
http://dx.doi.org/10.1126/scitranslmed.aaf1059 PMID: 27225182 people with a mental illness. Aust. J. Prim. Health, 2006, 12(2), 75-
[92] Stein, P.S.; Desrosiers, M.; Donegan, S.J.; Yepes, J.F.; Kryscio, R.J. 82.
Tooth loss, dementia and neuropathology in the Nun study. J. Am. http://dx.doi.org/10.1071/PY06025
Dent. Assoc., 2007, 138(10), 1314-1322. [105] Rekha, R.; Hiremath, S.S. Oral health status and treatment
http://dx.doi.org/10.14219/jada.archive.2007.0046 PMID: 17908844 requirments of confectionery workers in Bangalore city. A
[93] Kamer, A.R.; Pirraglia, E.; Tsui, W.; Rusinek, H.; Vallabhajosula, comparative study. Indian J. Dent. Res., 2002, 13(3-4), 161-165.
S.; Mosconi, L.; Yi, L.; McHugh, P.; Craig, R.G.; Svetcov, S.; PMID: 12765096
Linker, R.; Shi, C.; Glodzik, L.; Williams, S.; Corby, P.; Saxena, D.; [106] Tang, W.K.; Sun, F.C.; Ungvari, G.S.; O’Donnell, D. Oral health of
de Leon, M.J. Periodontal disease associates with higher brain psychiatric in-patients in Hong Kong. Int. J. Soc. Psychiatry, 2004,
amyloid load in normal elderly. Neurobiol. Aging, 2015, 36(2), 627- 50(2), 186-191.
633. http://dx.doi.org/10.1177/0020764004043134 PMID: 15293435
1380 Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 Wadhawan et al.

[107] Kisely, S.; Baghaie, H.; Lalloo, R.; Siskind, D.; Johnson, N.W. A [123] Cormac, I.; Jenkins, P. Understanding the importance of oral health
systematic review and meta-analysis of the association between in psychiatric patients. Adv. Psychiatr. Treat., 1999, 5(1), 53-60.
poor oral health and severe mental illness. Psychosom. Med., 2015, http://dx.doi.org/10.1192/apt.5.1.53
77(1), 83-92. [124] American Psychiatric Association. Diagnostic and Statistical
http://dx.doi.org/10.1097/PSY.0000000000000135 PMID: Manual of Mental Disorders, 5th ed.; American Psychiatric
25526527 Association: Washington, DC, 2013.
[108] Singh, A.; Mittal, P.; Goel, P.; Purohit, B.M.; Thukral, R. Severity [125] Beck, A.T. Cognitive therapy. A 30-year retrospective. Am.
of illness and extra pyramidal symptoms as predictors for oral Psychol., 1991, 46(4), 368-375.
diseases among patients with schizophrenia. Acta Odontol. Scand., http://dx.doi.org/10.1037/0003-066X.46.4.368 PMID: 2048795
2017, 75(3), 220-226. [126] O’Dowd, L.K.; Durham, J.; McCracken, G.I.; Preshaw, P.M.
http://dx.doi.org/10.1080/00016357.2017.1278789 PMID: Patients’ experiences of the impact of periodontal disease. J. Clin.
28116993 Periodontol., 2010, 37(4), 334-339.
[109] Shetty, S.; Bose, A. Schizophrenia and periodontal disease: An oro- http://dx.doi.org/10.1111/j.1600-051X.2010.01545.x PMID:
neural connection? A cross-sectional epidemiological study. J. 20447256
Indian Soc. Periodontol., 2014, 18(1), 69-73. [127] Bardow, A.; Nyvad, B.; Nauntofte, B. Relationships between
http://dx.doi.org/10.4103/0972-124X.128222 PMID: 24744548 medication intake, complaints of dry mouth, salivary flow rate and
[110] Cunha, F.A.; Cota, L.O.M.; Cortelli, S.C.; Miranda, T.B.; Neves, composition, and the rate of tooth demineralization in situ. Arch.
F.S.; Cortelli, J.R.; Costa, F.O. Periodontal condition and levels of Oral Biol., 2001, 46(5), 413-423.
bacteria associated with periodontitis in individuals with bipolar http://dx.doi.org/10.1016/S0003-9969(01)00003-6 PMID: 11286806
affective disorders: A case-control study. J. Periodontal Res., 2019, [128] Lewis, S.; Jagger, R.G.; Treasure, E. The oral health of psychiatric
54(1), 63-72. in-patients in South Wales. Spec. Care Dentist., 2001, 21(5), 182-
http://dx.doi.org/10.1111/jre.12605 PMID: 30207388 186.
[111] Gurbuz Oflezer, O.; Altinbas, K.; Delice, M.; Oflezer, C.; Kurt, E. http://dx.doi.org/10.1111/j.1754-4505.2001.tb00252.x PMID:
Oral health among patients with bipolar disorder. Oral Health Prev. 11803642
Dent., 2018, 16(6), 509-516. [129] Ramon, T.; Grinshpoon, A.; Zusman, S.P.; Weizman, A. Oral health
PMID: 30574605 and treatment needs of institutionalized chronic psychiatric patients
[112] Kopycka-Kedzierawski, D.T.; Li, D.; Xiao, J.; Billings, R.J.; Dye, in Israel. Eur. Psychiatry, 2003, 18(3), 101-105.
T.D. Association of periodontal disease with depression and adverse http://dx.doi.org/10.1016/S0924-9338(03)00023-3 PMID: 12763294
birth outcomes: Results from the Perinatal database; Finger Lakes [130] Wadhawan, A.; Daue, M.L.; Dagdag, A.; Makkar, H.; Ryan, K.A.;
region, New York State. PLoS One, 2019, 14(4), e0215440. Gragnoli, C.; Postolache, T.T. S144. Obesity is associated with
http://dx.doi.org/10.1371/journal.pone.0215440 PMID: 30998794 anhedonia only in the younger amish women. Biol. Psychiatry,
[113] Nascimento, G.G.; Gastal, M.T.; Leite, F.R.M.; Quevedo, L.A.; 2019, 85(10), S352.
Peres, K.G.; Peres, M.A.; Horta, B.L.; Barros, F.C.; Demarco, F.F. http://dx.doi.org/10.1016/j.biopsych.2019.03.895
Is there an association between depression and periodontitis? A [131] Postolache, T.T.; Del Bosque-Plata, L.; Jabbour, S.; Vergare, M.;
birth cohort study. J. Clin. Periodontol., 2019, 46(1), 31-39. Wu, R.; Gragnoli, C. Co-shared genetics and possible risk gene
PMID: 30499588 pathway partially explain the comorbidity of schizophrenia, major
[114] Dagdag, A.; Reynolds, M.A.; Daue, M.; Wadhawan, A.; Nijjar, G.; depressive disorder, type 2 diabetes, and metabolic syndrome. Am.
Ryan, K.A.; Fuchs, D.; Mitchell, B.; Postolache, T.T. F156. J. Med. Genet. B. Neuropsychiatr. Genet., 2019, 180(3), 186-203.
Anhedonia and Hopelessness/Dysphoria Associated With Tooth http://dx.doi.org/10.1002/ajmg.b.32712 PMID: 30729689
Loss in the Old Order Amish: Gender Differences and Neopterin [132] Marcotte, H.; Lavoie, M.C. Oral microbial ecology and the role of
Levels-Mediator or Confounder? Biol. Psychiatry, 2018, 83(9), salivary immunoglobulin A. Microbiol. Mol. Biol. Rev., 1998,
S298-S299. 62(1), 71-109.
http://dx.doi.org/10.1016/j.biopsych.2018.02.770 http://dx.doi.org/10.1128/MMBR.62.1.71-109.1998 PMID:
[115] Warren, K.R.; Postolache, T.T.; Groer, M.E.; Pinjari, O.; Kelly, 9529888
D.L.; Reynolds, M.A. Role of chronic stress and depression in [133] Pennisi, E. A mouthful of microbes. Science, 2005, 307(5717),
periodontal diseases. Periodontol. 2000, 2014, 64(1), 127-138. 1899-1901.
http://dx.doi.org/10.1111/prd.12036 PMID: 24320960 http://dx.doi.org/10.1126/science.307.5717.1899 PMID: 15790840
[116] Alkan, A.; Cakmak, O.; Yilmaz, S.; Cebi, T.; Gurgan, C. [134] Aas, J.A.; Paster, B.J.; Stokes, L.N.; Olsen, I.; Dewhirst, F.E.
Relationship between psychological factors and oral health status Defining the normal bacterial flora of the oral cavity. J. Clin.
and behaviours. Oral Health Prev. Dent., 2015, 13(4), 331-339. Microbiol., 2005, 43(11), 5721-5732.
PMID: 25197739 http://dx.doi.org/10.1128/JCM.43.11.5721-5732.2005 PMID:
[117] Little, J.W. Dental implications of mood disorders. Gen. Dent., 16272510
2004, 52(5), 442-450. [135] Paster, B.J.; Olsen, I.; Aas, J.A.; Dewhirst, F.E. The breadth of
PMID: 15544223 bacterial diversity in the human periodontal pocket and other oral
[118] Clark, D.B. Dental care for the patient with bipolar disorder. J. Can. sites. Periodontol. 2000, 2006, 42, 80-87.
Dent. Assoc., 2003, 69(1), 20-24. http://dx.doi.org/10.1111/j.1600-0757.2006.00174.x PMID:
PMID: 12556265 16930307
[119] Milosevic, A. Eating disorders and the dentist. Br. Dent. J., 1999, [136] Eloe-Fadrosh, E.A.; Rasko, D.A. The human microbiome: from
186(3), 109-113. symbiosis to pathogenesis. Annu. Rev. Med., 2013, 64, 145-163.
http://dx.doi.org/10.1038/sj.bdj.4800036 PMID: 10101906 http://dx.doi.org/10.1146/annurev-med-010312-133513 PMID:
[120] Bretz, W.A. Oral profiles of bulimic women: Diagnosis and 23327521
management. What is the evidence? J. Evid. Based Dent. Pract., [137] Socransky, S.S.; Haffajee, A.D.; Cugini, M.A.; Smith, C.; Kent,
2002, 2(4), 267-272. R.L., Jr Microbial complexes in subgingival plaque. J. Clin.
http://dx.doi.org/10.1016/S1532-3382(02)70078-X PMID: Periodontol., 1998, 25(2), 134-144.
22287937 http://dx.doi.org/10.1111/j.1600-051X.1998.tb02419.x PMID:
[121] Page, M.M. Psychotropic drugs and dentistry. Aust. Prescr., 2007, 9495612
30(4), 98-101. [138] Socransky, S.S.; Haffajee, A.D. Dental biofilms: difficult
http://dx.doi.org/10.18773/austprescr.2007.059 therapeutic targets. Periodontol 2000, 2002, 28, 12-55.
[122] Lalloo, R.; Kisely, S.; Amarasinghe, H.; Perera, R.; Johnson, N. http://dx.doi.org/10.1034/j.1600-0757.2002.280102.x PMID:
Oral health of patients on psychotropic medications: a study of 12013340
outpatients in Queensland. Australas. Psychiatry, 2013, 21(4), 338- [139] Carrouel, F.; Viennot, S.; Santamaria, J.; Veber, P.; Bourgeois, D.
342. Quantitative molecular detection of 19 major pathogens in the
http://dx.doi.org/10.1177/1039856213486308 PMID: 23671224 interdental biofilm of periodontally healthy young adults. Front.
Microbiol., 2016, 7, 840.
Periodontal Pathogens and Neuropsychiatric Health Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 1381

http://dx.doi.org/10.3389/fmicb.2016.00840 PMID: 27313576 [154] Holt, S.C.; Kesavalu, L.; Walker, S.; Genco, C.A. Virulence factors
[140] Socransky, S.S.; Haffajee, A.D. Periodontal microbial ecology. of Porphyromonas gingivalis. Periodontol. 2000, 1999, 20, 168-
Periodontol 2000, 2005, 38, 135-187. 238.
http://dx.doi.org/10.1111/j.1600-0757.2005.00107.x PMID: http://dx.doi.org/10.1111/j.1600-0757.1999.tb00162.x PMID:
15853940 10522227
[141] Yang, H.W.; Huang, Y.F.; Chou, M.Y. Occurrence of [155] Lamont, R.J.; Jenkinson, H.F. Life below the gum line: pathogenic
Porphyromonas gingivalis and Tannerella forsythensis in mechanisms of Porphyromonas gingivalis. Microbiol. Mol. Biol.
periodontally diseased and healthy subjects. J. Periodontol., 2004, Rev., 1998, 62(4), 1244-1263.
75(8), 1077-1083. http://dx.doi.org/10.1128/MMBR.62.4.1244-1263.1998 PMID:
http://dx.doi.org/10.1902/jop.2004.75.8.1077 PMID: 15455734 9841671
[142] Hwang, A.M.; Stoupel, J.; Celenti, R.; Demmer, R.T.; Papapanou, [156] Curtis, M.A.; Aduse-Opoku, J.; Rangarajan, M. Cysteine proteases
P.N. Serum antibody responses to periodontal microbiota in chronic of Porphyromonas gingivalis. Crit. Rev. Oral Biol. Med., 2001,
and aggressive periodontitis: a postulate revisited. J. Periodontol., 12(3), 192-216.
2014, 85(4), 592-600. http://dx.doi.org/10.1177/10454411010120030101 PMID:
http://dx.doi.org/10.1902/jop.2013.130172 PMID: 23725029 11497373
[143] Shah, H.; Collins, M. Proposal for reclassification of bacteroides [157] Travis, J.; Pike, R.; Imamura, T.; Potempa, J. Porphyromonas
asaccharolyticus, bacteroides gingivalis, and bacteroides gingivalis proteinases as virulence factors in the development of
endodontalis in a new genus, porphyromonas. Int. J. Syst. Evol. periodontitis. J. Periodontal Res., 1997, 32(1 Pt 2), 120-125.
Microbiol., 1988, 38(1), 128-131. http://dx.doi.org/10.1111/j.1600-0765.1997.tb01392.x PMID:
[144] Bostanci, N.; Belibasakis, G.N. Porphyromonas gingivalis: an 9085221
invasive and evasive opportunistic oral pathogen. FEMS Microbiol. [158] de Diego, I.; Veillard, F.; Sztukowska, M.N.; Guevara, T.; Potempa,
Lett., 2012, 333(1), 1-9. B.; Pomowski, A.; Huntington, J.A.; Potempa, J.; Gomis-Rüth, F.X.
http://dx.doi.org/10.1111/j.1574-6968.2012.02579.x PMID: Structure and mechanism of cysteine peptidase gingipain K (Kgp), a
22530835 major virulence factor of Porphyromonas gingivalis in periodontitis.
[145] Bodet, C.; Chandad, F.; Grenier, D. [Pathogenic potential of J. Biol. Chem., 2014, 289(46), 32291-32302.
Porphyromonas gingivalis, Treponema denticola and Tannerella http://dx.doi.org/10.1074/jbc.M114.602052 PMID: 25266723
forsythia, the red bacterial complex associated with periodontitis]. [159] Potempa, J.; Banbula, A.; Travis, J. Role of bacterial proteinases in
Pathol. Biol. (Paris), 2007, 55(3-4), 154-162. matrix destruction and modulation of host responses. Periodontol
http://dx.doi.org/10.1016/j.patbio.2006.07.045 PMID: 17049750 2000, 2000, 24, 153-192.
[146] Casarin, R.C.; Ribeiro, Edel.P.; Mariano, F.S.; Nociti, F.H., Jr; http://dx.doi.org/10.1034/j.1600-0757.2000.2240108.x PMID:
Casati, M.Z.; Gonçalves, R.B. Levels of Aggregatibacter 11276866
actinomycetemcomitans, Porphyromonas gingivalis, inflammatory [160] Kristoffersen, A.K.; Solli, S.J.; Nguyen, T.D.; Enersen, M.
cytokines and species-specific immunoglobulin G in generalized Association of the rgpB gingipain genotype to the major fimbriae
aggressive and chronic periodontitis. J. Periodontal Res., 2010, (fimA) genotype in clinical isolates of the periodontal pathogen
45(5), 635-642. Porphyromonas gingivalis. J. Oral Microbiol., 2015, 7, 29124.
http://dx.doi.org/10.1111/j.1600-0765.2010.01278.x PMID: http://dx.doi.org/10.3402/jom.v7.29124 PMID: 26387644
20546109 [161] Dubin, G.; Koziel, J.; Pyrc, K.; Wladyka, B.; Potempa, J. Bacterial
[147] Mahanonda, R.; Seymour, G.J.; Powell, L.W.; Good, M.F.; proteases in disease - role in intracellular survival, evasion of
Halliday, J.W. Effect of initial treatment of chronic inflammatory coagulation/ fibrinolysis innate defenses, toxicoses and viral
periodontal disease on the frequency of peripheral blood T- infections. Curr. Pharm. Des., 2013, 19(6), 1090-1113.
lymphocytes specific to periodontopathic bacteria. Oral Microbiol. http://dx.doi.org/10.2174/1381612811319060011 PMID: 23016681
Immunol., 1991, 6(4), 221-227. [162] Andrian, E.; Grenier, D.; Rouabhia, M. In vitro models of tissue
http://dx.doi.org/10.1111/j.1399-302X.1991.tb00481.x PMID: penetration and destruction by Porphyromonas gingivalis. Infect.
1667435 Immun., 2004, 72(8), 4689-4698.
[148] Moore, W.E.; Moore, L.H.; Ranney, R.R.; Smibert, R.M.; http://dx.doi.org/10.1128/IAI.72.8.4689-4698.2004 PMID:
Burmeister, J.A.; Schenkein, H.A. The microflora of periodontal 15271930
sites showing active destructive progression. J. Clin. Periodontol., [163] Imamura, T.; Travis, J.; Potempa, J. The biphasic virulence
1991, 18(10), 729-739. activities of gingipains: activation and inactivation of host proteins.
http://dx.doi.org/10.1111/j.1600-051X.1991.tb00064.x PMID: Curr. Protein Pept. Sci., 2003, 4(6), 443-450.
1752997 http://dx.doi.org/10.2174/1389203033487027 PMID: 14683429
[149] Schmidt, J.; Jentsch, H.; Stingu, C.S.; Sack, U. General immune [164] Haraguchi, A.; Miura, M.; Fujise, O.; Hamachi, T.; Nishimura, F.
status and oral microbiology in patients with different forms of Porphyromonas gingivalis gingipain is involved in the detachment
periodontitis and healthy control subjects. PLoS One, 2014, 9(10), and aggregation of Aggregatibacter actinomycetemcomitans
e109187. biofilm. Mol. Oral Microbiol., 2014, 29(3), 131-143.
http://dx.doi.org/10.1371/journal.pone.0109187 PMID: 25299619 http://dx.doi.org/10.1111/omi.12051 PMID: 24661327
[150] Baek, K.J.; Ji, S.; Kim, Y.C.; Choi, Y. Association of the invasion [165] Bao, K.; Belibasakis, G.N.; Thurnheer, T.; Aduse-Opoku, J.; Curtis,
ability of Porphyromonas gingivalis with the severity of M.A.; Bostanci, N. Role of Porphyromonas gingivalis gingipains in
periodontitis. Virulence, 2015, 6(3), 274-281. multi-species biofilm formation. BMC Microbiol., 2014, 14, 258.
http://dx.doi.org/10.1080/21505594.2014.1000764 PMID: http://dx.doi.org/10.1186/s12866-014-0258-7 PMID: 25270662
25616643 [166] Sroka, A.; Sztukowska, M.; Potempa, J.; Travis, J.; Genco, C.A.
[151] Dorn, B.R.; Burks, J.N.; Seifert, K.N.; Progulske-Fox, A. Invasion Degradation of host heme proteins by lysine- and arginine-specific
of endothelial and epithelial cells by strains of Porphyromonas cysteine proteinases (gingipains) of Porphyromonas gingivalis. J.
gingivalis. FEMS Microbiol. Lett., 2000, 187(2), 139-144. Bacteriol., 2001, 183(19), 5609-5616.
http://dx.doi.org/10.1111/j.1574-6968.2000.tb09150.x PMID: http://dx.doi.org/10.1128/JB.183.19.5609-5616.2001 PMID:
10856647 11544223
[152] Hajishengallis, G.; Darveau, R.P.; Curtis, M.A. The keystone- [167] Hajishengallis, G.; Abe, T.; Maekawa, T.; Hajishengallis, E.;
pathogen hypothesis. Nat. Rev. Microbiol., 2012, 10(10), 717-725. Lambris, J.D. Role of complement in host-microbe homeostasis of
http://dx.doi.org/10.1038/nrmicro2873 PMID: 22941505 the periodontium. Semin. Immunol., 2013, 25(1), 65-72.
[153] Hajishengallis, G.; Lamont, R.J. Breaking bad: manipulation of the http://dx.doi.org/10.1016/j.smim.2013.04.004 PMID: 23684627
host response by Porphyromonas gingivalis. Eur. J. Immunol., [168] Maekawa, T.; Krauss, J.L.; Abe, T.; Jotwani, R.; Triantafilou, M.;
2014, 44(2), 328-338. Triantafilou, K.; Hashim, A.; Hoch, S.; Curtis, M.A.; Nussbaum, G.;
http://dx.doi.org/10.1002/eji.201344202 PMID: 24338806 Lambris, J.D.; Hajishengallis, G. Porphyromonas gingivalis
manipulates complement and TLR signaling to uncouple bacterial
1382 Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 Wadhawan et al.

clearance from inflammation and promote dysbiosis. Cell Host [182] Beutler, B.; Du, X.; Poltorak, A. Identification of Toll-like receptor
Microbe, 2014, 15(6), 768-778. 4 (Tlr4) as the sole conduit for LPS signal transduction: genetic and
http://dx.doi.org/10.1016/j.chom.2014.05.012 PMID: 24922578 evolutionary studies. J. Endotoxin. Res., 2001, 7(4), 277-280.
[169] Bostanci, N.; Thurnheer, T.; Aduse-Opoku, J.; Curtis, M.A.; [183] Raetz, C.R.H.; Whitfield, C. Lipopolysaccharide endotoxins. Annu.
Zinkernagel, A.S.; Belibasakis, G.N. Porphyromonas gingivalis Rev. Biochem., 2002, 71, 635-700.
regulates TREM-1 in human polymorphonuclear neutrophils via its http://dx.doi.org/10.1146/annurev.biochem.71.110601.135414
gingipains. PLoS One, 2013, 8(10), e75784. PMID: 12045108
http://dx.doi.org/10.1371/journal.pone.0075784 PMID: 24124513 [184] Jain, S.; Darveau, R.P. Contribution of Porphyromonas gingivalis
[170] Belibasakis, G.N.; Bostanci, N.; Reddi, D. Regulation of protease- lipopolysaccharide to periodontitis. Periodontol 2000, 2010, 54(1),
activated receptor-2 expression in gingival fibroblasts and Jurkat T 53-70.
cells by Porphyromonas gingivalis. Cell Biol. Int., 2010, 34(3), 287- http://dx.doi.org/10.1111/j.1600-0757.2009.00333.x PMID:
292. 20712633
http://dx.doi.org/10.1042/CBI20090290 PMID: 19947912 [185] Mühlradt, P.F.; Golecki, J.R. Asymmetrical distribution and
[171] Hamedi, M.; Belibasakis, G.N.; Cruchley, A.T.; Rangarajan, M.; artifactual reorientation of lipopolysaccharide in the outer
Curtis, M.A.; Bostanci, N. Porphyromonas gingivalis culture membrane bilayer of Salmonella typhimurium. Eur. J. Biochem.,
supernatants differentially regulate interleukin-1beta and 1975, 51(2), 343-352.
interleukin-18 in human monocytic cells. Cytokine, 2009, 45(2), 99- http://dx.doi.org/10.1111/j.1432-1033.1975.tb03934.x PMID:
104. 807474
http://dx.doi.org/10.1016/j.cyto.2008.11.005 PMID: 19091595 [186] Hajjar, A.M.; Ernst, R.K.; Tsai, J.H.; Wilson, C.B.; Miller, S.I.
[172] Sheets, S.M.; Potempa, J.; Travis, J.; Casiano, C.A.; Fletcher, H.M. Human Toll-like receptor 4 recognizes host-specific LPS
Gingipains from Porphyromonas gingivalis W83 induce cell modifications. Nat. Immunol., 2002, 3(4), 354-359.
adhesion molecule cleavage and apoptosis in endothelial cells. http://dx.doi.org/10.1038/ni777 PMID: 11912497
Infect. Immun., 2005, 73(3), 1543-1552. [187] Miller, S.I.; Ernst, R.K.; Bader, M.W. LPS, TLR4 and infectious
http://dx.doi.org/10.1128/IAI.73.3.1543-1552.2005 PMID: disease diversity. Nat. Rev. Microbiol., 2005, 3(1), 36-46.
15731052 http://dx.doi.org/10.1038/nrmicro1068 PMID: 15608698
[173] Kinane, J.A.; Benakanakere, M.R.; Zhao, J.; Hosur, K.B.; Kinane, [188] Scott, A.J.; Oyler, B.L.; Goodlett, D.R.; Ernst, R.K. Lipid A
D.F. Porphyromonas gingivalis influences actin degradation within structural modifications in extreme conditions and identification of
epithelial cells during invasion and apoptosis. Cell. Microbiol., unique modifying enzymes to define the Toll-like receptor 4
2012, 14(7), 1085-1096. structure-activity relationship. Biochim. Biophys. Acta Mol. Cell
http://dx.doi.org/10.1111/j.1462-5822.2012.01780.x PMID: Biol. Lipids, 2017, 1862(11), 1439-1450.
22381126 http://dx.doi.org/10.1016/j.bbalip.2017.01.004 PMID: 28108356
[174] Stathopoulou, P.G.; Galicia, J.C.; Benakanakere, M.R.; Garcia, [189] Kumada, H.; Haishima, Y.; Umemoto, T.; Tanamoto, K. Structural
C.A.; Potempa, J.; Kinane, D.F. Porphyromonas gingivalis induce study on the free lipid a isolated from lipopolysaccharide of
apoptosis in human gingival epithelial cells through a gingipain- Porphyromonas gingivalis. J. Bacteriol., 1995, 177(8), 2098-2106.
dependent mechanism. BMC Microbiol., 2009, 9, 107. http://dx.doi.org/10.1128/JB.177.8.2098-2106.1995 PMID: 7721702
http://dx.doi.org/10.1186/1471-2180-9-107 PMID: 19473524 [190] Ogawa, T.; Asai, Y.; Hashimoto, M.; Takeuchi, O.; Kurita, T.;
[175] Guo, Y.; Nguyen, K.A.; Potempa, J. Dichotomy of gingipains action Yoshikai, Y.; Miyake, K.; Akira, S. Cell activation by
as virulence factors: from cleaving substrates with the precision of a Porphyromonas gingivalis lipid A molecule through Toll-like
surgeon’s knife to a meat chopper-like brutal degradation of receptor 4- and myeloid differentiation factor 88-dependent
proteins. Periodontol. 2000, 2010, 54(1), 15-44. signaling pathway. Int. Immunol., 2002, 14(11), 1325-1332.
http://dx.doi.org/10.1111/j.1600-0757.2010.00377.x PMID: http://dx.doi.org/10.1093/intimm/dxf097 PMID: 12407023
20712631 [191] Reife, R.A.; Coats, S.R.; Al-Qutub, M.; Dixon, D.M.; Braham, P.A.;
[176] Travis, J.; Potempa, J. Bacterial proteinases as targets for the Billharz, R.J.; Howald, W.N.; Darveau, R.P. Porphyromonas
development of second-generation antibiotics. Biochim. Biophys. gingivalis lipopolysaccharide lipid A heterogeneity: differential
Acta, 2000, 1477(1-2), 35-50. activities of tetra- and penta-acylated lipid A structures on E-
http://dx.doi.org/10.1016/S0167-4838(99)00278-2 PMID: 10708847 selectin expression and TLR4 recognition. Cell. Microbiol., 2006,
[177] Clatworthy, A.E.; Pierson, E.; Hung, D.T. Targeting virulence: a 8(5), 857-868.
new paradigm for antimicrobial therapy. Nat. Chem. Biol., 2007, http://dx.doi.org/10.1111/j.1462-5822.2005.00672.x PMID:
3(9), 541-548. 16611234
http://dx.doi.org/10.1038/nchembio.2007.24 PMID: 17710100 [192] Herath, T.D.K.; Darveau, R.P.; Seneviratne, C.J.; Wang, C-Y.;
[178] Supuran, C.T.; Scozzafava, A.; Mastrolorenzo, A. Bacterial Wang, Y.; Jin, L. Tetra- and penta-acylated lipid A structures of
proteases: current therapeutic use and future prospects for the Porphyromonas gingivalis LPS differentially activate TLR4-
development of new antibiotics. Expert Opin. Ther. Pat., 2001, mediated NF-κB signal transduction cascade and immuno-
11(2), 221-259. inflammatory response in human gingival fibroblasts. PLoS One,
http://dx.doi.org/10.1517/13543776.11.2.221 2013, 8(3), e58496-e17.
[179] Flemmig, T.F.; Milián, E.; Karch, H.; Klaiber, B. Differential http://dx.doi.org/10.1371/journal.pone.0058496 PMID: 23554896
clinical treatment outcome after systemic metronidazole and [193] Nativel, B.; Couret, D.; Giraud, P.; Meilhac, O. Porphyromonas
amoxicillin in patients harboring Actinobacillus gingivalis lipopolysaccharides act exclusively through TLR4 with a
actinomycetemcomitans and/or Porphyromonas gingivalis. J. Clin. resilience between mouse and human. Sci. Rep., 2017, 7(1), 1-12.
Periodontol., 1998, 25(5), 380-387. DOI: 10.1038/s41598-017-16190-y
http://dx.doi.org/10.1111/j.1600-051X.1998.tb02459.x PMID: [194] Hirschfeld, M.; Ma, Y.; Weis, J. H.; Vogel, S. N.; Weis, J. J.
9650874 Cutting edge: repurification of lipopolysaccharide eliminates
[180] Kadowaki, T.; Baba, A.; Abe, N.; Takii, R.; Hashimoto, M.; signaling through both human and murine toll-like receptor 2. J.
Tsukuba, T.; Okazaki, S.; Suda, Y.; Asao, T.; Yamamoto, K. Immunol., 2000, 165 (2), 618-622
Suppression of pathogenicity of Porphyromonas gingivalis by [195] Hirschfeld, M.; Weis, J.J.; Toshchakov, V.; Salkowski, C.A.; Cody,
newly developed gingipain inhibitors. Mol. Pharmacol., 2004, M.J.; Ward, D.C.; Qureshi, N.; Michalek, S.M.; Vogel, S.N.
66(6), 1599-1606. Signaling by toll-like receptor 2 and 4 agonists results in differential
http://dx.doi.org/10.1124/mol.104.004366 PMID: 15361547 gene expression in murine macrophages. Infect. Immun., 2001,
[181] Raetz, C.R.H. Biochemistry of endotoxins. Annu. Rev. Biochem., 69(3), 1477-1482.
1990, 59, 129-170. http://dx.doi.org/10.1128/IAI.69.3.1477-1482.2001 PMID:
http://dx.doi.org/10.1146/annurev.bi.59.070190.001021 PMID: 11179315
1695830 [196] Darveau, R.P.; Arbabi, S.; Garcia, I.; Bainbridge, B.; Maier, R.V.
Porphyromonas gingivalis lipopolysaccharide is both agonist and
Periodontal Pathogens and Neuropsychiatric Health Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 1383

antagonist for p38 mitogen-activated protein kinase activation. [209] Deleon-Pennell, K.Y.; de Castro Brás, L.E.; Lindsey, M.L.
Infect. Immun., 2002, 70(4), 1867-1873. Circulating Porphyromonas gingivalis lipopolysaccharide resets
http://dx.doi.org/10.1128/IAI.70.4.1867-1873.2002 PMID: cardiac homeostasis in mice through a matrix metalloproteinase-9-
11895949 dependent mechanism. Physiol. Rep., 2013, 1(5), e00079.
[197] Hashimoto, M.; Asai, Y.; Ogawa, T. Separation and structural http://dx.doi.org/10.1002/phy2.79 PMID: 24159380
analysis of lipoprotein in a lipopolysaccharide preparation from [210] Gokyu, M.; Kobayashi, H.; Nanbara, H.; Sudo, T.; Ikeda, Y.; Suda,
Porphyromonas gingivalis. Int. Immunol., 2004, 16(10), 1431-1437. T.; Izumi, Y. Thrombospondin-1 production is enhanced by
http://dx.doi.org/10.1093/intimm/dxh146 PMID: 15326096 Porphyromonas gingivalis lipopolysaccharide in THP-1 cells. PLoS
[198] Darveau, R.P.; Pham, T.T.; Lemley, K.; Reife, R.A.; Bainbridge, One, 2014, 9(12), e115107.
B.W.; Coats, S.R.; Howald, W.N.; Way, S.S.; Hajjar, A.M. http://dx.doi.org/10.1371/journal.pone.0115107 PMID: 25501558
Porphyromonas gingivalis lipopolysaccharide contains multiple [211] Na, H.S.; Lim, E.J.; Jeong, S.Y.; Ryu, M.H.; Park, M.H.; Chung, J.
lipid A species that functionally interact with both toll-like receptors Plasminogen activator inhibitor type 1 expression induced by
2 and 4. Infect. Immun., 2004, 72(9), 5041-5051. lipopolysaccharide of Porphyromonas gingivalis in human gingival
http://dx.doi.org/10.1128/IAI.72.9.5041-5051.2004 PMID: fibroblast. J. Microbiol., 2014, 52(2), 154-160.
15321997 http://dx.doi.org/10.1007/s12275-014-3022-7 PMID: 24500480
[199] Lee, H.-K.; Lee, J.; Tobias, P. S. Two lipoproteins extracted from [212] Frangogiannis, N.G.; Ren, G.; Dewald, O.; Zymek, P.; Haudek, S.;
escherichia coli K-12 LCD25 lipopolysaccharide are the major Koerting, A.; Winkelmann, K.; Michael, L.H.; Lawler, J.; Entman,
components responsible for toll-like receptor 2-mediated signaling M.L. Critical role of endogenous thrombospondin-1 in preventing
J. Immunol., 2002, 168 (8), 4012-4017 expansion of healing myocardial infarcts. Circulation, 2005,
[200] Asai, Y.; Hashimoto, M.; Fletcher, H.M.; Miyake, K.; Akira, S.; 111(22), 2935-2942.
Ogawa, T. Lipopolysaccharide preparation extracted from http://dx.doi.org/10.1161/CIRCULATIONAHA.104.510354 PMID:
Porphyromonas gingivalis lipoprotein-deficient mutant shows a 15927970
marked decrease in toll-like receptor 2-mediated signaling. Infect. [213] Kohler, H.P.; Grant, P.J. Plasminogen-activator inhibitor type 1 and
Immun., 2005, 73(4), 2157-2163. coronary artery disease. N. Engl. J. Med., 2000, 342(24), 1792-
http://dx.doi.org/10.1128/IAI.73.4.2157-2163.2005 PMID: 1801.
15784558 http://dx.doi.org/10.1056/NEJM200006153422406 PMID:
[201] Sawada, N.; Ogawa, T.; Asai, Y.; Makimura, Y.; Sugiyama, A. 10853003
Toll-like receptor 4-dependent recognition of structurally different [214] Nikaido, H. Molecular basis of bacterial outer membrane
forms of chemically synthesized lipid As of Porphyromonas permeability revisited. Microbiol. Mol. Biol. Rev., 2003, 67(4), 593-
gingivalis. Clin. Exp. Immunol., 2007, 148(3), 529-536. 656.
http://dx.doi.org/10.1111/j.1365-2249.2007.03346.x PMID: http://dx.doi.org/10.1128/MMBR.67.4.593-656.2003 PMID:
17335558 14665678
[202] Kumada, H.; Haishima, Y.; Watanabe, K.; Hasegawa, C.; Tsuchiya, [215] Bos, M.P.; Robert, V.; Tommassen, J. Biogenesis of the gram-
T.; Tanamoto, K.; Umemoto, T. Biological properties of the native negative bacterial outer membrane. Annu. Rev. Microbiol., 2007, 61,
and synthetic lipid A of Porphyromonas gingivalis 191-214.
lipopolysaccharide. Oral Microbiol. Immunol., 2008, 23(1), 60-69. http://dx.doi.org/10.1146/annurev.micro.61.080706.093245 PMID:
http://dx.doi.org/10.1111/j.1399-302X.2007.00392.x PMID: 17506684
18173800 [216] Gonzales, J.R.; Groeger, S.; Johansson, A.; Meyle, J. T helper cells
[203] Al-Qutub, M.N.; Braham, P.H.; Karimi-Naser, L.M.; Liu, X.; from aggressive periodontitis patients produce higher levels of
Genco, C.A.; Darveau, R.P. Hemin-dependent modulation of the interleukin-1 beta and interleukin-6 in interaction with
lipid A structure of Porphyromonas gingivalis lipopolysaccharide. Porphyromonas gingivalis. Clin. Oral Investig., 2014, 18(7), 1835-
Infect. Immun., 2006, 74(8), 4474-4485. 1843.
http://dx.doi.org/10.1128/IAI.01924-05 PMID: 16861633 http://dx.doi.org/10.1007/s00784-013-1162-5 PMID: 24352581
[204] Olsen, I.; Singhrao, S.K. Importance of heterogeneity in [217] Watanabe, K.; Yamaji, Y.; Umemoto, T. Correlation between cell-
Porhyromonas gingivalis lipopolysaccharide lipid A in tissue adherent activity and surface structure in Porphyromonas gingivalis.
specific inflammatory signalling. J. Oral Microbiol., 2018, 10(1), Oral Microbiol. Immunol., 1992, 7(6), 357-363.
1440128. http://dx.doi.org/10.1111/j.1399-302X.1992.tb00636.x PMID:
http://dx.doi.org/10.1080/20002297.2018.1440128 PMID: 1363734
29503705 [218] Saito, S.; Hiratsuka, K.; Hayakawa, M.; Takiguchi, H.; Abiko, Y.
[205] Zenobia, C.; Hasturk, H.; Nguyen, D.; Van Dyke, T.E.; Kantarci, Inhibition of a Porphyromonas gingivalis colonizing factor between
A.; Darveau, R.P.; Bäumler, A.J. Porphyromonas gingivalis lipid A Actinomyces viscosus ATCC 19246 by monoclonal antibodies
phosphatase activity is critical for colonization and increasing the against recombinant 40-kDa outer-membrane protein. Gen.
commensal load in the rabbit ligature model. Infect. Immun., 2014, Pharmacol., 1997, 28(5), 675-680.
82(2), 650-659. http://dx.doi.org/10.1016/S0306-3623(96)00366-7 PMID: 9184801
http://dx.doi.org/10.1128/IAI.01136-13 PMID: 24478080 [219] Chen, Y.Y.; Peng, B.; Yang, Q.; Glew, M.D.; Veith, P.D.; Cross,
[206] Darveau, R.P.; Belton, C.M.; Reife, R.A.; Lamont, R.J. Local K.J.; Goldie, K.N.; Chen, D.; O’Brien-Simpson, N.; Dashper, S.G.;
chemokine paralysis, a novel pathogenic mechanism for Reynolds, E.C. The outer membrane protein LptO is essential for
Porphyromonas gingivalis. Infect. Immun., 1998, 66(4), 1660-1665. the O-deacylation of LPS and the co-ordinated secretion and
http://dx.doi.org/10.1128/IAI.66.4.1660-1665.1998 PMID: 9529095 attachment of A-LPS and CTD proteins in Porphyromonas
[207] Yoshimura, A.; Hara, Y.; Kaneko, T.; Kato, I. Secretion of IL-1 gingivalis. Mol. Microbiol., 2011, 79(5), 1380-1401.
beta, TNF-alpha, IL-8 and IL-1ra by human polymorphonuclear http://dx.doi.org/10.1111/j.1365-2958.2010.07530.x PMID:
leukocytes in response to lipopolysaccharides from 21244528
periodontopathic bacteria. J. Periodontal Res., 1997, 32(3), 279- [220] Saiki, K.; Konishi, K. Identification of a novel Porphyromonas
286. gingivalis outer membrane protein, PG534, required for the
http://dx.doi.org/10.1111/j.1600-0765.1997.tb00535.x PMID: production of active gingipains. FEMS Microbiol. Lett., 2010,
9138193 310(2), 168-174.
[208] Kato, H.; Taguchi, Y.; Tominaga, K.; Umeda, M.; Tanaka, A. http://dx.doi.org/10.1111/j.1574-6968.2010.02059.x PMID:
Porphyromonas gingivalis LPS inhibits osteoblastic differentiation 20695897
and promotes proinflammatory cytokine production in human [221] Handley, P.S.; Tipler, L.S. An electron microscope survey of the
periodontal ligament stem cells. Arch. Oral Biol., 2014, 59(2), 167- surface structures and hydrophobicity of oral and non-oral species
175. of the bacterial genus Bacteroides. Arch. Oral Biol., 1986, 31(5),
http://dx.doi.org/10.1016/j.archoralbio.2013.11.008 PMID: 325-335.
24370188 http://dx.doi.org/10.1016/0003-9969(86)90047-6 PMID: 2875705
1384 Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 Wadhawan et al.

[222] Nagano, K.; Hasegawa, Y.; Abiko, Y.; Yoshida, Y.; Murakami, Y.; lecithinolyticum, the phylogenetic group IV oral spirochetes
Yoshimura, F. Porphyromonas gingivalis FimA fimbriae: fimbrial associated with periodontitis and endodontic infections. Infect.
assembly by fimA alone in the fim gene cluster and differential Immun., 2005, 73(1), 268-276.
antigenicity among fimA genotypes. PLoS One, 2012, 7(9), e43722. http://dx.doi.org/10.1128/IAI.73.1.268-276.2005 PMID: 15618163
http://dx.doi.org/10.1371/journal.pone.0043722 PMID: 22970139 [237] Bolstad, A.I.; Jensen, H.B.; Bakken, V. Taxonomy, biology, and
[223] Amano, A. Bacterial adhesins to host components in periodontitis. periodontal aspects of Fusobacterium nucleatum. Clin. Microbiol.
Periodontol. 2000, 2010, 52(1), 12-37. Rev., 1996, 9(1), 55-71.
http://dx.doi.org/10.1111/j.1600-0757.2009.00307.x PMID: http://dx.doi.org/10.1128/CMR.9.1.55-71.1996 PMID: 8665477
20017793 [238] Skaar, E.P. The battle for iron between bacterial pathogens and their
[224] Amano, A.; Nakagawa, I.; Okahashi, N.; Hamada, N. Variations of vertebrate hosts. PLoS Pathog., 2010, 6(8), e1000949.
Porphyromonas gingivalis fimbriae in relation to microbial http://dx.doi.org/10.1371/journal.ppat.1000949 PMID: 20711357
pathogenesis. J. Periodontal Res., 2004, 39(2), 136-142. [239] Ebersole, J.L.; Cappelli, D. Acute-phase reactants in infections and
http://dx.doi.org/10.1111/j.1600-0765.2004.00719.x PMID: inflammatory diseases. Periodontol. 2000, 2000, 23, 19-49.
15009522 http://dx.doi.org/10.1034/j.1600-0757.2000.2230103.x PMID:
[225] Lee, J.Y.; Sojar, H.T.; Bedi, G.S.; Genco, R.J. Synthetic peptides 11276764
analogous to the fimbrillin sequence inhibit adherence of [240] Loos, B.G. Systemic markers of inflammation in periodontitis. J.
Porphyromonas gingivalis. Infect. Immun., 1992, 60(4), 1662-1670. Periodontol., 2005, 76(11)(Suppl.), 2106-2115.
http://dx.doi.org/10.1128/IAI.60.4.1662-1670.1992 PMID: 1347762 http://dx.doi.org/10.1902/jop.2005.76.11-S.2106
[226] Amano, A. Disruption of epithelial barrier and impairment of [241] Khader, Y.S.; Albashaireh, Z.S.; Alomari, M.A. Periodontal
cellular function by Porphyromonas gingivalis. Front. Biosci., diseases and the risk of coronary heart and cerebrovascular diseases:
2007, 12, 3965-3974. a meta-analysis. J. Periodontol., 2004, 75(8), 1046-1053.
http://dx.doi.org/10.2741/2363 PMID: 17485350 http://dx.doi.org/10.1902/jop.2004.75.8.1046 PMID: 15455730
[227] Sharma, A.; Sojar, H.T.; Lee, J.Y.; Genco, R.J. Expression of a [242] Lafon, A.; Pereira, B.; Dufour, T.; Rigouby, V.; Giroud, M.; Bejot,
functional Porphyromonas gingivalis fimbrillin polypeptide in Y.; Tubert-Jeannin, S. Periodontal disease and stroke: a meta-
Escherichia coli: purification, physicochemical and analysis of cohort studies. Eur. J. Neurol, 2014, 21(9), 1155-e67.
immunochemical characterization, and binding characteristics. http://dx.doi.org/10.1111/ene.12415
Infect. Immun., 1993, 61(8), 3570-3573. [243] Mustapha, I.Z.; Debrey, S.; Oladubu, M.; Ugarte, R. Markers of
http://dx.doi.org/10.1128/IAI.61.8.3570-3573.1993 PMID: 8392975 systemic bacterial exposure in periodontal disease and
[228] Zenobia, C.; Hajishengallis, G. Porphyromonas gingivalis virulence cardiovascular disease risk: a systematic review and meta-analysis.
factors involved in subversion of leukocytes and microbial J. Periodontol., 2007, 78(12), 2289-2302.
dysbiosis. Virulence, 2015, 6(3), 236-243. http://dx.doi.org/10.1902/jop.2007.070140 PMID: 18052701
http://dx.doi.org/10.1080/21505594.2014.999567 PMID: 25654623 [244] Kweider, M.; Lowe, G.D.; Murray, G.D.; Kinane, D.F.; McGowan,
[229] Yang, J.; Wu, J.; Liu, Y.; Huang, J.; Lu, Z.; Xie, L.; Sun, W.; Ji, Y. D.A. Dental disease, fibrinogen and white cell count; links with
Porphyromonas gingivalis infection reduces regulatory T cells in myocardial infarction? Scott. Med. J., 1993, 38(3), 73-74.
infected atherosclerosis patients. PLoS One, 2014, 9(1), e86599- http://dx.doi.org/10.1177/003693309303800304 PMID: 8356427
e86599. [245] Slade, G.D.; Offenbacher, S.; Beck, J.D.; Heiss, G.; Pankow, J.S.
http://dx.doi.org/10.1371/journal.pone.0086599 PMID: 24466164 Acute-phase inflammatory response to periodontal disease in the US
[230] Maiden, M.F.; Cohee, P.; Tanner, A.C. Proposal to conserve the population. J. Dent. Res., 2000, 79(1), 49-57.
adjectival form of the specific epithet in the reclassification of http://dx.doi.org/10.1177/00220345000790010701 PMID:
Bacteroides forsythus Tanner et al. 1986 to the genus Tannerella 10690660
Sakamoto et al. 2002 as Tannerella forsythia corrig., gen. nov., [246] Wu, T.; Trevisan, M.; Genco, R.J.; Falkner, K.L.; Dorn, J.P.;
comb. nov. Request for an Opinion. Int. J. Syst. Evol. Microbiol., Sempos, C.T. Examination of the relation between periodontal
2003, 53(Pt 6), 2111-2112. health status and cardiovascular risk factors: serum total and high
http://dx.doi.org/10.1099/ijs.0.02641-0 PMID: 14657155 density lipoprotein cholesterol, C-reactive protein, and plasma
[231] Hajishengallis, G.; Lamont, R.J. Beyond the red complex and into fibrinogen. Am. J. Epidemiol., 2000, 151(3), 273-282.
more complexity: the polymicrobial synergy and dysbiosis (PSD) http://dx.doi.org/10.1093/oxfordjournals.aje.a010203 PMID:
model of periodontal disease etiology. Mol. Oral Microbiol., 2012, 10670552
27(6), 409-419. [247] Liljestrand, J.M.; Paju, S.; Pietiäinen, M.; Buhlin, K.; Persson, G.R.;
http://dx.doi.org/10.1111/j.2041-1014.2012.00663.x PMID: Nieminen, M.S.; Sinisalo, J.; Mäntylä, P.; Pussinen, P.J.
23134607 Immunologic burden links periodontitis to acute coronary
[232] Dashper, S.G.; Seers, C.A.; Tan, K.H.; Reynolds, E.C. Virulence syndrome. Atherosclerosis, 2018, 268, 177-184.
factors of the oral spirochete Treponema denticola. J. Dent. Res., http://dx.doi.org/10.1016/j.atherosclerosis.2017.12.007 PMID:
2011, 90(6), 691-703. 29232563
http://dx.doi.org/10.1177/0022034510385242 PMID: 20940357 [248] Hajishengallis, G.; Liang, S.; Payne, M.A.; Hashim, A.; Jotwani, R.;
[233] Polak, D.; Wilensky, A.; Shapira, L.; Halabi, A.; Goldstein, D.; Eskan, M.A.; McIntosh, M.L.; Alsam, A.; Kirkwood, K.L.;
Weiss, E.I.; Houri-Haddad, Y. Mouse model of experimental Lambris, J.D.; Darveau, R.P.; Curtis, M.A. Low-abundance biofilm
periodontitis induced by Porphyromonas gingivalis/Fusobacterium species orchestrates inflammatory periodontal disease through the
nucleatum infection: bone loss and host response. J. Clin. commensal microbiota and complement. Cell Host Microbe, 2011,
Periodontol., 2009, 36(5), 406-410. 10(5), 497-506.
http://dx.doi.org/10.1111/j.1600-051X.2009.01393.x PMID: http://dx.doi.org/10.1016/j.chom.2011.10.006 PMID: 22036469
19419440 [249] Walsh, M.C.; Lee, J.; Choi, Y. Tumor necrosis factor receptor-
[234] Kesavalu, L.; Sathishkumar, S.; Bakthavatchalu, V.; Matthews, C.; associated factor 6 (TRAF6) regulation of development, function,
Dawson, D.; Steffen, M.; Ebersole, J.L. Rat model of polymicrobial and homeostasis of the immune system. Immunol. Rev., 2015,
infection, immunity, and alveolar bone resorption in periodontal 266(1), 72-92.
disease. Infect. Immun., 2007, 75(4), 1704-1712. http://dx.doi.org/10.1111/imr.12302 PMID: 26085208
http://dx.doi.org/10.1128/IAI.00733-06 PMID: 17210663 [250] Tang, L.; Zhou, X.D.; Wang, Q.; Zhang, L.; Wang, Y.; Li, X.Y.;
[235] Deng, Z-L.; Sztajer, H.; Jarek, M.; Bhuju, S.; Wagner-Döbler, I. Huang, D.M. Expression of TRAF6 and proinflammatory cytokines
Worlds apart - transcriptome profiles of key oral microbes in the through activation of TLR2, TLR4, NOD1, and NOD2 in human
periodontal pocket compared to single laboratory culture reflect periodontal ligament fibroblasts. Arch. Oral Biol., 2011, 56(10),
synergistic interactions. Front. Microbiol., 2018, 9, 124-124. 1064-1072.
http://dx.doi.org/10.3389/fmicb.2018.00124 PMID: 29467738 http://dx.doi.org/10.1016/j.archoralbio.2011.02.020 PMID:
[236] Lee, S.H.; Kim, K.K.; Choi, B.K. Upregulation of intercellular 21457942
adhesion molecule 1 and proinflammatory cytokines by the major [251] Tang, L.; Zhou, X.D.; Wang, Q.; Zhang, L.; Wang, Y.; Huang,
surface proteins of Treponema maltophilum and Treponema D.M. TNF receptor-associated factor 6 suppression inhibits
Periodontal Pathogens and Neuropsychiatric Health Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 1385

inflammatory response to Porphyromonas gingivialis in human [265] Choi, J-I.; Chung, S-W.; Kang, H-S.; Rhim, B.Y.; Park, Y-M.; Kim,
periodontal ligament cells. Quintessence Int., 2011, 42(9), 787-796. U-S.; Kim, S-J. Epitope mapping of Porphyromonas gingivalis
PMID: 21909504 heat-shock protein and human heat-shock protein in human
[252] Ikeda, U.; Ito, T.; Shimada, K. Interleukin-6 and acute coronary atherosclerosis. J. Dent. Res., 2004, 83(12), 936-940.
syndrome. Clin. Cardiol., 2001, 24(11), 701-704. http://dx.doi.org/10.1177/154405910408301209 PMID: 15557401
http://dx.doi.org/10.1002/clc.4960241103 PMID: 11714126 [266] Hinode, D.; Nakamura, R.; Grenier, D.; Mayrand, D. Cross-
[253] Bagavad Gita, J.; George, A.V.; Pavithra, N.; Chandrasekaran, S.C.; reactivity of specific antibodies directed to heat shock proteins from
Latchumanadhas, K.; Gnanamani, A. Dysregulation of miR-146a by periodontopathogenic bacteria and of human origin [corrected].
periodontal pathogens: A risk for acute coronary syndrome. J. Oral Microbiol. Immunol., 1998, 13(1), 55-58.
Periodontol., 2019, 90(7), 756-765. http://dx.doi.org/10.1111/j.1399-302X.1998.tb00752.x PMID:
http://dx.doi.org/10.1002/JPER.18-0466 PMID: 30618100 9573824
[254] Shimada, Y.; Komatsu, Y.; Ikezawa-Suzuki, I.; Tai, H.; Sugita, N.; [267] Yamazaki, K.; Ohsawa, Y.; Itoh, H.; Ueki, K.; Tabeta, K.; Oda, T.;
Yoshie, H. The effect of periodontal treatment on serum leptin, Nakajima, T.; Yoshie, H.; Saito, S.; Oguma, F.; Kodama, M.;
interleukin-6, and C-reactive protein. J. Periodontol., 2010, 81(8), Aizawa, Y.; Seymour, G.J. T-cell clonality to Porphyromonas
1118-1123. gingivalis and human heat shock protein 60s in patients with
http://dx.doi.org/10.1902/jop.2010.090741 PMID: 20370420 atherosclerosis and periodontitis. Oral Microbiol. Immunol., 2004,
[255] Sabatine, M.S.; Morrow, D.A.; Cannon, C.P.; Murphy, S.A.; 19(3), 160-167.
Demopoulos, L.A.; DiBattiste, P.M.; McCabe, C.H.; Braunwald, E.; http://dx.doi.org/10.1111/j.0902-0055.2004.00134.x PMID:
Gibson, C.M. Relationship between baseline white blood cell count 15107067
and degree of coronary artery disease and mortality in patients with [268] Ford, P.; Gemmell, E.; Walker, P.; West, M.; Cullinan, M.;
acute coronary syndromes: a TACTICS-TIMI 18 (Treat Angina Seymour, G. Characterization of heat shock protein-specific T cells
with Aggrastat and determine Cost of Therapy with an Invasive or in atherosclerosis. Clin. Diagn. Lab. Immunol., 2005, 12(2), 259-
Conservative Strategy- Thrombolysis in Myocardial Infarction 18 267.
trial)substudy. J. Am. Coll. Cardiol., 2002, 40(10), 1761-1768. PMID: 15699420
http://dx.doi.org/10.1016/S0735-1097(02)02484-1 PMID: 12446059 [269] Fuchs, D.; Hausen, A.; Reibnegger, G.; Werner, E.R.; Dierich,
[256] Takeda, Y.; Suzuki, S.; Fukutomi, T.; Kondo, H.; Sugiura, M.; M.P.; Wachter, H. Neopterin as a marker for activated cell-mediated
Suzumura, H.; Murasaki, G.; Okutani, H.; Itoh, M. Elevated white immunity: application in HIV infection. Immunol. Today, 1988,
blood cell count as a risk factor of coronary artery disease: 9(5), 150-155.
inconsistency between forms of the disease. Jpn. Heart J., 2003, http://dx.doi.org/10.1016/0167-5699(88)91203-0 PMID: 3076770
44(2), 201-211. [270] Huber, C.; Batchelor, J.R.; Fuchs, D.; Hausen, A.; Lang, A.;
http://dx.doi.org/10.1536/jhj.44.201 PMID: 12718482 Niederwieser, D.; Reibnegger, G.; Swetly, P.; Troppmair, J.;
[257] Pitsavos, C.; Kourlaba, G.; Panagiotakos, D.B.; Tsamis, E.; Kogias, Wachter, H. Immune response-associated production of neopterin.
Y.; Stravopodis, P.; Stefanadis, C. Does smoking status affect the Release from macrophages primarily under control of interferon-
association between baseline white blood cell count and in-hospital gamma. J. Exp. Med., 1984, 160(1), 310-316.
mortality of patients presented with acute coronary syndrome? The http://dx.doi.org/10.1084/jem.160.1.310 PMID: 6429267
Greek study of acute coronary syndromes (GREECS). Int. J. [271] Wirleitner, B.; Reider, D.; Ebner, S.; Böck, G.; Widner, B.; Jaeger,
Cardiol., 2008, 125(1), 94-100. M.; Schennach, H.; Romani, N.; Fuchs, D. Monocyte-derived
http://dx.doi.org/10.1016/j.ijcard.2007.05.030 PMID: 17655949 dendritic cells release neopterin. J. Leukoc. Biol., 2002, 72(6), 1148-
[258] Johannsen, A.; Susin, C.; Gustafsson, A. Smoking and 1153.
inflammation: evidence for a synergistic role in chronic disease. PMID: 12488496
Periodontol. 2000, 2014, 64(1), 111-126. [272] Werner-Felmayer, G.; Werner, E.R.; Fuchs, D.; Hausen, A.;
http://dx.doi.org/10.1111/j.1600-0757.2012.00456.x PMID: Reibnegger, G.; Wachter, H. Tumour necrosis factor-alpha and
24320959 lipopolysaccharide enhance interferon-induced tryptophan
[259] Srivastava, P. Roles of heat-shock proteins in innate and adaptive degradation and pteridine synthesis in human cells. Biol. Chem.
immunity. Nat. Rev. Immunol., 2002, 2(3), 185-194. Hoppe Seyler, 1989, 370(9), 1063-1069.
http://dx.doi.org/10.1038/nri749 PMID: 11913069 http://dx.doi.org/10.1515/bchm3.1989.370.2.1063 PMID: 2482041
[260] Lamb, D.J.; El-Sankary, W.; Ferns, G.A. Molecular mimicry in [273] Murr, C.; Widner, B.; Wirleitner, B.; Fuchs, D. Neopterin as a
atherosclerosis: a role for heat shock proteins in immunisation. marker for immune system activation. Curr. Drug Metab., 2002,
Atherosclerosis, 2003, 167(2), 177-185. 3(2), 175-187.
http://dx.doi.org/10.1016/S0021-9150(02)00301-5 PMID: 12818399 http://dx.doi.org/10.2174/1389200024605082 PMID: 12003349
[261] Rizzo, M.; Cappello, F.; Marfil, R.; Nibali, L.; Marino Gammazza, [274] Nathan, C.F. Peroxide and pteridine: a hypothesis on the regulation
A.; Rappa, F.; Bonaventura, G.; Galindo-Moreno, P.; O’Valle, F.; of macrophage antimicrobial activity by interferon gamma.
Zummo, G.; Conway de Macario, E.; Macario, A.J.; Mesa, F. Heat- Interferon, 1986, 7, 125-143.
shock protein 60 kDa and atherogenic dyslipidemia in patients with PMID: 3102389
untreated mild periodontitis: a pilot study. Cell Stress Chaperones, [275] Ozmeriç, N.; Baydar, T.; Bodur, A.; Engin, A.B.; Uraz, A.; Eren,
2012, 17(3), 399-407. K.; Sahin, G. Level of neopterin, a marker of immune cell activation
http://dx.doi.org/10.1007/s12192-011-0315-1 PMID: 22215516 in gingival crevicular fluid, saliva, and urine in patients with
[262] Maeda, H.; Miyamoto, M.; Hongyo, H.; Nagai, A.; Kurihara, H.; aggressive periodontitis. J. Periodontol., 2002, 73(7), 720-725.
Murayama, Y. Heat shock protein 60 (GroEL) from Porphyromonas http://dx.doi.org/10.1902/jop.2002.73.7.720 PMID: 12146530
gingivalis: molecular cloning and sequence analysis of its gene and [276] Vrecko, K.; Staedtler, P.; Mischak, I.; Maresch, L.; Reibnegger, G.
purification of the recombinant protein. FEMS Microbiol. Lett., Periodontitis and concentrations of the cellular immune activation
1994, 119(1-2), 129-135. marker neopterin in saliva and urine. Clin. Chim. Acta, 1997, 268(1-
http://dx.doi.org/10.1111/j.1574-6968.1994.tb06879.x PMID: 2), 31-40.
7913687 http://dx.doi.org/10.1016/S0009-8981(97)00154-X PMID: 9495569
[263] Vayssier, C.; Mayrand, D.; Grenier, D. Detection of stress proteins [277] Pradeep, A.R.; Kumar, M.S.; Ramachandraprasad, M.V.; Shikha, C.
in Porphyromonas gingivalis and other oral bacteria by western Gingival crevicular fluid levels of neopterin in healthy subjects and
immunoblotting analysis. FEMS Microbiol. Lett., 1994, 121(3), in patients with different periodontal diseases. J. Periodontol., 2007,
303-307. 78(10), 1962-1967.
http://dx.doi.org/10.1111/j.1574-6968.1994.tb07117.x PMID: http://dx.doi.org/10.1902/jop.2007.070096 PMID: 18062118
7926686 [278] Bartel, D.P. MicroRNAs: genomics, biogenesis, mechanism, and
[264] Lu, B.; McBride, B.C. Stress response of Porphyromonas function. Cell, 2004, 116(2), 281-297.
gingivalis. Oral Microbiol. Immunol., 1994, 9(3), 166-173. http://dx.doi.org/10.1016/S0092-8674(04)00045-5 PMID: 14744438
http://dx.doi.org/10.1111/j.1399-302X.1994.tb00054.x PMID: [279] Ebert, M.S.; Sharp, P.A. Roles for microRNAs in conferring
7936723 robustness to biological processes. Cell, 2012, 149(3), 515-524.
1386 Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 Wadhawan et al.

http://dx.doi.org/10.1016/j.cell.2012.04.005 PMID: 22541426 http://dx.doi.org/10.1155/2013/172351 PMID: 23983402


[280] Lee, R.C.; Feinbaum, R.L.; Ambros, V. The C. elegans [298] Slota, J.A.; Booth, S.A. MicroRNAs in neuroinflammation:
heterochronic gene lin-4 encodes small RNAs with antisense implications in disease pathogenesis, biomarker discovery and
complementarity to lin-14. Cell, 1993, 75(5), 843-854. therapeutic applications. Noncoding RNA, 2019, 5(2), E35.
http://dx.doi.org/10.1016/0092-8674(93)90529-Y PMID: 8252621 http://dx.doi.org/10.3390/ncrna5020035 PMID: 31022830
[281] Hamar, P. Role of regulatory micro RNAs in type 2 diabetes [299] Su, W.; Aloi, M.S.; Garden, G.A. MicroRNAs mediating CNS
mellitus-related inflammation. Nucleic Acid Ther., 2012, 22(5), 289- inflammation: Small regulators with powerful potential. Brain
294. Behav. Immun., 2016, 52, 1-8.
http://dx.doi.org/10.1089/nat.2012.0381 PMID: 22950794 http://dx.doi.org/10.1016/j.bbi.2015.07.003 PMID: 26148445
[282] Kloosterman, W.P.; Plasterk, R.H. The diverse functions of [300] Thounaojam, M.C.; Kaushik, D.K.; Basu, A. MicroRNAs in the
microRNAs in animal development and disease. Dev. Cell, 2006, brain: it’s regulatory role in neuroinflammation. Mol. Neurobiol.,
11(4), 441-450. 2013, 47(3), 1034-1044.
http://dx.doi.org/10.1016/j.devcel.2006.09.009 PMID: 17011485 http://dx.doi.org/10.1007/s12035-013-8400-3 PMID: 23315269
[283] Kantharidis, P.; Wang, B.; Carew, R.M.; Lan, H.Y. Diabetes [301] Guo, Y.; Hong, W.; Wang, X.; Zhang, P.; Körner, H.; Tu, J.; Wei,
complications: the microRNA perspective. Diabetes, 2011, 60(7), W. MicroRNAs in microglia: how do microRNAs affect activation,
1832-1837. inflammation, polarization of microglia and mediate the interaction
http://dx.doi.org/10.2337/db11-0082 PMID: 21709278 between microglia and glioma? Front. Mol. Neurosci., 2019, 12,
[284] Baulina, N.M.; Kulakova, O.G.; Favorova, O.O. MicroRNAs: The 125.
Role in Autoimmune Inflammation. Acta Naturae, 2016, 8(1), 21- http://dx.doi.org/10.3389/fnmol.2019.00125 PMID: 31133802
33. [302] Gaudet, A.D.; Fonken, L.K.; Watkins, L.R.; Nelson, R.J.; Popovich,
http://dx.doi.org/10.32607/20758251-2016-8-1-21-33 PMID: P.G. MicroRNAs: Roles in regulating neuroinflammation.
27099782 Neuroscientist, 2018, 24(3), 221-245.
[285] Raisch, J.; Darfeuille-Michaud, A.; Nguyen, H.T.T. Role of http://dx.doi.org/10.1177/1073858417721150 PMID: 28737113
microRNAs in the immune system, inflammation and cancer. World [303] Taganov, K.D.; Boldin, M.P.; Chang, K.J.; Baltimore, D. NF-
J. Gastroenterol., 2013, 19(20), 2985-2996. kappaB-dependent induction of microRNA miR-146, an inhibitor
http://dx.doi.org/10.3748/wjg.v19.i20.2985 PMID: 23716978 targeted to signaling proteins of innate immune responses. Proc.
[286] Dai, R.; Ahmed, S.A.; Micro, R.N.A. MicroRNA, a new paradigm Natl. Acad. Sci. USA, 2006, 103(33), 12481-12486.
for understanding immunoregulation, inflammation, and http://dx.doi.org/10.1073/pnas.0605298103 PMID: 16885212
autoimmune diseases. Transl. Res., 2011, 157(4), 163-179. [304] Lawrence, T. The nuclear factor NF-kappaB pathway in
http://dx.doi.org/10.1016/j.trsl.2011.01.007 PMID: 21420027 inflammation. Cold Spring Harb. Perspect. Biol., 2009, 1(6),
[287] Lu, L.F.; Liston, A. MicroRNA in the immune system, microRNA a001651.
as an immune system. Immunology, 2009, 127(3), 291-298. http://dx.doi.org/10.1101/cshperspect.a001651 PMID: 20457564
http://dx.doi.org/10.1111/j.1365-2567.2009.03092.x PMID: [305] Meisgen, F.; Xu Landén, N.; Wang, A.; Réthi, B.; Bouez, C.;
19538248 Zuccolo, M.; Gueniche, A.; Ståhle, M.; Sonkoly, E.; Breton, L.;
[288] Roy, B.; Dunbar, M.; Shelton, R. C.; Dwivedi, Y. Identification of Pivarcsi, A. MiR-146a negatively regulates TLR2-induced
MicroRNA-124-3p as a putative epigenetic signature of major inflammatory responses in keratinocytes. J. Invest. Dermatol., 2014,
depressive disorder. Neuropsychopharmacology, 2017, 42 (4), 864- 134(7), 1931-1940.
875. http://dx.doi.org/10.1038/jid.2014.89 PMID: 24670381
[289] Roy, B.; Dwivedi, Y. Understanding the neuroepigenetic [306] Bartel, D.P. MicroRNAs: target recognition and regulatory
constituents of suicide brain. Prog. Mol. Biol. Transl. Sci., 2018, functions. Cell, 2009, 136(2), 215-233.
157, 233-262. http://dx.doi.org/10.1016/j.cell.2009.01.002 PMID: 19167326
http://dx.doi.org/10.1016/bs.pmbts.2018.01.007 PMID: 29933952 [307] Qin, Z.; Wang, P.Y.; Su, D.F.; Liu, X. miRNA-124 in Immune
[290] Roy, B.; Shelton, R.C.; Dwivedi, Y. DNA methylation and System and Immune Disorders. Front. Immunol., 2016, 7, 406.
expression of stress related genes in PBMC of MDD patients with http://dx.doi.org/10.3389/fimmu.2016.00406 PMID: 27757114
and without serious suicidal ideation. J. Psychiatr. Res., 2017, 89, [308] Hutchison, E.R.; Kawamoto, E.M.; Taub, D.D.; Lal, A.;
115-124. Abdelmohsen, K.; Zhang, Y.; Wood, W.H., III; Lehrmann, E.;
http://dx.doi.org/10.1016/j.jpsychires.2017.02.005 PMID: 28246044 Camandola, S.; Becker, K.G.; Gorospe, M.; Mattson, M.P. Evidence
[291] Smalheiser, N.R.; Lugli, G.; Rizavi, H.S.; Torvik, V.I.; Turecki, G.; for miR-181 involvement in neuroinflammatory responses of
Dwivedi, Y. MicroRNA expression is down-regulated and astrocytes. Glia, 2013, 61(7), 1018-1028.
reorganized in prefrontal cortex of depressed suicide subjects. PLoS http://dx.doi.org/10.1002/glia.22483 PMID: 23650073
One, 2012, 7(3), e33201. [309] Sebastiani, G.; Grieco, F.A.; Spagnuolo, I.; Galleri, L.; Cataldo, D.;
http://dx.doi.org/10.1371/journal.pone.0033201 PMID: 22427989 Dotta, F. Increased expression of microRNA miR-326 in type 1
[292] Smalheiser, N.R.; Lugli, G.; Zhang, H.; Rizavi, H.; Cook, E.H.; diabetic patients with ongoing islet autoimmunity. Diabetes Metab.
Dwivedi, Y. Expression of microRNAs and other small RNAs in Res. Rev., 2011, 27(8), 862-866.
prefrontal cortex in schizophrenia, bipolar disorder and depressed http://dx.doi.org/10.1002/dmrr.1262 PMID: 22069274
subjects. PLoS One, 2014, 9(1)e86469 [310] Shi, C.; Zhu, L.; Chen, X.; Gu, N.; Chen, L.; Zhu, L.; Yang, L.;
http://dx.doi.org/10.1371/journal.pone.0086469 PMID: 24475125 Pang, L.; Guo, X.; Ji, C.; Zhang, C. IL-6 and TNF-α induced
[293] Wang, Q.; Roy, B.; Turecki, G.; Shelton, R.C.; Dwivedi, Y. Role of obesity-related inflammatory response through transcriptional
complex epigenetic switching in tumor necrosis factor-α regulation of miR-146b. J. Interferon Cytokine Res., 2014, 34(5),
upregulation in the prefrontal cortex of suicide subjects. Am. J. 342-348.
Psychiatry, 2018, 175(3), 262-274. http://dx.doi.org/10.1089/jir.2013.0078 PMID: 24428800
http://dx.doi.org/10.1176/appi.ajp.2017.16070759 PMID: 29361849 [311] Li, J.J.; Wang, B.; Kodali, M.C.; Chen, C.; Kim, E.; Patters, B.J.;
[294] Xiao, C.; Rajewsky, K. MicroRNA control in the immune system: Lan, L.; Kumar, S.; Wang, X.; Yue, J.; Liao, F.F. In vivo evidence
basic principles. Cell, 2009, 136(1), 26-36. for the contribution of peripheral circulating inflammatory
http://dx.doi.org/10.1016/j.cell.2008.12.027 PMID: 19135886 exosomes to neuroinflammation. J. Neuroinflammation, 2018,
[295] Sochocka, M.; Diniz, B.S.; Leszek, J. Inflammatory response in the 15(1), 8.
CNS: friend or foe? Mol. Neurobiol., 2017, 54(10), 8071-8089. http://dx.doi.org/10.1186/s12974-017-1038-8 PMID: 29310666
http://dx.doi.org/10.1007/s12035-016-0297-1 PMID: 27889895 [312] Honda, T.; Takahashi, N.; Miyauchi, S.; Yamazaki, K.
[296] Soreq, H.; Wolf, Y. NeurimmiRs: microRNAs in the neuroimmune Porphyromonas gingivalis lipopolysaccharide induces miR-146a
interface. Trends Mol. Med., 2011, 17(10), 548-555. without altering the production of inflammatory cytokines.
http://dx.doi.org/10.1016/j.molmed.2011.06.009 PMID: 21813326 Biochem. Biophys. Res. Commun., 2012, 420(4), 918-925.
[297] Ksiazek-Winiarek, D.J.; Kacperska, M.J.; Glabinski, A. http://dx.doi.org/10.1016/j.bbrc.2012.03.102 PMID: 22480686
MicroRNAs as novel regulators of neuroinflammation. Mediators
Inflamm., 2013, 2013, 172351.
Periodontal Pathogens and Neuropsychiatric Health Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 1387

[313] Xie, Y.F.; Shu, R.; Jiang, S.Y.; Liu, D.L.; Zhang, X.L. Comparison Hernando, C.; Suárez, Y. Macrophage deficiency of miR-21
of microRNA profiles of human periodontal disease and healthy promotes apoptosis, plaque necrosis, and vascular inflammation
gingival tissues. Int. J. Oral Sci., 2011, 3(3), 125-134. during atherogenesis. EMBO Mol. Med., 2017, 9(9), 1244-1262.
http://dx.doi.org/10.4248/IJOS11046 PMID: 21789961 http://dx.doi.org/10.15252/emmm.201607492 PMID: 28674080
[314] Stoecklin-Wasmer, C.; Guarnieri, P.; Celenti, R.; Demmer, R.T.; [328] Yuan, J.; Chen, H.; Ge, D.; Xu, Y.; Xu, H.; Yang, Y.; Gu, M.; Zhou,
Kebschull, M.; Papapanou, P.N. MicroRNAs and their target genes Y.; Zhu, J.; Ge, T.; Chen, Q.; Gao, Y.; Wang, Y.; Li, X.; Zhao, Y.
in gingival tissues. J. Dent. Res., 2012, 91(10), 934-940. Mir-21 promotes cardiac fibrosis after myocardial infarction via
http://dx.doi.org/10.1177/0022034512456551 PMID: 22879578 targeting smad7. Cell. Physiol. Biochem., 2017, 42(6), 2207-2219.
[315] Tang, L.; Li, X.; Bai, Y.; Wang, P.; Zhao, Y. MicroRNA-146a http://dx.doi.org/10.1159/000479995 PMID: 28817807
negatively regulates the inflammatory response to Porphyromonas [329] Wang, Z.H.; Sun, X.Y.; Li, C.L.; Sun, Y.M.; Li, J.; Wang, L.F.; Li,
gingivalis in human periodontal ligament fibroblasts via Z.Q. miRNA-21 expression in the serum of elderly patients with
TRAF6/p38 pathway. J. Periodontol., 2019, 90(4), 391-399. acute myocardial infarction. Med. Sci. Monit., 2017, 23, 5728-5734.
http://dx.doi.org/10.1002/JPER.18-0190 PMID: 30378773 http://dx.doi.org/10.12659/MSM.904933 PMID: 29197221
[316] An, F.; Gong, B.; Wang, H.; Yu, D.; Zhao, G.; Lin, L.; Tang, W.; [330] Du, A.; Zhao, S.; Wan, L.; Liu, T.; Peng, Z.; Zhou, Z.; Liao, Z.;
Yu, H.; Bao, S.; Xie, Q. miR-15b and miR-16 regulate TNF Fang, H. MicroRNA expression profile of human periodontal
mediated hepatocyte apoptosis via BCL2 in acute liver failure. ligament cells under the influence of Porphyromonas gingivalis
Apoptosis, 2012, 17(7), 702-716. LPS. J. Cell. Mol. Med., 2016, 20(7), 1329-1338.
http://dx.doi.org/10.1007/s10495-012-0704-7 PMID: 22374434 http://dx.doi.org/10.1111/jcmm.12819 PMID: 26987780
[317] Lezina, L.; Purmessur, N.; Antonov, A.V.; Ivanova, T.; Karpova, [331] Yao, Q.; Xu, H.; Zhang, Q.Q.; Zhou, H.; Qu, L.H. MicroRNA-21
E.; Krishan, K.; Ivan, M.; Aksenova, V.; Tentler, D.; Garabadgiu, promotes cell proliferation and down-regulates the expression of
A.V.; Melino, G.; Barlev, N.A. miR-16 and miR-26a target programmed cell death 4 (PDCD4) in HeLa cervical carcinoma
checkpoint kinases Wee1 and Chk1 in response to p53 activation by cells. Biochem. Biophys. Res. Commun., 2009, 388(3), 539-542.
genotoxic stress. Cell Death Dis., 2013, 4, e953. http://dx.doi.org/10.1016/j.bbrc.2009.08.044 PMID: 19682430
http://dx.doi.org/10.1038/cddis.2013.483 PMID: 24336073 [332] Venugopal, P.; Koshy, T.; Lavu, V.; Ranga Rao, S.; Ramasamy, S.;
[318] Zhang, X.; Wan, G.; Mlotshwa, S.; Vance, V.; Berger, F.G.; Chen, Hariharan, S.; Venkatesan, V. Differential expression of
H.; Lu, X. Oncogenic Wip1 phosphatase is inhibited by miR-16 in microRNAs let-7a, miR-125b, miR-100, and miR-21 and interaction
the DNA damage signaling pathway. Cancer Res., 2010, 70(18), with NF-kB pathway genes in periodontitis pathogenesis. J. Cell.
7176-7186. Physiol., 2018, 233(8), 5877-5884.
http://dx.doi.org/10.1158/0008-5472.CAN-10-0697 PMID: http://dx.doi.org/10.1002/jcp.26391 PMID: 29226952
20668064 [333] Zhou, W.; Su, L.; Duan, X.; Chen, X.; Hays, A.; Upadhyayula, S.;
[319] Zhan, X-H.; Xu, Q-Y.; Tian, R.; Yan, H.; Zhang, M.; Wu, J.; Wang, Shivde, J.; Wang, H.; Li, Y.; Huang, D.; Liang, S. MicroRNA-21
W.; He, J. MicroRNA16 regulates glioma cell proliferation, down-regulates inflammation and inhibits periodontitis. Mol.
apoptosis and invasion by targeting Wip1-ATM-p53 feedback loop. Immunol., 2018, 101, 608-614.
Oncotarget, 2017, 8(33), 54788-54798. http://dx.doi.org/10.1016/j.molimm.2018.05.008 PMID: 29884447
http://dx.doi.org/10.18632/oncotarget.18510 PMID: 28903382 [334] Miguel-Hidalgo, J. J.; Hall, K. O.; Bonner, H.; Roller, A. M.; Syed,
[320] Besnier, M.; Shantikumar, S.; Anwar, M.; Dixit, P.; Chamorro- M.; Park, C. J.; Ball, J. P.; Rothenberg, M. E.; Stockmeier, C. A.;
Jorganes, A.; Sweaad, W.; Sala-Newby, G.; Madeddu, P.; Thomas, Romero, D. G. MicroRNA-21: Expression in oligodendrocytes and
A.C.; Howard, L.; Mushtaq, S.; Petretto, E.; Caporali, A.; Emanueli, correlation with low myelin mRNAs in depression and alcoholism
C. miR-15a/-16 inhibit angiogenesis by targeting the Tie2 coding Prog. Neuropsychopharmacol. Biol. Psychiatry, 2017, 79 (Pt B),
sequence: therapeutic potential of a miR-15a/16 decoy system in 503-514
limb ischemia. Mol. Ther. Nucleic Acids, 2019, 17, 49-62. [335] Ouyang, Y.B.; Xu, L.; Lu, Y.; Sun, X.; Yue, S.; Xiong, X.X.;
http://dx.doi.org/10.1016/j.omtn.2019.05.002 PMID: 31220779 Giffard, R.G. Astrocyte-enriched miR-29a targets PUMA and
[321] Song, M.F.; Dong, J.Z.; Wang, Y.W.; He, J.; Ju, X.; Zhang, L.; reduces neuronal vulnerability to forebrain ischemia. Glia, 2013,
Zhang, Y.H.; Shi, J.F.; Lv, Y.Y. CSF miR-16 is decreased in major 61(11), 1784-1794.
depression patients and its neutralization in rats induces depression- http://dx.doi.org/10.1002/glia.22556 PMID: 24038396
like behaviors via a serotonin transmitter system. J. Affect. Disord., [336] Yang, Z.; Wu, L.; Zhu, X.; Xu, J.; Jin, R.; Li, G.; Wu, F. MiR-29a
2015, 178, 25-31. modulates the angiogenic properties of human endothelial cells.
http://dx.doi.org/10.1016/j.jad.2015.02.022 PMID: 25779937 Biochem. Biophys. Res. Commun., 2013, 434(1), 143-149.
[322] Gheysarzadeh, A.; Sadeghifard, N.; Afraidooni, L.; Pooyan, F.; http://dx.doi.org/10.1016/j.bbrc.2013.03.054 PMID: 23541945
Mofid, M.R.; Valadbeigi, H.; Bakhtiari, H.; Keikhavani, S. Serum- [337] Kebschull, M.; Papapanou, P.N. Mini but mighty: microRNAs in
based microRNA biomarkers for major depression: MiR-16, miR- the pathobiology of periodontal disease. Periodontol. 2000, 2015,
135a, and miR-1202. J. Res. Med. Sci., 2018, 23, 69. 69(1), 201-220.
http://dx.doi.org/10.4103/jrms.JRMS_879_17 PMID: 30181751 http://dx.doi.org/10.1111/prd.12095 PMID: 26252410
[323] Zhong, J.; He, Y.; Chen, W.; Shui, X.; Chen, C.; Lei, W. Circulating [338] Roncarati, R.; Viviani Anselmi, C.; Losi, M.A.; Papa, L.;
microRNA-19a as a potential novel biomarker for diagnosis of acute Cavarretta, E.; Da Costa Martins, P.; Contaldi, C.; Saccani Jotti, G.;
myocardial infarction. Int. J. Mol. Sci., 2014, 15(11), 20355-20364. Franzone, A.; Galastri, L.; Latronico, M.V.; Imbriaco, M.; Esposito,
http://dx.doi.org/10.3390/ijms151120355 PMID: 25383678 G.; De Windt, L.; Betocchi, S.; Condorelli, G. Circulating miR-29a,
[324] Chen, H.; Li, X.; Liu, S.; Gu, L.; Zhou, X. MircroRNA-19a among other up-regulated microRNAs, is the only biomarker for
promotes vascular inflammation and foam cell formation by both hypertrophy and fibrosis in patients with hypertrophic
targeting HBP-1 in atherogenesis. Sci. Rep., 2017, 7(1), 12089. cardiomyopathy. J. Am. Coll. Cardiol., 2014, 63(9), 920-927.
http://dx.doi.org/10.1038/s41598-017-12167-z PMID: 28935967 http://dx.doi.org/10.1016/j.jacc.2013.09.041 PMID: 24161319
[325] Marques, F.Z.; Eikelis, N.; Bayles, R.G.; Lambert, E.A.; Straznicky, [339] Wang, P.; Gu, Y.; Zhang, Q.; Han, Y.; Hou, J.; Lin, L.; Wu, C.;
N.E.; Hering, D.; Esler, M.D.; Head, G.A.; Barton, D.A.; Schlaich, Bao, Y.; Su, X.; Jiang, M.; Wang, Q.; Li, N.; Cao, X. Identification
M.P.; Lambert, G.W. A polymorphism in the norepinephrine of resting and type I IFN-activated human NK cell miRNomes
transporter gene is associated with affective and cardiovascular reveals microRNA-378 and microRNA-30e as negative regulators
disease through a microRNA mechanism. Mol. Psychiatry, 2017, of NK cell cytotoxicity. J. Immunol., 2012, 189(1), 211-221.
22(1), 134-141. http://dx.doi.org/10.4049/jimmunol.1200609 PMID: 22649192
http://dx.doi.org/10.1038/mp.2016.40 PMID: 27046647 [340] Perri, R.; Nares, S.; Zhang, S.; Barros, S.P.; Offenbacher, S.
[326] Li, X.; Teng, C.; Ma, J.; Fu, N.; Wang, L.; Wen, J.; Wang, T.Y. MicroRNA modulation in obesity and periodontitis. J. Dent. Res.,
miR-19 family: A promising biomarker and therapeutic target in 2012, 91(1), 33-38.
heart, vessels and neurons. Life Sci., 2019, 232, 116651. http://dx.doi.org/10.1177/0022034511425045 PMID: 22043006
http://dx.doi.org/10.1016/j.lfs.2019.116651 PMID: 31302195 [341] Wang, X.T.; Wu, X.D.; Lu, Y.X.; Sun, Y.H.; Zhu, H.H.; Liang,
[327] Canfrán-Duque, A.; Rotllan, N.; Zhang, X.; Fernández-Fuertes, M.; J.B.; He, W.K.; Zeng, Z.Y.; Li, L. Potential involvement of MiR-
Ramírez-Hidalgo, C.; Araldi, E.; Daimiel, L.; Busto, R.; Fernández- 30e-3p in myocardial injury induced by coronary microembolization
1388 Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 Wadhawan et al.

via autophagy activation. Cell. Physiol. Biochem., 2017, 44(5), D.R.; Simionescu, M.; Sima, A.V. MiR-486 and miR-92a identified
1995-2004. in circulating HDL discriminate between stable and vulnerable
http://dx.doi.org/10.1159/000485905 PMID: 29237156 coronary artery disease patients. PLoS One, 2015, 10(10),
[342] Gorinski, N.; Bijata, M.; Prasad, S.; Wirth, A.; Abdel Galil, D.; e0140958.
Zeug, A.; Bazovkina, D.; Kondaurova, E.; Kulikova, E.; Ilchibaeva, http://dx.doi.org/10.1371/journal.pone.0140958 PMID: 26485305
T.; Zareba-Koziol, M.; Papaleo, F.; Scheggia, D.; [354] Song, L.; Lin, C.; Gong, H.; Wang, C.; Liu, L.; Wu, J.; Tao, S.; Hu,
Kochlamazashvili, G.; Dityatev, A.; Smyth, I.; Krzystyniak, A.; B.; Cheng, S.Y.; Li, M.; Li, J. miR-486 sustains NF-κB activity by
Wlodarczyk, J.; Richter, D.W.; Strekalova, T.; Sigrist, S.; Bang, C.; disrupting multiple NF-κB-negative feedback loops. Cell Res.,
Hobuß, L.; Fiedler, J.; Thum, T.; Naumenko, V.S.; Pandey, G.; 2013, 23(2), 274-289.
Ponimaskin, E. Attenuated palmitoylation of serotonin receptor 5- http://dx.doi.org/10.1038/cr.2012.174 PMID: 23247627
HT1A affects receptor function and contributes to depression-like [355] Mailhot, J.M.; Schuster, G.S.; Garnick, J.J.; Hanes, P.J.; Lapp, C.A.;
behaviors. Nat. Commun., 2019, 10(1), 3924. Lewis, J.B. Human periodontal ligament and gingival fibroblast
http://dx.doi.org/10.1038/s41467-019-11876-5 PMID: 31477731 response to TGF-beta 1 stimulation. J. Clin. Periodontol., 1995,
[343] Raitoharju, E.; Lyytikäinen, L.P.; Levula, M.; Oksala, N.; 22(9), 679-685.
Mennander, A.; Tarkka, M.; Klopp, N.; Illig, T.; Kähönen, M.; http://dx.doi.org/10.1111/j.1600-051X.1995.tb00826.x PMID:
Karhunen, P.J.; Laaksonen, R.; Lehtimäki, T. miR-21, miR-210, 7593697
miR-34a, and miR-146a/b are up-regulated in human atherosclerotic [356] Liu, Z.; Lu, C.L.; Cui, L.P.; Hu, Y.L.; Yu, Q.; Jiang, Y.; Ma, T.;
plaques in the tampere vascular study. Atherosclerosis, 2011, Jiao, D.K.; Wang, D.; Jia, C.Y. MicroRNA-146a modulates TGF-
219(1), 211-217. β1-induced phenotypic differentiation in human dermal fibroblasts
http://dx.doi.org/10.1016/j.atherosclerosis.2011.07.020 PMID: by targeting SMAD4. Arch. Dermatol. Res., 2012, 304(3), 195-202.
21820659 http://dx.doi.org/10.1007/s00403-011-1178-0 PMID: 21968601
[344] Radović, N.; Nikolić Jakoba, N.; Petrović, N.; Milosavljević, A.; [357] Xu, B.; Wang, N.; Wang, X.; Tong, N.; Shao, N.; Tao, J.; Li, P.;
Brković, B.; Roganović, J. MicroRNA-146a and microRNA-155 as Niu, X.; Feng, N.; Zhang, L.; Hua, L.; Wang, Z.; Chen, M. MiR-
novel crevicular fluid biomarkers for periodontitis in non-diabetic 146a suppresses tumor growth and progression by targeting EGFR
and type 2 diabetic patients. J. Clin. Periodontol., 2018, 45(6), 663- pathway and in a p-ERK-dependent manner in castration-resistant
671. prostate cancer. Prostate, 2012, 72(11), 1171-1178.
http://dx.doi.org/10.1111/jcpe.12888 PMID: 29517812 http://dx.doi.org/10.1002/pros.22466 PMID: 22161865
[345] Chen, Y.; Li, L.; Lu, Y.; Su, Q.; Sun, Y.; Liu, Y.; Yang, D. [358] O’Neill, L.A. When signaling pathways collide: positive and
Upregulation of miR-155 in CD4(+) t cells promoted th1 bias in negative regulation of toll-like receptor signal transduction.
patients with unstable angina. J. Cell. Physiol., 2015, 230(10), Immunity, 2008, 29(1), 12-20.
2498-2509. http://dx.doi.org/10.1016/j.immuni.2008.06.004 PMID: 18631453
http://dx.doi.org/10.1002/jcp.24987 PMID: 25760478 [359] Kondo, T.; Kawai, T.; Akira, S. Dissecting negative regulation of
[346] Corsten, M.F.; Papageorgiou, A.; Verhesen, W.; Carai, P.; Lindow, Toll-like receptor signaling. Trends Immunol., 2012, 33(9), 449-
M.; Obad, S.; Summer, G.; Coort, S.L.; Hazebroek, M.; van 458.
Leeuwen, R.; Gijbels, M.J.; Wijnands, E.; Biessen, E.A.; De http://dx.doi.org/10.1016/j.it.2012.05.002 PMID: 22721918
Winther, M.P.; Stassen, F.R.; Carmeliet, P.; Kauppinen, S.; [360] Lopez, J.P.; Fiori, L.M.; Cruceanu, C.; Lin, R.; Labonte, B.; Cates,
Schroen, B.; Heymans, S. MicroRNA profiling identifies H.M.; Heller, E.A.; Vialou, V.; Ku, S.M.; Gerald, C.; Han, M.H.;
microRNA-155 as an adverse mediator of cardiac injury and Foster, J.; Frey, B.N.; Soares, C.N.; Müller, D.J.; Farzan, F.; Leri,
dysfunction during acute viral myocarditis. Circ. Res., 2012, 111(4), F.; MacQueen, G.M.; Feilotter, H.; Tyryshkin, K.; Evans, K.R.;
415-425. Giacobbe, P.; Blier, P.; Lam, R.W.; Milev, R.; Parikh, S.V.;
http://dx.doi.org/10.1161/CIRCRESAHA.112.267443 PMID: Rotzinger, S.; Strother, S.C.; Lewis, C.M.; Aitchison, K.J.;
22715471 Wittenberg, G.M.; Mechawar, N.; Nestler, E.J.; Uher, R.; Kennedy,
[347] Wang, X.; Wang, B.; Zhao, J.; Liu, C.; Qu, X.; Li, Y. MiR-155 is S.H.; Turecki, G. MicroRNAs 146a/b-5 and 425-3p and 24-3p are
involved in major depression disorder and antidepressant treatment markers of antidepressant response and regulate MAPK/Wnt-system
via targeting SIRT1. Biosci. Rep., 2018, 38(6), BSR20181139 genes. Nat. Commun., 2017, 8, 15497.
http://dx.doi.org/10.1042/BSR20181139 PMID: 30482883 http://dx.doi.org/10.1038/ncomms15497 PMID: 28530238
[348] Li, T.; Cao, H.; Zhuang, J.; Wan, J.; Guan, M.; Yu, B.; Li, X.; [361] Kim, H.K.; Tyryshkin, K.; Elmi, N.; Dharsee, M.; Evans, K.R.;
Zhang, W. Identification of miR-130a, miR-27b and miR-210 as Good, J.; Javadi, M.; McCormack, S.; Vaccarino, A.L.; Zhang, X.;
serum biomarkers for atherosclerosis obliterans. Clin. Chim. Acta, Andreazza, A.C.; Feilotter, H. Plasma microRNA expression levels
2011, 412(1-2), 66-70. and their targeted pathways in patients with major depressive
http://dx.doi.org/10.1016/j.cca.2010.09.029 PMID: 20888330 disorder who are responsive to duloxetine treatment. J. Psychiatr.
[349] Schulte, C.; Molz, S.; Appelbaum, S.; Karakas, M.; Ojeda, F.; Lau, Res., 2019, 110, 38-44.
D.M.; Hartmann, T.; Lackner, K.J.; Westermann, D.; Schnabel, http://dx.doi.org/10.1016/j.jpsychires.2018.12.007 PMID: 30580082
R.B.; Blankenberg, S.; Zeller, T. miRNA-197 and miRNA-223 [362] Lagos-Quintana, M.; Rauhut, R.; Yalcin, A.; Meyer, J.; Lendeckel,
predict cardiovascular death in a cohort of patients with W.; Tuschl, T. Identification of tissue-specific microRNAs from
symptomatic coronary artery disease. PLoS One, 2015, 10(12), mouse. Curr. Biol., 2002, 12(9), 735-739.
e0145930. http://dx.doi.org/10.1016/S0960-9822(02)00809-6 PMID: 12007417
http://dx.doi.org/10.1371/journal.pone.0145930 PMID: 26720041 [363] Zhang, C. MicroRNomics: a newly emerging approach for disease
[350] Li, C.; Fang, Z.; Jiang, T.; Zhang, Q.; Liu, C.; Zhang, C.; Xiang, Y. biology. Physiol. Genomics, 2008, 33(2), 139-147.
Serum microRNAs profile from genome-wide serves as a http://dx.doi.org/10.1152/physiolgenomics.00034.2008 PMID:
fingerprint for diagnosis of acute myocardial infarction and angina 18303086
pectoris. BMC Med. Genomics, 2013, 6, 16. [364] Roy, S.; Khanna, S.; Hussain, S.R.; Biswas, S.; Azad, A.; Rink, C.;
http://dx.doi.org/10.1186/1755-8794-6-16 PMID: 23641832 Gnyawali, S.; Shilo, S.; Nuovo, G.J.; Sen, C.K. MicroRNA
[351] Yuan, X.; Berg, N.; Lee, J.W.; Le, T.T.; Neudecker, V.; Jing, N.; expression in response to murine myocardial infarction: miR-21
Eltzschig, H. MicroRNA miR-223 as regulator of innate immunity. regulates fibroblast metalloprotease-2 via phosphatase and tensin
J. Leukoc. Biol., 2018, 104(3), 515-524. homologue. Cardiovasc. Res., 2009, 82(1), 21-29.
http://dx.doi.org/10.1002/JLB.3MR0218-079R PMID: 29969525 http://dx.doi.org/10.1093/cvr/cvp015 PMID: 19147652
[352] Camkurt, M.A.; Acar, Ş.; Coşkun, S.; Güneş, M.; Güneş, S.; [365] Ji, R.; Cheng, Y.; Yue, J.; Yang, J.; Liu, X.; Chen, H.; Dean, D.B.;
Yılmaz, M.F.; Görür, A.; Tamer, L. Comparison of plasma Zhang, C. MicroRNA expression signature and antisense-mediated
MicroRNA levels in drug naive, first episode depressed patients and depletion reveal an essential role of MicroRNA in vascular
healthy controls. J. Psychiatr. Res., 2015, 69, 67-71. neointimal lesion formation. Circ. Res., 2007, 100(11), 1579-1588.
http://dx.doi.org/10.1016/j.jpsychires.2015.07.023 PMID: 26343596 http://dx.doi.org/10.1161/CIRCRESAHA.106.141986 PMID:
[353] Niculescu, L.S.; Simionescu, N.; Sanda, G.M.; Carnuta, M.G.; 17478730
Stancu, C.S.; Popescu, A.C.; Popescu, M.R.; Vlad, A.; Dimulescu,
Periodontal Pathogens and Neuropsychiatric Health Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 1389

[366] Suárez, Y.; Fernández-Hernando, C.; Pober, J.S.; Sessa, W.C. Dicer [380] Dwivedi, Y. MicroRNAs in depression and suicide: Recent insights
dependent microRNAs regulate gene expression and functions in and future perspectives. J. Affect. Disord., 2018, 240, 146-154.
human endothelial cells. Circ. Res., 2007, 100(8), 1164-1173. http://dx.doi.org/10.1016/j.jad.2018.07.075 PMID: 30071418
http://dx.doi.org/10.1161/01.RES.0000265065.26744.17 PMID: [381] Chen, S.D.; Sun, X.Y.; Niu, W.; Kong, L.M.; He, M.J.; Fan, H.M.;
17379831 Li, W.S.; Zhong, A.F.; Zhang, L.Y.; Lu, J. A preliminary analysis of
[367] Cheng, Y.; Ji, R.; Yue, J.; Yang, J.; Liu, X.; Chen, H.; Dean, D.B.; microRNA-21 expression alteration after antipsychotic treatment in
Zhang, C. MicroRNAs are aberrantly expressed in hypertrophic patients with schizophrenia. Psychiatry Res., 2016, 244, 324-332.
heart: do they play a role in cardiac hypertrophy? Am. J. Pathol., http://dx.doi.org/10.1016/j.psychres.2016.04.087 PMID: 27512922
2007, 170(6), 1831-1840. [382] Beech, R.D.; Leffert, J.J.; Lin, A.; Hong, K.A.; Hansen, J.; Umlauf,
http://dx.doi.org/10.2353/ajpath.2007.061170 PMID: 17525252 S.; Mane, S.; Zhao, H.; Sinha, R. Stress-related alcohol
[368] Al-Hayali, M.A.; Sozer, V.; Durmus, S.; Erdenen, F.; Altunoglu, E.; consumption in heavy drinkers correlates with expression of miR-
Gelisgen, R.; Atukeren, P.; Atak, P.G.; Uzun, H. Clinical value of 10a, miR-21, and components of the TAR-RNA-binding protein-
circulating microribonucleic acids miR-1 and miR-21 in evaluating associated complex. Alcohol. Clin. Exp. Res., 2014, 38(11), 2743-
the diagnosis of acute heart failure in asymptomatic type 2 diabetic 2753.
patients. Biomolecules, 2019, 9(5), E193. http://dx.doi.org/10.1111/acer.12549 PMID: 25421511
http://dx.doi.org/10.3390/biom9050193 PMID: 31109008 [383] Friedman, M.; Levin, C.E. Nutritional and medicinal aspects of D-
[369] Thum, T.; Gross, C.; Fiedler, J.; Fischer, T.; Kissler, S.; Bussen, M.; amino acids. Amino Acids, 2012, 42(5), 1553-1582.
Galuppo, P.; Just, S.; Rottbauer, W.; Frantz, S.; Castoldi, M.; http://dx.doi.org/10.1007/s00726-011-0915-1 PMID: 21519915
Soutschek, J.; Koteliansky, V.; Rosenwald, A.; Basson, M.A.; Licht, [384] Richard, D.M.; Dawes, M.A.; Mathias, C.W.; Acheson, A.; Hill-
J.D.; Pena, J.T.; Rouhanifard, S.H.; Muckenthaler, M.U.; Tuschl, T.; Kapturczak, N.; Dougherty, D.M. L-tryptophan: basic metabolic
Martin, G.R.; Bauersachs, J.; Engelhardt, S. MicroRNA-21 functions, behavioral research and therapeutic indications. Int. J.
contributes to myocardial disease by stimulating MAP kinase Tryptophan Res., 2009, 2, 45-60.
signalling in fibroblasts. Nature, 2008, 456(7224), 980-984. http://dx.doi.org/10.4137/IJTR.S2129 PMID: 20651948
http://dx.doi.org/10.1038/nature07511 PMID: 19043405 [385] Ruddick, J.P.; Evans, A.K.; Nutt, D.J.; Lightman, S.L.; Rook, G.A.;
[370] Thum, T.; Galuppo, P.; Wolf, C.; Fiedler, J.; Kneitz, S.; van Laake, Lowry, C.A. Tryptophan metabolism in the central nervous system:
L.W.; Doevendans, P.A.; Mummery, C.L.; Borlak, J.; Haverich, A.; medical implications. Expert Rev. Mol. Med., 2006, 8(20), 1-27.
Gross, C.; Engelhardt, S.; Ertl, G.; Bauersachs, J. MicroRNAs in the http://dx.doi.org/10.1017/S1462399406000068 PMID: 16942634
human heart: a clue to fetal gene reprogramming in heart failure. [386] Stone, T.W.; Darlington, L.G. Endogenous kynurenines as targets
Circulation, 2007, 116(3), 258-267. for drug discovery and development. Nat. Rev. Drug Discov., 2002,
http://dx.doi.org/10.1161/CIRCULATIONAHA.107.687947 PMID: 1(8), 609-620.
17606841 http://dx.doi.org/10.1038/nrd870 PMID: 12402501
[371] Wei, F.; Liu, D.; Feng, C.; Zhang, F.; Yang, S.; Hu, Y.; Ding, G.; [387] Vécsei, L.; Szalárdy, L.; Fülöp, F.; Toldi, J. Kynurenines in the
Wang, S. microRNA-21 mediates stretch-induced osteogenic CNS: recent advances and new questions. Nat. Rev. Drug Discov.,
differentiation in human periodontal ligament stem cells. Stem Cells 2013, 12(1), 64-82.
Dev., 2015, 24(3), 312-319. http://dx.doi.org/10.1038/nrd3793 PMID: 23237916
http://dx.doi.org/10.1089/scd.2014.0191 PMID: 25203845 [388] Chen, Y.; Guillemin, G. J. Kynurenine pathway metabolites in
[372] Li, C.; Li, C.; Yue, J.; Huang, X.; Chen, M.; Gao, J.; Wu, B. miR-21 humans: disease and healthy states. Intl. J. Tryptophan Res., 2009,
and miR-101 regulate PLAP-1 expression in periodontal ligament 2, 1-9.
cells. Mol. Med. Rep., 2012, 5(5), 1340-1346. http://dx.doi.org/10.4137/IJTR.S2097
PMID: 22367347 [389] Grant, R.S.; Coggan, S.E.; Smythe, G.A. The physiological action
[373] Wei, F.; Yang, S.; Guo, Q.; Zhang, X.; Ren, D.; Lv, T.; Xu, X. of picolinic Acid in the human brain. Int. J. Tryptophan Res., 2009,
MicroRNA-21 regulates osteogenic differentiation of periodontal 2, 71-79.
ligament stem cells by targeting smad5. Sci. Rep., 2017, 7(1), http://dx.doi.org/10.4137/IJTR.S2469 PMID: 22084583
16608. [390] Sublette, M.E.; Postolache, T.T. Neuroinflammation and
http://dx.doi.org/10.1038/s41598-017-16720-8 PMID: 29192241 depression: the role of indoleamine 2,3-dioxygenase (IDO) as a
[374] Li, X.; Guo, L.; Liu, Y.; Su, Y.; Xie, Y.; Du, J.; Wang, S.; Wang, molecular pathway. Psychosom. Med., 2012, 74(7), 668-672.
H.; Liu, Y. MicroRNA-21 promotes wound healing via the Smad7- http://dx.doi.org/10.1097/PSY.0b013e318268de9f PMID: 22923699
Smad2/3-Elastin pathway. Exp. Cell Res., 2018, 362(2), 245-251. [391] Serafini, G.; Adavastro, G.; Canepa, G.; Capobianco, L.; Conigliaro,
http://dx.doi.org/10.1016/j.yexcr.2017.11.019 PMID: 29154818 C.; Pittaluga, F.; Murri, M.B.; Valchera, A.; De Berardis, D.;
[375] Niu, J.; Shi, Y.; Tan, G.; Yang, C.H.; Fan, M.; Pfeffer, L.M.; Wu, Pompili, M.; Lindqvist, D.; Brundin, L.; Amore, M. Abnormalities
Z.H. DNA damage induces NF-κB-dependent microRNA-21 up- in kynurenine pathway metabolism in treatment-resistant depression
regulation and promotes breast cancer cell invasion. J. Biol. Chem., and suicidality: a systematic review. CNS Neurol. Disord. Drug
2012, 287(26), 21783-21795. Targets, 2017, 16(4), 440-453.
http://dx.doi.org/10.1074/jbc.M112.355495 PMID: 22547075 http://dx.doi.org/10.2174/1871527316666170413110605 PMID:
[376] Kocerha, J.; Dwivedi, Y.; Brennand, K.J. Noncoding RNAs and 28412922
neurobehavioral mechanisms in psychiatric disease. Mol. [392] Skolnick, P.; Layer, R.T.; Popik, P.; Nowak, G.; Paul, I.A.; Trullas,
Psychiatry, 2015, 20(6), 677-684. R. Adaptation of N-methyl-D-aspartate (NMDA) receptors
http://dx.doi.org/10.1038/mp.2015.30 PMID: 25824307 following antidepressant treatment: implications for the
[377] Dwivedi, Y. Emerging role of microRNAs in major depressive pharmacotherapy of depression. Pharmacopsychiatry, 1996, 29(1),
disorder: diagnosis and therapeutic implications. Dialogues Clin. 23-26.
Neurosci., 2014, 16(1), 43-61. http://dx.doi.org/10.1055/s-2007-979537 PMID: 8852530
PMID: 24733970 [393] Trullas, R.; Skolnick, P. Functional antagonists at the NMDA
[378] Miranda, R.C.; Pietrzykowski, A.Z.; Tang, Y.; Sathyan, P.; receptor complex exhibit antidepressant actions. Eur. J. Pharmacol.,
Mayfield, D.; Keshavarzian, A.; Sampson, W.; Hereld, D. 1990, 185(1), 1-10.
MicroRNAs: master regulators of ethanol abuse and toxicity? http://dx.doi.org/10.1016/0014-2999(90)90204-J PMID: 2171955
Alcohol. Clin. Exp. Res., 2010, 34(4), 575-587. [394] Nowak, G.; Ordway, G.A.; Paul, I.A. Alterations in the N-methyl-
http://dx.doi.org/10.1111/j.1530-0277.2009.01126.x PMID: D-aspartate (NMDA) receptor complex in the frontal cortex of
20102566 suicide victims. Brain Res., 1995, 675(1-2), 157-164.
[379] Issler, O.; Haramati, S.; Paul, E.D.; Maeno, H.; Navon, I.; Zwang, http://dx.doi.org/10.1016/0006-8993(95)00057-W PMID: 7796124
R.; Gil, S.; Mayberg, H.S.; Dunlop, B.W.; Menke, A.; Awatramani, [395] Krystal, J.H.; Sanacora, G.; Blumberg, H.; Anand, A.; Charney,
R.; Binder, E.B.; Deneris, E.S.; Lowry, C.A.; Chen, A. MicroRNA D.S.; Marek, G.; Epperson, C.N.; Goddard, A.; Mason, G.F.
135 is essential for chronic stress resiliency, antidepressant efficacy, Glutamate and GABA systems as targets for novel antidepressant
and intact serotonergic activity. Neuron, 2014, 83(2), 344-360. and mood-stabilizing treatments. Mol. Psychiatry, 2002, 7(Suppl.
http://dx.doi.org/10.1016/j.neuron.2014.05.042 PMID: 24952960 1), S71-S80.
1390 Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 Wadhawan et al.

http://dx.doi.org/10.1038/sj.mp.4001021 PMID: 11986998 [408] Schwarcz, R.; Rassoulpour, A.; Wu, H.Q.; Medoff, D.; Tamminga,
[396] Zarate, C.A.; Quiroz, J.; Payne, J.; Manji, H.K. Modulators of the C.A.; Roberts, R.C. Increased cortical kynurenate content in
glutamatergic system: implications for the development of improved schizophrenia. Biol. Psychiatry, 2001, 50(7), 521-530.
therapeutics in mood disorders. Psychopharmacol. Bull., 2002, http://dx.doi.org/10.1016/S0006-3223(01)01078-2 PMID: 11600105
36(4), 35-83. [409] Erhardt, S.; Blennow, K.; Nordin, C.; Skogh, E.; Lindström, L.H.;
PMID: 12858143 Engberg, G. Kynurenic acid levels are elevated in the cerebrospinal
[397] Erhardt, S.; Lim, C.K.; Linderholm, K.R.; Janelidze, S.; Lindqvist, fluid of patients with schizophrenia. Neurosci. Lett., 2001, 313(1-2),
D.; Samuelsson, M.; Lundberg, K.; Postolache, T.T.; Träskman- 96-98.
Bendz, L.; Guillemin, G.J.; Brundin, L. Connecting inflammation http://dx.doi.org/10.1016/S0304-3940(01)02242-X PMID:
with glutamate agonism in suicidality. Neuropsychopharmacology, 11684348
2013, 38(5), 743-752. [410] Nilsson, L.K.; Linderholm, K.R.; Engberg, G.; Paulson, L.;
http://dx.doi.org/10.1038/npp.2012.248 PMID: 23299933 Blennow, K.; Lindström, L.H.; Nordin, C.; Karanti, A.; Persson, P.;
[398] Bryleva, E. Y.; Brundin, L. Kynurenine pathway metabolites and Erhardt, S. Elevated levels of kynurenic acid in the cerebrospinal
suicidality. Neuropharmacology , 2017, 112 (Pt B), 324-330 fluid of male patients with schizophrenia. Schizophr. Res., 2005,
http://dx.doi.org/10.1016/j.neuropharm.2016.01.034 80(2-3), 315-322.
[399] Sublette, M.E.; Galfalvy, H.C.; Fuchs, D.; Lapidus, M.; Grunebaum, http://dx.doi.org/10.1016/j.schres.2005.07.013 PMID: 16125901
M.F.; Oquendo, M.A.; Mann, J.J.; Postolache, T.T. Plasma [411] Linderholm, K.R.; Alm, M.T.; Larsson, M.K.; Olsson, S.K.; Goiny,
kynurenine levels are elevated in suicide attempters with major M.; Hajos, M.; Erhardt, S.; Engberg, G. Inhibition of kynurenine
depressive disorder. Brain Behav. Immun., 2011, 25(6), 1272-1278. aminotransferase II reduces activity of midbrain dopamine neurons.
http://dx.doi.org/10.1016/j.bbi.2011.05.002 PMID: 21605657 Neuropharmacology, 2016, 102, 42-47.
[400] Brundin, L.; Sellgren, C.M.; Lim, C.K.; Grit, J.; Pålsson, E.; http://dx.doi.org/10.1016/j.neuropharm.2015.10.028 PMID:
Landén, M.; Samuelsson, M.; Lundgren, K.; Brundin, P.; Fuchs, D.; 26514401
Postolache, T.T.; Traskman-Bendz, L.; Guillemin, G.J.; Erhardt, S. [412] Sathyasaikumar, K.V.; Stachowski, E.K.; Wonodi, I.; Roberts, R.C.;
An enzyme in the kynurenine pathway that governs vulnerability to Rassoulpour, A.; McMahon, R.P.; Schwarcz, R. Impaired
suicidal behavior by regulating excitotoxicity and kynurenine pathway metabolism in the prefrontal cortex of
neuroinflammation. Transl. Psychiatry, 2016, 6(8), e865. individuals with schizophrenia. Schizophr. Bull., 2011, 37(6), 1147-
http://dx.doi.org/10.1038/tp.2016.133 PMID: 27483383 1156.
[401] Reininghaus, E.Z.; McIntyre, R.S.; Reininghaus, B.; Geisler, S.; http://dx.doi.org/10.1093/schbul/sbq112 PMID: 21036897
Bengesser, S.A.; Lackner, N.; Hecht, K.; Birner, A.; Kattnig, F.; [413] Plitman, E.; Iwata, Y.; Caravaggio, F.; Nakajima, S.; Chung, J.K.;
Unterweger, R.; Kapfhammer, H.P.; Zelzer, S.; Fuchs, D.; Mangge, Gerretsen, P.; Kim, J.; Takeuchi, H.; Chakravarty, M.M.;
H. Tryptophan breakdown is increased in euthymic overweight Remington, G.; Graff-Guerrero, A. Kynurenic acid in
individuals with bipolar disorder: a preliminary report. Bipolar schizophrenia: a systematic review and meta-analysis. Schizophr.
Disord., 2014, 16(4), 432-440. Bull., 2017, 43(4), 764-777.
http://dx.doi.org/10.1111/bdi.12166 PMID: 24330408 http://dx.doi.org/10.1093/schbul/sbw221 PMID: 28187219
[402] Birner, A.; Platzer, M.; Bengesser, S.A.; Dalkner, N.; Fellendorf, [414] Giil, L.M.; Midttun, Ø.; Refsum, H.; Ulvik, A.; Advani, R.; Smith,
F.T.; Queissner, R.; Pilz, R.; Rauch, P.; Maget, A.; Hamm, C.; A.D.; Ueland, P.M. Kynurenine pathway metabolites in alzheimer’s
Herzog-Eberhard, S.; Mangge, H.; Fuchs, D.; Moll, N.; Zelzer, S.; disease. J. Alzheimers Dis., 2017, 60(2), 495-504.
Schütze, G.; Schwarz, M.; Reininghaus, B.; Kapfhammer, H.P.; http://dx.doi.org/10.3233/JAD-170485 PMID: 28869479
Reininghaus, E.Z. Increased breakdown of kynurenine towards its [415] Busse, M.; Hettler, V.; Fischer, V.; Mawrin, C.; Hartig, R.;
neurotoxic branch in bipolar disorder. PLoS One, 2017, 12(2), Dobrowolny, H.; Bogerts, B.; Frodl, T.; Busse, S. Increased
e0172699. quinolinic acid in peripheral mononuclear cells in Alzheimer’s
http://dx.doi.org/10.1371/journal.pone.0172699 PMID: 28241062 dementia. Eur. Arch. Psychiatry Clin. Neurosci., 2018, 268(5), 493-
[403] Maget, A.; Platzer, M.; Bengesser, S.A.; Fellendorf, F.T.; Birner, 500.
A.; Queissner, R.; Hamm, C.; Reininghaus, B.; Hecker, A.; http://dx.doi.org/10.1007/s00406-017-0785-y PMID: 28386767
Tomberger, L.; Pilz, R.; Dalkner, N.; Moll, N.; Schutze, G.; [416] Buczko, P.; Stokowska, W.; Górska, M.; Kucharewicz, I.; Pawlak,
Schwarz, M.; Kapfhammer, H.P.; Reininghaus, E.Z. Differences in D.; Buczko, W. Tryptophan metabolites via kynurenine pathway in
kynurenine metabolism during depressive, manic, and euthymic saliva of diabetic patients. Dent. Med. Probl, 2006, 43(1), 21-25.
phases of bipolar affective disorder. Curr. Top. Med. Chem., 2019. [417] Buczko, W.; Cylwik, D.; Stokowska, W. [Metabolism of tryptophan
[Epub ahead of print] via the kynurenine pathway in saliva]. Postepy Hig. Med. Dosw.,
http://dx.doi.org/10.2174/1568026619666190802145128 2005, 59, 283-289.
[404] Platzer, M.; Dalkner, N.; Fellendorf, F.T.; Birner, A.; Bengesser, PMID: 15995595
S.A.; Queissner, R.; Kainzbauer, N.; Pilz, R.; Herzog-Eberhard, S.; [418] Nisapakultorn, K.; Makrudthong, J.; Sa-Ard-Iam, N.; Rerkyen, P.;
Hamm, C.; Hörmanseder, C.; Maget, A.; Rauch, P.; Mangge, H.; Mahanonda, R.; Takikawa, O. Indoleamine 2,3-dioxygenase
Fuchs, D.; Zelzer, S.; Schütze, G.; Moll, N.; Schwarz, M.J.; Mansur, expression and regulation in chronic periodontitis. J. Periodontol.,
R.B.; McIntyre, R.S.; Reininghaus, E.Z. Tryptophan breakdown and 2009, 80(1), 114-121.
cognition in bipolar disorder. Psychoneuroendocrinology, 2017, 81, http://dx.doi.org/10.1902/jop.2009.080315 PMID: 19228097
144-150. [419] Moon, J.S.; Cheong, N.R.; Yang, S.Y.; Kim, I.S.; Chung, H.J.;
http://dx.doi.org/10.1016/j.psyneuen.2017.04.015 PMID: 28482311 Jeong, Y.W.; Park, J.C.; Kim, M.S.; Kim, S.H.; Ko, H.M.
[405] Erhardt, S.; Schwieler, L.; Imbeault, S.; Engberg, G. The Lipopolysaccharide-induced indoleamine 2,3-dioxygenase
kynurenine pathway in schizophrenia and bipolar disorder. expression in the periodontal ligament. J. Periodontal Res., 2013,
Neuropharmacology , 2017, 112 (Pt B), 297-306 48(6), 733-739.
http://dx.doi.org/10.1016/j.neuropharm.2016.05.020 http://dx.doi.org/10.1111/jre.12063 PMID: 23488665
[406] Stone, T.W. Neuropharmacology of quinolinic and kynurenic acids. [420] Wolowczuk, I.; Hennart, B.; Leloire, A.; Bessede, A.; Soichot, M.;
Pharmacol. Rev., 1993, 45(3), 309-379. Taront, S.; Caiazzo, R.; Raverdy, V.; Pigeyre, M.; Guillemin, G.J.;
PMID: 8248282 Allorge, D.; Pattou, F.; Froguel, P.; Poulain-Godefroy, O. ABOS
[407] Hilmas, C.; Pereira, E.F.; Alkondon, M.; Rassoulpour, A.; Consortium. Tryptophan metabolism activation by indoleamine 2,3-
Schwarcz, R.; Albuquerque, E.X. The brain metabolite kynurenic dioxygenase in adipose tissue of obese women: an attempt to
acid inhibits alpha7 nicotinic receptor activity and increases non- maintain immune homeostasis and vascular tone. Am. J. Physiol.
alpha7 nicotinic receptor expression: physiopathological Regul. Integr. Comp. Physiol., 2012, 303(2), R135-R143.
implications. J. Neurosci., 2001, 21(19), 7463-7473. http://dx.doi.org/10.1152/ajpregu.00373.2011 PMID: 22592557
http://dx.doi.org/10.1523/JNEUROSCI.21-19-07463.2001 PMID: [421] Favennec, M.; Hennart, B.; Caiazzo, R.; Leloire, A.; Yengo, L.;
11567036 Verbanck, M.; Arredouani, A.; Marre, M.; Pigeyre, M.; Bessede, A.;
Guillemin, G.J.; Chinetti, G.; Staels, B.; Pattou, F.; Balkau, B.;
Allorge, D.; Froguel, P.; Poulain-Godefroy, O. The kynurenine
Periodontal Pathogens and Neuropsychiatric Health Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 1391

pathway is activated in human obesity and shifted toward [435] McGeer, P.L.; Itagaki, S.; Tago, H.; McGeer, E.G. Reactive
kynurenine monooxygenase activation. Obesity (Silver Spring), microglia in patients with senile dementia of the Alzheimer type are
2015, 23(10), 2066-2074. positive for the histocompatibility glycoprotein HLA-DR. Neurosci.
http://dx.doi.org/10.1002/oby.21199 PMID: 26347385 Lett., 1987, 79(1-2), 195-200.
[422] Dalkner, N.; Platzer, M.; Bengesser, S.A.; Birner, A.; Fellendorf, http://dx.doi.org/10.1016/0304-3940(87)90696-3 PMID: 3670729
F.T.; Queissner, R.; Painold, A.; Mangge, H.; Fuchs, D.; [436] Torres-Platas, S.G.; Cruceanu, C.; Chen, G.G.; Turecki, G.;
Reininghaus, B.; Kapfhammer, H.P.; Holasek, S.J.; Reininghaus, Mechawar, N. Evidence for increased microglial priming and
E.Z. The role of tryptophan metabolism and food craving in the macrophage recruitment in the dorsal anterior cingulate white
relationship between obesity and bipolar disorder. Clin. Nutr., 2018, matter of depressed suicides. Brain Behav. Immun., 2014, 42, 50-59.
37(5), 1744-1751. http://dx.doi.org/10.1016/j.bbi.2014.05.007 PMID: 24858659
http://dx.doi.org/10.1016/j.clnu.2017.06.024 PMID: 28712531 [437] Su, L.; Faluyi, Y.O.; Hong, Y.T.; Fryer, T.D.; Mak, E.; Gabel, S.;
[423] Mangge, H.; Stelzer, I.; Reininghaus, E.Z.; Weghuber, D.; Hayes, L.; Soteriades, S.; Williams, G.B.; Arnold, R.; Passamonti,
Postolache, T.T.; Fuchs, D. Disturbed tryptophan metabolism in L.; Rodríguez, P.V.; Surendranathan, A.; Bevan-Jones, R.W.; Coles,
cardiovascular disease. Curr. Med. Chem., 2014, 21(17), 1931- J.; Aigbirhio, F.; Rowe, J.B.; O’Brien, J.T. Neuroinflammatory and
1937. morphological changes in late-life depression: the NIMROD study.
http://dx.doi.org/10.2174/0929867321666140304105526 PMID: Br. J. Psychiatry, 2016, 209(6), 525-526.
24606499 http://dx.doi.org/10.1192/bjp.bp.116.190165 PMID: 27758838
[424] Zuo, H.; Ueland, P.M.; Ulvik, A.; Eussen, S.J.; Vollset, S.E.; [438] van Berckel, B.N.; Bossong, M.G.; Boellaard, R.; Kloet, R.;
Nygård, O.; Midttun, Ø.; Theofylaktopoulou, D.; Meyer, K.; Tell, Schuitemaker, A.; Caspers, E.; Luurtsema, G.; Windhorst, A.D.;
G.S. Plasma biomarkers of inflammation, the kynurenine pathway, Cahn, W.; Lammertsma, A.A.; Kahn, R.S. Microglia activation in
and risks of all-cause, cancer, and cardiovascular disease mortality: recent-onset schizophrenia: a quantitative (R)-[11C]PK11195
the hordaland health study. Am. J. Epidemiol., 2016, 183(4), 249- positron emission tomography study. Biol. Psychiatry, 2008, 64(9),
258. 820-822.
http://dx.doi.org/10.1093/aje/kwv242 PMID: 26823439 http://dx.doi.org/10.1016/j.biopsych.2008.04.025 PMID: 18534557
[425] Niinisalo, P.; Oksala, N.; Levula, M.; Pelto-Huikko, M.; Järvinen, [439] Fillman, S.G.; Cloonan, N.; Catts, V.S.; Miller, L.C.; Wong, J.;
O.; Salenius, J.P.; Kytömäki, L.; Soini, J.T.; Kähönen, M.; McCrossin, T.; Cairns, M.; Weickert, C.S. Increased inflammatory
Laaksonen, R.; Hurme, M.; Lehtimäki, T. Activation of indoleamine markers identified in the dorsolateral prefrontal cortex of
2,3-dioxygenase-induced tryptophan degradation in advanced individuals with schizophrenia. Mol. Psychiatry, 2013, 18(2), 206-
atherosclerotic plaques: Tampere vascular study. Ann. Med., 2010, 214.
42(1), 55-63. http://dx.doi.org/10.1038/mp.2012.110 PMID: 22869038
http://dx.doi.org/10.3109/07853890903321559 PMID: 19941414 [440] Doorduin, J.; de Vries, E.F.; Willemsen, A.T.; de Groot, J.C.;
[426] Niinisalo, P.; Raitala, A.; Pertovaara, M.; Oja, S.S.; Lehtimäki, T.; Dierckx, R.A.; Klein, H.C. Neuroinflammation in schizophrenia-
Kähönen, M.; Reunanen, A.; Jula, A.; Moilanen, L.; Kesäniemi, related psychosis: a PET study. J. Nucl. Med., 2009, 50(11), 1801-
Y.A.; Nieminen, M.S.; Hurme, M. Indoleamine 2,3-dioxygenase 1807.
activity associates with cardiovascular risk factors: the Health 2000 http://dx.doi.org/10.2967/jnumed.109.066647 PMID: 19837763
study. Scand. J. Clin. Lab. Invest., 2008, 68(8), 767-770. [441] Kessler, R.C. The effects of stressful life events on depression.
http://dx.doi.org/10.1080/00365510802245685 PMID: 18622801 Annu. Rev. Psychol., 1997, 48, 191-214.
[427] Pedersen, E.R.; Tuseth, N.; Eussen, S.J.; Ueland, P.M.; Strand, E.; http://dx.doi.org/10.1146/annurev.psych.48.1.191 PMID: 9046559
Svingen, G.F.; Midttun, Ø.; Meyer, K.; Mellgren, G.; Ulvik, A.; [442] Hammen, C. Stress and depression. Annu. Rev. Clin. Psychol., 2005,
Nordrehaug, J.E.; Nilsen, D.W.; Nygård, O. Associations of plasma 1, 293-319.
kynurenines with risk of acute myocardial infarction in patients with http://dx.doi.org/10.1146/annurev.clinpsy.1.102803.143938 PMID:
stable angina pectoris. Arterioscler. Thromb. Vasc. Biol., 2015, 17716090
35(2), 455-462. [443] Krishnan, V.; Nestler, E.J. The molecular neurobiology of
http://dx.doi.org/10.1161/ATVBAHA.114.304674 PMID: 25524770 depression. Nature, 2008, 455(7215), 894-902.
[428] Ormstad, H.; Verkerk, R.; Amthor, K.F.; Sandvik, L. Activation of http://dx.doi.org/10.1038/nature07455 PMID: 18923511
the kynurenine pathway in the acute phase of stroke and its role in [444] Cohen, S.; Tyrrell, D.A.; Smith, A.P. Psychological stress and
fatigue and depression following stroke. J. Mol. Neurosci., 2014, susceptibility to the common cold. N. Engl. J. Med., 1991, 325(9),
54(2), 181-187. 606-612.
http://dx.doi.org/10.1007/s12031-014-0272-0 PMID: 24664436 http://dx.doi.org/10.1056/NEJM199108293250903 PMID: 1713648
[429] Gold, A.B.; Herrmann, N.; Swardfager, W.; Black, S.E.; Aviv, R.I.; [445] Genco, R.J.; Ho, A.W.; Grossi, S.G.; Dunford, R.G.; Tedesco, L.A.
Tennen, G.; Kiss, A.; Lanctôt, K.L. The relationship between Relationship of stress, distress and inadequate coping behaviors to
indoleamine 2,3-dioxygenase activity and post-stroke cognitive periodontal disease. J. Periodontol., 1999, 70(7), 711-723.
impairment. J. Neuroinflammation, 2011, 8, 17. http://dx.doi.org/10.1902/jop.1999.70.7.711 PMID: 10440631
http://dx.doi.org/10.1186/1742-2094-8-17 PMID: 21324164 [446] Vedhara, K.; Bennett, P.D.; Clark, S.; Lightman, S.L.; Shaw, S.;
[430] Singhrao, S.K.; Olsen, I. Assessing the role of Porphyromonas Perks, P.; Hunt, M.A.; Philip, J.M.; Tallon, D.; Murphy, P.J.; Jones,
gingivalis in periodontitis to determine a causative relationship with R.W.; Wilcock, G.K.; Shanks, N.M. Enhancement of antibody
Alzheimer’s disease. J. Oral Microbiol., 2019, 11(1), 1563405. responses to influenza vaccination in the elderly following a
http://dx.doi.org/10.1080/20002297.2018.1563405 PMID: cognitive-behavioural stress management intervention. Psychother.
30728914 Psychosom., 2003, 72(5), 245-252.
[431] Sochocka, M.; Zwolińska, K.; Leszek, J. The Infectious Etiology of http://dx.doi.org/10.1159/000071895 PMID: 12920328
Alzheimer’s Disease. Curr. Neuropharmacol., 2017, 15(7), 996- [447] Nadkarni, R.B.; Fristad, M.A. Stress and support for parents of
1009. youth with bipolar disorder. Isr. J. Psychiatry Relat. Sci., 2012,
http://dx.doi.org/10.2174/1570159X15666170313122937 PMID: 49(2), 104-110.
28294067 PMID: 22801289
[432] Hashioka, S.; Inoue, K.; Hayashida, M.; Wake, R.; Oh-Nishi, A.; [448] Caserta, M.T.; O’Connor, T.G.; Wyman, P.A.; Wang, H.;
Miyaoka, T. Implications of systemic inflammation and Moynihan, J.; Cross, W.; Tu, X.; Jin, X. The associations between
periodontitis for major depression. Front. Neurosci., 2018, 12, 483. psychosocial stress and the frequency of illness, and innate and
http://dx.doi.org/10.3389/fnins.2018.00483 PMID: 30072865 adaptive immune function in children. Brain Behav. Immun., 2008,
[433] McGeer, P.L.; McGeer, E.G. History of innate immunity in 22(6), 933-940.
neurodegenerative disorders. Front. Pharmacol., 2011, 2, 77. http://dx.doi.org/10.1016/j.bbi.2008.01.007 PMID: 18308510
http://dx.doi.org/10.3389/fphar.2011.00077 PMID: 22144960 [449] Kiecolt-Glaser, J.K.; Page, G.G.; Marucha, P.T.; MacCallum, R.C.;
[434] McGeer, P.L.; McGeer, E.; Rogers, J.; Sibley, J. Anti-inflammatory Glaser, R. Psychological influences on surgical recovery.
drugs and Alzheimer disease. Lancet, 1990, 335(8696), 1037. Perspectives from psychoneuroimmunology. Am. Psychol., 1998,
http://dx.doi.org/10.1016/0140-6736(90)91101-F PMID: 1970087 53(11), 1209-1218.
1392 Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 Wadhawan et al.

http://dx.doi.org/10.1037/0003-066X.53.11.1209 PMID: 9830373 [465] Sheline, Y.I.; Mittler, B.L.; Mintun, M.A. The hippocampus and
[450] Kiecolt-Glaser, J.K.; Marucha, P.T.; Malarkey, W.B.; Mercado, depression. Eur. Psychiatry, 2002, 17(Suppl. 3), 300-305.
A.M.; Glaser, R. Slowing of wound healing by psychological stress. http://dx.doi.org/10.1016/S0924-9338(02)00655-7 PMID: 15177085
Lancet, 1995, 346(8984), 1194-1196. [466] Maes, M.; Yirmyia, R.; Noraberg, J.; Brene, S.; Hibbeln, J.; Perini,
http://dx.doi.org/10.1016/S0140-6736(95)92899-5 PMID: 7475659 G.; Kubera, M.; Bob, P.; Lerer, B.; Maj, M. The inflammatory &
[451] Kiecolt-Glaser, J.K.; Loving, T.J.; Stowell, J.R.; Malarkey, W.B.; neurodegenerative (I&ND) hypothesis of depression: leads for
Lemeshow, S.; Dickinson, S.L.; Glaser, R. Hostile marital future research and new drug developments in depression. Metab.
interactions, proinflammatory cytokine production, and wound Brain Dis., 2009, 24(1), 27-53.
healing. Arch. Gen. Psychiatry, 2005, 62(12), 1377-1384. http://dx.doi.org/10.1007/s11011-008-9118-1 PMID: 19085093
http://dx.doi.org/10.1001/archpsyc.62.12.1377 PMID: 16330726 [467] Myint, A.M.; Kim, Y.K. Cytokine-serotonin interaction through
[452] Marucha, P.T.; Kiecolt-Glaser, J.K.; Favagehi, M. Mucosal wound IDO: a neurodegeneration hypothesis of depression. Med.
healing is impaired by examination stress. Psychosom. Med., 1998, Hypotheses, 2003, 61(5-6), 519-525.
60(3), 362-365. http://dx.doi.org/10.1016/S0306-9877(03)00207-X PMID:
http://dx.doi.org/10.1097/00006842-199805000-00025 PMID: 14592780
9625226 [468] Khabazghazvini, B.; Groer, M.; Fuchs, D.; Strassle, P.; Lapidus, M.;
[453] Pariante, C.M.; Miller, A.H. Glucocorticoid receptors in major Sleemi, A.; Cabassa, J.B.; Postolache, T.T. Psychiatric
depression: relevance to pathophysiology and treatment. Biol. manifestations of latent toxoplasmosis. Potential mediation by
Psychiatry, 2001, 49(5), 391-404. indoleamine 2, 3-dioxygenase. Int. J. Disabil. Hum. Dev., 2010,
http://dx.doi.org/10.1016/S0006-3223(00)01088-X PMID: 9(1), 3-10.
11274650 http://dx.doi.org/10.1515/IJDHD.2010.002
[454] Raison, C.L.; Capuron, L.; Miller, A.H. Cytokines sing the blues: [469] Wadhawan, A.; Daue, M.L.; Brenner, L.A.; Lowry, C.A.; Dagdag,
inflammation and the pathogenesis of depression. Trends Immunol., A.; Stiller, J.W.; Benros, M.E.; Erlangsen, A.; Baca-Garcia, E.;
2006, 27(1), 24-31. Hoisington, A.J. F158. Toxoplasma Gondii-Oocyst Seropositivity
http://dx.doi.org/10.1016/j.it.2005.11.006 PMID: 16316783 and Depression in the Old Order Amish. Biol. Psychiatry, 2018,
[455] Pollak, Y.; Yirmiya, R. Cytokine-induced changes in mood and 83(9), S299-S300.
behaviour: implications for ‘depression due to a general medical http://dx.doi.org/10.1016/j.biopsych.2018.02.772
condition’, immunotherapy and antidepressive treatment. Int. J. [470] Wadhawan, A.; Dagdag, A.; Duffy, A.; Daue, M.L.; Ryan, K.A.;
Neuropsychopharmacol., 2002, 5(4), 389-399. Brenner, L.A.; Stiller, J.W.; Pollin, T.I.; Groer, M.W.; Huang, X.;
http://dx.doi.org/10.1017/S1461145702003152 PMID: 12466037 Lowry, C.A.; Mitchell, B.D.; Postolache, T.T. Positive association
[456] Leonard, B.E. Inflammation and depression: a causal or coincidental between Toxoplasma gondii IgG serointensity and current
link to the pathophysiology? Acta Neuropsychiatr., 2018, 30(1), 1- dysphoria/hopelessness scores in the Old Order Amish: a
16. preliminary study. Pteridines, 2017, 28(3-4), 185-194.
http://dx.doi.org/10.1017/neu.2016.69 PMID: 28112061 http://dx.doi.org/10.1515/pterid-2017-0019 PMID: 29657363
[457] Sugama, S.; Fujita, M.; Hashimoto, M.; Conti, B. Stress induced [471] Postolache, T.; Constantine, N.; Daue, M.; Dagdag, A.; Wadhawan,
morphological microglial activation in the rodent brain: A.; Brenner, L.A.; Lowry, C.A.; Makkar, H.; Reynolds, M.A. S91.
involvement of interleukin-18. Neuroscience, 2007, 146(3), 1388- P. Gingivalis and Cardinal Symptoms of Depression. Biol.
1399. Psychiatry, 2019, 85(10), S332.
http://dx.doi.org/10.1016/j.neuroscience.2007.02.043 PMID: http://dx.doi.org/10.1016/j.biopsych.2019.03.842
17433555 [472] Myint, A-M.; Leonard, B.E.; Steinbusch, H.W.; Kim, Y-K. Th1,
[458] Steiner, J.; Bielau, H.; Brisch, R.; Danos, P.; Ullrich, O.; Mawrin, Th2, and Th3 cytokine alterations in major depression. J. Affect.
C.; Bernstein, H.G.; Bogerts, B. Immunological aspects in the Disord., 2005, 88(2), 167-173.
neurobiology of suicide: elevated microglial density in http://dx.doi.org/10.1016/j.jad.2005.07.008 PMID: 16126278
schizophrenia and depression is associated with suicide. J. [473] Kaestner, F.; Hettich, M.; Peters, M.; Sibrowski, W.; Hetzel, G.;
Psychiatr. Res., 2008, 42(2), 151-157. Ponath, G.; Arolt, V.; Cassens, U.; Rothermundt, M. Different
http://dx.doi.org/10.1016/j.jpsychires.2006.10.013 PMID: 17174336 activation patterns of proinflammatory cytokines in melancholic and
[459] Glaser, R.; Robles, T.F.; Sheridan, J.; Malarkey, W.B.; Kiecolt- non-melancholic major depression are associated with HPA axis
Glaser, J.K. Mild depressive symptoms are associated with activity. J. Affect. Disord., 2005, 87(2-3), 305-311.
amplified and prolonged inflammatory responses after influenza http://dx.doi.org/10.1016/j.jad.2005.03.012 PMID: 15951024
virus vaccination in older adults. Arch. Gen. Psychiatry, 2003, [474] Schiepers, O.J.; Wichers, M.C.; Maes, M. Cytokines and major
60(10), 1009-1014. depression. Prog. Neuropsychopharmacol. Biol. Psychiatry, 2005,
http://dx.doi.org/10.1001/archpsyc.60.10.1009 PMID: 14557146 29(2), 201-217.
[460] Ongür, D.; Drevets, W.C.; Price, J.L. Glial reduction in the http://dx.doi.org/10.1016/j.pnpbp.2004.11.003 PMID: 15694227
subgenual prefrontal cortex in mood disorders. Proc. Natl. Acad. [475] Maes, M. Cytokines and major depression. Prog. Neuropsycho-
Sci. USA, 1998, 95(22), 13290-13295. pharmacol. Biol. Psychiatry, 2005, 9(2), 201-217.
http://dx.doi.org/10.1073/pnas.95.22.13290 PMID: 9789081 http://dx.doi.org/10.1016/0006-3223(94)90652-1
[461] Myint, A.M. Inflammation, neurotoxins and psychiatric disorders. [476] Kim, Y-K.; Myint, A-M.; Lee, B-H.; Han, C-S.; Lee, H-J.; Kim, D-
Mod. Trends Pharmacopsychiatry, 2013, 28, 61-74. J.; Leonard, B.E. Th1, Th2 and Th3 cytokine alteration in
http://dx.doi.org/10.1159/000343968 PMID: 25224891 schizophrenia. Prog. Neuropsychopharmacol. Biol. Psychiatry,
[462] Duman, R.S.; Heninger, G.R.; Nestler, E.J. A molecular and cellular 2004, 28(7), 1129-1134.
theory of depression. Arch. Gen. Psychiatry, 1997, 54(7), 597-606. http://dx.doi.org/10.1016/j.pnpbp.2004.05.047 PMID: 15610925
http://dx.doi.org/10.1001/archpsyc.1997.01830190015002 PMID: [477] Rapaport, M.H.; McAllister, C.G.; Pickar, D.; Nelson, D.L.; Paul,
9236543 S.M. Elevated levels of soluble interleukin 2 receptors in
[463] Yeager, M.P.; Pioli, P.A.; Guyre, P.M. Cortisol exerts bi-phasic schizophrenia. Arch. Gen. Psychiatry, 1989, 46(3), 291-292.
regulation of inflammation in humans. Dose Response, 2011, 9(3), http://dx.doi.org/10.1001/archpsyc.1989.01810030097017 PMID:
332-347. 2784047
http://dx.doi.org/10.2203/dose-response.10-013.Yeager PMID: [478] Cazzullo, C.L.; Sacchetti, E.; Galluzzo, A.; Panariello, A.;
22013396 Colombo, F.; Zagliani, A.; Clerici, M. Cytokine profiles in drug-
[464] Smyth, G.P.; Stapleton, P.P.; Freeman, T.A.; Concannon, E.M.; naive schizophrenic patients. Schizophr. Res., 2001, 47(2-3), 293-
Mestre, J.R.; Duff, M.; Maddali, S.; Daly, J.M. Glucocorticoid 298.
pretreatment induces cytokine overexpression and nuclear factor- http://dx.doi.org/10.1016/S0920-9964(00)00046-3 PMID: 11278147
kappaB activation in macrophages. J. Surg. Res., 2004, 116(2), 253- [479] Moises, H.W.; Schindler, L.; Leroux, M.; Kirchner, H. Decreased
261. production of interferon alpha and interferon gamma in leucocyte
http://dx.doi.org/10.1016/S0022-4804(03)00300-7 PMID: 15013364 cultures of schizophrenic patients. Acta Psychiatr. Scand., 1985,
72(1), 45-50.
Periodontal Pathogens and Neuropsychiatric Health Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 1393

http://dx.doi.org/10.1111/j.1600-0447.1985.tb02569.x PMID: T.T. Elevated cytokine expression in the orbitofrontal cortex of


3929565 victims of suicide. Acta Psychiatr. Scand., 2008, 117(3), 198-206.
[480] Stertz, L.; Magalhães, P.V.; Kapczinski, F. Is bipolar disorder an http://dx.doi.org/10.1111/j.1600-0447.2007.01128.x PMID:
inflammatory condition? The relevance of microglial activation. 18081924
Curr. Opin. Psychiatry, 2013, 26(1), 19-26. [494] Mann, J.J.; Arango, V.A.; Avenevoli, S.; Brent, D.A.; Champagne,
http://dx.doi.org/10.1097/YCO.0b013e32835aa4b4 PMID: F.A.; Clayton, P.; Currier, D.; Dougherty, D.M.; Haghighi, F.;
23196997 Hodge, S.E.; Kleinman, J.; Lehner, T.; McMahon, F.; Mościcki,
[481] Goldstein, B.I.; Kemp, D.E.; Soczynska, J.K.; McIntyre, R.S. E.K.; Oquendo, M.A.; Pandey, G.N.; Pearson, J.; Stanley, B.;
Inflammation and the phenomenology, pathophysiology, Terwilliger, J.; Wenzel, A. Candidate endophenotypes for genetic
comorbidity, and treatment of bipolar disorder: a systematic review studies of suicidal behavior. Biol. Psychiatry, 2009, 65(7), 556-563.
of the literature. J. Clin. Psychiatry, 2009, 70(8), 1078-1090. http://dx.doi.org/10.1016/j.biopsych.2008.11.021 PMID: 19201395
http://dx.doi.org/10.4088/JCP.08r04505 PMID: 19497250 [495] Hassanain, M.; Bhatt, S.; Zalcman, S.; Siegel, A. Potentiating role
[482] Berk, M.; Kapczinski, F.; Andreazza, A.C.; Dean, O.M.; Giorlando, of interleukin-1beta (IL-1beta) and IL-1beta type 1 receptors in the
F.; Maes, M.; Yücel, M.; Gama, C.S.; Dodd, S.; Dean, B.; medial hypothalamus in defensive rage behavior in the cat. Brain
Magalhães, P.V.; Amminger, P.; McGorry, P.; Malhi, G.S. Res., 2005, 1048(1-2), 1-11.
Pathways underlying neuroprogression in bipolar disorder: focus on http://dx.doi.org/10.1016/j.brainres.2005.04.086 PMID: 15919060
inflammation, oxidative stress and neurotrophic factors. Neurosci. [496] Bhatt, S.; Bhatt, R.; Zalcman, S.S.; Siegel, A. Role of IL-1 beta and
Biobehav. Rev., 2011, 35(3), 804-817. 5-HT2 receptors in midbrain periaqueductal gray (PAG) in
http://dx.doi.org/10.1016/j.neubiorev.2010.10.001 PMID: 20934453 potentiating defensive rage behavior in cat. Brain Behav. Immun.,
[483] Brietzke, E.; Stertz, L.; Fernandes, B.S.; Kauer-Sant’anna, M.; 2008, 22(2), 224-233.
Mascarenhas, M.; Escosteguy Vargas, A.; Chies, J.A.; Kapczinski, http://dx.doi.org/10.1016/j.bbi.2007.07.011 PMID: 17890051
F. Comparison of cytokine levels in depressed, manic and euthymic [497] Dunn, A.J. Effects of cytokines and infections on brain
patients with bipolar disorder. J. Affect. Disord., 2009, 116(3), 214- neurochemistry. Clin. Neurosci. Res., 2006, 6(1-2), 52-68.
217. http://dx.doi.org/10.1016/j.cnr.2006.04.002 PMID: 18079991
http://dx.doi.org/10.1016/j.jad.2008.12.001 PMID: 19251324 [498] Pandey, G.N.; Rizavi, H.S.; Ren, X.; Fareed, J.; Hoppensteadt,
[484] Akiyama, H.; Barger, S.; Barnum, S.; Bradt, B.; Bauer, J.; Cole, D.A.; Roberts, R.C.; Conley, R.R.; Dwivedi, Y. Proinflammatory
G.M.; Cooper, N.R.; Eikelenboom, P.; Emmerling, M.; Fiebich, cytokines in the prefrontal cortex of teenage suicide victims. J.
B.L.; Finch, C.E.; Frautschy, S.; Griffin, W.S.; Hampel, H.; Hull, Psychiatr. Res., 2012, 46(1), 57-63.
M.; Landreth, G.; Lue, L.; Mrak, R.; Mackenzie, I.R.; McGeer, P.L.; http://dx.doi.org/10.1016/j.jpsychires.2011.08.006 PMID: 21906753
O’Banion, M.K.; Pachter, J.; Pasinetti, G.; Plata-Salaman, C.; [499] Lindqvist, D.; Janelidze, S.; Hagell, P.; Erhardt, S.; Samuelsson, M.;
Rogers, J.; Rydel, R.; Shen, Y.; Streit, W.; Strohmeyer, R.; Minthon, L.; Hansson, O.; Björkqvist, M.; Träskman-Bendz, L.;
Tooyoma, I.; Van Muiswinkel, F.L.; Veerhuis, R.; Walker, D.; Brundin, L. Interleukin-6 is elevated in the cerebrospinal fluid of
Webster, S.; Wegrzyniak, B.; Wenk, G.; Wyss-Coray, T. suicide attempters and related to symptom severity. Biol.
Inflammation and Alzheimer’s disease. Neurobiol. Aging, 2000, Psychiatry, 2009, 66(3), 287-292.
21(3), 383-421. http://dx.doi.org/10.1016/j.biopsych.2009.01.030 PMID: 19268915
http://dx.doi.org/10.1016/S0197-4580(00)00124-X PMID: [500] Wadhawan, A.; Stiller, J.W.; Potocki, E.; Okusaga, O.; Dagdag, A.;
10858586 Lowry, C.A.; Benros, M.E.; Postolache, T.T. Traumatic brain injury
[485] Wyss-Coray, T. Inflammation in Alzheimer disease: driving force, and suicidal behavior: a review. J. Alzheimers Dis., 2019, 68(4),
bystander or beneficial response? Nat. Med., 2006, 12(9), 1005- 1339-1370.
1015. http://dx.doi.org/10.3233/JAD-181055 PMID: 30909230
PMID: 16960575 [501] Valkanova, V.; Ebmeier, K.P.; Allan, C.L. CRP, IL-6 and
[486] McGeer, P.L.; Rogers, J.; McGeer, E.G. Inflammation, depression: a systematic review and meta-analysis of longitudinal
Antiinflammatory Agents, and Alzheimer’s Disease: The Last 22 studies. J. Affect. Disord., 2013, 150(3), 736-744.
Years. J. Alzheimers Dis., 2016, 54(3), 853-857. http://dx.doi.org/10.1016/j.jad.2013.06.004 PMID: 23870425
http://dx.doi.org/10.3233/JAD-160488 PMID: 27716676 [502] Howren, M.B.; Lamkin, D.M.; Suls, J. Associations of depression
[487] McGeer, P.L.; Schulzer, M.; McGeer, E.G. Arthritis and anti- with C-reactive protein, IL-1, and IL-6: a meta-analysis.
inflammatory agents as possible protective factors for Alzheimer’s Psychosom. Med., 2009, 71(2), 171-186.
disease: a review of 17 epidemiologic studies. Neurology, 1996, http://dx.doi.org/10.1097/PSY.0b013e3181907c1b PMID:
47(2), 425-432. 19188531
http://dx.doi.org/10.1212/WNL.47.2.425 PMID: 8757015 [503] Liu, Y.; Ho, R.C-M.; Mak, A. Interleukin (IL)-6, tumour necrosis
[488] Dregan, A.; Chowienczyk, P.; Gulliford, M.C. Are inflammation factor alpha (TNF-α) and soluble interleukin-2 receptors (sIL-2R)
and related therapy associated with all-cause dementia in a primary are elevated in patients with major depressive disorder: a meta-
care population? J. Alzheimers Dis., 2015, 46(4), 1039-1047. analysis and meta-regression. J. Affect. Disord., 2012, 139(3), 230-
http://dx.doi.org/10.3233/JAD-150171 PMID: 26402631 239.
[489] Brundin, L.; Erhardt, S.; Bryleva, E.Y.; Achtyes, E.D.; Postolache, http://dx.doi.org/10.1016/j.jad.2011.08.003 PMID: 21872339
T.T. The role of inflammation in suicidal behaviour. Acta Psychiatr. [504] Dowlati, Y.; Herrmann, N.; Swardfager, W.; Liu, H.; Sham, L.;
Scand., 2015, 132(3), 192-203. Reim, E.K.; Lanctôt, K.L. A meta-analysis of cytokines in major
http://dx.doi.org/10.1111/acps.12458 PMID: 26256862 depression. Biol. Psychiatry, 2010, 67(5), 446-457.
[490] Black, C.; Miller, B.J. Meta-analysis of cytokines and chemokines http://dx.doi.org/10.1016/j.biopsych.2009.09.033 PMID: 20015486
in suicidality: distinguishing suicidal versus nonsuicidal patients. [505] Fleshner, M.; Frank, M.; Maier, S.F. Danger signals and
Biol. Psychiatry, 2015, 78(1), 28-37. inflammasomes: stress-evoked sterile inflammation in mood
http://dx.doi.org/10.1016/j.biopsych.2014.10.014 PMID: 25541493 disorders. Neuropsychopharmacology, 2017, 42(1), 36-45.
[491] Ducasse, D.; Olié, E.; Guillaume, S.; Artéro, S.; Courtet, P. A meta- http://dx.doi.org/10.1038/npp.2016.125 PMID: 27412959
analysis of cytokines in suicidal behavior. Brain Behav. Immun., [506] O’Donovan, A.; Rush, G.; Hoatam, G.; Hughes, B.M.; McCrohan,
2015, 46, 203-211. A.; Kelleher, C.; O’Farrelly, C.; Malone, K.M. Suicidal ideation is
http://dx.doi.org/10.1016/j.bbi.2015.02.004 PMID: 25678163 associated with elevated inflammation in patients with major
[492] Serafini, G.; Pompili, M.; Elena Seretti, M.; Stefani, H.; Palermo, depressive disorder. Depress. Anxiety, 2013, 30(4), 307-314.
M.; Coryell, W.; Girardi, P. The role of inflammatory cytokines in http://dx.doi.org/10.1002/da.22087 PMID: 23504697
suicidal behavior: a systematic review. Eur. [507] Janelidze, S.; Mattei, D.; Westrin, Å.; Träskman-Bendz, L.;
Neuropsychopharmacol., 2013, 23(12), 1672-1686. Brundin, L. Cytokine levels in the blood may distinguish suicide
http://dx.doi.org/10.1016/j.euroneuro.2013.06.002 PMID: 23896009 attempters from depressed patients. Brain Behav. Immun., 2011,
[493] Tonelli, L.H.; Stiller, J.; Rujescu, D.; Giegling, I.; Schneider, B.; 25(2), 335-339.
Maurer, K.; Schnabel, A.; Möller, H.J.; Chen, H-H.; Postolache, http://dx.doi.org/10.1016/j.bbi.2010.10.010 PMID: 20951793
1394 Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 Wadhawan et al.

[508] Yang, C.; Tiemessen, K.M.; Bosker, F.J.; Wardenaar, K.J.; Lie, J.; Giuseppe, B.; Moschetta, F.S.; Cotellessa, C.; Fulcheri, M.; Salerno,
Schoevers, R.A. Interleukin, tumor necrosis factor-α and C-reactive R.M.; Ferro, F.M. The role of C-reactive protein in mood disorders.
protein profiles in melancholic and non-melancholic depression: A Int. J. Immunopathol. Pharmacol., 2006, 19(4), 721-725.
systematic review. J. Psychosom. Res., 2018, 111, 58-68. http://dx.doi.org/10.1177/039463200601900402 PMID: 17166394
http://dx.doi.org/10.1016/j.jpsychores.2018.05.008 PMID: [523] Endres, D.; Perlov, E.; Dersch, R.; Baumgartner, A.; Hottenrott, T.;
29935756 Berger, B.; Stich, O.; Tebartz van Elst, L. Evidence of cerebrospinal
[509] Adhikari, A.; Dikshit, R.; Karia, S.; Sonavane, S.; Shah, N.; De fluid abnormalities in patients with depressive syndromes. J. Affect.
Sousa, A. Neutrophil-lymphocyte ratio and c-reactive protein level Disord., 2016, 198, 178-184.
in patients with major depressive disorder before and after http://dx.doi.org/10.1016/j.jad.2016.03.030 PMID: 27017374
pharmacotherapy. East Asian Arch. Psychiatry, 2018, 28(2), 53-58. [524] Sutin, A.R.; Milaneschi, Y.; Cannas, A.; Ferrucci, L.; Uda, M.;
PMID: 29921741 Schlessinger, D.; Zonderman, A.B.; Terracciano, A. Impulsivity-
[510] Hoekstra, R.; Fekkes, D.; Pepplinkhuizen, L.; Loonen, A.J.; Tuinier, related traits are associated with higher white blood cell counts. J.
S.; Verhoeven, W.M. Nitric oxide and neopterin in bipolar affective Behav. Med., 2012, 35(6), 616-623.
disorder. Neuropsychobiology, 2006, 54(1), 75-81. http://dx.doi.org/10.1007/s10865-011-9390-0 PMID: 22190235
http://dx.doi.org/10.1159/000096042 PMID: 17028447 [525] Mann, J.J.; Waternaux, C.; Haas, G.L.; Malone, K.M. Toward a
[511] Korte, S.; Arolt, V.; Peters, M.; Weitzsch, C.; Rothermundt, M.; clinical model of suicidal behavior in psychiatric patients. Am. J.
Kirchner, H. Increased serum neopterin levels in acutely ill and Psychiatry, 1999, 156(2), 181-189.
recovered schizophrenic patients. Schizophr. Res., 1998, 32(1), 63- PMID: 9989552
67. [526] Batty, G.D.; Jung, K.J.; Lee, S.; Back, J.H.; Jee, S.H. Systemic
http://dx.doi.org/10.1016/S0920-9964(98)00037-1 PMID: 9690336 inflammation and suicide risk: cohort study of 419 527 Korean men
[512] Sperner-Unterweger, B.; Barnas, C.; Fleischhacker, W.W.; Fuchs, and women. J. Epidemiol. Community Health, 2018, 72(7), 572-
D.; Meise, U.; Reibnegger, G.; Wachter, H. Is schizophrenia linked 574.
to alteration in cellular immunity? Schizophr. Res., 1989, 2(4-5), http://dx.doi.org/10.1136/jech-2017-210086 PMID: 29572361
417-421. [527] Keaton, S.A.; Madaj, Z.B.; Heilman, P.; Smart, L.; Grit, J.;
http://dx.doi.org/10.1016/0920-9964(89)90035-2 PMID: 2487182 Gibbons, R.; Postolache, T.T.; Roaten, K.; Achtyes, E.D.; Brundin,
[513] Celik, C.; Erdem, M.; Cayci, T.; Ozdemir, B.; Ozgur Akgul, E.; L. An inflammatory profile linked to increased suicide risk. J.
Kurt, Y.G.; Yaman, H.; Isintas, M.; Ozgen, F.; Ozsahin, A. The Affect. Disord., 2019, 247, 57-65.
association between serum levels of neopterin and number of http://dx.doi.org/10.1016/j.jad.2018.12.100 PMID: 30654266
depressive episodes of major depression. Prog. [528] Miná, V.A.; Lacerda-Pinheiro, S.F.; Maia, L.C.; Pinheiro, R.F., Jr;
Neuropsychopharmacol. Biol. Psychiatry, 2010, 34(2), 372-375. Meireles, C.B.; de Souza, S.I.; Reis, A.O.; Bianco, B.; Rolim, M.L.
http://dx.doi.org/10.1016/j.pnpbp.2010.01.002 PMID: 20074610 The influence of inflammatory cytokines in physiopathology of
[514] Dunbar, P.R.; Hill, J.; Neale, T.J.; Mellsop, G.W. Neopterin suicidal behavior. J. Affect. Disord., 2015, 172, 219-230.
measurement provides evidence of altered cell-mediated immunity http://dx.doi.org/10.1016/j.jad.2014.09.057 PMID: 25451421
in patients with depression, but not with schizophrenia. Psychol. [529] Isung, J.; Aeinehband, S.; Mobarrez, F.; Nordström, P.; Runeson,
Med., 1992, 22(4), 1051-1057. B.; Asberg, M.; Piehl, F.; Jokinen, J. High interleukin-6 and
http://dx.doi.org/10.1017/S0033291700038629 PMID: 1488478 impulsivity: determining the role of endophenotypes in attempted
[515] Taymur, I.; Özdel, K.; Özen, N.E.; Güngör, B.B.; Atmaca, M. suicide. Transl. Psychiatry, 2014, 4, e470.
Urinary neopterine levels in patients with major depressive disorder: http://dx.doi.org/10.1038/tp.2014.113 PMID: 25335166
alterations after treatment with paroxetine and comparison with [530] Zimmermann, M.; Arruda-Silva, F.; Bianchetto-Aguilera, F.;
healthy controls. Psychiatr. Danub., 2015, 27(1), 25-30. Finotti, G.; Calzetti, F.; Scapini, P.; Lunardi, C.; Cassatella, M.A.;
PMID: 25751446 Tamassia, N. IFNα enhances the production of IL-6 by human
[516] Abou-Saleh, M.T.; Anderson, D.N.; Collins, J.; Hughes, K.; Cattell, neutrophils activated via TLR8. Sci. Rep., 2016, 6, 19674.
R.J.; Hamon, C.G.; Blair, J.A. The role of pterins in depression and http://dx.doi.org/10.1038/srep19674 PMID: 26790609
the effects of antidepressive therapy. Biol. Psychiatry, 1995, 38(7), [531] Ericson, S.G.; Zhao, Y.; Gao, H.; Miller, K.L.; Gibson, L.F.; Lynch,
458-463. J.P.; Landreth, K.S. Interleukin-6 production by human neutrophils
http://dx.doi.org/10.1016/0006-3223(94)00323-U PMID: 8672606 after Fc-receptor cross-linking or exposure to granulocyte colony-
[517] Tang, C.Z.; Zhang, Y.L.; Wang, W.S.; Li, W.G.; Shi, J.P. Elevated stimulating factor. Blood, 1998, 91(6), 2099-2107.
serum levels of neopterin at admission predicts depression after http://dx.doi.org/10.1182/blood.V91.6.2099 PMID: 9490696
acute ischemic stroke: a 6-month follow-up study. Mol. Neurobiol., [532] Oishi, K.; Machida, K. Some plasma component is essential for IL-
2016, 53(5), 3194-3204. 6 secretion by Neutrophils. Environ. Health Prev. Med., 1997, 2(2),
http://dx.doi.org/10.1007/s12035-015-9220-4 PMID: 26041659 89-92.
[518] Chittiprol, S.; Venkatasubramanian, G.; Neelakantachar, N.; Babu, http://dx.doi.org/10.1007/BF02931972 PMID: 21432460
S.V.; Reddy, N.A.; Shetty, K.T.; Gangadhar, B.N. Oxidative stress [533] Lyngsø, D.; Simonsen, L.; Bülow, J. Interleukin-6 production in
and neopterin abnormalities in schizophrenia: a longitudinal study. human subcutaneous abdominal adipose tissue: the effect of
J. Psychiatr. Res., 2010, 44(5), 310-313. exercise. J. Physiol., 2002, 543(Pt 1), 373-378.
http://dx.doi.org/10.1016/j.jpsychires.2009.09.002 PMID: 19850302 http://dx.doi.org/10.1113/jphysiol.2002.019380 PMID: 12181307
[519] Park, R.J.; Kim, Y.H. Association between high sensitivity CRP and [534] Makki, K.; Froguel, P.; Wolowczuk, I. Adipose tissue in obesity-
suicidal ideation in the Korean general population. Eur. related inflammation and insulin resistance: cells, cytokines, and
Neuropsychopharmacol., 2017, 27(9), 885-891. chemokines. ISRN Inflamm., 2013, 2013, 139239.
http://dx.doi.org/10.1016/j.euroneuro.2017.06.010 PMID: 28663123 http://dx.doi.org/10.1155/2013/139239 PMID: 24455420
[520] Barnes, J.; Mondelli, V.; Pariante, C.M. Genetic contributions of [535] Hallberg, L.; Janelidze, S.; Engstrom, G.; Wisén, A.G.; Westrin, A.;
inflammation to depression. Neuropsychopharmacology, 2017, Brundin, L. Exercise-induced release of cytokines in patients with
42(1), 81-98. major depressive disorder. J. Affect. Disord., 2010, 126(1-2), 262-
http://dx.doi.org/10.1038/npp.2016.169 PMID: 27555379 267.
[521] De Berardis, D.; Fornaro, M.; Orsolini, L.; Iasevoli, F.; Tomasetti, http://dx.doi.org/10.1016/j.jad.2010.02.133 PMID: 20347489
C.; de Bartolomeis, A.; Serroni, N.; De Lauretis, I.; Girinelli, G.; [536] Kern, P.A.; Ranganathan, S.; Li, C.; Wood, L.; Ranganathan, G.
Mazza, M.; Valchera, A.; Carano, A.; Vellante, F.; Matarazzo, I.; Adipose tissue tumor necrosis factor and interleukin-6 expression in
Perna, G.; Martinotti, G.; Di Giannantonio, M. Effect of human obesity and insulin resistance. Am. J. Physiol. Endocrinol.
agomelatine treatment on C-reactive protein levels in patients with Metab., 2001, 280(5), E745-E751.
major depressive disorder: an exploratory study in “real-world,” http://dx.doi.org/10.1152/ajpendo.2001.280.5.E745 PMID:
everyday clinical practice. CNS Spectr., 2017, 22(4), 342-347. 11287357
http://dx.doi.org/10.1017/S1092852916000572 PMID: 27702411 [537] Broz, P.; Monack, D.M. Newly described pattern recognition
[522] De Berardis, D.; Campanella, D.; Gambi, F.; La Rovere, R.; Carano, receptors team up against intracellular pathogens. Nat. Rev.
A.; Conti, C.M.; Sivestrini, C.; Serroni, N.; Piersanti, D.; Di Immunol., 2013, 13(8), 551-565.
Periodontal Pathogens and Neuropsychiatric Health Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 1395

http://dx.doi.org/10.1038/nri3479 PMID: 23846113 M.R.L.; Sousa, F.B.; Alves, A.P.N.N. Molecular analysis of oral
[538] Pandey, G.N.; Rizavi, H.S.; Bhaumik, R.; Ren, X. Innate immunity bacteria in heart valve of patients with cardiovascular disease by
in the postmortem brain of depressed and suicide subjects: Role of real-time polymerase chain reaction. Medicine (Baltimore), 2015,
Toll-like receptors. Brain Behav. Immun., 2019, 75, 101-111. 94(47), e2067.
http://dx.doi.org/10.1016/j.bbi.2018.09.024 PMID: 30266463 http://dx.doi.org/10.1097/MD.0000000000002067 PMID: 26632711
[539] Hung, Y.Y.; Kang, H.Y.; Huang, K.W.; Huang, T.L. Association [553] Teles, R.; Wang, C.Y. Mechanisms involved in the association
between toll-like receptors expression and major depressive between periodontal diseases and cardiovascular disease. Oral Dis.,
disorder. Psychiatry Res., 2014, 220(1-2), 283-286. 2011, 17(5), 450-461.
http://dx.doi.org/10.1016/j.psychres.2014.07.074 PMID: 25155940 http://dx.doi.org/10.1111/j.1601-0825.2010.01784.x PMID:
[540] Griffen, A.L.; Becker, M.R.; Lyons, S.R.; Moeschberger, M.L.; 21223455
Leys, E.J. Prevalence of Porphyromonas gingivalis and periodontal [554] Kuramitsu, H.K.; Qi, M.; Kang, I.C.; Chen, W. Role for periodontal
health status. J. Clin. Microbiol., 1998, 36(11), 3239-3242. bacteria in cardiovascular diseases. Ann. Periodontol., 2001, 6(1),
http://dx.doi.org/10.1128/JCM.36.11.3239-3242.1998 PMID: 41-47.
9774572 http://dx.doi.org/10.1902/annals.2001.6.1.41 PMID: 11887470
[541] Ishikawa, M.; Yoshida, K.; Okamura, H.; Ochiai, K.; Takamura, H.; [555] Herzberg, M.C.; Meyer, M.W. Effects of oral flora on platelets:
Fujiwara, N.; Ozaki, K. Oral Porphyromonas gingivalis translocates possible consequences in cardiovascular disease. J. Periodontol.,
to the liver and regulates hepatic glycogen synthesis through the 1996, 67(10)(Suppl.), 1138-1142.
Akt/GSK-3β signaling pathway. Biochim. Biophys. Acta, 2013, http://dx.doi.org/10.1902/jop.1996.67.10s.1138
1832(12), 2035-2043. [556] D’Aiuto, F.; Parkar, M.; Tonetti, M.S. Periodontal therapy: a novel
http://dx.doi.org/10.1016/j.bbadis.2013.07.012 PMID: 23899607 acute inflammatory model. Inflamm. Res., 2005, 54(10), 412-414.
[542] Mahendra, J.; Mahendra, L.; Kurian, V.M.; Jaishankar, K.; Mythilli, http://dx.doi.org/10.1007/s00011-005-1375-4 PMID: 16283108
R. Prevalence of periodontal pathogens in coronary atherosclerotic [557] Tonetti, M.S.; D’Aiuto, F.; Nibali, L.; Donald, A.; Storry, C.;
plaque of patients undergoing coronary artery bypass graft surgery. Parkar, M.; Suvan, J.; Hingorani, A.D.; Vallance, P.; Deanfield, J.
J. Maxillofac. Oral Surg., 2009, 8(2), 108-113. Treatment of periodontitis and endothelial function. N. Engl. J.
http://dx.doi.org/10.1007/s12663-009-0028-5 PMID: 23139486 Med., 2007, 356(9), 911-920.
[543] Katz, J.; Chegini, N.; Shiverick, K.T.; Lamont, R.J. Localization of http://dx.doi.org/10.1056/NEJMoa063186 PMID: 17329698
P. gingivalis in preterm delivery placenta. J. Dent. Res., 2009, [558] Tonetti, M.S.; Van Dyke, T.E. Working group 1 of the joint
88(6), 575-578. EFP/AAP workshop. Periodontitis and atherosclerotic
http://dx.doi.org/10.1177/0022034509338032 PMID: 19587165 cardiovascular disease: consensus report of the Joint EFP/AAP
[544] Mougeot, J.C.; Stevens, C.B.; Paster, B.J.; Brennan, M.T.; Lockhart, Workshop on Periodontitis and Systemic Diseases. J. Clin.
P.B.; Mougeot, F.K. Porphyromonas gingivalis is the most Periodontol., 2013, 40(Suppl. 14), S24-S29.
abundant species detected in coronary and femoral arteries. J. Oral http://dx.doi.org/10.1111/jcpe.12089 PMID: 23627332
Microbiol., 2017, 9(1)1281562 [559] Minassian, C.; D’Aiuto, F.; Hingorani, A.D.; Smeeth, L. Invasive
http://dx.doi.org/10.1080/20002297.2017.1281562 PMID: dental treatment and risk for vascular events: a self-controlled case
28326156 series. Ann. Intern. Med., 2010, 153(8), 499-506.
[545] Hayashi, C.; Gudino, C.V.; Gibson, F.C., III; Genco, C.A. Review: http://dx.doi.org/10.7326/0003-4819-153-8-201010190-00006
Pathogen-induced inflammation at sites distant from oral infection: PMID: 20956706
bacterial persistence and induction of cell-specific innate immune [560] Hotamisligil, G.S. Inflammation and metabolic disorders. Nature,
inflammatory pathways. Mol. Oral Microbiol., 2010, 25(5), 305- 2006, 444(7121), 860-867.
316. http://dx.doi.org/10.1038/nature05485 PMID: 17167474
http://dx.doi.org/10.1111/j.2041-1014.2010.00582.x PMID: [561] Burcelin, R.; Garidou, L.; Pomié, C. Immuno-microbiota cross and
20883220 talk: the new paradigm of metabolic diseases. Semin. Immunol.,
[546] Gibson, F.C., III; Yumoto, H.; Takahashi, Y.; Chou, H.H.; Genco, 2012, 24(1), 67-74.
C.A. Innate immune signaling and Porphyromonas gingivalis- http://dx.doi.org/10.1016/j.smim.2011.11.011 PMID: 22265028
accelerated atherosclerosis. J. Dent. Res., 2006, 85(2), 106-121. [562] Nicholson, J.K.; Holmes, E.; Kinross, J.; Burcelin, R.; Gibson, G.;
http://dx.doi.org/10.1177/154405910608500202 PMID: 16434728 Jia, W.; Pettersson, S. Host-gut microbiota metabolic interactions.
[547] Gibson, F.C., III; Genco, C.A. Porphyromonas gingivalis mediated Science, 2012, 336(6086), 1262-1267.
periodontal disease and atherosclerosis: disparate diseases with http://dx.doi.org/10.1126/science.1223813 PMID: 22674330
commonalities in pathogenesis through TLRs. Curr. Pharm. Des., [563] Kolb, H.; Eizirik, D.L. Resistance to type 2 diabetes mellitus: a
2007, 13(36), 3665-3675. matter of hormesis? Nat. Rev. Endocrinol., 2011, 8(3), 183-192.
http://dx.doi.org/10.2174/138161207783018554 PMID: 18220804 http://dx.doi.org/10.1038/nrendo.2011.158 PMID: 22024974
[548] Chiu, B. Multiple infections in carotid atherosclerotic plaques. Am. [564] Clemente, J.C.; Ursell, L.K.; Parfrey, L.W.; Knight, R. The impact
Heart J., 1999, 138(5 Pt 2), S534-S536. of the gut microbiota on human health: an integrative view. Cell,
http://dx.doi.org/10.1016/S0002-8703(99)70294-2 PMID: 10539867 2012, 148(6), 1258-1270.
[549] Kozarov, E.V.; Dorn, B.R.; Shelburne, C.E.; Dunn, W.A., Jr; http://dx.doi.org/10.1016/j.cell.2012.01.035 PMID: 22424233
Progulske-Fox, A. Human atherosclerotic plaque contains viable [565] Furusho, H.; Miyauchi, M.; Hyogo, H.; Inubushi, T.; Ao, M.;
invasive Actinobacillus actinomycetemcomitans and Ouhara, K.; Hisatune, J.; Kurihara, H.; Sugai, M.; Hayes, C.N.;
Porphyromonas gingivalis. Arterioscler. Thromb. Vasc. Biol., 2005, Nakahara, T.; Aikata, H.; Takahashi, S.; Chayama, K.; Takata, T.
25(3), e17-e18. Dental infection of Porphyromonas gingivalis exacerbates high fat
http://dx.doi.org/10.1161/01.ATV.0000155018.67835.1a PMID: diet-induced steatohepatitis in mice. J. Gastroenterol., 2013, 48(11),
15662025 1259-1270.
[550] Tomás, I.; Diz, P.; Tobías, A.; Scully, C.; Donos, N. Periodontal http://dx.doi.org/10.1007/s00535-012-0738-1 PMID: 23307045
health status and bacteraemia from daily oral activities: systematic [566] Qin, N.; Yang, F.; Li, A.; Prifti, E.; Chen, Y.; Shao, L.; Guo, J.; Le
review/meta-analysis. J. Clin. Periodontol., 2012, 39(3), 213-228. Chatelier, E.; Yao, J.; Wu, L.; Zhou, J.; Ni, S.; Liu, L.; Pons, N.;
http://dx.doi.org/10.1111/j.1600-051X.2011.01784.x PMID: Batto, J.M.; Kennedy, S.P.; Leonard, P.; Yuan, C.; Ding, W.; Chen,
22092606 Y.; Hu, X.; Zheng, B.; Qian, G.; Xu, W.; Ehrlich, S.D.; Zheng, S.;
[551] Nakano, K.; Inaba, H.; Nomura, R.; Nemoto, H.; Takeda, M.; Li, L. Alterations of the human gut microbiome in liver cirrhosis.
Yoshioka, H.; Matsue, H.; Takahashi, T.; Taniguchi, K.; Amano, Nature, 2014, 513(7516), 59-64.
A.; Ooshima, T. Detection of cariogenic Streptococcus mutans in http://dx.doi.org/10.1038/nature13568 PMID: 25079328
extirpated heart valve and atheromatous plaque specimens. J. Clin. [567] Hotamisligil, G.S.; Budavari, A.; Murray, D.; Spiegelman, B.M.
Microbiol., 2006, 44(9), 3313-3317. Reduced tyrosine kinase activity of the insulin receptor in obesity-
http://dx.doi.org/10.1128/JCM.00377-06 PMID: 16954266 diabetes. Central role of tumor necrosis factor-alpha. J. Clin. Invest.,
[552] Oliveira, F.A.F.; Forte, C.P.F.; Silva, P.G.; Lopes, C.B.; 1994, 94(4), 1543-1549.
Montenegro, R.C.; Santos, Â.K.; Sobrinho, C.R.M.R.; Mota, http://dx.doi.org/10.1172/JCI117495 PMID: 7523453
1396 Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 Wadhawan et al.

[568] Arimatsu, K.; Yamada, H.; Miyazawa, H.; Minagawa, T.; Nakajima, Normal gut microbiota modulates brain development and behavior.
M.; Ryder, M.I.; Gotoh, K.; Motooka, D.; Nakamura, S.; Iida, T.; Proc. Natl. Acad. Sci. USA, 2011, 108(7), 3047-3052.
Yamazaki, K. Oral pathobiont induces systemic inflammation and http://dx.doi.org/10.1073/pnas.1010529108 PMID: 21282636
metabolic changes associated with alteration of gut microbiota. Sci. [582] Ait-Belgnaoui, A.; Colom, A.; Braniste, V.; Ramalho, L.; Marrot,
Rep., 2014, 4, 4828. A.; Cartier, C.; Houdeau, E.; Theodorou, V.; Tompkins, T. Probiotic
http://dx.doi.org/10.1038/srep04828 PMID: 24797416 gut effect prevents the chronic psychological stress-induced brain
[569] Nakajima, M.; Arimatsu, K.; Kato, T.; Matsuda, Y.; Minagawa, T.; activity abnormality in mice. Neurogastroenterol. Motil., 2014,
Takahashi, N.; Ohno, H.; Yamazaki, K. Oral Administration of P. 26(4), 510-520.
gingivalis Induces Dysbiosis of Gut Microbiota and Impaired http://dx.doi.org/10.1111/nmo.12295 PMID: 24372793
Barrier Function Leading to Dissemination of Enterobacteria to the [583] Hemarajata, P.; Versalovic, J. Effects of probiotics on gut
Liver. PLoS One, 2015, 10(7), e0134234. microbiota: mechanisms of intestinal immunomodulation and
http://dx.doi.org/10.1371/journal.pone.0134234 PMID: 26218067 neuromodulation. Therap. Adv. Gastroenterol., 2013, 6(1), 39-51.
[570] Darveau, R.P. Periodontitis: a polymicrobial disruption of host http://dx.doi.org/10.1177/1756283X12459294 PMID: 23320049
homeostasis. Nat. Rev. Microbiol., 2010, 8(7), 481-490. [584] Cryan, J.F.; Dinan, T.G. Mind-altering microorganisms: the impact
http://dx.doi.org/10.1038/nrmicro2337 PMID: 20514045 of the gut microbiota on brain and behaviour. Nat. Rev. Neurosci.,
[571] Ritchie, C.S.; Kinane, D.F. Nutrition, inflammation, and periodontal 2012, 13(10), 701-712.
disease. Nutrition, 2003, 19(5), 475-476. http://dx.doi.org/10.1038/nrn3346 PMID: 22968153
http://dx.doi.org/10.1016/S0899-9007(02)01043-2 PMID: 12714106 [585] Galland, L. The gut microbiome and the brain. J. Med. Food, 2014,
[572] Kau, A.L.; Ahern, P.P.; Griffin, N.W.; Goodman, A.L.; Gordon, J.I. 17(12), 1261-1272.
Human nutrition, the gut microbiome and the immune system. http://dx.doi.org/10.1089/jmf.2014.7000 PMID: 25402818
Nature, 2011, 474(7351), 327-336. [586] Painold, A.; Mörkl, S.; Kashofer, K.; Halwachs, B.; Dalkner, N.;
http://dx.doi.org/10.1038/nature10213 PMID: 21677749 Bengesser, S.; Birner, A.; Fellendorf, F.; Platzer, M.; Queissner, R.;
[573] Kelly, J.R.; Kennedy, P.J.; Cryan, J.F.; Dinan, T.G.; Clarke, G.; Schütze, G.; Schwarz, M.J.; Moll, N.; Holzer, P.; Holl, A.K.;
Hyland, N.P. Breaking down the barriers: the gut microbiome, Kapfhammer, H.P.; Gorkiewicz, G.; Reininghaus, E.Z. A step
intestinal permeability and stress-related psychiatric disorders. ahead: Exploring the gut microbiota in inpatients with bipolar
Front. Cell. Neurosci., 2015, 9, 392. disorder during a depressive episode. Bipolar Disord., 2019, 21(1),
http://dx.doi.org/10.3389/fncel.2015.00392 PMID: 26528128 40-49.
[574] Messaoudi, M.; Lalonde, R.; Violle, N.; Javelot, H.; Desor, D.; http://dx.doi.org/10.1111/bdi.12682 PMID: 30051546
Nejdi, A.; Bisson, J-F.; Rougeot, C.; Pichelin, M.; Cazaubiel, M.; [587] Talamo, B.R.; Feng, W.H.; Perez-Cruet, M.; Adelman, L.; Kosik,
Cazaubiel, J.M. Assessment of psychotropic-like properties of a K.; Lee, M.Y.; Cork, L.C.; Kauer, J.S. Pathologic changes in
probiotic formulation (Lactobacillus helveticus R0052 and olfactory neurons in Alzheimer’s disease. Ann. N. Y. Acad. Sci.,
Bifidobacterium longum R0175) in rats and human subjects. Br. J. 1991, 640(1), 1-7.
Nutr., 2011, 105(5), 755-764. http://dx.doi.org/10.1111/j.1749-6632.1991.tb00182.x PMID:
http://dx.doi.org/10.1017/S0007114510004319 PMID: 20974015 1776726
[575] Ong, I.M.; Gonzalez, J.G.; McIlwain, S.J.; Sawin, E.A.; Schoen, [588] Coureuil, M.; Lécuyer, H.; Bourdoulous, S.; Nassif, X. A journey
A.J.; Adluru, N.; Alexander, A.L.; Yu, J.J. Gut microbiome into the brain: insight into how bacterial pathogens cross blood-
populations are associated with structure-specific changes in white brain barriers. Nat. Rev. Microbiol., 2017, 15(3), 149-159.
matter architecture. Transl. Psychiatry, 2018, 8(1), 6. http://dx.doi.org/10.1038/nrmicro.2016.178 PMID: 28090076
http://dx.doi.org/10.1038/s41398-017-0022-5 PMID: 29317592 [589] Giacona, M.B.; Papapanou, P.N.; Lamster, I.B.; Rong, L.L.;
[576] Bercik, P.; Park, A.J.; Sinclair, D.; Khoshdel, A.; Lu, J.; Huang, X.; D’Agati, V.D.; Schmidt, A.M.; Lalla, E. Porphyromonas gingivalis
Deng, Y.; Blennerhassett, P.A.; Fahnestock, M.; Moine, D.; Berger, induces its uptake by human macrophages and promotes foam cell
B.; Huizinga, J.D.; Kunze, W.; McLean, P.G.; Bergonzelli, G.E.; formation in vitro. FEMS Microbiol. Lett., 2004, 241(1), 95-101.
Collins, S.M.; Verdu, E.F. The anxiolytic effect of Bifidobacterium http://dx.doi.org/10.1016/j.femsle.2004.10.009 PMID: 15556715
longum NCC3001 involves vagal pathways for gut-brain [590] Li, L.; Michel, R.; Cohen, J.; Decarlo, A.; Kozarov, E. Intracellular
communication. Neurogastroenterol. Motil., 2011, 23(12), 1132- survival and vascular cell-to-cell transmission of Porphyromonas
1139. gingivalis. BMC Microbiol., 2008, 8, 26.
http://dx.doi.org/10.1111/j.1365-2982.2011.01796.x PMID: http://dx.doi.org/10.1186/1471-2180-8-26 PMID: 18254977
21988661 [591] Cope, T.E.; Rittman, T.; Borchert, R.J.; Jones, P.S.; Vatansever, D.;
[577] Lyte, M.; Li, W.; Opitz, N.; Gaykema, R.P.; Goehler, L.E. Induction Allinson, K.; Passamonti, L.; Vazquez Rodriguez, P.; Bevan-Jones,
of anxiety-like behavior in mice during the initial stages of infection W.R.; O’Brien, J.T.; Rowe, J.B. Tau burden and the functional
with the agent of murine colonic hyperplasia Citrobacter rodentium. connectome in Alzheimer’s disease and progressive supranuclear
Physiol. Behav., 2006, 89(3), 350-357. palsy. Brain, 2018, 141(2), 550-567.
http://dx.doi.org/10.1016/j.physbeh.2006.06.019 PMID: 16887154 http://dx.doi.org/10.1093/brain/awx347 PMID: 29293892
[578] Zheng, P.; Zeng, B.; Zhou, C.; Liu, M.; Fang, Z.; Xu, X.; Zeng, L.; [592] Frister, A.; Schmidt, C.; Schneble, N.; Brodhun, M.; Gonnert, F.A.;
Chen, J.; Fan, S.; Du, X.; Zhang, X.; Yang, D.; Yang, Y.; Meng, H.; Bauer, M.; Hirsch, E.; Müller, J.P.; Wetzker, R.; Bauer, R.
Li, W.; Melgiri, N.D.; Licinio, J.; Wei, H.; Xie, P. Gut microbiome Phosphoinositide 3-kinase γ affects LPS-induced disturbance of
remodeling induces depressive-like behaviors through a pathway blood-brain barrier via lipid kinase-independent control of cAMP in
mediated by the host’s metabolism. Mol. Psychiatry, 2016, 21(6), microglial cells. Neuromolecular Med., 2014, 16(4), 704-713.
786-796. http://dx.doi.org/10.1007/s12017-014-8320-z PMID: 25033932
http://dx.doi.org/10.1038/mp.2016.44 PMID: 27067014 [593] Wu, Z.; Ni, J.; Liu, Y.; Teeling, J.L.; Takayama, F.; Collcutt, A.;
[579] Burokas, A.; Arboleya, S.; Moloney, R.D.; Peterson, V.L.; Murphy, Ibbett, P.; Nakanishi, H. Cathepsin B plays a critical role in
K.; Clarke, G.; Stanton, C.; Dinan, T.G.; Cryan, J.F. Targeting the inducing Alzheimer’s disease-like phenotypes following chronic
microbiota-gut-brain axis: prebiotics have anxiolytic and systemic exposure to lipopolysaccharide from Porphyromonas
antidepressant-like effects and reverse the impact of chronic stress gingivalis in mice. Brain Behav. Immun., 2017, 65, 350-361.
in mice. Biol. Psychiatry, 2017, 82(7), 472-487. http://dx.doi.org/10.1016/j.bbi.2017.06.002 PMID: 28610747
http://dx.doi.org/10.1016/j.biopsych.2016.12.031 PMID: 28242013 [594] Liu, Y.; Wu, Z.; Nakanishi, Y.; Ni, J.; Hayashi, Y.; Takayama, F.;
[580] Desbonnet, L.; Clarke, G.; Traplin, A.; O’Sullivan, O.; Crispie, F.; Zhou, Y.; Kadowaki, T.; Nakanishi, H. Infection of microglia with
Moloney, R.D.; Cotter, P.D.; Dinan, T.G.; Cryan, J.F. Gut Porphyromonas gingivalis promotes cell migration and an
microbiota depletion from early adolescence in mice: Implications inflammatory response through the gingipain-mediated activation of
for brain and behaviour. Brain Behav. Immun., 2015, 48, 165-173. protease-activated receptor-2 in mice. Sci. Rep., 2017, 7(1), 11759.
http://dx.doi.org/10.1016/j.bbi.2015.04.004 PMID: 25866195 http://dx.doi.org/10.1038/s41598-017-12173-1 PMID: 28924232
[581] Diaz Heijtz, R.; Wang, S.; Anuar, F.; Qian, Y.; Björkholm, B.; [595] Wu, Z.; Zhang, J.; Nakanishi, H. Leptomeningeal cells activate
Samuelsson, A.; Hibberd, M.L.; Forssberg, H.; Pettersson, S. microglia and astrocytes to induce IL-10 production by releasing
Periodontal Pathogens and Neuropsychiatric Health Current Topics in Medicinal Chemistry, 2020, Vol. 20, No. 15 1397

proinflammatory cytokines during systemic inflammation. J. [607] Yiemwattana, I.; Kaomongkolgit, R. Alpha-mangostin suppresses
Neuroimmunol., 2005, 167(1-2), 90-98. IL-6 and IL-8 expression in P. gingivalis LPS-stimulated human
http://dx.doi.org/10.1016/j.jneuroim.2005.06.025 PMID: 16095726 gingival fibroblasts. Odontology, 2015, 103(3), 348-355.
[596] Liu, Y.; Wu, Z.; Zhang, X.; Ni, J.; Yu, W.; Zhou, Y.; Nakanishi, H. http://dx.doi.org/10.1007/s10266-014-0160-7 PMID: 24888491
Leptomeningeal cells transduce peripheral macrophages [608] Derradjia, A.; Alanazi, H.; Park, H.J.; Djeribi, R.; Semlali, A.;
inflammatory signal to microglia in reponse to Porphyromonas Rouabhia, M. α -tocopherol decreases interleukin-1β and -6 and
gingivalis LPS. Mediators Inflamm., 2013, 2013, 407562. increases human β -defensin-1 and -2 secretion in human gingival
http://dx.doi.org/10.1155/2013/407562 PMID: 24363500 fibroblasts stimulated with Porphyromonas gingivalis
[597] D’Mello, C.; Swain, M.G. Immune-to-brain communication lipopolysaccharide. J. Periodontal Res., 2016, 51(3), 295-303.
pathways in inflammation-associated sickness and depression In http://dx.doi.org/10.1111/jre.12308 PMID: 26214284
Inflammation-Associated Depression: Evidence, Mechanisms and [609] Jian, C.X.; Li, M.Z.; Zheng, W.Y.; He, Y.; Ren, Y.; Wu, Z.M.; Fan,
Implications. Springer: Berlin, 2016, pp. 73-94. Q.S.; Hu, Y.H.; Li, C.J. Tormentic acid inhibits LPS-induced
http://dx.doi.org/10.1007/7854_2016_37 inflammatory response in human gingival fibroblasts via inhibition
[598] Perry, V.H. The influence of systemic inflammation on of TLR4-mediated NF-κB and MAPK signalling pathway. Arch.
inflammation in the brain: implications for chronic Oral Biol., 2015, 60(9), 1327-1332.
neurodegenerative disease. Brain Behav. Immun., 2004, 18(5), 407- http://dx.doi.org/10.1016/j.archoralbio.2015.05.005 PMID:
413. 26123747
http://dx.doi.org/10.1016/j.bbi.2004.01.004 PMID: 15265532 [610] Ci, X.; Chen, L.; Ou, X. Grape seed proanthocyanidin extracts
[599] D’Mello, C.; Le, T.; Swain, M.G. Cerebral microglia recruit inhibit lipopolysaccharide of Porphyromonas gingivalis. Shanghai
monocytes into the brain in response to tumor necrosis factoralpha kou qiang yi xue, 2015, 24 (4), 433-436
signaling during peripheral organ inflammation. J. Neurosci., 2009, [611] Azelmat, J.; Larente, J.F.; Grenier, D. The anthraquinone rhein
29(7), 2089-2102. exhibits synergistic antibacterial activity in association with
http://dx.doi.org/10.1523/JNEUROSCI.3567-08.2009 PMID: metronidazole or natural compounds and attenuates virulence gene
19228962 expression in Porphyromonas gingivalis. Arch. Oral Biol., 2015,
[600] Capuron, L.; Miller, A.H. Immune system to brain signaling: 60(2), 342-346.
neuropsychopharmacological implications. Pharmacol. Ther., 2011, http://dx.doi.org/10.1016/j.archoralbio.2014.11.006 PMID:
130(2), 226-238. 25463909
http://dx.doi.org/10.1016/j.pharmthera.2011.01.014 PMID: [612] Kong, L.; Qi, X.; Huang, S.; Chen, S.; Wu, Y.; Zhao, L. Theaflavins
21334376 inhibit pathogenic properties of P. gingivalis and MMPs production
[601] Bluthé, R-M.; Walter, V.; Parnet, P.; Layé, S.; Lestage, J.; Verrier, in P. gingivalis-stimulated human gingival fibroblasts. Arch. Oral
D.; Poole, S.; Stenning, B.E.; Kelley, K.W.; Dantzer, R. Biol., 2015, 60(1), 12-22.
Lipopolysaccharide induces sickness behaviour in rats by a vagal http://dx.doi.org/10.1016/j.archoralbio.2014.08.019 PMID:
mediated mechanism. C. R. Acad. Sci. III, 1994, 317(6), 499-503. 25244614
PMID: 7987701 [613] Kataoka, S.; Baba, A.; Suda, Y.; Takii, R.; Hashimoto, M.;
[602] GAIN Trial: Phase 2/3 Study of COR388 in Subjects With Kawakubo, T.; Asao, T.; Kadowaki, T.; Yamamoto, K. A novel,
Alzheimer's Disease. potent dual inhibitor of Arg-gingipains and Lys-gingipain as a
https://clinicaltrials.gov/ct2/show/NCT03823404 (Accessed promising agent for periodontal disease therapy. FASEB J., 2014,
September 26, 2019). 28(8), 3564-3578.
[603] Fornicola, W.; Pelcovits, A.; Li, B.X.; Heath, J.; Perry, G.; http://dx.doi.org/10.1096/fj.14-252130 PMID: 24776743
Castellani, R.J. Alzheimer disease pathology in middle age reveals a [614] Löhr, G.; Beikler, T.; Podbielski, A.; Standar, K.; Redanz, S.;
spatial-temporal disconnect between amyloid-β and Phosphorylated Hensel, A. Polyphenols from Myrothamnus flabellifolia Welw.
Tau. Open Neurol. J., 2014, 8, 22-26. inhibit in vitro adhesion of Porphyromonas gingivalis and exert
http://dx.doi.org/10.2174/1874205X01408010022 PMID: 25628768 anti-inflammatory cytoprotective effects in KB cells. J. Clin.
[604] Yamasaki, Y.; Nomura, R.; Nakano, K.; Naka, S.; Matsumoto- Periodontol., 2011, 38(5), 457-469.
Nakano, M.; Asai, F.; Ooshima, T. Distribution of periodontopathic http://dx.doi.org/10.1111/j.1600-051X.2010.01654.x PMID:
bacterial species in dogs and their owners. Arch. Oral Biol., 2012, 21158896
57(9), 1183-1188. [615] Medawar, P.B. In Some immunological and endocrinological
http://dx.doi.org/10.1016/j.archoralbio.2012.02.015 PMID: problems raised by the evolution of viviparity in vertebrates. Symp.
22417880 Soc. Exp. Biol., 1953, 320-337.
[605] Murakami, Y.; Kawata, A.; Ito, S.; Katayama, T.; Fujisawa, S. The [616] Scrandis, D.A.; Langenberg, P.; Tonelli, L.H.; Sheikh, T.M.;
radical scavenging activity and cytotoxicity of resveratrol, orcinol Manogura, A.C.; Alberico, L.A.; Hermanstyne, T.; Fuchs, D.;
and 4-allylphenol and their inhibitory effects on cox-2 gene Mighty, H.; Hasday, J.D.; Boteva, K.; Postolache, T.T. Prepartum
expression and nf-κb activation in raw264.7 cells stimulated with depressive symptoms correlate positively with c-reactive protein
porphyromonas gingivalis-fimbriae. In Vivo, 2015, 29(3), 341-349. levels and negatively with tryptophan levels: a preliminary report.
PMID: 25977379 Int. J. Child Health Hum. Dev., 2008, 1(2), 167-174.
[606] Murakami, Y.; Kawata, A.; Ito, S.; Katayama, T.; Fujisawa, S. PMID: 18924606
Radical-scavenging and anti-inflammatory activity of quercetin and [617] Groer, M.; Fuchs, D.; Duffy, A.; Louis-Jacques, A.; D’Agata, A.;
related compounds and their combinations against raw264.7 cells Postolache, T.T. Associations among obesity, inflammation, and
stimulated with porphyromonas gingivalis fimbriae. relationships tryptophan catabolism in pregnancy. associations among obesity,
between anti-inflammatory activity and quantum chemical inflammation, and tryptophan catabolism in pregnancy. Biol. Res.
parameters. In Vivo, 2015, 29(6), 701-710. Nurs., 2018, 20(3), 284-291.
PMID: 26546527 http://dx.doi.org/10.1177/1099800417738363 PMID: 29141444

You might also like