Science - USA (2022-05-27)

(Maropa) #1

GRAPHIC: KELLIE HOLOSKI/


SCIENCE


SCIENCE science.org 27 MAY 2022 • VOL 376 ISSUE 6596 935

receptor–mediated inflammatory cytokine
production by neutrophils. Concurrently, P.
gingivalis inhibits neutrophil bacterial kill-
ing activity by disarming the host-protective
Toll-like receptor pathways. P. gingivalis
thus creates the inflammatory conditions
that it needs to thrive, yet avoids the anti-
bacterial response, thereby perpetuating
dysbiosis and periodontitis ( 5 ). Gingipains
also induce vascular permeability, which
thus grants P. gingivalis, and other bacteria,
systemic reach. Notably, gingipain genes of
P. gingivalis are detectable in calcified tooth
tartar samples from ancient humans ( 6 ),

suggesting that these virulence factors have
aided P. gingivalis to persist in the oral mi-
crobiota for 100,000 years.
In a pathological context, oral cavity–
associated microbes have been detected in
distant organs in humans, including the
intestine, lungs, heart, and brain. Indeed,
numerous studies have suggested a relation-
ship between oral hygiene and respiratory
diseases, including asthma, chronic obstruc-
tive pulmonary disease, and pneumonia
( 7 ). Although these findings demonstrate
associations rather than causality, oral mi-
crobiota are likely microaspirated and af-
fect the lung microbiome. Oral bacteria are
also swallowed with saliva (1 ml of saliva can
contain up to 10^7 to 10^8 microbes) but are
exposed to gastric acid, bile acids, anaerobic
environments, and numerous other colo-
nization resistance mechanisms conferred
by the gut microbiota. Therefore, intestinal
colonization by oral microbiota is limited in
healthy individuals. However, an increased

proportion of oral microbes among the in-
testinal microbiota can be observed in sev-
eral diseases. For example, oral microbes are
significantly more abundant in the fecal mi-
crobiota of patients with IBD, primary scle-
rosing cholangitis (bile duct inflammation),
multiple sclerosis, and gastroesophageal re-
flux disease compared with healthy controls
( 8 ). Although the enrichment of oral bacte-
ria in the intestine could be a consequence
of a depleted gut commensal population,
several reports demonstrate the causal con-
tribution of oral microbes to disease patho-
genesis. For example, Klebsiella strains car-

rying antibiotic-resistance genes isolated
from saliva samples from patients with IBD
can persist in the intestine of antibiotics-
treated mice and drive intestinal inflamma-
tion ( 8 ). Consistently, intestinal enrichment
of Klebsiella species has been associated with
IBD in multiple human studies ( 9 ).
Opportunistic infections by oral microbes
may also contribute to the development of
cancer in the alimentary tract. For exam-
ple, oral microbes such as Fusobacterium,
Parvimonas, and Peptostreptococcus have
been associated with colorectal cancer (CRC)
development. In mice, F. nucleatum colonizes
CRC tissues through FadA adhesin–mediated
binding to host E-cadherin and accelerates
proliferation of tumor cells through activa-
tion of the Wnt– b-catenin signaling pathway
( 10 ). Moreover, Fusobacterium species can
migrate with metastasizing CRC cells, and
intracellular Fusobacterium-positive CRC
cells established metastatic lesions more ef-
fectively than Fusobacterium-negative cells

in mice ( 11 ). Therefore, the oral cavity could
act as a reservoir for opportunistic pathogens
that can colonize the intestine and might
causally affect chronic inflammation, as well
as cancer development and metastasis.
The mouth is a highly vascularized organ.
As a result, oral microbes and microbial mol-
ecules might directly enter the bloodstream
and contribute to the pathogenesis of sys-
temic diseases. The development of rheuma-
toid arthritis is promoted through a loss of
immune tolerance to citrullinated proteins.
Concordantly, amounts of anti-citrullinated
protein antibodies (ACPAs) are predictive
for current and future disease onset. ACPA-
positive individuals exhibit a higher relative
abundance of P. gingivalis in their oral mi-
crobiota. Although it is unclear whether P.
gingivalis plays a causal role, it may contrib-
ute to disease pathogenesis through the gen-
eration of citrullinated proteins. Gingipains
proteolytically cleave bacterial and host
proteins at arginine residues, and the short
peptides with carboxyl-terminal arginine are
immediately citrullinated by bacterial pepti-
dylarginine deiminase (PAD) that is secreted
with gingipains. It has been suggested that
citrullinated proteins elicit the generation of
antibodies that cross-react with bacterial and
host citrullinated molecules through molecu-
lar mimicry ( 12 ). Aggregatibacter actinomy-
cetemcomitans, associated with periodontitis,
also has the capacity to produce citrullinated
autoantigens through a mechanism distinct
from that of P. gingivalis, thereby also induc-
ing ACPAs through molecular mimicry ( 13 ).
Epidemiological studies have identi-
fied an association between periodontitis,
cognitive function, and dementia. Recent
work using brain specimens and cerebro-
spinal fluid from individuals diagnosed
with Alzheimer’s disease suggested that
P. gingivalis could colonize the brain and
induce neurodegeneration ( 14 ). P. gingi-
valis gingipains are found in association
with neurons, tau tangles, and amyloid-b
(Ab1-42) depositions in Alzheimer’s disease
specimens. Although further investiga-
tions will be required, P. gingivalis might
translocate to the brain by hematogenous
routes, leading to activation of the comple-
ment cascade, increasing amyloid- b pro-
duction, augmenting proinflammatory
cytokine expression, and causing neuroin-
flammation and neurodegeneration ( 14 ).
These studies suggest that oral microbiota
dysbiosis may have systemic ramifications
that promote development of autoimmune
and neurodegenerative diseases.
The oral microbiota is of particular in-
terest in the search for taxonomic and
molecular biomarkers of oral and systemic
diseases owing to the ease of sampling and
its compositional stability. Although the

Supragingival biofilm

Oral microbiota inuence disease

fungiform

Papillae:
filiform

Dorsal tongue biofilm

Fa p 2 Fa d A

E-cadherin

Fa d A , F. nucleatum adhesin;
Gal-GalNAc, N-acetylgalactosamine

Fusobacterium

Porphyromonas
gingivalis

Ta r g e t
protein

Peptidylarginine
deiminase,
gingipains

O
HN

H 2 N

Citrullinated
proteins

Production of
anti-citrullinated
protein antibodies

Wnt pathway

Corynebacterium
Streptococcus
Neisseria
Fusobacterium
Leptotrichia
Porphyromonas
Haemophilus

Rothia
Veillonella
Actinomyces
Streptococcus
Neisseria

Colorectal cancer Rheumatoid arthritis

Tumor cell proliferation

Gal-GalNAc

Cross-react to
citrullinated
α-enolase

O
HN

H 2 N

Oral microbiota in health and disease
Oral microbiota form distinct biofilms in different regions of the mouth that protect host tissues [based on ( 2 , 3 )].
When homeostasis between host and microbiota is lost, oral microbiota can affect distant tissues and
influence, for example, colorectal cancer cell proliferation and autoimmunity that leads to rheumatoid arthritis.
Free download pdf