STREPTOCOCCUS MUTANS OMZ175 INVASION OF TISSUES IN CULTURE

By

EDITH MARION SAMPSON

A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF FLORIDA IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY

UNIVERSITY OF FLORIDA

2014

© 2014 Edith Marion Sampson

To my family

ACKNOWLEDGMENTS

I thank my husband, children, and extended family for their support. I thank my colleague and partner in science, Dr. Priscilla Phillips, for daring me to chase after my dreams. I thank Drs. Paulette Robinson, Leticia Reyes, and Brian Bainbridge for their support and kindness. I thank the APF laboratory past and present lab members, including Jacob Burks, Joan Whitlock, Amandeep Chadda, Dr. Amanda Barrett, and

Ryan Chastain-Gross for their support and friendship. I thank my undergraduate research assistants, especially Ionut “Johnny” Albu, Bryce Bergeron, Whitney Swonger,

Meaghan Foti, and Maria Zimmerman for their time and attention to detail. I thank my mentor, Dr. Ann Progulske-Fox, for her example of a successful academic scientist and for guiding me through thoughtful conversation and constructive criticism. I thank my committee for their high expectations and guidance: Dr. Bill Dunn for his criticism and guidance during lab meetings on my research; Dr. Bob Burne for sharing his knowledge on streptococci and asking hard questions; Dr. Alex Lucas for sharing her knowledge of the cardiovascular system and cardiovascular research; and Dr. Scott Grieshaber for his assistance with confocal microscopy and criticism of the literature and my data. I thank our collaborators Dr. Henry Baker and Cecilia Lopez for completing the microarray chemistry and data analysis, and I thank Dr. Jacqueline Abranches, James

Miller, and Dr. Jeanne Brady for sharing bacterial strains, specimens, reagents, and ideas. I would like to thank Karen Kelley, Kim Backer-Kelley, and Neal Benson at the

ICBR core facilities. Finally, I would like to thank the Oral Biology T32DE-007200

Training Grant and the Basic Microbiology and Infectious Diseases (BMID) T32AI-7110-

29 Training Grant for the financial support to complete this work.

4

TABLE OF CONTENTS

page

ACKNOWLEDGMENTS ...... 4

LIST OF TABLES ...... 7

LIST OF FIGURES ...... 8

LIST OF ABBREVIATIONS ...... 10

ABSTRACT ...... 13

CHAPTER

1 LITERATURE REVIEW ...... 15

Introduction ...... 15 Streptococcus mutans ...... 17 S. mutans and the Oral Cavity ...... 23 S. mutans and Cardiovascular Disease ...... 27 Circulatory System ...... 36 Arteries ...... 36 Endothelial Cells ...... 37 Pathogen-Host Interaction ...... 40 Summary ...... 42

2 STREPTOCOCCUS MUTANS OMZ175 INVASION OF HUMAN ORAL AND CARDIOVASCULAR CELLS ...... 49

Introduction ...... 49 Materials and Methods...... 52 Cell Culture ...... 52 Bacterial Culture ...... 53 Attachment Assay ...... 53 Antibiotic Protection Assays ...... 54 Transmission Electron Microscopy (TEM) ...... 55 Relative Real-Time PCR ...... 56 Host Cell Cycling ...... 58 Bacterial Intercellular Spreading ...... 59 Expression Analysis ...... 59 Statistical Analysis ...... 61 Results ...... 61 S. mutans OMZ175 Invades Human Oral Cells in Culture...... 61 S. mutans Does Not Replicate Intracellularly...... 63 Intracellular S. mutans Exit the Host and Remain Infectious...... 65

5

Cnm Affects the Expression of Endothelial Cell Cytoskeleton Linked ...... 66 S. mutans OMZ175 Invades Human Coronary Artery Smooth Muscle Cells. ... 67 Discussion ...... 68

3 GENE EXPRESSION CHANGES IN HCAEC DURING INFECTION WITH STREPTOCOCCUS MUTANS OMZ175 ...... 92

Introduction ...... 92 Materials and Methods...... 95 Preparation of Confluent Human Cells in Culture...... 95 Bacterial Culture ...... 95 Gene Expression Analysis ...... 95 RT2 Profiler™ PCR Array ...... 97 RT-PCR Validation ...... 97 Enzyme Linked Immunosorbant Assay (ELISA) ...... 98 Caspase Activity Assay ...... 99 Assays to Determine the Ratio of Necrosis/Apoptosis ...... 99 Microscopy Analysis of Apoptosis ...... 100 FACS Analysis of Apoptosis ...... 101 Results ...... 101 Microarray Summary ...... 101 Ontology Analysis ...... 102 RT-PCR Validation of Microarray ...... 103 Cytokines ...... 103 Cytoskeletal Elements and Adhesion Molecules ...... 105 Apoptosis ...... 107 Discussion ...... 110

4 DISCUSSION AND FUTURE DIRECTIONS ...... 140

General Summary and Discussion ...... 140 Route from Oral Cavity to Cardiovascular Tissues ...... 140 Mechanisms of Dissemination ...... 142 Endothelial Response to S. mutans Infection ...... 142 Future Work ...... 144 Introduction ...... 144 Entry and Intracellular Trafficking ...... 145 Role of Cnm in S. mutans Entry ...... 146 Summary ...... 147

APPENDIX: MICROARRAY EXPRESSION DATA AND KEGG PATHWAYS ...... 148

LIST OF REFERENCES ...... 166

BIOGRAPHICAL SKETCH ...... 194

6

LIST OF TABLES

Table page

1-1 Summary of common characteristics in the genus Streptococcus* ...... 44

1-2 Mutans Group Characteristics ...... 47

1-3 Fast facts about S. mutans ...... 48

2-1 S. mutans strains used in this study ...... 75

2-2 Microarray differential gene expression in HCAEC due to Cnm interactions 1 h post infection ...... 75

3-1 List of primers used in this study ...... 119

3-2 Supervised analysis of pairwise comparisons ...... 119

3-3 Ontology analysis of endothelial cell pathways impacted by infection with S. mutans OMZ175 over time ...... 120

3-4 Gene expression changes of growth factor receptors and ligands 5 h post infection with S. mutans OMZ175 compared to uninfected control (p< 0.001) . 121

3-5 Microarray gene expression changes of chemokines and cytokines 5 h post infection with S. mutans OMZ175 compared to uninfected control (p< 0.001) . 122

3-6 Differential expression of involved in actin cytoskeleton organization, vesicle transport, cell proliferation, migration, and survival of HCAEC ...... 124

3-7 Microarray gene expression changes of leukocyte adhesion molecules 5 h post infection with S. mutans OMZ175 compared to uninfected control ...... 127

3-8 Human RT2 Profiler™ PCR array of adhesion molecule gene expression ...... 127

3-9 Microarray gene expression changes of genes involved in apoptotic pathways ...... 128

A-1 Microarray gene expression changes of genes with fold changes greater than 2 fold ...... 148

A-2 Microarray gene expression changes of genes with fold reductions less than -2 fold ...... 156

7

LIST OF FIGURES

Figure page

2-1 S. mutans attached and intracellular A) HOK and B) HGF-1 ...... 76

2-2 Electron micrograph of HGF-1 invasion by S. mutans OMZ175 after 2 h of infection ...... 77

2-3 Electron micrograph of S. mutans OMZ175 invasion of HGF-1 ...... 78

2-4 Electron micrograph of uninfected HGF-1 ...... 79

2-5 Intracellular S. mutans OMZ175 growth curve...... 80

2-6 Molecular methods to measure bacterial replication over time using RT-PCR ... 81

2-7 Sequential antibiotic protection assays to determine if host cell passaged S. mutans remain viable and infectious ...... 82

2-8 Micrographs of sequential antibiotic protection assays to determine if HCAEC passaged S. mutans remain viable and infectious ...... 83

2-9 S. mutans A) attachment and B) invasion of CASMC ...... 84

2-10 Electron micrograph of CASMC invasion by S. mutans OMZ175 after 2 h of infection ...... 85

2-11 Electron micrograph of S. mutans OMZ175 invasion of CASMC after 2 h infection ...... 86

2-12 Electron micrograph of uninfected CASMC ...... 87

2-13 Molecular methods to measure bacterial replication over time using RT-PCR ... 88

2-14 Reservoir Model: Oral Tissue ...... 89

2-15 Overview of S. mutans invasion of human tissues ...... 90

2-16 Invasion Cycle Model: Cardiovascular Tissue ...... 91

3-1 Hierarchical clustering of variance-normalized gene expression data ...... 129

3-2 RT-PCR validation of microarray results ...... 130

3-3 Enzyme linked immunosorbant assays to measure the levels of VEGF A) and PDGF B) in HCAEC lysates and conditioned media 24 h post infection with S. mutans ...... 131

8

3-4 Enzyme linked immunosorbant assays to measure the levels of A) IL1A, B) IL1B; C) IL6; and D) IL8 in culture conditioned media 5 and 24 h post infection with S. mutans ...... 132

3-5 Caspase 3/7 activity to determine activation of the apoptotic pathway presented as a percent of the uninfected HCAEC control ...... 133

3-6 Flow cytometric analysis of HCAEC fate during infection with S. mutans OMZ175 over time ...... 134

3-7 Percentage of HCAEC among the four fate groups as determined by flow cytometry after infection with S. mutans OMZ175 over time ...... 135

3-8 Flow cytometric analysis of HCAEC fate after infection with S. mutans OMZ175 at MOI = 1 and MOI = 100 ...... 136

3-9 Percentage of HCAEC among the four fate groups as determined by flow cytometry after infection with S. mutans OMZ175 ...... 137

3-10 Microscopic analysis of HCAEC to differentiate between apoptosis, necrosis, and healthy cells 5 h post infection with S. mutans OMZ175 ...... 138

3-11 Percentage of HCAEC among the fate groups as determined by microscopy after 5 h infection with S. mutans ...... 139

9

LIST OF ABBREVIATIONS

ASVD Atherosclerotic vascular disease

oC Degrees Celsius

CASMC Coronary artery smooth muscle cell

Cbm Collagen binding motif

CD Cluster of differentiation

CFU Colony forming units

Cnm Cold-agglutination negative mutant

CO2 Carbon dioxide

CSP Complement stimulating peptide

DMEM Dulbecco's Modified Eagle Medium

DNA Deoxyribonucleic acids

dTDP Deoxythymidine diphosphate

gDNA Genomic deoxyribonucleic acids

EPS Extrapolymeric substance

Fc receptor Eukaryotic immune cell surface that binds to the fragment crystallizable region (Fc region) of antibodies

GAG Glycosaminoglycan

GAS Group A streptococci

GTF Glucosyltransferase h Hours

HBSS Hank’s buffered saline solution

HCAEC Human coronary artery endothelial cell

HGF-1 Human gingival fibroblast-1

HOK Human oral keratinocyte

10

ICBR Interdisciplinary Center for Biotechnology Research a research support center for University of Florida

IL- Interleukin

l Liter

LDH Lactate dehydrogenase

LDL Low density lipid

M Molarity

MEROPS Peptidase database

mg Milligram

min Minutes

ml Milliliter

mm Millimeter

mM Millimolarity

ORF Open reading frame

PBS Phosphate buffered saline without calcium and magnesium

PCR Polymerase chain reaction

PD Periodontal disease

pH Negative base-10 logarithm of the molar concentration of hydrogen ions in a solution

pKa Negative base-10 logarithm of the acid dissociation constant in a solution

RGP Rhamnose glucose polymers

RNA Ribonucleic acids

rRNA Ribosomal ribonucleic acids

rt room temperature

TH T helper cells

11

TNF Tumor necrosis factor

UDP Uridine diphosphate x g Relative centrifugal force

16S rRNA Component of the 30S small subunit of prokaryotic ribosomes

18S rRNA Component of the 40S small subunit of eukaryotic ribosomes

µg Microgram

µl Microliter

µm Micrometer

µM Micromolarity

12

Abstract of Dissertation Presented to the Graduate School of the University of Florida in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy

STREPTOCOCCUS MUTANS OMZ175 INVASION OF HUMAN TISSUES IN CULTURE

By

Edith Marion Sampson

May 2014

Chair: Ann Progulske-Fox Major: Medical Sciences — Immunology and Microbiology

Studies suggest that Streptococcus mutans, an etiological agent of dental caries, may be associated with cardiovascular disease. Our , S. mutans

OMZ175, expresses the collagen binding protein Cnm, a virulence factor for invasion, and has been shown to invade human coronary artery endothelial cells (HCAEC) in culture, and to lead to cardiovascular disease in an atherogenic mouse model. The purpose of this study was to gain a better understanding of S. mutans pathogenesis by determining the host tissue tropism, invasion cycle, and the ability of this organism to replicate intracellularly. Furthermore, the effects of invasion on host gene expression and the fate of HCAEC after infection were investigated. It was found that S. mutans invades oral and cardiovascular tissues and remains infectious upon exiting the host cell. No evidence was found to indicate that S. mutans replicates intracellularly; however, S. mutans persisted for >30 h post-infection. Upon infection, HCAEC underwent a change in gene expression, as determined by microarray analysis, indicative of endothelial dysfunction. Key findings include down regulation of genes involved in apoptosis in infected HCAEC and up-regulation of growth factors indicative of angiogenesis. This finding is contrary to the expectation of S. mutans as an organism

13

that disseminates by sepsis. The results of this study support the hypothesis that strains of S. mutans with Cnm possess the pathogenic characteristics (tropism, cycling, persistence) required to transition from an oral site of infection to cardiovascular tissue infection.

14

CHAPTER 1 LITERATURE REVIEW

Introduction

Cardiovascular diseases such as hypertension and atherosclerosis are a heterogeneous group of conditions that are the leading cause of death in the United

States and in developed countries [1, 2]. However, classic risk factors such as obesity

and physical inactivity do not account for all cases [3-5]. For example, almost 25 % and

15 % of coronary deaths in males and females, respectively, occur in persons in the

lowest two quintiles of the multivariate Framingham Heart Study risk scores [6]. These

and other epidemiological studies have led to the hypothesis of an infectious theory of

atherosclerosis, which suggests that a chronic inflammatory response leading to

atherosclerosis is caused by a localized infection [7, 8]. A significant list of

epidemiological studies provides evidence for an association between cardiovascular

disease and oral infectious diseases [9-20]. Other studies have shown an association

between oral infectious diseases and inflammatory factors such as C-reactive protein,

tissue plasminogen activator, TNF-α (tumor necrosis factor), and low density lipid (LDL)

cholesterol in relation to cardiovascular diseases [21-24].

In addition to the data indicating an epidemiological relationship between oral

infectious diseases and cardiovascular disease, there is experimental evidence for such

a relationship including histological and/or genomic DNA detection of oral pathogens

such as Streptococcus mutans, Bacteroides forsythus, Porphyromonas gingivalis,

Aggregatibacter actinomycetemcomitans, and Prevotella intermedia in atheromas

dissected from human vascular tissues [9, 10, 25, 26]. Furthermore, live P. gingivalis

and A. actinomycetemcomitans have been recovered from excised atheromatous

15

human tissues [11, 27]. Animal studies also provide evidence for a role for oral pathogens in cardiovascular disease. For instance, S. mutans and P. gingivalis were

found to accelerate atherosclerosis in an apolipoprotein E null mouse as evidenced by

increased atherosclerotic plaque formation and expression of innate immune response

markers in the aortic tissues of infected animals [28-31].

The current American Heart Association (AHA) stance on the role of periodontal

disease (PD) pathogens in atherosclerotic vascular disease (ASVD) was published in

April 2012 in Circulation, a journal of the AHA [12]. In this scientific statement, the AHA

states that 1) “Observational studies to date support an association between PD and

ASVD independent of known confounders”, and 2) “Although periodontal interventions

result in a reduction in systemic inflammation and endothelial dysfunction in short-term

studies, there is no evidence that they prevent ASVD or modify its outcomes.” However,

the AHA also states that there are potential associations between PD and ASVD as

supported by multiple randomized clinical trials or meta-analyses (Level of Evidence A).

Specifically, they state that 1) “An association between PD and ASVD is supported by

evidence that meets standards for Level of Evidence A” and 2) “A benefit of periodontal

intervention in decreasing local periodontal inflammation is also supported by level A

evidence”; however, “Causation of ASVD by PD is not supported.” While S. mutans is

generally characterized as a dental caries pathogen and not a periodontal pathogen, the

statement published by the AHA includes data reporting the identification of S. mutans

DNA within human atherosclerotic plaque and heart valve specimens at higher

frequency than other bacteria (74 % and 69 % of specimens, respectively). Such

studies have been the rationale for investigating the potential of S. mutans in the

16

development or exacerbation of ASVD. This chapter will be focused on reviewing the association of an oral pathogen S. mutans with cardiovascular diseases and the related

pathological states.

Streptococcus mutans

The human oral cavity has over 700 species of bacteria in subgingival plaque that

have been identified using 16S ribosomal RNA (rRNA) gene sequences, though only

about two thirds of the oral microbiota have been cultivated [32-34]. In prokaryotes, the

hypervariable regions of 16S rRNA genes (a highly conserved ribosomal component of

the 30S small subunit of prokaryotic ribosomes) impart species-specific signature

sequences; consequently, polymerase chain reaction (PCR) techniques can be used as

a tool to identify/distinguish between bacterial species [35]. Oral biofilms are initiated

through attachment of colonizer bacteria to the host soft and hard tissues, colonized

through cell-to-cell interactions and coadhesion via an acquired pellicle. The oral

bacterial community is established after birth with the introduction of the early colonizing

species (e.g., Streptococcus and Actinomyces) followed by late colonizers (e.g.,

Prevotella, Fusobacterium, and Selenomonas) acquired after tooth eruption [36-39]. Of

37 known genera of bacteria found in the oral cavity, there are 18 genera isolated most

frequently from dental plaque, which associate through cell surface-associated

adherence proteins [40, 41]. Cell-to-cell interaction is thought to be a property of

essentially all oral bacteria and that the various bacterial adhesins recognize

glycoprotein, protein, or polysaccharide receptors found on human oral and bacterial

cell surfaces [40]. The numerous interactions that can occur between multiple surfaces

and cell types is thought to afford late colonizers the same advantages of colonization

17

as the early colonizers [40]. It has been shown that 47 % - 85 % of the cultivable cells

isolated 4 h after a professional dental cleaning are of the Genus Streptococcus.

S. mutans is a member of the phylum Firmicutes, Class Bacilli, Order

Lactobacillales, and Family Streptococcaceae. According to Bergey’s Manual of

Systematic Bacteriology [42], the genus Streptococcus contains over 50 different

species. Streptococcus species have been classified classically based on the Lancefield

scheme according to cell surface antigens (e.g., carbohydrates and teichoic acids),

blood hemolytic properties, biotyping, and more recently into “species groups” according

to 16S rRNA sequence (e.g., “Pyogenic”, “Bovis”, “Mitis”, “Anginosus”, “Salivarius”, and

“Mutans”). Oral streptococci (viridans streptococci) are divided into five groups: Mutans,

Anginosus, Salivarius, Sanguinis, and Mitis [43], and do not include any members of the

“Pyogenic” or “Bovis” groups, and generally, neither the species S. pneumoniae,

S. thermophilus, S. suis, or S. acidominimus. Members of this genus have

characteristic features (Table 1-1). Most notably, the bacteria are non-motile, Gram

positive microorganisms with a coccoid morphology, and found in chains or pairs. The

cell wall consists mainly of peptidoglycan, carbohydrates, teichoic acids, and protein

antigens. Some species form capsules which protect against the host immune response

(e.g., phagocytosis). Adhesins expressed on the cell surface are essential for

colonization and confer the ability to attach to certain host proteins and carbohydrates

that compose saliva, serum, glycocalyx, extracellular matrix, and basement membrane

components (reviewed in Nobbs et al. [44]. For instance, species in the Mutans group

bind to salivary glycoproteins via species specific adhesins, which facilitate attachment

to the salivary pellicle on tooth surfaces. The majority of streptococci are catalase

18

negative, facultative anaerobes that ferment a wide variety of carbohydrates to lactic

(major bi-product), acetic, or formic acids, ethanol and CO2. Most streptococci are

commensal organisms of the oral cavity, upper respiratory tract, and gastrointestinal

tract of mammals and birds; however, some strains are animal pathogens or

opportunistic pathogens causing systemic and localized infections.

S. mutans is one of the six primary members included in the Mutans group based

on 16S rRNA sequence similarity and is typically located within the dental plaque of

and other animals (Table 1-2). Basic characteristics of S. mutans strains are

provided in Table 1-3. S. mutans grows in chains without a capsule and can be coccoid

or rod shaped, depending on the growth conditions. The colonies on blood agar tend to

be small, circular and irregularly shaped (approximately 0.5 - 1.0 mm in diameter) and

grow into the media, pitting the surface of the agarose. Optimal growth is obtained at

o 37 C in air containing 5 % CO2. One interesting characteristic of S. mutans is its

adaptive ability to grow optimally (with respect to glucose uptake and glycolysis) at a

wide pH range (5.1 – 7.5) by a mechanism of generating intracellular pH gradients [45].

Many strains are naturally competent, though some strains are more efficient at

transformation than others [46]. Virulence factors for S. mutans include: 1)

environmental sensing [44], 2) acidogenicity [47], 3) stress tolerance [48], 4) adherence

to host cells and components [44], 5) and biofilm production [49].

S. mutans is a master at resource acquisition, able to sense and respond to

changes in its environment, devoting nearly 15 % of the genome to transport [50].

S. mutans metabolizes a greater variety of carbohydrates than any other known

sequenced Gram positive organism, and the fermentation of sugars is the main source

19

of energy production [50]. For example, S. mutans ferments a wide variety of sugars

including: glucose, fructose, sucrose, lactose, galactose, mannose, D-cellobiose, β-

glucosides, trehalose, maltose/maltodextrin, raffinose, ribulose, melibiose starch,

isomaltosaccharides, sorbose, and sugar-alcohols mannitol and sorbitol [42]. True to its

genus, the major end product of glucose fermentation is lactic acid when cultured under

aerobic conditions, and its ability to produce acid and survive in an acidic environment

contribute to its pathogenicity [48, 50, 51]. In addition to carbohydrate acquisition and

utilization, the MEROPS peptidase database [52, 53] predicts 66 putative peptidases

and 20 “other” proteins; each of the “other” 20 proteins could be placed in a peptidase

family based on except that one or more of the catalytic residues is

lacking. Among the identified peptidases are those predicted to degrade host

extracellular matrix proteins, inactivate host immune proteins, or cleave transmembrane

proteins. The tricarboxylic acid cycle is incomplete in this organism and postulated to be

used to produce amino acid precursors, as S. mutans contains all of the amino acid

biosynthetic pathways [50]. The metabolic diversity of S. mutans not only provides it

with a competitive advantage over other oral microbes, fermentative byproducts (such

as lactic acid) produced by S. mutans lower the pH of the oral cavity, inhibiting the

growth of many competing microbial species [38, 48, 50].

Besides the abilities to acquire and metabolize a wide variety of substrates,

S. mutans uses a number of strategies to colonize the oral cavity. Streptococci are

found in different tissues and locations in the human body in part due to a variety of

adhesins on the cell surface that facilitate biofilm formation. Biofilm can be described as

bacteria irreversibly attached to a surface (viable or nonviable) and embedded in an

20

extrapolymeric substance (EPS) composed of polysaccharides, proteins, glycolipids, and extracellular DNA (eDNA) [54-56]. Biofilm structures are produced through a series

of sequential steps characterized by: 1) reversible adherence of planktonic bacteria to a

surface; bacterial replication; small colony aggregates [55, 57]; 2) irreversible

adherence; communication through quorum sensing [58]; gene expression changes to

cope with environmental changes such as nutrient and oxygen limitations; phenotypic

differentiation by becoming metabolically sessile [55, 58-60]; 3) production of EPS [56];

formation of complex structures [54]; tolerance to antibiotics, biocides, antiseptics, and

disinfectants [61, 62]; protection from host immune factors (e.g., phagocytosis,

antibodies, defensins, complement) [59]; cooperative ecology [57].

S. mutans accomplishes the above steps by first adhering to surfaces and other

microbes through both sucrose independent and sucrose dependent mechanisms [44,

63]. S. mutans adherence to hard and soft tissues in the oral cavity, which can be

independent of the presence of sucrose, is accomplished via the expression of adhesins

on the bacterial surface. One such adhesin is Protein Antigen c (PAc), an important

adhesin that facilitates both attachment to host cells and surfaces as well as bacterial

autoaggregation, co-aggregation and co-adhesion [44, 50, 64, 65]. Aliases for PAc

reported in the literature include Ag I/II, P1, SpaP, B, SR, MSL-1, and IF. As a member

of the antigen (Ag) I/II family, PAc contains multiple domains which mediate binding to

tooth surfaces, extracellular matrix, serum, and saliva components such as collagen,

fibronectin, fibrinogen, and glycoprotein 340 (reviewed in Brady et al. [66]). Other strain

specific S. mutans adhesins are wall associated antigen A (WapA) and collagen binding

proteins (Cnm and Cbm). WapA is a protein adhesin involved in cell to cell aggregation

21

and biofilm production [44], and Cnm (cold-agglutination-negative mutant, with respect

to its phenotype after mutation) and Cbm (collagen binding motif) are protein adhesins

that mediate cell to cell, as well as, cell to host extracellular matrix and basement

membrane attachment [67, 68]. In addition to protein antigens, the cell wall serotype

specific polysaccharides, rhamnose glucose polymers, also facilitate bacterial

attachment to host tissues [69].

When grown in the presence of sucrose, S. mutans produces water insoluble

sugar polymers, α(1,3) and α(1, 6) glucans and β(2,1) and β(2,6) fructans, which

promote bacterial attachment, aggregation, and biofilm formation in addition to serving

as an external food source. Glucosyltransferases and fructosyltransferases

extracellularly convert sucrose into glucans or fructans, respectively. Glucan binding

proteins are virulence factors of dental caries and mediate glucan-dependent

aggregation [40]. Furthermore, glucans contribute to biofilm structures in a sucrose

dependent manner, as S. mutans production of cell wall anchored or excreted glucan

binding proteins facilitate cell to cell and cell to host interactions (for review see

reference [63]). Thus, it may also function as a potential virulence factor for attachment

to host cells. There are 4 strain specific glucan binding proteins (GbpA, GbpB, GbpC,

GbpD) that function in biofilm structure, peptidoglycan synthesis, bacterial aggregation,

and plaque cohesion, respectively. Other exoenzymes involved in extracellular sucrose

metabolism are dextranase and fructase. Dextranase is a secreted α(1,6)

glucanohydrolase that cleaves glucose from α(1,6) linkages of glucans contributing to

acid generation and water-insolubility. Fructanases are secreted β(2,1) and β(2,6)

fructan hydrolases, which release fructose from plaque fructans, contributing to the

22

duration of an acid challenge. The next section will discuss the pathological consequences of S. mutans dental plaque on oral cavity health.

S. mutans and the Oral Cavity

The oral cavity or mouth contains tissues and structures that permit humans to

taste, swallow, and chew food, as well as, speak. Some oral structures include the

tongue, teeth, supporting tissues of the teeth (e.g., periodontal ligament and

cementum), oral mucosa (e.g., gingiva and salivary glands), and jaw bones. Though

preventable with prescribed oral hygiene practices, bacterial oral infections are a

common health problem and when left untreated can result in severe pain and mortality.

Two common oral infectious diseases include dental caries and gum disease (e.g.,

gingivitis and periodontitis). Because the primary etiological agents differ for these

diseases, it is possible to develop these diseases independently [70]. However,

bacterial colonization of the oral cavity important for the establishment of these diseases

generally progresses in a similar manner through the formation of polymicrobial biofilm,

otherwise known as dental plaque (reviewed in Kolenbrander et al. in [71]).

Dental plaque initiation begins with the attachment of early colonizing bacteria to

the tooth pellicle (predominately facultative anaerobic Gram positive cocci), followed by

secondary colonizers (facultative anaerobic Gram positive and negative rods), and then

late colonizers (anaerobic Gram negative motile rods). Dental plaque initially forms

above the gingiva (supragingival plaque) and may lead to the formation of carious

lesions, as well as, progress to the gingival crevice eliciting an inflammatory response.

Once the gingival tissues become inflamed, gingivitis is established and manifested

clinically through the bleeding of gingival tissues. When gingivitis goes untreated or

treatment is prolonged, periodontitis may develop, which is histologically characterized

23

by the recession of the junctional epithelial attachment to the tooth, deepening of the gingival crevice, and degradation of connective tissue and alveolar bone [72]. In the

beginning stages of periodontal disease, gingival epithelial cells (e.g., gingival junctional, sulcus, and keratinocytes) are exposed to subgingival plaque and respond by producing chemokines and cytokines that attract and activate gingival fibroblasts, neutrophils, and T-cells [72-75]. As a consequence of immune activation, the gingival

tissues become engorged with serum due to the increased permeability of the blood

vessels. The leakiness of the blood vessels provides oral bacteria a route to the

circulatory system. In the later stages of disease progression, ulcers develop, breaching

the continuity of the epithelial layer and exposing the underlying connective tissue. The

development of ulcers provides bacteria further access to the blood stream [72]. Though

S. mutans is typically associated with supragingival plaque and dental caries, there is

no doubt that it gains access to the circulatory system as it is a causative agent of

endocarditis.

Dental caries is an infectious and transmissible disease that is dependent on diet

[38], since a diet rich in fermentable carbohydrates (like sucrose) is necessary for the

initiation and progression of caries [38]. According to the National Center for Health

Statistics, 92% of adults between the ages of 20 and 64 have had dental caries; more

than 20 % of the United States population has untreated caries, and 23 % of adults over

the age of 65 are edentulous [76]. S. mutans is the “prototypic caries pathogen”

because of its ability to form biofilm through sucrose independent and dependent

adherence, produce acid, and tolerate an acidic environment [48]. However, S. mutans

colonization does not necessarily result in caries formation (reviewed in these

24

references [38, 44, 77]). A bacterial load of > 106 S. mutans in a milliliter of stimulated

saliva is a clinical indicator of future caries, and antimicrobial therapy is recommended

before orthodontic treatments, such as, the placement of dental devices or restorations

(e.g., dental crowns, bridges, and braces) [70]. This section will focus on the role of

S. mutans in dental caries.

The amount of damage done to the tooth during a carbohydrate challenge is

dependent upon the depth and duration of plaque acidification (pH modeling of dental

plaque - reviewed in this reference [78]).One way to describe changes in plaque acidity

(measured from saliva specimens) over time is the use of "Stephan Curves". First described in 1943 by Robert Stephan, Stephan Curves predict whether an oral environment is conducive to dental caries formation following a carbohydrate challenge,

as solubilization of enamel increases exponentially as the pH in the oral cavity drops

[79, 80]. Carious lesions usually develop slowly from repeated cycles of acidification

and alkalinization of oral biofilms. Extended periods of low pH exposure influence the

oral microbiome by selecting for acid tolerant organisms. Less acid tolerant organisms

maintain a relatively alkaline environment through the production of ammonia and other

high pKa chemicals and when the pH is lowered they begin to be eliminated or poorly

represented. The ability of S. mutans to lower the pH and survive in an acidic

environment provides an advantage over acid sensitive commensal organisms [70].

S. mutans produces multiple sucrase enzymes which catalyze the hydrolysis of sucrose

to intra- and extracellular glucose and fructose and extracellular glucose and fructose

polymers, which facilitate biofilm formation [81]. When sucrose is available to

S. mutans, it is fermented rapidly to lactic acid, and the pH of dental plaque is reduced

25

to a pH of 5 or lower [82]. When the dental plaque pH drops below 5.0 for prolonged periods of time (sustained by between meal carbohydrate snacks), the buffering and mineral replenishing capacity of saliva is diminished and prolonged localized acid production results in the loss of calcium and phosphates from the external tooth enamel resulting in eventual tooth cavitation [38].

Shallow compared to deep carious lesions contain different concentrations of

bacterial species and elicit different types of T-helper (TH1 or TH2) cell-mediated

immune responses. The type of immune response is driven by the cytokine profile that

exists within the lesion. A TH1 or type 1 response is primarily characterized by the recruitment and activation of cluster of differentiation 8 positive (CD8+) cytotoxic T-cells that induce cell death of infected host cells generally in response to intracellular pathogens (e.g., viruses). A TH2 or type 2 response is primarily characterized by the

recruitment and activation of B cells to proliferate and differentiate into plasma cells

generally in response to extracellular pathogens. Plasma cells produce copious

amounts of antibodies (~2000 antibody molecules per second) that function to

neutralize antigens [83, 84].

S. mutans is typically the dominant bacterial species associated with shallow

lesions and Lactobacillus casei with deep lesions. In shallow lesions, CD8+ T cells are

the predominant form of immune cells localized to the dental pulp through an

inflammatory response to bacterial antigens. This type 1 immune response is

characterized by -gamma (IFNγ), tumor necrosis factor-beta (TNF-β), and

interleukin-2 (IL) production. In deep lesions, the bacteria reach the inflamed dental pulp

and a type 2 immune response emerges with localization of CD4+ T cells, B cells, and

26

plasma cells and the production of IL-4, IL-5, IL-6, IL-10, and IL-13 in addition to the type 1 response. The presence of S. mutans in human carious lesions has been shown to correlate with IFN-γ mRNA production and the presence of CD8+ T cells in human specimens [85].

In summary, S. mutans is an etiologic agent of dental caries. Virulence factors

attributed to S. mutans include its ability to: 1) sense and respond to environmental

changes in the oral cavity, 2) express adhesins that promote attachment and

colonization, 3) produce and tolerate acid, and 4) form biofilm. Dental caries is a

preventable disease that is prevalent in the United States that impacts both oral and

systemic health.

S. mutans and Cardiovascular Disease

S. mutans has long been recognized as a major pathogen of dental caries, one of

the most common infectious diseases in humans [38, 86, 87]. What is much less

appreciated is its role in extra-oral diseases [88-97]. S. mutans is an opportunistic

pathogen that causes extra-oral infections that are typically associated with co-

morbidities such as pregnancy [93-95, 98], post surgical infection [99], congenital heart

defects[100], atrial myxoma [91, 101, 102], kidney disease [89], autoimmune disease

(i.e. Sjogren’s syndrome) [103, 104], and chronic infections [88].

A model has been proposed to describe how oral bacteria enter the blood stream

and lead to cardiovascular pathology [105]. The pathway begins with poor oral hygiene

which results in access to the vascular system through bleeding gums, tooth abscesses

or direct tissue invasion. Once in the blood stream, systemic exposure to the bacteria

and bacterial components results in an inflammatory response and subsequent damage

27

to heart tissues. Transient bacteremias following routine dental procedures such as brushing and scaling [106] offer oral pathogens like S. mutans a direct route to the circulatory system. In fact, dissemination of oral bacteria into the bloodstream is common in humans and has been calculated to account for 3 h per day of transient bacteremia on average [106-110]. Thus, the direct interactions between oral-derived

bacteria and cells of the cardiovascular system may have a significant effect on the

progression of cardiovascular diseases (reviewed in these references [111-113]).

S. mutans has four serotypes (c, e, f and k) with serotype c being the most

common in the oral cavities of healthy individuals and f and k being the most rare [112,

114]. This serotype classification is different from the classic streptococci Lancefield

group serotyping used to distinguish streptococci genus and species. The oral cavity

and cardiovascular tissues of patients with cardiovascular disease compared to healthy

individuals were reported to have different S. mutans serotype distributions with an

increase of rare and untypable serotypes in atherosclerotic diseased cardiovascular

tissues. Serological classification of S. mutans strains is dependent upon the rhamnose-

glucose polysaccharides (RGP) located in the cell wall [reviewed in this reference

[114]]. Each of the four serotypes have a rhamnose backbone composed of alternating

α 1,2- and α 1,3- linkages [115], but it is the type of or absence of glucose side chain

linkage that is used to distinguish between serotypes. The genes involved in the

biosynthesis, transport, and assembly of RGP are located at four different

loci. RmlABCD and GluA function to synthesize RGP nucleotide precursors,

deoxythymidine diphosphate (dTDP)-L-rhamnose [116, 117] and uridine diphosphate

(UDP)-D-glucose [118]; RgpABCDEF likely function in the transport and assembly of

28

RGP [119]; and RgpG, a putative glycosyl transferase N-acetylglucosaminyltransferase, may function to initiate RGP synthesis [120].

Serotype classification can be accomplished using immunodiffusion methodology

with rabbit antisera specific for the different serotype antigens or with PCR primers

specific for the variable region between rgpF and ORF12 involved in glucose side chain

formation [121]. However, serotype classification may only be relevant to the clonal

lineage of S. mutans strains and the association between specific serotypes and

disease may be coincidental with respect to the presence and type of glucose side-

chains found on the bacterial cell surface and the strain’s ability to invade. The cell wall

characteristics that specifically distinguish the serotypes may have no functional

importance relevant to the virulence of the invasive strains being investigated. While it is

possible that there is a link between serotype and strain virulence, the purpose of this

study is not to determine if these characteristics are functionally linked, rather serotype

is only used as a way to classify these strains. In support of an argument for a role of

S. mutans in infectious cardiovascular disease, monocytes that were harvested from the

blood of healthy human donors and treated with purified serotype f RGP were shown to

have increased expression of Fc receptors for immunoglobulin G (IgG) on their surface

and secreted pro-inflammatory cytokines: tumor necrosis factor-α (TNF-α) and

interleukin 1β (IL-1β) [122]. The stimulation of monocytes to secrete TNF-α has been

reported to be mediated by interactions with CD14, a toll-like receptor 4 (TLR-4) co- receptor [123]. Unfortunately, other studies have not been done with other purified

RGPs from other serotypes to determine if the cell surface sugars play a direct role in

the host immune response. Overall these data suggest that each S. mutans serotype

29

may have factors that contribute to their ability to survive under different physiological conditions and that some of these factors may contribute to cardiovascular disease [114].

About 10% of S. mutans clinical strains have a gene that encodes for a collagen binding protein (Cnm), and the frequency of cnm is 2 to 10 times higher in the f and k rare serotype strains [67, 89, 124, 125]. Recent reports have shown that strains of

S. mutans expressing Cnm are able to invade and persist in human coronary artery endothelial cells (HCAEC), while Cnm cognate mutant strains were found to be invasion deficient [126, 127]. A serotype f strain, S. mutans OMZ175, was determined to be the

most invasive strain and was found to express Cnm [126, 127]. The intracellular location

of S. mutans OMZ175 was confirmed using transmission electron microscopy at 0.5, 5,

and 29 h post infection [127]. The results showed that S. mutans OMZ175 entered the

HCAEC in vacuoles; some bacteria escaped to the cytoplasm by 5 h post infection, and

those bacteria in vacuoles appeared intact after 29 h [127]. Once inside the host cells,

some pathogens (e.g. S. pyogenes) are destroyed by exposure to increasingly acidic

and proteolytic conditions of the early endosome, late endosome, and lysosomal

compartments, while other pathogens are able to subvert this degradative pathway. A

function of endosomes is to sort material to be degraded in lysosomes, but they also

possess enzymes with protease and carboxypeptidase activity [128]. The pH of early

endosomes is typically near 6, late endosomes near 5, and lysosomes even lower [129,

130]. S. mutans is acid tolerant being able to grow at pH 5.5 under conditions that allow

adaptation [131]; therefore, S. mutans may be expected to survive within early

endosomes with an adequate carbohydrate source such as the carbohydrates found on

30

endothelial membrane-associated glycoproteins and internalized during the formation of vesicles. The principle sugars (e.g., glucose, galactose, mannose, N- acetylglucosamine) found in glycoproteins, if and when made available, may serve as a nutrient source for S. mutans [132] [reviewed in [128]].

Infectious endocarditis is a serious cardiovascular disease with often fatal outcomes. Moreover, the onset of infectious endocarditis following dental treatment procedures has been reported [105, 133, 134]. This infectious disease is characterized

by masses of platelets, fibrin, bacteria, and inflammatory cells (called vegetations) and

disease symptoms or signs include fever, anemia, general weakness, and heart

murmur. Infective endocarditis arises from adherence of bacteria to activated platelet

masses that accumulate on the surfaces of heart valves as a result of injury or disease

[135, 136]. Virulence factors that contribute to infectious endocarditis pathogenesis are

the abilities of microorganisms 1) to survive in the blood for an extended time by

evading host immune factors, 2) to adhere to host cells or host cell components through

bacterial adhesins, 3) or to elicit platelet aggregation [135]. S. mutans, an etiologic

agent of infectious endocarditis, has each of the three phenotypes for contributing to

endocarditis virulence. The role of cell surface adhesins in S. mutans induced

bacteremia and endocarditis has been investigated. Out of 100 S. mutans strains

isolated from 100 Japanese children, 7% of the strains showed an increased ability to

survive in the blood, which correlated with the absence of PAc expression or presence

of a truncated form of PAc [137, 138]. Additionally, S. mutans and other oral

streptococci have been reported to promote the differentiation of monocytes to short-

lived dendritic cells rather than longer-lived macrophages [139], suggesting that they

31

use monocytes to carry them to diseased sites and establish infections by directing monocyte differentiation. Surface adhesins such as PAc, WapA, collagen binding proteins, and serotype polysaccharides have also been reported to facilitate adherence to host cells and extracellular matrix proteins [66, 94, 123, 126, 140-144]. Finally, the

expression of PAc, glucosyltransferases, and serotype polysaccharides have been

reported to contribute to human platelet aggregation [145-147]. S. mutans is the

causative agent in 14.2% of all patients with streptococcal valvular disease. In the

United States, oral streptococci are estimated to be responsible for 35 – 45% of

infectious endocarditis cases each year with increasing incidence [148].

Hemorrhagic stroke, which occurs when a blood vessel ruptures in the brain,

accounts for 10% of all strokes and has a 40% fatality rate, resulting in 24,000 deaths

per year in the United States [149]. In 2011, Nakano et al. [150] demonstrated that

S. mutans strains isolated from hemorrhagic stroke patients were capable of inducing

hemorrhagic stroke in mice following a procedure that photochemically induced

endothelial injury of the middle cerebral artery. The pathogenic S. mutans localized to

the site of injury, disrupted blood vessel barriers by activating host collagenases, and

interacted with collagen fibers on the surface of damaged vessels, thereby blocking

platelet activation through the collagen binding protein, Cnm. Using this animal model,

Cnm was shown to be directly involved in reducing platelet aggregation by suppressing

the interaction between platelets and collagen; typically, exposed collagen in a vessel

activates platelets through the interaction of the collagen receptor GPVI, and platelet

activation is associated with clot formation [151]. Unexpectedly, purified recombinant

Cnm was able to induce hemorrhagic disease in this mouse model, suggesting that the

32

increased cranial bleeding was likely protein adhesin mediated. An epidemiological link

between cnm-expressing S. mutans strains and cerebral hemorrhagic stroke was

established by observing that the frequency of cnm-expressing strains from age

matched, healthy control subjects was significantly lower than that of patients with

cerebral hemorrhage (odds ratio, 5.4). This study provided evidence that S. mutans

collagen binding proteins may contribute to hemorrhagic stroke risk and that endothelial

injury was necessary for this adverse sequelae.

Epidemiological and DNA sequencing evidence supports a contributing role of

S. mutans to atherosclerosis [124, 152-154]. Atherosclerosis is a complex inflammatory

disease afflicting medium and large sized arteries and is the leading cause of death in

the United States [155]. Atherosclerosis results from an excessive, inflammatory

fibroproliferative response to various forms of insult to the endothelium and smooth

muscle of the artery wall. An early event in atheroma development is endothelial

dysfunction, a pathological state of the endothelium characterized by reduced nitric

oxide, increased reactive oxygen species, secretion of pro-inflammatory molecules, and

production of leukocyte adhesion molecules (reviewed in this reference [156]).

Endothelial dysfunction contributes to various types of vascular disease such as,

hypertension, coronary artery disease, peripheral artery disease, and chronic heart

failure. Disease onset may be present before symptoms are apparent. For instance, the

progeny of hypertensive subjects were reported to have endothelial dysfunction without

hypertension [157], and children and young adults at risk for developing atherosclerosis

were shown to have endothelial dysfunction without symptoms [158]. The initial vessel

33

cell type with which bacteremic bacteria interact is the endothelial cell layer, and these interactions appear to be key to ultimate disease outcome.

S. mutans has been reported to be the predominant bacterial species isolated from diseased heart valve tissues and in atheromatous plaques [10, 152, 153]. Non-c

serotype isolates of S. mutans were identified with a significantly higher frequency (70 -

75 %) in diseased S. mutans positive cardiovascular specimens than were serotype c

isolates [154]. Most significantly, atherogenic prone mice in a restenosis injury model

developed atherosclerotic plaques by 18 weeks post surgery when also infected with a

cnm-positive strain of S. mutans [31]. The arterial injury was produced using an

angioplasty balloon catheter introduced through the femoral artery and inflated in the

distal abdominal aorta, advanced retrograde to the distal thoracic aorta, and then withdrawn. Macrophage infiltration and elevated TLR-4 gene expression indicative of an innate immune response was observed in the injured abdominal aorta tissues when infected with a strain of S. mutans that expresses Cnm compared to injury only and infected only control mice [31]. The increased presence of macrophages and the recovery of S. mutans DNA from balloon angioplasty injured abdominal aortas collected

in this study suggests that the bacteria colonized the damaged artery and/or were

carried there by macrophages, though live cell culture was not used to confirm bacterial

viability [31].

The ability of a pathogen to attach to and invade endothelial tissues has previously

been shown to be required for virulence in animal models of atherosclerosis and

infectious endocarditis [30, 159-164] and the ability to invade endothelial cells has been

shown to correlate with infectious endocarditis severity [165]. For example, some strains

34

of Lancefield Group A streptococci invade epithelial and endothelial cells through caveolae, resulting in non-phagocytic uptake and persistence in nonlysosomal compartments inside the cells [166]. In contrast, other strains of Group A streptococci invade via a zipper mechanism and localized F-actin accumulation, resulting in trafficking to lysosomes [167]. Also, two partially homologous invasins that have been shown to direct different entry mechanisms have been identified in Group A and Group

G streptococci where caveolae mediated entry was found to facilitate greater bacterial persistence than entry and trafficking through the classical endocytic pathway [168].

Thus the mode of entry plays a major role in cell trafficking, bacterial fate and, very likely, disease outcome. The mechanisms of oral streptococcal species entry into any cell type are not well characterized. Invasion of host cells consists of an active bacterially-driven process whereby signal transduction pathways of otherwise non- phagocytic cells are subverted to accommodate bacterial entry [169]. Species of other groups of streptococci that have been reported to invade host cells include S. uberis

[170], S. pyogenes (GAS) [171, 172], Group B streptococci [173], S. pneumoniae [174],

S. suis [175] and S. gordonii [176, 177]. Stinson and colleagues reported that some oral

streptococci were capable of invading human umbilical vein endothelial cells (HUVEC)

[177]. S. gordonii was considered the most invasive species of those tested, but

differences in invasion efficiency were observed among strains. The authors tested only

one strain of S. mutans, UA159, a serotype c strain, which was found not to invade

HUVECs.

S. mutans adherence to cardiovascular tissues may be enhanced during

injury/healing when extracellular matrix and basement membrane molecules are

35

exposed, since S. mutans adheres selectively to the cell membranes of skeletal, cardiac, and smooth muscle cells (sarcolemmal sheaths) and capillaries of cardiac muscle in monkeys [178].

The next section was assembled to provide the reader background knowledge of

the basic anatomy of the artery with emphasis on endothelial cells which were used as

a model of infectious cardiovascular disease in this study.

Circulatory System

Blood travels throughout the body through a closed system of interconnecting

vessels in a circuit that travels from the heart to the tissues and then back to the heart

[179]. Arteries are blood vessels that transport oxygenated blood from the heart to

tissues throughout the body, and venules are vessels that are responsible for

transporting deoxygenated blood back to the heart. There are different types of arteries.

The large elastic arteries are connected to the heart while the medium muscular arteries

branch out from the elastic arteries to varying regions of the body. The medium arteries

are further divided into smaller arteries which truncate into even smaller arteries that

enter tissues called arterioles. The arterioles further delineate into capillaries where

oxygen, nutrients, and other substances are exchanged between the blood and the

tissues. Capillaries connect to venules, and the blood begins to move back to the heart

from the venules to increasingly larger vessels called veins. Like other tissues, blood

vessels require oxygen and nutrients to survive which are supplied by the vasculature of

vessels, the vasa vasorum.

Arteries

The walls of arteries are composed of three layers called the tunica interna

(intima), tunica media, and tunica externa (adventitia)[179]. The intima is the innermost

36

layer and is in direct contact with the blood in the lumen of the artery through the endothelium (specialized epithelial tissues derived from the mesoderm that coat the lumen of the artery in a single layer and connect to the basement membrane and internal elastic lamina, see below). The media is the middle layer of the artery and is made up of smooth muscle cells and elastic fibers. The adventitia is the outermost layer

and contains fibroblasts and connective tissue composed principally of elastic and

collagen fibers and nourished by the vasa vasorum [179].

The anatomy of the artery provides elasticity and contractility properties which are

essential for function. When the heart contracts, the aortic valve of the heart opens and

blood is forced out of the ventricle chamber into the aortic artery which expands to make

room for the influx of blood. As the ventricle relaxes, the aortic valve closes, preventing

blood backflow into ventricles, and the elastic recoil of the artery forces the blood away

from the heart. The arterial smooth muscle cells, controlled by sympathetic fibers of the

autonomous nervous system, are responsible for the contractility properties of the artery

and are arranged longitudinally and in rings around the artery to perform this function. In

addition to the nervous system, chemical substances secreted by endothelial cells (e.g.,

nitric oxide) can control arterial vasodilation and vasoconstriction [179].

Endothelial Cells

The simple squamous epithelial layer that lines the heart, blood vessels, and

lymphatic vessels is known as the endothelium. The endothelium is avascular, arranged

in a continuous single layer of tightly packed cells with an apical surface in contact with

the blood in the lumen of the artery and a basal surface attached to the basement

membrane. The molecular composition of endothelial surfaces is of significance since

extracellular matrix synthesis and deposition are essential for normal artery function and

37

facilitate adherence and invasion of certain infectious agents in cardiovascular disease

[180].

On the apical side, endothelial cells produce a carbohydrate rich extracellular

matrix called the glycocalyx. The glycocalyx consists of membrane bound and secreted

molecules that act as a permeability barrier limiting the access of various blood

components to the endothelial cells and vice versa (reviewed in this reference [181]).

Because the biosynthesis and shedding of the proteoglycans and glycoproteins that

make up the glycocalyx is a dynamic process, the exact composition and thickness of

the glycocalyx matrix are ever-changing and difficult to define. In healthy individuals, the

glycocalyx may extend 2 to 3 µm in small arteries and 4.5 µm in medium arteries [181].

Vascular abnormalities can occur when the glycocalyx is incomplete or absent from the

endothelial surface and may result in increased vascular permeability, adhesion of

mononuclear cells and platelets, and reduced vasodilatation capabilities. Particular

components of the glycocalyx that contribute to its thickness are the heavily

glycosylated proteoglycans. The core proteins of proteoglycans are either anchored to

the plasma membrane (e.g., syndecan, glypican) or secreted into the glycocalyx matrix

and bloodstream (e.g., mimecan, perlecan, and biglycan). Core proteins are decorated

with one or more of the five types of glycosaminoglycans (GAGs): heparin sulfate,

chondroitin sulfate, dermatan sulfate, keratin sulfate, and hyaluronan. Many of the

sugars of the GAGs contain carboxyl and sulfate groups, which impart a highly negative

charge to the glycocalyx. The negative charges of the GAGs attract water molecules

forming a hydrated gel, which limits the movement of bacteria and large molecules but

allows diffusion of water soluble molecules in healthy tissue. Certain bacteria produce

38

enzymes that degrade glycocalyx proteoglycans, which impart a competitive edge through creating a niche for attachment and a food source for extracellular growth and persistence.

On the basal side, the endothelium uses focal adhesions and hemidesmosomes to adhere firmly to the basement membrane to keep from moving or being torn away by

the shear stress that is associated with blood flow. The basement membrane consists of

two layers including the basal lamina composed of collagen, laminin, and proteoglycans

and the reticular lamina composed of reticular fibers, fibronectin, and glycoproteins.

Focal adhesins connect actin filaments to fibronectin through cell adhesion molecules

called integrins. Integrins consist of an α and β subunit, and mammals have 17 types of

α subunits and 8 types of β subunits that combine to form at least 22 heterodimers.

Though integrins lack enyzymatic activity, they facilitate communication through

conformational changes in a process called outside-in integrin signal transduction [182].

As integrins localize to both the apical and basal sides of endothelial cells, some

bacterial pathogens use integrin ligand molecules as molecular bridges to gain entry

into host cells [183, 184]. Hemidesmosomes connect intermediate filaments (e.g.,

vimentin and keratin) in the cytosol to the basal lamina, increasing the overall rigidity of

the endothelial cells.

Primary human coronary artery endothelial cells (HCAEC) have been used for in

vitro studies of bacterial attachment and invasion, thrombosis, atherosclerosis, and

hypertension. Because of the deficient glycocalyx made by HCAEC on polystyrene, our

invasion model more closely represents a diseased population [185].

39

Pathogen-Host Interaction

A fibronectin-binding protein of Group A streptococci (GAS) is required and sufficient to determine/direct the mechanism of and complete entry of GAS into endothelial cells

[167]. S. mutans also has surface protein adhesins (PAc, Cbm, and WapA) that have been reported to bind to host basement membrane proteins i.e. fibronectin and collagen

[186-188]. PAc has been shown to be an important molecule in host cell adherence, as it directly binds to the α5β1 integrins [140, 189]. Additionally, a recent report has identified and characterized a new collagen binding protein, Cbm, found in patients with infectious endocarditis and approximately 2% of systemically healthy patients [188,

190]. The Cbm collagen binding domain was shown to have 78% identity and 90% similarity to the collagen binding domain of Cnm, and cbm-positive strains were primarily identified in the serotype k group whereas the cnm-positive strains were predominately found in the serotype f group [188]. None of the S. mutans strains identified carried both the cnm and cbm genes in the same organism. Although the adherence rates to human umbilical endothelial cells (HUVEC) were shown to be similar and dependent on either the presence of cnm or cbm, the differences in host cell invasion were not reported [188]. All of the cbm-positive strains in this 2005 report were isolated from the oral cavities of healthy subjects; however, Nomura et al. report that cbm may be important virulence factor associated with S. mutans pathogenesis of infectious endocarditis in 2013 [188, 190]. Other studies investigating the role of

S. mutans adhesion surface molecules such as PAc and WapA have not shown that these molecules contribute to the invasive phenotype in endothelial cells [126]. While

PAc may not be critical for an invasive phenotype, there is no doubt that it plays a role in adherence.

40

Two important pathways for bacterial internalization into eukaryotic cells are the caveolae/lipid raft-mediated endocytosis pathway and the receptor-mediated (clathrin-

dependent) endocytosis pathway. Entry via caveolae and/or lipid rafts has been

reported for several Gram-positive bacteria including S. uberis [191, 192] and GAS

[166], but S. suis [175] is believed to enter epithelial cells through both pathways.

During the internalization of S. suis and S. pyogenes, large invaginations, through which

bacteria enter the cells, have been described [166, 175].

Knowledge of the mechanisms of oral streptococcal species internalization is

limited, and no studies have been done using cells from the adult human cardiovascular

system. Internalization of streptococci including S. suis and S. pyogenes (GAS) in other

cell types has been shown to progress starting with large invaginations, through which

bacteria enter the cells [166, 175]. Caveolae-like structures have been observed close

to these invaginations during entry of S. pyogenes [166]. Upon invasion, some

Streptococcus species avoid lysosomal death and survive in many diverse host cells.

For example, S. uberis internalizes within epithelial cells using GPI-anchored molecules

and then exits the phagosome avoiding intravesicular acidification and lysosome fusion

[191], while S. suis survives within acidified phagolysosome-like vacuoles [175].

S. pyogenes exploits caveolae to gain entry into both epithelial (HEp-2) and endothelial

(HUVEC) cells to replicate within caveosomes [166]. However, in HeLa cells,

S. pyogenes exits the phagosome to replicate within the cytoplasm, but these

cytoplasmic bacteria eventually become sequestered in LC3-positive autophagosome-

like vacuoles that are approximately 5 to 10 times larger than the standard

autophagosomes [193]. A significant decrease in bacterial viability was observed after

41

fusion of these GAS-autophagosomes with lysosomes [193]. Thus the mode and

mechanism of entry likely sets the course for intracellular trafficking and ultimate

outcome.

The eukaryotic intracellular environment provides bacterial pathogens protection

against host immune responses and antibiotics, which may lead to long-term carriage,

recurrent infection, and chronic inflammation. Invasion of human coronary artery

endothelial cells (HCAEC) by S. mutans can occur within 1 h and persist at least 29 h

post infection under continuous antibiotic pressure [127]. S. mutans invasion of

cardiovascular endothelial cells is Cnm dependent. As provided above, it is estimated

that approximately 10-20% of all S. mutans strains are cnm-positive and that these

strains are more likely to belong to the serotype f group. An increase in serotype f

strains was shown to be present in the oral cavities and tissues of patients with

cardiovascular disease. Recently, two independent studies have reported that systemic

infection with S. mutans strains expressing Cnm increased the risk of cerebral

hemorrhagic stroke and accelerated atherosclerosis in mice. In both cases, endothelial

cells lining blood vessels were damaged leading to exposure of the underlying

extracellular matrix. Furthermore, S. mutans Cnm shares protein sequence identity with

collagen-binding proteins in S. suis [194, 195], Enterococcus faecalis [196-198], and

Staphylococcus aureus [199, 200]: organisms known to cause bacteremia and

endocarditis.

Summary

S. mutans is an oral pathogen of dental caries with access to the bloodstream

through every day routine activities such as chewing and personal hygiene regimens

such as brushing and flossing. Once in the bloodstream, S. mutans has been reported

42

to promote platelet aggregation, influence monocyte differentiation to the dendritic cell

lineage, and adhere to extracellular matrix proteins and cells of the cardiovascular

system in a strain specific manner. In age groups at increased risk for cardiovascular

disease (>45 years), the prevalence of untreated caries is approximately 21.5% in the

U.S. [76]. S. mutans is known to be a causative agent of endocarditis, a potential risk

factor for hemorrhagic stroke, and linked to atherosclerosis—all diseases of the

cardiovascular system. According to the Centers for Disease Control and Prevention, cardiovascular disease morbidity and mortality is an enormous burden on the American public and economy with more than 4 million people disabled and accounting for more than 33.6% of all deaths in the U.S. and costs exceeding $444 billion in 2010 [201]. The

objective of the research presented in this dissertation is to determine the invasion

tropism, define the invasion cycle, and to investigate the pathogenic potential of

S. mutans invasive strains on endothelial cells.

43

Table 1-1. Summary of common characteristics in the genus Streptococcus* Property Description

Cell Spherical, or ovoid, less than 2 µm in diameter Morphology S. mutans: 0.5 – 0.75 µm in diameter or may form rods 1.5 – 3.0 µm in length Cell-Wall Peptidoglycan Composition Peptidoglycan Group A with L-lysine as the diamino acid in position 3 of the peptide subunit Most common form is Lys-Alan (n < or equal to 4) Ala can be replaced by L-Thr or L-Ser S. mutans and S. bovis have L-Thr Carbohydrate Rhamnose is a common constituent of almost all streptococci cell walls Amino sugars glucosamine and muramic acid are always present Galactosamine is a variable component Common reducing sugars are glucose, galactose and rhamnose Rhamnose absent in pneumonia, oralis, and mitis Polyols glucitols in S. agalactiae and glycerol present in S. ratti Adhesins Strains with PAc include: mutans, sobrinus, gordonii, oralis, intermedius Bind extracellular matrix and serum components (fibronectin, and plasminogen) Often contain repeating blocks of amino acids Nutrition and Facultative anaerobic Growth Some require extra CO2 (S. mutans) Susceptible to vancomicin Variable reactions for growth in 6.5% NaCl containing media Require amino acids, peptides, purines, pyrimidines, and vitamins in complex media Colonial and Growth enhanced by addition of 5% sheep blood, serum, or glucose Cultural 0.5-1.0 mm in diameter on glucose at 24 h Features Non-pigmented Genomes S. mutans ~ 2.03 Mb UA159 does not contain temperate bacteriophage DNA Horizontal Competence is not constitutive but regulated by genes comA-comE Gene from 2 com operons Transfer comC: Competence Stimulating Peptide (CSP) comA and comB encoding a CSP-secretion apparatus comD and comE encode a two-component regulatory system

44

Table 1-1. Continued Property Description

Bacterio- Active temperate and virulent phages have been described. Phages are phages present within a high proportion of group A streptococci and contribute to the pathogenic potential of the species. The pyrogenic exotoxins SpeA and SpeC are bacteriophage encoded Sequencing of S. pyogenes genome has revealed the presence of complete or partial sequences of four bacteriophage genomes containing genes for one or more previously undiscovered super antigen-like proteins Genes encoding proteins Pb1A and Pb1B involved in the binding of S. mitis to human platelets, with obvious relevance to pathogenesis of infective endocarditis, are encoded by lysogenic bacteriophage Antibiotic In general susceptible to most antibiotics Sensitivity Macrolide resistance has been increasing Increase in penicillin resistance by S. pneumonia due to altered forms of penicillin binding proteins PBP1a, PBP2x, and PBP2b due to interspecies recombination events involving viridians streptococci Ecology Streptococci are associated with warm blooded animals and birds Most species are commensal organisms Colonize mucosal surfaces in the oral cavity, URT, and GI tract, under certain conditions can cause local and systemic infections Adherence to many surfaces present in their natural environment Ability to rapidly utilize available nutrients Ability to tolerate, resist, or destroy host immune defenses Selective For isolation of cariogenic S. mutans: tryptone yeast cystine media and media mitis salivarius media. Members of Anginosus species group can be isolated on semi-selective agar medium containing 40 g/l selective agar (Lab M) + 30 µg/ml nalidixic acid, 1 mg/ml sulfamethazine, and 5% (v/v) defibrinated horse blood (NAS). NAS will also select for S. mutans and S. sobrinus, although recovery is strain-dependent. Hemolysis Determined on a blood agar base medium such as Todd-Hewitt, brain heart infusion, proteose peptone, plus 5% defibrinated horse or sheep blood. Anaerobic incubation is recommended. S. pyogenes is divided into serotypes on the basis of streptococcal Serological Determination surface M proteins with more than 80 types known, with 1, 3, 11, 12, and 28 as the most common in invasive and toxic infections

45

Table 1-1. Continued Property Description

Biochemical Carbohydrate fermentation, production of acetyl methyl carbinol from and glucose in the Voges-Proskauer reaction, production of ammonia Physiological from arginine, hydrolysis of esculin, hippurate, and starch, reduction Tests of litmus milk, production of H2O2, tolerance to NaCl and bile, and formation of extracellular polysaccharide from sucrose Hydrolysis of sodium hippurate is shared by several species The formation of extracellular polysaccharides is an important characteristic of several of the oral streptococci including S. mutans, S. sangius, S. salivarius, S. oralis *Information and categories collected from Bergey’s Manual of Determinative Bacteriology [42]

46

Table 1-2. Mutans Group Characteristics* Species Animal host

S. mutans Human S. sobrinus Human, Rats S. criceti Hamsters, wild rats, and occasionally humans S. ratti Laboratory rats S. downei Monkeys (Macaca fascicularis ) and occasionally humans S. macacae Monkeys (Macaca fascicularis). S. ferus* Wild rats and pigs

*Information and categories collected from Bergey’s Manual of Determinative Bacteriology [42] **S. ferus is a peripheral member of the group due to DNA-DNA hybridization studies.

47

Table 1-3. Fast facts about S. mutans* Characteristic Description

Cell Size 0.5 – 0.75 mm in diameter Typically coccoid; however, short rods may form under acidic Cell Shape conditions (1.5-3.0 mm in length)

DNA G+C content 36-38 (mol%)

Lancefield grouping Non-grouping antiserum

Oxygen requirements Facultative anaerobe, some strains require CO2 o o Temperature 37 C optimum; no growth at 10 C Usually α-hemolytic or non-hemolytic—some strains β- Growth on blood agar hemolytic Under anaerobic conditions: white or gray colonies ATP generation Substrate-level phosphorylation Metabolic by products Lactic acid, acetate, ethanol, formate, acetoin Glucose, fructose, sucrose, lactose, galactose, mannose, D- cellobiose, β-glucosides, trehalose, maltose/maltodextrin, Acid production from D-raffinose, ribulose, melibiose starch, growth on these isomaltosaccharides, sorbose, N-acetylglucosamine, molecules esculin, arbutin, inulin, salicin and sugar-alcohols mannitol and sorbitol Glycerol, glycogen, starch, rhamnose, adonitol, arabinose, No acid production cyclodextrin, dulcitol, erythritol, gluconate, inositol, methyl from these molecules D-glucoside, methyl D-mannoside, methyl D-xyloside, melezitose, ribose, sorbose, xylose Ability to hydrolyze: Esculin, starch Inability to hydrolyze Arginine, hippurate, and urea Catalase Negative Hydrogen peroxide Negative production Peptidoglycan type Group A peptidoglycan Serotypes c, e, f, k, and serologically untypable *Information and categories collected from Bergey’s Manual of Determinative Bacteriology [42]

48

CHAPTER 2 STREPTOCOCCUS MUTANS OMZ175 INVASION OF HUMAN ORAL AND CARDIOVASCULAR CELLS

Introduction

The Centers for Disease Control (CDC) and Prevention report that half of

American adults over the age of 20 have at least one of the risk factors (high blood pressure, high low density lipid cholesterol levels, and smoking) for heart disease, the leading cause of death in the United States [202]. The CDC also reports that half of

American adults over 30 suffer from periodontitis [203]. The American Heart Association

has recently reported that heart disease and periodontitis share common risk factors

and that there is a potential link between oral health and development of cardiovascular

disease (CVD); however, a causative link has yet to be established [12]. Oral bacteria

induced bacteremia is common in humans with inflamed gingival tissues. Typically,

transient bacteremia episodes caused by oral bacteria are short-lived, without obvious

health consequences [204]. However, there are certain co-morbidities (e.g., diabetes,

rheumatoid arthritis, malignancies), which have been linked to oral bacterial infections in

sites outside of the mouth [105]. The increased risk of developing these infections may

be due to the cardiovascular tissue dysfunction typically sustained in these patients.

Streptococcus mutans, a member of the Mutans group of oral streptococci, is an

etiological agent of dental caries (one of the most common human infectious diseases),

bacteremia, and infectious endocarditis [205]. Interestingly, the serotype distributions of

S. mutans differ in the oral cavities and cardiovascular tissues of healthy versus

individuals with cardiovascular disease. This suggests that the strains within each

serotype may have unique virulence factors in common, and these factors may

contribute to the ability of certain strains to enter and survive in the bloodstream.

49

Recently, certain strains of S. mutans have been shown to invade human gingival fibroblasts (HGF) and cardiovascular endothelial cells [127, 206]. The ability of

S. mutans to invade endothelial cells has been linked to the expression of a cell surface

collagen binding protein, Cnm [126]. Strains of S. mutans expressing cnm have been

linked to endocarditis [207, 208], hemorrhagic stroke [150], and atherosclerosis [152,

153]. This cell surface adhesin/virulence factor is estimated to be present in 10-12% of

all S. mutans strains, and it is more frequently detected in strains belonging to the rarer

serotype f and k groups, whose prevalence was reported to increase when isolated from

the oral cavities and the cardiovascular tissues of patients with CVD [114]. One

potential route for S. mutans to enter the cardiovascular system from the oral cavity is

through bleeding gingival tissues, a symptom of periodontal disease. The gingiva is a

vascular rich tissue at the site of tooth eruption where gingival epithelial cells (e.g.,

junctional, sulcal, and keratinocytes) serve as a protective barrier of the underlying

connective tissue from plaque biofilm and HGFs maintain the connective tissues during

health and healing. The evidence indicating S. mutans invades HGFs suggests that

S. mutans has the potential to access the bloodstream through the gingiva. The

contribution of cnm-expressing strains of S. mutans to the invasion of oral cells has not

yet been investigated, so it is unknown whether oral epithelial cells or gingival

fibroblasts can serve as a reservoir for extra oral infections caused by cnm-expressing

strains of S. mutans.

Previously, the ability of a bacterium to attach to and invade endothelial tissues

has been shown to be required for the induction of atherosclerotic plaque formation and

the establishment of infection in animal models of atherosclerosis and infectious

50

endocarditis, respectively [30, 159, 160, 162]. Furthermore, the ability to invade

endothelial cells has been shown to correlate with infectious endocarditis severity [165,

209]. Bacteria that are able to invade and persist within endothelial cells are protected

from phagocytosis; consequently, intracellular bacteria may have a selective advantage

at remote sites and an opportunity to disseminate into deeper cardiovascular tissues

(e.g., smooth muscle cells) [210, 211]. Our lab has previously investigated intracellular

trafficking of S. mutans OMZ175 in human endothelial cells using transmission electron

microscopy [127]. Within 5 h, S. mutans localized to single and double membrane

vacuoles, suggesting that both the endocytic and autophagic pathways are important for

intracellular trafficking. After 24 h, large numbers of S. mutans were observed in the

cytoplasm of infected endothelial cells, suggesting that S. mutans escaped the vacuoles

and replicated inside of the cytoplasm of the HCAEC. However, it was also reported that

bacterial plate counts from antibiotic protection assays showed little change in bacterial

load 29 h post invasion. In contrast, when repeating the antibiotic protection assays for

the work reported here, cultivable S. mutans OMZ175 significantly decreased at 30 h

post infection, while there was no statistically significant change in bacterial load

observed at earlier time points. Before investigating whether there is a causative link

between the increased incidence of extra-oral infections by oral pathogens and these

co-morbidities, we must first develop a model of the probable route these organisms

take from the oral cavity to these sites, define whether there is a biological potential for

these organisms to invade and persist, and determine the dissemination potential to

other relevant cell types.

51

The work described here reports the first evidence that S. mutans attaches to and

invades human oral cells that reside in the gingiva (HOK and HGF-1) in a cnm-

dependent manner, supporting the epidemiological link between S. mutans and

cardiovascular disease. Additionally, the fate of S. mutans within endothelial cells was

investigated, specifically its ability to replicate intracellularly and the infection cycle,

including cytoskeletal gene expression changes attributed to Cnm early in the invasion

cycle. Furthermore, the potential of S. mutans dissemination to deeper arterial tissues

was investigated by assessing its ability to attach to and invade arterial smooth muscle

cells.

Materials and Methods

The follow materials and methods were used in this study. The rationale for the

methods and cell types selected to investigate each proposed hypothesis are explained

in detail in the results section.

Cell Culture

Primary human coronary artery endothelial cells (HCAEC; Lonza, Allendale, NJ),

primary coronary artery smooth muscle cells (CASMC; Lonza), primary oral

keratinocytes (HOK; ScienCell, Carlsbad, CA), and primary human gingival fibroblasts

(HGF-1; ATCC®, Manassas, VA) were cultured for invasion assays. The HCAEC and

CASMC were cultured in endothelial cell basal medium-2 (EBM-2; Lonza)

supplemented with EGM-2MV single-use aliquots (Lonza) or Smooth Muscle Cell Basal

Medium (SmBm Media; Lonza) supplemented with SmGM-2 BulletKit (Lonza),

respectively. The HGF-1 cells were maintained in Dulbecco's Modified Eagle Medium

(DMEM; Corning Inc., Manassas, VA) containing glucose (4.5 g/l), L-glutamine (2 mM), and sodium pyruvate (1 mM), and supplemented with 10 % fetal bovine serum (FBS;

52

Atlantic Biological; Flowery Branch, GA). The HOK were maintained in serum free OKM media (OKM; ScienCel) supplemented with OKGS. The primary cells were maintained at 37°C in a humidified, 5% carbon dioxide (CO2) atmosphere. The cells were detached

using Accutase™ and washed in cell appropriate media. The cells were counted using a

Coulter Counter (Beckman Coulter Model Z1; Brea, CA) and seeded in flat-bottom tissue culture treated plates (Corning Inc.) followed by overnight incubation at 37°C in a

5% CO2 atmosphere (4). Tissue culture plates used to culture CASMC and HGF-1 were

coated with rat tail, type I collagen (Invitrogen™; Grand Island, NY), and those used to

culture HOK were coated with poly-L lysine (Sigma-Aldrich®; Saint Louis, MO) per

manufacturer instruction.

Bacterial Culture

The bacterial strains used in this study are listed in Table 1. S. mutans strains

were cultured in either brain heart infusion (BHI; Becton Dickinson & Co., Franklin

Lakes, NJ) broth or Todd Hewitt (TH; Becton Dickinson & Co.) broth overnight. Bacterial

cultures were then pelleted at 4,629 x g for 5 min at room temperature (rt), the culture

conditioned media were aspirated, and the pellets were resuspended in sterile

phosphate-buffered saline (PBS, pH 7.2; Corning Inc.) to break up aggregates. Next the

bacteria were diluted in host cell appropriate media without antibiotics to obtain bacterial

suspensions containing 100 colony forming units (CFU) per well.

Attachment Assay

Attachment assays were used to measure the ability of S. mutans to attach to

clinically relevant cardiovascular and oral host cell types (HCAEC, CASMC, HOK, and

HGF-1) by measuring live attached and intracellular bacteria as previously described

with some modifications [212]. Prior to infection, host cells were either placed on ice for

53

20 min or treated with cytochalasin D (5ug/ml) (Sigma-Aldrich Corp.) for 30 min. Ice

treatment promotes the disassembly of microtubules and actin filaments, while

cytochalasin D inhibits actin polymerization. Both treatments were continued throughout

the entire assay. HCAEC were infected at 100 bacteria CFU per host cell. After 30 min,

the host cells were washed three times with Hank’s buffered saline solution (HBSS;

Corning Inc.). Preliminary experiments showed that concentrations of dimethyl sulfoxide

had no noticeable effect on bacterial internalization and that concentrations of

cytochalasin D had no effect on bacterial viability. Cell lysates were diluted in PBS and

plated in duplicate. Efficiency of attachment was calculated by taking the ratio of the

attached bacteria and the inoculum and multiplying by 100. A sample size of four was

used for each condition per experiment, and each experiment was repeated a minimum

of two times.

Antibiotic Protection Assays

Antibiotic protection assays were used to measure the ability of S. mutans to

invade clinically relevant cardiovascular and oral host cell types: HCAEC, CASMC,

HOK, and HGF-1 [213, 214]. S. mutans OMZ175 was used as a model for S. mutans

invasion, since this strain expresses the Cnm protein and has been reported to invade

and persist within HCAEC [127]. Primary cells were infected at 100 bacteria CFU per

host cell. After 2 h, the host cells were washed three times with HBSS and replaced with

cell appropriate media supplemented with 300 microgram ml-1 gentamicin and 10

microgram ml-1 penicillin G for a minimum of 3 h to kill extracellular bacteria. All

antibiotics used in this study were supplied by Sigma-Aldrich. Prior to host cell lysis,

host cells were washed three times with HBSS. Cell lysates were diluted in PBS and

plated in duplicate. The efficiency of invasion was calculated by taking the ratio of the

54

intracellular bacteria and the attached bacteria and multiplying by 100. A sample size of four was used for each condition per experiment, and each experiment was repeated a minimum of three times.

Invasion deficient S. mutans strains functioned as the negative control (e.g.,

S. mutans UA159 and/or S. mutans OMZ175:cnm, a collagen binding protein deletion

mutant) [126]. To assess the ability of S. mutans to replicate intracellularly, infected

HCAEC were transferred to media containing gentamicin at a concentration of 50 µg ml-

1 with and without chloramphenicol at 10 µg ml-1 after the standard antibiotic protection

assay was completed. The gentamicin kills bacteria that exit the host, and

chloramphenicol inhibits bacterial protein synthesis of intracellular and extracellular

bacteria.

Transmission Electron Microscopy (TEM)

TEM was used to observe S. mutans internalization and intracellular location

within HGF-1 and CASMC. Host cells were seeded into 6-well plates to confluence and

infected with S. mutans OMZ175 at 100 CFU per host cell for 2 h. Post invasion, human

cells were washed 3X with 3ml HBSS per well. Cells were detached with Accutase™,

collected, and mixed with an equal volume of cell appropriate media. Cells were

pelleted at 90 x g for 5 min. Culture conditioned media were aspirated, and the pellets

resuspended in PBS. The cells in PBS were centrifuged at 90 x g over a FBS cushion

(5x the volume of PBS). This centrifugation step is to separate host cells from

unattached bacteria and dead host cells. After the FBS was aspirated, the cell pellet

was fixed with 4 % paraformaldehyde and 2 % glutaraldehyde in 0.1 M cacodylate

buffer, pH 7.2. Fixed tissues were processed with the aid of a Pelco BioWave laboratory

microwave (Ted Pella, Redding, CA, USA). The samples were washed in 0.1 M sodium

55

cacodylate pH 7.24, post fixed with buffered 2 % OsO4, washed with H2O and

dehydrated in a graded ethanol series 25 %, 50 %, 75 %, 95 %, 100 %. Dehydrated

samples were infiltrated in ethanol/LR White Resin (Electron Microscopy Sciences,

Hatfield, PA) 50 %, 100 %, and cured at 60 oC. Cured resin blocks were trimmed, thin

sectioned and collected on Formvar copper coated grids, post-stained with 2% aqueous uranyl acetate and Reynold’s lead citrate. Sections were examined with a Hitachi H-

7000 TEM (Hitachi High Technologies America, Inc. Schaumburg, IL) and digital images acquired with a Veleta 2k x 2k camera and iTEM software (Olympus Soft-Imaging

Solutions Corp, Lakewood, CO).

Relative Real-Time PCR

To investigate intracellular bacterial replication with molecular methods, RT-PCR was used to detect bacterial and host cell nucleic acids following S. mutans OMZ175 invasion of HCAEC or CASMC. Primers used in this study include universal 16S-sense

5’ACTACGTGCCAGCAGCC3’; 16S-antisense 5’GGACTACCAGGGTATCTAATCC3’;

18S-sense 5’CGCCGCTAGAGGTGAAATTCT3’; and 18S-antisense

5’CGAACCTCCGACTTTCGTTCT3’ [215]. Molecular methods were used to measure

bacterial replication over time using RT-PCR. Purified gDNA was used as a template.

RT-PCR was used to amplify the bacterial 16S and human 18S ribosomal gene

sequences using IQ™ SYBR® green supermix (Bio-Rad; Hercules, CA) detection and

specific primers provided above. RT-PCR data was analyzed by the comparative Ct method [187]. Assay controls include: 1) HCAEC or CASMC gDNA amplified with 16S and 18S primer sets in separate reactions; 2) S. mutans OMZ175 gDNA amplified with

16S and 18S primer sets in separate reactions; 3) no template control used with both

56

16S and 18S primer sets in separate reactions. The average Ct value of replicates was

determined per sample.

Calculation 1: ΔCt, average Ct of bacterial 16S gene – average of human 18S Ct,

was calculated to normalize the data. The ΔΔ Ct (Δ Ct n –Δ Ct 5h) was calculated and

used to calculate the fold change (2-ΔΔCt) in bacterial load per host cell. If the fold

change value was less than 1, then there was a reduction in bacterial load.

To measure bacterial replication in HCAEC, antibiotic protection assays were

performed on HCAEC seeded in 6-well plates to confluence. The harvesting times were

5, 7.5, 10, 12.5, 15, 17.5, and 20 h post infection, and the sample size per condition was

3. After the 5 h time point, the wells were washed with HBSS, and replaced with media

supplemented with and without the following antibiotics: gentamicin (300 µg ml-1) with

penicillin G (10 µg ml-1) in the presence or absence of chloramphenicol (10 µg ml-1).

Chloramphenicol was used as a negative control for bacterial replication, since it is

bacteriostatic on extracellular and intracellular bacteria. Total nucleic acids were

isolated using Trizol® reagent (Invitrogen™) per manufacturer instruction. Purified

gDNA was used as a template, after template quality control measurements were

obtained using NanoDrop 2000c UV-Vis Spectrophotometer (Thermo Scientific). Real

time PCR was used to amplify the bacterial 16S and human 18S ribosomal gene

sequences using IQ™ SYBR® green supermix (Bio-Rad; Hercules, CA) detection and

specific primers. The data was analyzed as described above.

To measure bacterial replication in CASMC, antibiotic protection assays were

performed on HCAEC seeded in 6-well plates to confluence. The harvesting times were

5, 10, 24, and 48 h post infection, and the sample size per condition was 3. After the 5 h

57

time point, the wells were washed with HBSS, and replaced with media supplemented with and without the following antibiotics: gentamicin (300 µg ml-1) with penicillin G (10

µg ml-1) in the presence or absence of chloramphenicol (10 µg ml-1). Genomic DNA

(gDNA) was extracted and purified using QiaAmp DNA extraction kit (Qiagen). Purified

gDNA was used as a template, after template quality control measurements were

obtained using NanoDrop 2000c. RT-PCR was used to amplify the bacterial 16S and human 18S ribosomal gene sequences using IQ™ SYBR® green supermix (Bio-Rad;

Hercules, CA) detection and specific primers. The data was analyzed as described

above.

Host Cell Cycling

Sequential antibiotic protection assays were performed to determine if intracellular

S. mutans OMZ175 exited endothelial cells and maintained infectivity. HCAEC were

seeded to confluence and infected with bacteria for 2 h, and the following day, washed,

and treated with media containing gentamicin 300 µg ml-1 and penicillin G 100 µg ml-1

for 24 h to kill extracellular bacteria (n=6). Next, the infected HCAEC were enzymatically

detached, diluted, and counted, and then equal volumes of infected HCAEC were

overlayed into duplicate wells onto attached uninfected HCAEC. An aliquot of the

infected HCAEC from each of the original wells was lysed, diluted, and plated to

determine the baseline intracellular bacterial load.

After 24 h, one well from each of the infected 6 samples was washed then

incubated for 3 h in the presence or absence of gentamicin 300 µg ml-1 and penicillin G

100 µg ml-1. Viable, cultivable intracellular bacteria were enumerated from cell lysates

through dilution and plating. The remaining well was processed for total extracellular

58

and intracellular bacteria by diluting and plating media conditioned media and cell lysates.

Bacterial Intercellular Spreading

This experiment was designed to determine whether host cell passaged S. mutans remain viable and infectious. HCAEC were seeded into 6 well plates with and without poly-L lysine coated 25 mm coverslips (Electron Microscopy Sciences; Hatfield, PA) at

1.5 x 105 cells per well for 24 h. The wells containing the coverslips were infected with

adenovirus expressing green fluorescent protein (GFP) (Welgen, Inc., Worcester, MA)

at an MOI of 10 virus particles per host cell for 24 h. An antibiotic protection assay was

completed on HCAEC seeded in wells without coverslips using S. mutans OMZ175 (2 h

infection followed by 3 h gentamicin 300 µg ml-1 and penicillin G 100 µg ml-1 treatment

in media), then the infected HCAEC were washed, detached, and overlayed onto the

washed monolayer of GFP expressing HCAEC in the presence and absence of

gentamicin (50 µg ml-1). After 24 h, the infected cells were fixed with 4 %

paraformaldehyde in PBS, counterstained with the DNA stain Draq 5™ (Cell Signaling

Technology®; Danvers, MA), and mounted using Prolong Gold (Invitrogen™). Controls

included slides that contained HCAEC that were only expressing GFP or only infected

with S. mutans. The microscopy was performed using a Leica inverted fluorescent

microscope with a Yokagowa spinning disk confocal scan head and a Roper Cascade II

EMCCD 512b camera set up for 3 channel fluorescent imaging.

Gene Expression Analysis

Microarray technology was used to explore the gene expression changes

attributed to pathogen associated molecular patterns, attachment, or attachment with

Cnm mediated/associated invasion by S. mutans collagen binding protein, Cnm, during

59

attachment and invasion of HCAEC using GeneChip® Human U133 Plus 2.0 Array

(Affymetrix, Inc; Cleveland, OH). For this assay, S. mutans strains, OMZ175 or

OMZ175:cnm, were used to infect HCAEC at 100 CFU per host cell for comparison to

non-infected controls. Co-cultures were carried out in quadruplicate. After 1 h post

infection, total RNA was extracted (RNeasy® mini kit; Qiagen), treated with DNase I

(Qiagen), eluted, quantified by using standard methods, and submitted to Dr. Henry A.

Baker’s laboratory, where the microarray chemistry and data analysis procedures were

completed. Double stranded cDNA was synthesized using the GeneChip® Array

protocol from Affymetrix (SuperScript® Double-Stranded cDNA Synthesis Kit; Invitrogen)

using 5 to 8 µg of total cellular RNA as a template. Double-stranded cDNA was purified

and used as a template for labeled cRNA synthesis. In vitro was performed

using a BioArray™ High-Yield® RNA Transcript Labeling Kit (T7; Enzo® Life Science,

Farmingdale, NY) to incorporate biotinylated nucleotides. cRNA was subsequently

fragmented and hybridized on microarray chips with proper controls. The microarrays

were hybridized for 16 h at 45 °C, stained with phycoerythrin-conjugated streptavidin,

and washed using the Affymetrix® protocol (EukGE-WS2v4) with an Affymetrix® fluidics

station, and followed by scanning with an Affymetrix® GeneChip® III Scanner.

Microarray data analysis was performed as previously described [216].

Affymetrix controls and probe sets whose signals were not detected in all samples

were removed from the analysis using expression filters. The remaining data set of

intensity values were variance normalized, mean centered, and ranked by their

coefficients of variation. Unsupervised hierarchical cluster analysis was performed on

the data set with the most variation across samples to reduce the background signal

60

variation on the analysis using Cluster software [217]. Treeview software was used to generate the heat map and cluster dendrograms [217]. Analysis was completed to

assess the extent of HCAEC responses to the S. mutans OMZ175 challenge over time.

Next, a supervised analysis was completed using log transformed raw signal

intensities for the probe sets that passed the initial expression filters in order to

investigate the gene regulation differences among treatments. BRB Array Tools (R.

Simon and A. Peng-Lam, National Cancer Institute, Rockville, MD) were used to

correlate the log transformed signal intensities. In each supervised analysis, biological

replicates were grouped into classes based on treatment, and probe sets significant at

the P <0.001 level for the class were identified. Next, leave-one-out cross-validation

(LOOCV) studies were used to compute the misclassification rate. The significant probe

sets were then used with nearest-neighbor predictor analysis.

Statistical Analysis

One-way analysis of variance (ANOVA) statistical analysis with Tukey’s post hoc

tests was performed using GraphPad Prism version 5.00 for Windows, GraphPad

Software, (San Diego, CA; http://www.graphpad.com/). Two-way analysis of variance

statistical analysis with Bonferroni post hoc tests was used to determine mean

differences over time. Bacterial CFU were log transformed prior to statistical analysis.

Statistical significance was defined as p value less than 0.05.

Results

S. mutans OMZ175 Invades Human Oral Cells in Culture.

This laboratory and our collaborators have reported that S. mutans OMZ175

invades and persists within primary endothelial cells in culture and that the bacterial cell

surface protein, Cnm, may contribute to systemic disease. Therefore, we wanted to

61

investigate whether cnm-expressing S. mutans could potentially use host cells that reside in the gingival tissues as a reservoir for S. mutans induced systemic diseases

[31, 126]. To this end, primary human oral cell lines (HOK and HGF-1) were infected with invasive S. mutans OMZ175 and invasion deficient strains OMZ175:cnm and

UA159.

The data indicate that the presence of Cnm contributed to S. mutans attachment and invasion of HOK when compared to strains without cnm. In a representative experiment, all of the S. mutans strains tested were able to attach to HOK (OMZ175,

5.8 E5 ± 2.0 E5 CFU; S. mutans OMZ175:cnm, 9.1 E4 ± 2.3 E4 CFU; UA159, 1.8 E4 ±

3.4 E3 CFU; p< 0.001), though at different efficiencies, (OMZ175, 5.8 %; S. mutans

OMZ175:cnm, 0.76 %; UA159, 0.22 %). The number of intracellular bacteria were

enumerated after completing a 5 h antibiotic protection assay, and the results indicate

that cnm contributes to invasion of HOK (9.5 % of attached OMZ175 invaded HOK) but

is not required for invasion, since 0.64 % of the OMZ175:cnm invaded the host cell,

(OMZ175, 5.6 E4 ± 8.3 E3 CFU; OMZ175:cnm, 5.8 E2 ± 1.6 E2; UA159, 3.1 ± 6.3;

p<0.001).

In a representative experiment, there was no difference in the ability of the

S. mutans strains to attach to HGF-1 in 30 min (OMZ175, 9.0 E3 ± 2.0 E3 CFU;

OMZ175:cnm, 2.4 E3 ± 1.9 E2; UA159, 5.2 E3 ± 1.8 E3 p < 0.348). Despite the fact

that all of the S. mutans strain attached to the HGF-1, only the strain with cnm was able

to invade HGF-1 efficiently (OMZ175, 2.3 E4 ± 9.2 E3 CFU; OMZ175:cnm, 3.3 E1 ± 4.2

E1; UA159, 2.3 E1 ± 1.6 E1 p < 0.001), Figure 2-1B. These results demonstrate that

62

different S. mutans strains are able to adhere to host oral cells, but only the strain with cnm was able to invade efficiently.

S. mutans invasion of HGF cells during epithelial injury may select for bacterial strains that are more commonly found in the blood as HGF reside in the vascularized connective tissue of the gingiva. Therefore, transmission electron microscopy was used to assess S. mutans contact with the HGF-1 surface and to observe S. mutans localization within HGF-1 cells, Figure 2-2 and Figure 2-3. Electron micrographs

revealed that S. mutans OMZ175 was engulfed by filipodia-like extensions on the

surface of the host, and that the intracellular bacteria localized to single membrane

vacuoles indicative of the endocytic pathway (Figure 2-2). Furthermore, bacterial

surface extensions were visible and appeared to come into direct contact with the host

membrane, indicating that the space between the bacterial cell and the vacuolar

membrane may be a space where bacterial cell surface molecules were washed away

during sample processing (Figure 2-3). The control noninfected HGF-1 cells contained

vacuoles and filipodia-like extensions similar to those observed in the presence of the

bacteria (Figure 2-4).

S. mutans Does Not Replicate Intracellularly.

Previously, our laboratory and collaborators assessed HCAEC intracellular

trafficking and persistence of S. mutans OMZ175 with transmission electron microscopy

[127]. After 24 h, large numbers of S. mutans were observed in the cytoplasm of

infected cells, suggesting that S. mutans escape the endosome and multiply within the

cytoplasm of the HCAEC. However, bacterial counts did not confirm bacterial

intracellular replication as there was a net loss (approximately 1 log) in bacterial load at

29 h post infection when compared to the bacterial load at 5 h post infection [127].

63

Since the ability of a bacterial pathogen to replicate intracellularly contributes to its persistence by providing a protective niche for growth and dissemination, we wanted to

determine if intracellular S. mutans was able to replicate within endothelial cells. In this

study, intracellular bacterial growth curves were completed to determine the intracellular

fate of S. mutans OMZ175 using continuous gentamicin treatment (50 µg ml-1) to kill

bacteria that exited the host and chloramphenicol treatment (10 µg ml-1) to inhibit

intracellular bacterial replication in the negative control. Results showed a reduction in

viable colony counts from antibiotic protection assays in both antibiotic treatment groups

over 25 h, with a one log reduction in cultivable intracellular S. mutans OMZ175

(Figure 2-5). There was no statistical difference in intracellular bacterial load between

treatment groups for each of the assay time points (5, 10, 15, 20, and 25 h post

infection), indicating that bacteria were likely not replicating intracellularly. This

experiment was repeated with similar results.

Since S. mutans normally grows in chains, the cell counts may have been

underestimated, or the intracellular S. mutans may have remained viable but become

uncultivable over time, which is a phenomenon that has been shown for other

intracellular bacterial species isolated from human atheromas [11, 27]. Therefore,

molecular techniques were used to measure relative amounts of bacterial 16S RNA

transcripts in HCAEC compared to HCAEC 18S transcripts during S. mutans OMZ175

invasion over time (Figure 2-6). Similar to the viable colony counts, the relative amount

of bacterial load decreased over time under continuous antibiotic treatment in both

treatment groups. These results indicate that intracellular S. mutans does not replicate

within HCAEC, and that S. mutans intracellular numbers begin to decline after 20 h.

64

Intracellular S. mutans Exit the Host and Remain Infectious.

Results obtained from bacterial growth curves indicated that intracellular

S. mutans numbers decreased slowly over time (Figure 2-5). Thus we wanted to

investigate whether intracellular S. mutans was able to exit endothelial cells and remain

infectious, allowing the invasion of other host cells. Therefore, sequential antibiotic

protection assays were performed with a sample size of 6 as described in the methods.

Briefly, HCAEC were infected with S. mutans OMZ175 for 2 h and then treated with

antibiotics for 24 h to kill extracellular bacteria. The infected HCAEC were washed,

detached, and overlayed onto uninfected HCAEC and co-cultured in media without

antibiotics. After 24 h, bacteria were enumerated from the co-culture conditioned media

(8.2 E5 ± 6.0 E5 CFU, n=12). These results demonstrated that S. mutans was viable

after being passaged through a host cell. To determine whether host cell passaged

bacteria were able to enter new cells, we compared the average initial intracellular bacterial load (1.0 E3 ± 0.45 E3 CFU, n=6) to the average intracellular bacterial load after overlaying infected HCAEC onto uninfected HCAEC (5.7 E4 ± 5.5 E4 CFU, n=6)

for 24 h (Figure 2-7). The results demonstrated that the total intracellular bacterial load

increased, and this increase was statistically significant (p< 0.005). Since S. mutans

does not replicate intracellularly, the bacteria must have exited the host, replicated in

the media, and entered into new cells. However, this data does not eliminate the

possibility that S. mutans may enter new host cell through intercellular spread. To

determine if the host cell passaged bacteria that had propagated and aggregated in the

media conditioned media were still infectious, conditioned media were collected,

pelleted, resuspended in fresh culture media, and used to infect HCAEC. An antibiotic

protection assay was performed: 1 h infection and 3 h antibiotic treatment. The host cell

65

twice, passaged bacteria were still infectious (2.0 E5 ± 0.32 E5 CFU, n=4). Additionally,

the bacteria in the conditioned media were tested for antibiotic susceptibility, since

bacterial aggregates were visible. The aggregated bacteria were killed with 3 h of

antibiotic treatment.

Confocal microscopy was also used to confirm that host cell passaged S. mutans

were still infectious. HCAEC attached to coverslips were transfected with nonreplicating

adenovirus expressing GFP. After 24 h, separate HCAEC seeded in culture flasks were

infected with S. mutans, treated with antibiotics to kill extracellular bacteria, detached,

and seeded over the HCAEC transfected with the viral vector. This viral vector

expresses GFP maximally 48 h post transfection. S. mutans was observed in the GFP

expressing HCAEC in the presence and absence of antibiotics suggesting that

S. mutans entered into new cells directly from cell to cell contact and confirmed that

exited bacteria were still infectious (Figure 2-8).

Cnm Affects the Expression of Endothelial Cell Cytoskeleton Linked Proteins.

To determine if invasive S. mutans strain OMZ175 elicited a different host

response than the invasion deficient strain S. mutans OMZ175:cnm, microarray analysis

was performed. Table 2-2 contains a list of genes linked to cytoskeleton

rearrangements that were differentially expressed after 1 h of infection with OMZ175

compared to the noninfected control (p<0.001) but were not differentially expressed

after 1 h of infection with the OMZ175:cnm mutant compared to the noninfected control

(p<0.001) as these genes are likely to be involved in Cnm induced pathways. Since

cytoskeletal rearrangements can be associated with mechanisms of endocytosis, this

data suggests that future work should be conducted to investigate the role Cnm may

have in host cell responses in terms of bacterial induced entry.

66

S. mutans OMZ175 Invades Human Coronary Artery Smooth Muscle Cells.

Now that there was evidence that S. mutans OMZ175 was able to exit HCAEC

and remain infectious, we wanted to determine if S. mutans was able to invade arterial

smooth muscle cells, as they are in close proximity to endothelial cells, typically

separated by a basement membrane. However, stimulated endothelial cells can induce

a cytokine response that may ultimately result in smooth muscle proliferation, as

manifested in the disease atherosclerosis. Furthermore, an ability to invade smooth

muscle cells would indicate the potential for S. mutans to traverse the endothelial lining

and disseminate into deeper tissues. Four wildtype strains of S. mutans and their

cognate cnm mutants: OMZ175, OM50E, LM7, and NCTC11060 were used to

determine the role of Cnm in CASMC adherence and invasion, as Cnm has already

been reported to have a role in the ability of these S. mutans strain to invade HCAEC

[126]. The mutant strains were provided by our collaborator Dr. Jacqueline Abranches.

S. mutans UA159, a natural cnm negative strain, was used as a negative control. All of

the S. mutans strains tested were able to adhere to CASMC, and there was no

statistical difference between the adherence efficiency of each wildtype and cognate

cnm mutant strain pair (Figure 2-9). This indicates that each of the S. mutans strains

was able to attach to CASMC in a Cnm independent manner. However, invasion results

showed that wildtype S. mutans strains expressing cnm were more successful at

invading CASMC by a factor of ~2 logs with a statistical difference of p< 0.05 (Figure 2-

9). Additionally, the cnm mutant strains’ abilities to invade were not significantly different

than that of the natural cnm negative strain UA159.

Transmission electron microscopy was used to confirm S. mutans OMZ175

invasion of CASMC, (Figure 2-10, 2-11). CASMC were infected with 100 CFU per host

67

cell. After 2 h, the CASMC were processed as described in the methods. Electron micrographs reveal that S. mutans OMZ175 was engulfed by filipodia-like extensions on the surface of the host, and that the intracellular bacteria localized to single membrane vacuoles (Figure 2-10). Similar to the HGF-1 results, bacterial adhesins were visible and appeared to come into contact with the host membrane (Figure 2-11). The control noninfected CASMC contained vacuoles and filipodia-like extensions similar to those observed in the presence of bacteria (Figure 2-12).

With evidence of S. mutans OMZ175 CASMC invasion, we wanted to know if

S. mutans was able to replicate intracellularly. Molecular techniques were used to measure relative amounts of bacteria gDNA normalized to CASMC gDNA over time

(Figure 2-13). The relative amount of bacterial load decreased over time under continuous antibiotic treatment. These results do not support the hypothesis that intracellular S. mutans replicates in CASMC. In contrast, bacterial gDNA in the pulse

antibiotic treated samples increased 5 fold; the pulse antibiotic treated group served as

the positive control, since this group was cultured in antibiotic free media after the initial

5 h antibiotic protection assay. Since there is no evidence of intracellular replication,

these results support the evidence that S. mutans is capable of exiting the host in a

viable state. The negative fold change in CASMC gDNA at 10 h post infection was likely

due to the wash steps and media replacement subsequent to the antibiotic protection

assay.

Discussion

Human oral keratinocytes and gingival fibroblasts have an important role in

immune surveillance of the gingiva by functioning as the first line of defense against

microbial subgingival colonizers. During periodontal disease, activated HOKs recruit

68

neutrophils to the gingival crevice to mount an immune response against the subgingival plaque. During this immune attack, HOKs may become the indiscriminate target of neutrophil degranulation, causing nonlytic detachment injury, which leaves the underlying connective tissue unprotected [218-221]. Once the connective tissue is exposed to infected gingival crevicular fluids, clotting cascades are activated, closely followed by HGF activation, migration, and proliferation, in an effort to prepare the wound for re-epithelialization. Since keratinized epithelium continually grows upward from the basal cell layer, shedding dead cells into the oral cavity, S. mutans invasion of

HOK cells may act as a reservoir for HGF infection, but would not be expected to directly bring S. mutans closer to the circulatory system Figure 2-14. Thus for the

purpose of this study, HGF cells, rather that HOK cells, were selected to investigate the

potential of S. mutans to use oral host cells to gain access to the circulatory system.

Attachment and invasion of host cells by bacteria typically involve specific

adhesins expressed on the surface of bacterial cells that recognize specific tissue types.

Thus, bacteria are known to have a predilection for specific tissue cell types and

environments. For example, S. mutans is abundant in dental plaque but is not found in

high numbers on the epithelial surfaces of the tongue; however, S. salivarius is typically

absent in dental plaque, but colonizes epithelial cells of the tongue in high numbers

[222]. Moreover, most bacterial pathogens have evolved to express adhesins that bind to specific host cell receptors. For example, adhesins, such as FbaB, expressed by certain strains of Group A Streptococci have been shown to be involved in endothelial

cell invasion and linked to invasive diseases such as necrotizing fasciitis [167].

However, some bacterial adhesins are less discriminating and bind to certain categories

69

or types of host cell surface molecules (e.g., carbohydrate and protein motifs). For example, the S. mutans PAc protein has been shown to specifically bind to α5β1 integrins expressed on endothelial cells, yet this adhesin has also been shown to specifically bind to additional host protein motifs such as collagen and fibronectin. While

studies have demonstrated that PAc is involved in host cell adhesion, it does not appear

to efficiently trigger invasion in endothelial cells or gingival fibroblasts. Thus, other bacterial adhesins are suspected to be involved in the mechanism of host cell invasion.

Epidemiological studies have reported that the S. mutans serotype distribution in

the oral cavity of healthy individuals differs from that of patients with cardiovascular

disease [88, 154, 223, 224]. Furthermore, the oral cavity and cardiovascular tissues of patients with cardiovascular disease were reported to have different S. mutans serotype

distributions with an increase of rare and untypable serotypes in both the oral cavities

and atherosclerotic tissues of patients with cardiovascular disease. These tissue

serotype distribution differences suggest that certain serotypes of S. mutans are more

likely to enter the bloodstream, encounter, adhere to, and multiply within cardiovascular

tissues than other serotypes. Other studies have reported that Cnm is expressed by

about 10% of S. mutans clinical strains, and that there is also a rare serotype bias for

the presence of this gene [67, 89, 124, 125]. Furthermore, recent reports have shown

that strains of S. mutans that express Cnm are able to invade and persist in human

coronary artery endothelial cells (HCAEC), while the Cnm negative strains were found

to be invasion deficient [126, 127].

The results of this study demonstrate that S. mutans OMZ175 invasion of oral

keratinocytes, gingival fibroblasts, and smooth muscle cells is dependent on the

70

presence of Cnm. Additionally, this study found no evidence that S. mutans replicates intracellularly in endothelial cells but it is possible that there are other cell types or cellular conditions in which intracellular S. mutans can replicate. The study also demonstrates that host cell passaged S. mutans remain viable and infectious after exiting endothelial cells. These results suggest that S. mutans is able to enter the oral epithelium to reside within cells found in deeper gingival tissues which may allow

S. mutans increased access to the vasculature of the gingiva. Consequently, the ability of S. mutans to invade oral cells may contribute to its ability to cause systemic disease.

Thus, individuals with invasive strains of S. mutans may be at increased risk of

infectious systemic diseases, especially if they have other co-morbidities that contribute

to inflammation such as gingivitis, diabetes or obesity. As this is the first study to

demonstrate that S. mutans invades coronary artery smooth muscle cells, it

demonstrates the potential of S. mutans to traverse the endothelium and disseminate

into deeper arterial tissues where it may contribute to chronic cardiovascular diseases

such as atherosclerosis, consistent with S. mutans exiting host cells having the ability to

infect other cells.

This study presents data that demonstrates that Cnm is key to the ability of

S. mutans to both attach to and enter oral keratinocytes, in contrast to gingival

fibroblasts, with which Cnm is significantly involved in the invasion of but not the

adherence. Thus, Cnm appears to be an adhesin for S. mutans attachment to oral

keratinocytes but not to gingival fibroblasts. Oral streptococci including S. mutans have

been previously reported to attach to and inhabit human buccal cells [225-227]. Reports

have also suggested that the ability of S. mutans to enter oral cells may provide a

71

competitive advantage for colonizing tooth surfaces in children, as S. mutans has been isolated from the oral cavities of predentate children [228-230]. S. mutans and

S. sobrinus have both been previously reported to adhere to and invade gingival

fibroblasts, especially when present in biofilms [206]. Fibroblasts are located in the

connective tissues under the epithelium and function to form extracellular matrix

substances such as collagen and elastin. When gingival tissues become inflamed, the

vasculature becomes more permeable and leaky and the gingival crevice can deepen,

acting as a reservoir for multiple bacterial species. The results of the study reported

here suggest that individuals with cnm-expressing strains of S. mutans may be at an

increased risk of bacteremia and other diseases outside the oral cavity. Based on the

results, we propose a model for S. mutans route from the gingival tissues to the

circulatory system (Figure 2-15).

The intracellular location of S. mutans OMZ175 was, previously, confirmed in

endothelial cells using transmission electron microscopy at 30 min, 5 h, and 29 h post

infection [127]. The Abranches et al. 2009 study demonstrated that S. mutans entered

HCAEC and were initially found in single and multiple membrane vacuoles, but at 5 and

29 h post infection, some bacteria escaped to the cytoplasm, while those bacteria in

vacuoles appeared intact, suggesting the absence of lysosomal fusion [127]. Other

studies have shown that once inside the host cell, some pathogens (e.g., S. pyogenes)

are destroyed by exposure to increasingly acidic and proteolytic conditions of the early,

late, and lysosomal compartments, while other pathogens are able to subvert this

degradative pathway. Interestingly, we found that after 29 h, large numbers of

S. mutans were found in the cytoplasm of infected cells, suggesting that S. mutans

72

replicates inside HCAEC and escape the endocytic pathway before the late endosome and lysosome fuse. However, bacterial plate counts from antibiotic protection assays did not support this in that CFUs did not increase over time.

Attachment, entry, trafficking, persistence, and exit are all important for successful

bacterial invasion of host cells [231]. Previous reports described each of these stages to

occur during S. mutans invasion of endothelial cells except the abilities to replicate

intracellularly (potentially contributing to persistence) and to exit the host [127]. Thus we

investigated the ability of S. mutans OMZ175 to replicate within endothelial cells and

determined whether S. mutans was able to exit the host. We also sought to determine if

bacteria that exit a cell are still infectious. The results of intracellular bacterial growth

curves and molecular methods to study intracellular replication (Figure 2-5 and 2-6)

indicate that S. mutans was able to persist within primary endothelial cells, but there

was no evidence of intracellular replication. S. mutans is dependent on carbohydrates

for energy; however, a ready source of unphosphorylated carbohydrate would not likely

be available inside of the host, which is consistent with our findings. The study reported

here also demonstrates that S. mutans OMZ175 is able to exit host cells and that host

cell passaged bacteria are still infectious. Additionally, microarray analysis found that

the presence of Cnm changes endothelial gene expression patterns in HCAEC

including genes linked to cytoskeletal rearrangements. However, future studies must be

performed to determine if cytoskeletal rearrangements are involved in Cnm-mediated

entry. Reports on Group A Streptococci mechanisms of invasion provide evidence that

different cell adhesins direct entry of endothelial and epithelial cells through multiple

73

pathways [166, 232-234]. Based of the results of our study, we propose an invasion cycle model of cardiovascular tissues (Figure 2-16).

The limitations of this study can be attributed by in large to the in vitro model(s) used to investigate S. mutans’ invasion and trafficking. For instance, the 1-2 log difference in bacterial adherence and invasion between HOK and HGF-1 may be due to their morphological difference as HOK have a cobblestone-like appearance and HGF-1 are spindle shaped. Although the confluent monolayers used in the assays are more physiologically relevant when studying host cell endothelium and epithelium interactions with bacteria, as bacteria might be expected to come into direct contact with these cell types in the oral cavity or the bloodstream, the limitations of the model must be considered when interpreting fibroblast and smooth muscle cell invasion data as these cells are typically surrounded by collagen, a likely competitive inhibitor for Cnm interactions with host cell surface molecules. Furthermore, it is unknown whether

S. mutans enters the bloodstream in a planktonic or biofilm state. This study was

completed using planktonic S. mutans.

Future studies will be directed toward understanding the mechanism of S. mutans

entry into endothelial cells, as the mode of entry may influence the intracellular

trafficking and fate of the bacteria.

74

Table 2-1. S. mutans strains used in this study Strain Origin Serotype Source UA159 Dental plaque c University of Alabama OMZ175 Dental plaque f B. Guggenheim LM7 Dental plaque e P. Caufield OM50E Dental plaque e P. Caufield NCTC11060 Dental plaque f University of Rochester* OMZ175:cnm cnm knockout University of Rochester* OM50E:cnm cnm knockout University of Rochester* LM7:cnm cnm knockout University of Rochester* NCTC11060:cnm cnm knockout University of Rochester* *Strains obtained from J. Abranches at the U niversity of Rochester [126]

Table 2-2. Microarray differential gene expression in HCAEC due to Cnm interactions 1 h post infection

Cytoskeleton rearrangements Endocytic pathway Upregulated ARF6, ARHGAP42, CDC42SE2, AP1S3*, AP3B2*, APPL2*, CTNNB1, C2CD4B, DOCK10, ARF6*, CLINT1*, CPNE3, EHBP1, EZR, GIT2, GBP1, CPNE8, EHBP1*, LDLR*, GIT2, ITGB3, MERTK PRKCE, MERTK, MAP3K2**, PHLDB2, PIP5K1A, RGL1, SORBS1, PICALM*, PIP5K1A, RHOB*, SOS2, SPIRE1, SPTAN1, RHOBTB3, SCIN, SCLT1*, SWAP70, TBC1D1, TRIO SCYL2*, SNX18*, WDFY1

Downregulated AAGAB*, AAGAB, ANXA2P1, ANXA2P3, ATG16L2, ACTR2, ITGA7, PRKAA1 CEACAM3, EHD4*, SDPR**, VPS13A* Genes linked specifically to *clathrin and **caveolin mediated endocytosis

75

Figure 2-1. S. mutans attached and intracellular A) HOK and B) HGF-1. Host cells were infected for 30 min (adherence assay) or 5 h (antibiotic protection assay) with 100 CFUs per host cell. The number of viable S. mutans CFUs recovered is shown ± SD. S. mutans OMZ175 (cnm-positive strain) attached and invaded HOK and invaded HGF than more efficiently than the cnm-negative control strains: OMZ175:cnm and UA159. However, the presence of cnm did not appear to contribute to HGF attachment. This figure is representative of at least 3 separate experiments. One-way analysis of variance (ANOVA) statistical analysis with Tukey’s post hoc tests was used to determine mean differences of log transformed data points. (***, p< 0.001)

76

Figure 2-2. Electron micrograph of HGF-1 invasion by S. mutans OMZ175 after 2 h of infection. The HGF-1 were detached, fixed and prepared for transmission electron microscopy prior to being examined with a Hitachi H-7000 TEM. Attached bacteria appeared to be engulfed by filipodia-like extensions (black arrows). Upon entry, intracellular bacteria were found in membrane bound vacuoles (white arrows). None of the bacteria were observed free in the cytoplasm.

77

Figure 2-3. Electron micrograph of S. mutans OMZ175 invasion of HGF-1 after 2 h infection. Bacterial cells entered through a vacuole with a single membrane. Structures that appear to be bacterial adhesins were visible (white arrows) and appeared to be in contact with the HGF-1 membrane (black arrows).

78

Figure 2-4. Electron micrograph of uninfected HGF-1. Vacuoles and filipodia like extensions can be observed.

79

Figure 2-5. Intracellular S. mutans OMZ175 growth curve. The number of S. mutans CFUs recovered from intracellular compartments. Gentamicin was used to kill any bacteria that exited the host, and chloramphenicol was used to inhibit bacterial replication within the host cell. Two-way ANOVA with Bonferroni post hoc tests was used to calculate mean differences in log transformed data points per antibiotic treatment for each individual time point. ANOVA statistical analysis with Tukey’s post hoc tests was used to determine mean differences of log transformed data points within in the same treatment group over time. There was no difference in intracellular bacterial load when comparing mean values between treatment groups at each of the time points. However, there was a statistical difference in bacterial load within the same treatment group between times 5 and 25 h and 5 and 30 h (p< 0.001). This figure is representative of 2 separate experiments.

80

Figure 2-6. Molecular methods to measure bacterial replication over time using RT- PCR. HCAEC were infected for 2 h, washed, and delivered a 3 h pulse of media with gentamicin and penicillin G antibiotic to kill extracellular bacteria. Next, the antibiotic media was aspirated, washed, and replaced with gentamicin (50 µg ml-1) ± chloramphenicol (10 µg ml-1) for continuous antibiotic treatment. gDNA was isolated every 2.5 h for a total of 25 h. RT- PCR data was analyzed by the comparative Ct method [235]. The average Ct value of replicates was determined per sample. A) ΔCt, average Ct of bacterial 16S gene – average Ct of host 18S, was calculated to normalize the data. The ΔΔCt (ΔCt n –ΔCt 5h) was calculated and used to calculate the fold change (2-ΔΔCt) in bacterial load per host cell. If the fold change value was less than 1, then there was a reduction in bacterial load. For all calculations n is the value at time points after 5 h.

81

Figure 2-7. Sequential antibiotic protection assays to determine if host cell passaged S. mutans remain viable and infectious. Comparison of initial intracellular bacterial load to the intracellular bacterial load after overlaying infected HCAEC onto uninfected HCAEC. Student’s t-test was used to determine statistical significance between the log transformed recovered CFUs. (p< 0.005, n=6).

82

A)

B)

Figure 2-8. Micrographs of sequential antibiotic protection assays to determine if HCAEC passaged S. mutans remain viable and infectious. Overlaying S. mutans OMZ175 infected HCAEC onto adenovirus GFP transfected HCAEC in the A) absence B) presence of antibiotics. S. mutans OMZ175 infected HCAEC exit the host to infect Ad-GFP expressing host, since bacteria were found in the GFP-labeled cells. Green color indicates fluorescence at 488 nm and red at 647 nm.

83

A)

B)

Figure 2-9. S. mutans A) attachment and B) invasion of CASMC. Results indicate that cnm expression does not contribute to attachment, but does contribute to invasion of CASMC. The number of viable S. mutans CFUs recovered is shown ± SD. One-way analysis of variance (ANOVA) statistical analysis with Tukey’s post hoc tests was used to determine mean differences of log transformed data points (ns, not significant, ***, p< 0.001). This figure is representative of three separate experiments.

84

Figure 2-10. Electron micrograph of CASMC invasion by S. mutans OMZ175 after 2 h of infection. The CASMC were detached, fixed and prepared for transmission electron microscopy prior to being examined with a Hitachi H-7000 TEM. Attached bacteria appeared to be engulfed by filipodia-like extensions (black arrows). None of the bacteria were observed free in the cytoplasm.

85

Figure 2-11. Electron micrograph of S. mutans OMZ175 invasion of CASMC after 2 h infection. Bacterial cells entered through a vacuole with a single membrane. Structures that appear to be bacterial adhesins were visible (red arrows) and appeared to be in contact with the CASMC membrane (black arrows).

86

Figure 2-12. Electron micrograph of uninfected CASMC. Vacuoles and filipodia like extensions can be observed.

87

Figure 2-13. Molecular methods to measure bacterial replication over time using RT- PCR. CASMC were infected for 2 h and treated with continuous or 3 h pulsed antibiotics. gDNA was isolated at 5, 10, 24, and 48 h post infection. The relative fold change of 16S to 18S rRNA gene copies over time was calculated using the comparative Ct method, normalized to 5 h data [235]. S. mutans OMZ175 replication was observed in the positive pulsed antibiotic control at 24 and 48 h post infection but not in the continuous antibiotic treatment groups.

88

Figure 2-14. Reservoir Model: Oral Tissue. This diagram depicts our reservoir model for S. mutans infection of oral epithelium and dissemination to gingival fibroblasts. In this model, S. mutans attaches to and may invade oral epithelial cells and these cells become a reservoir for future oral infections (i.e. dental caries). As epithelial cells serve as a regenerative protective barrier, the infected epithelial cell is likely to undergo shedding or cell death. Under conditions of periodontal disease or mechanical injury, the connective tissues and supportive cells are exposed. Once the connective tissue is exposed to oral bacteria, gingival fibroblasts become activated; and these cells proliferate, fill in the wound bed with extra cellular matrix (e.g., collagen), and initiate an immune response through the production of cytokines. Once the connective tissue is exposed, invasive S. mutans have an opportunity to enter gingival fibroblasts, which then become a reservoir for dissemination, and these bacteria may gain access to the circulatory system. HOK- human oral keratinocytes; HGF- human gingival fibroblasts; OMZ175- invasive strain of S. mutans

89

Figure 2-15. Overview of S. mutans invasion of human tissues. Flow chart of the proposed route of infection from the oral cavity to the artery and disease outcome with graphic diagram to illustrate the proposed route of infection. G means gingival epithelium.

90

Figure. 2-16. Invasion Cycle Model: Cardiovascular Tissue. This diagram depicts our invasion cycle model for S. mutans infection of human coronary artery endothelial cells (HCAEC) and dissemination to intimal resident arterial smooth muscle cells (CASMC). In this model, S. mutans attaches to and may invade HCAEC. The intracellular bacteria then cycle-out of the HCAEC into the tunica intima where they can replicate and disseminate to intimal resident CASMC. Once invasive S. mutans have an opportunity to enter CASMC, they may become a reservoir for persistence. OMZ175- invasive strain of S. mutans

91

CHAPTER 3 GENE EXPRESSION CHANGES IN HCAEC DURING INFECTION WITH STREPTOCOCCUS MUTANS OMZ175

Introduction

In healthy tissues, vascular endothelial cells maintain blood fluidity, regulate blood flow, control vessel-wall permeability and quiesce circulating leukocytes (reviewed in

[236-238]. Failure of endothelial cells to adequately perform any of these basal functions constitutes endothelial dysfunction, a contributing factor to atherosclerosis and other types of cardiovascular disease. Atherosclerosis is a progressive disease that results from an excessive, inflammatory-fibroproliferative response to various forms of insult to the endothelium and smooth muscle of the artery wall [239]. It is characterized by the formation of plaques consisting of foam cells, immune cells, vascular endothelial cells, smooth muscle cells, platelets, extracellular matrix, and a lipid-rich core with extensive necrosis and fibrosis of surrounding tissues [239]. In the initial stages of lesion formation, activated endothelial cells present several types of leukocyte adhesion molecules at their membrane surface and secrete cytokines [240]. Cytokines, small soluble proteins that include interleukins, , growth factors, colony-stimulating factors, cytotoxic factors, activating or inhibitory factors and chemokines, play important roles in not only tissue homeostasis but in regulating the intensity and duration of the immune response in many infectious diseases. Various cytokines found to be expressed by endothelial cells in atherosclerotic lesions include interleukin-1α (IL-1α),

IL-1β, IL-5, IL-6, IL-8 (CXCL8), IL-11, IL-14, IL-15, GM-CSF, M-CSF, and CD40L [241,

242] and are reviewed in Tedgui and Mallat [243].

Leukocyte recruitment into sites of inflammation is a tightly regulated process.

Although cytokines are crucial in this process, adhesion molecules present in the

92

membrane of endothelial cells are also equally important as several of these molecules have been shown to be detected in atherosclerotic lesions [244, 245]. The role of

adhesion molecules in the development and progression of atherosclerosis is reviewed

in Blankenberg et al. [246]. Briefly, activated endothelial cells express both soluble and

membrane anchored adhesion molecules including P-selectin, E-selectin, vascular cell adhesion molecule 1 (VCAM-1) and intercellular adhesion molecule 1 (ICAM-1), allowing for the adhesion and diapedesis of leukocytes to the site of inflammation/infection [244, 247].

Different studies have shown modulation of cytokines and adhesion molecules in endothelial cells in response to stimulation by multiple microorganisms including

S. mutans [248-252]. The purified recombinant PAc protein of S. mutans OMZ175 has

been shown to directly upregulate E-selectin, ICAM-1, and VCAM-1 mediated by lectin activity for N-acetylneuraminic acid and L-fucose containing receptors [253, 254].

Studies have shown that purified recombinant PAc specifically binds to α5β1 integrins

on the surface of human saphenous vein endothelial cells (HSVEC) in an N-

acetylneuraminic acid dependent manner [140, 189]. Furthermore, the purified

recombinant PAc protein and the purified rhamnose glucose polymer of S. mutans

OMZ175 have been shown to bind and signal through toll like receptors TLR2 and TLR4

and the membrane anchored co-receptor CD14 [255-257]. Endothelial cells

constitutively express TLR4 and increased expression levels of TLR4 in atherosclerotic

lesions has been shown to correlate with plaque destabilization and plaque progression

[149, 258-261]. To date, studies directly investigating the role of other S. mutans

93

adhesins, like Cnm, in modulating the expression of endothelial adhesion molecules have not been reported.

Human aortic endothelial cells infected with S. mutans up-regulate inflammatory cytokine production (IL-6, IL-8, CCL2), consistent with the hypothesis that S. mutans is

involved in the pathogenesis of atherosclerosis [253, 262, 263]. Using an in vivo model

of restenosis in an atherogenic mouse model, S. mutans mediated an exacerbation of

disease pathology demonstrated by atherosclerotic plaque, increased inflammation in

the outer adventitial area, and macrophage infiltration [31]. In this study microarray

technology was used to investigate the global transcriptional responses of primary

human coronary artery endothelial cell (HCAEC) to S. mutans OMZ175 challenged at 1

and 5 h compared to uninfected control. There were 7,550 probe sets reported to be

differentially expressed between the groups (false detection rate; FDR <0.05).

There is evidence that apoptotic cells are found within atheromatous tissue (e.g.,

foam cells) and that apoptosis of endothelial cells may have a role in the progression of

atherosclerosis [264-266]. Many bacterial species, including streptococci, can modulate

host cell apoptosis [177, 267-272]. For example, S. gordonii and S. pyogenes have

been shown to induce apoptosis in epithelial and endothelial cells, respectively [177,

269]. In addition, lipoteichoic acids of S. mutans have been shown to affect the growth

and the mitotic activity of epithelial cells [273], and to induce apoptosis in dental pulp

cells [274]. However, information regarding the effects on the fate of HCAEC upon

contact/invasion with live S. mutans to this point had been lacking.

94

Materials and Methods

Preparation of Confluent Human Cells in Culture.

Primary human coronary artery endothelial cells (HCAEC; Lonza, Allendale, NJ), were cultured in endothelial cell basal medium-2 (EBM-2; Lonza) supplemented with

EGM-2MV single-use aliquots (Lonza) for a maximum of 8 passages. The primary cells

were maintained at 37 °C in a humidified, 5 % CO2 atmosphere. The cells were washed

with Hanks buffered saline solution (HBSS; Corning Inc., Manassas, VA), detached

using Accutase™ (Innovative Cell Tech., Inc., San Diego, CA), and washed in cell

appropriate media. The cells were counted using a Coulter Counter (Beckman Coulter

Model Z1; Brea, CA) and seeded in flat-bottom tissue culture treated plates (Corning)

followed by overnight incubation at 37°C in a 5% CO2 atmosphere.

Bacterial Culture

S. mutans was cultured in Todd Hewitt broth overnight (TH; Becton Dickinson &

Co.). Bacterial cultures were pelleted at 4,629 x g for 5 min using a table top centrifuge

at rt. Culture conditioned media were aspirated, and the pellets were resuspended in

sterile phosphate-buffered saline (PBS, pH 7.2; Corning). Next the bacteria were diluted

in EBM2 complete media without antibiotics to obtain bacterial suspensions containing

100 colony-forming units (CFU) per host cell.

Gene Expression Analysis

This experiment was designed to evaluate gene expression changes of HCAEC

infected by S. mutans OMZ175 over time using GeneChip® Human U133 Plus 2.0

Array (Affymetrix, Inc; Cleveland, OH). HCAEC were co-cultured with bacteria at 100

cfu per host cell. Quadruplicate repeats were carried out for each treatment. After either

1 or 5 h post infection, total RNA was extracted using the RNeasy® mini kit from

95

Qiagen, treated with DNase I, purified, and quantified by using standard methods. cDNA was synthesized using the GeneChip® Array protocol from Affymetrix (SuperScript®

Double-Stranded cDNA Synthesis Kit; Invitrogen); 5 to 8 µg of total cellular RNA was used as a template to amplify mRNA species for detection. Double-stranded cDNA was purified and used as a template for labeled cRNA synthesis. In vitro transcription was performed using a BioArray™ High-Yield® RNA Transcript Labeling Kit (T7; Enzo® Life

Science, Farmingdale, NY) to incorporate biotinylated nucleotides. cRNA was subsequently fragmented and hybridized on GeneChip® Human U133 Plus 2.0 Array

(Affymetrix) with proper controls. The microarrays were hybridized for 16 h at 45 °C,

stained with phycoerythrin-conjugated streptavidin, and washed using the Affymetrix®

protocol (EukGE-WS2v4) with an Affymetrix® fluidics station, and then followed by

scanning with an Affymetrix® GeneChip® III Scanner.

Microarray data analysis was performed as previously described [216]. Affymetrix

controls and probe sets whose signals were not detected in all samples were removed

from the analysis using expression filters. The remaining data set of intensity values

were variance normalized, mean centered, and ranked by their coefficients of variation.

Unsupervised hierarchical cluster analysis was performed on the data set with the most

variation across samples to reduce the background signal variation on the analysis

using Cluster software [217]. Treeview software was used to generate the heat map and

cluster dendrograms [217]. Analysis was completed to assess the extent of HCAEC

responses to the S. mutans OMZ175 challenge over time.

Next, a supervised analysis was completed using log transformed raw signal

intensities for the probe sets that passed the initial expression filters in order to

96

investigate the gene regulation differences among treatments. BRB Array Tools (R.

Simon and A. Peng-Lam, National Cancer Institute, Rockville, MD) were used to

correlate the log transformed signal intensities. In each supervised analysis, biological

replicates were grouped into classes based on treatment, and probe sets significant at

the P <0.001 level for the class were identified. Next, leave-one-out cross-validation

(LOOCV) studies were used to compute the misclassification rate. The significant probe

sets were then used with nearest-neighbor predictor analysis.

RT2 Profiler™ PCR Array

This assay was performed using RNA that was collected for the microarray assay

as directed by manufacturer’s protocol, and was used to validate gene expression levels

for human adhesion molecules 5 hrs post-infection. RNA (0.5 ug) from each replicate

(n=4) was pooled, and single strand cDNA was synthesized using the RT2 First Strand

Kit (Qiagen). The data was analyzed using the web based data analysis tool, RT²

Profiler™ PCR Array Data Analysis version 3.5 [275]. Expression levels were

normalized to the average CT values generated by HPRT1, RPL13A, GAPDH, and

ACTB.

RT-PCR Validation

HCAEC were co-cultured with bacteria at 100 CFU per host cell. Co-cultures and

uninfected control were carried out in triplicate. After 5 h infection, total RNA was

extracted using the RNeasy® mini kit from Qiagen, treated with DNase I, purified, and

quantified by using standard methods. Single strand cDNA was generated from RNA

samples using iScript™ cDNA synthesis kit (Bio-Rad) as per the manufacturer’s

instructions. The RT-PCR reactions were prepared using IQ SYBR Green RT-PCR kit

(Bio-Rad) as per the manufacturer’s instructions with custom primer pairs (Table 3-1) to

97

gene targets and run on an iQ5-icycler (Bio-Rad). RT-PCR data was analyzed by the comparative Ct method. The average Ct value of replicates was determined per target.

ΔCt, average Ct of target gene – average Ct of beta-actin control gene, was calculated

to normalize the data. The ΔΔCt (ΔCt infected – ΔCt uninfected) was calculated to

determine the difference in expression of target genes. To determine the fold change in

expression value for each target gene, the 2-ΔΔCt was calculated.

Enzyme Linked Immunosorbant Assay (ELISA)

The level of cytokine proteins (IL-6, IL-8, IL-1α, IL-1β) secreted by HCAEC during

invasion with S. mutans OMZ175 was investigated using capture ELISA. For this,

HCAEC were seeded in 24 well plates to confluence for 24 h. HCAEC were infected

with S. mutans for 5 h at 100 CFU per host cell in EBM2 complete media without

antibiotics. The conditioned media were collected and stored at -80 oC. For ELISA,

culture conditioned media were thawed on ice and assayed according to the

manufacture’s instruction. Human IL-6 CytoSet™, Human IL-8 CytoSet™, and Human

IL-1β CytoSet™ (Invitrogen, Camarillo, CA) ELISA kits were used with the manufacturer

recommended Buffer Kit for Antibody Pairs (Novex®). Human IL-1α/IL-1F1 Quantikine®

ELISA kit supplied all assay materials (R&D Systems, Inc., Minneapolis, MN).

The level of vascular endothelial growth factors (VEGF) and platelet derived

growth factor (PDGF AA) produced by HCAEC after 24 h of infection with S. mutans

OMZ175 was investigated using capture ELISA. For this, HCAEC were seeded in 24

well plates to confluence for 24 h. The standard media (EBM-2) for culturing endothelial

cells contains growth factor rich fetal bovine serum (FBS) and additional growth factor supplements, such as VEGF; therefore the media used to infect HCAEC for ELISA analysis was RPMI media supplemented with 0.1% FBS (Atlanta Biologicals, Inc.,

98

Flowery Branch, GA) and 1% bovine serum albumin (Sigma). HCAEC were infected with S. mutans for 5 h at 100 CFU per host cell in RPMI media with supplements without antibiotics. Conditioned media were aspirated, and the HCAEC were washed 2x with HBSS and placed in RPMI media with supplements and containing gentamicin

(300 µg ml-1) and penicillin G (100 µg ml-1). The conditioned media were collected and

stored at -80 oC. For VEGF and PDGF AA ELISA (Thermo Scientific, Rockford, IL)

(Abcam, Cambridge, MA), conditioned media were thawed on ice and assayed

according to the manufacture’s instruction.

Caspase Activity Assay

Caspase activity was measured to determine the induction of apoptosis of HCAEC

by S. mutans. HCAEC extracts were collected at each time point of invasion and

analyzed for caspase 3/7 activity using a colorimetric caspase assay (Apo-ONE®

Homogeneous Caspase-3/7 Assay; Promega, Madison, WI) according to the

manufacturer’s instructions. The caspase substrate rhodamine 110, bis-(N-CBZ-L-

aspatyl-L-glutamyl-L-valyl-L-aspartic acid amide; Z-DEVD-R110) is sequentially

cleaved; and with the removal of DEVD peptides, the rhodamine 110 leaving group

becomes intensely fluorescent. The caspase activity was measured using the BioTek®

Synergy™ 2 Multi-Mode Microplate Reader (Bio Tek Instruments, Winooski, VT). The

amount of fluorescent product produced is proportional to the amount of caspase 3/7

activity. Caspase involvement was evaluated by treating S. mutans-challenged HCAEC

with the irreversible broad-spectrum caspase inhibitor z-VAD-FMK (Calbiochem,).

Assays to Determine the Ratio of Necrosis/Apoptosis

Two assays were used to determine the number of necrotic versus apoptotic infected cells. For the first assay, S. mutans-infected HCAEC, at the indicated time

99

points, were treated with propidium iodide (PI) and the nuclei were counterstained with

Hoechst 33258 (Sigma-Aldrich®), which labels DNA. Condensed chromatins are a universal marker of apoptotic cells. The number of PI-positive cells (necrotic cells) and the number of condensed chromatin-cells (apoptotic cells) relative to the total number of nuclei per field were counted by fluorescence microscopy in a blind manner. Each condition was assayed in triplicate, and at least 300 cells were counted in each well. A topoisomerase inhibitor, Camptothecin (Sigma-Aldrich®), was used as a positive control for induction of apoptosis, and unchallenged HCAEC were used as the negative control.

Microscopy Analysis of Apoptosis

Early in the apoptotic cascade of events, phosphatidyl serine residues are translocated from the internal to external leaflet of the cell membrane, while the cell membrane retains its integrity. Accordingly, Annexin V binds specifically to phosphatidyl serine residues and was used as a probe for apoptosis [276]. Ethidium homodimer III

(EthD-III) was used as an exclusion method for plasma membrane integrity of cells.

Annexin V and EthD-III were used in association to determine if dead cells were necrotic (only PI-labeled cells) or apoptotic (Annexin V-labeled cells). HCAEC were seeded onto coverslips with poly-L-lysine coating. Coverslips with S. mutans-infected

HCAEC were collected 5 h post infection, and the cells were stained using a kit from

PromoKine (Heidelberg, Germany) per the manufacturer’s instruction. Coverslips were mounted onto microscope slides with ProLong® Gold antifade reagent. The slides were observed with a Leica DM LB2 upright fluorescent scope (Leica Microsystems; Wetzlar,

Germany), and micrograph images were captured with a QImaging camera and

QCapture Pro software and analyzed using ImageJ software [277, 278]. Camptothecin

(10 µM) was used as a positive control for the induction of apoptosis; hydrogen peroxide

100

was used as a positive control for necrosis, and unchallenged HCAEC were used as a negative control.

FACS Analysis of Apoptosis

For FACS analysis infected and control cells were stained as above, using

Annexin V and ethidium homodimer III. Cells were then analyzed using a FACSCalibur flow cytometer (BD Biosciences). The cytometric data produced was analyzed using the

FACSDiva™ Software Version 6.1.2 (BD Biosciences). Camptothecin (10 µM) was used as a positive control for the induction of apoptosis; hydrogen peroxide (2 mM) was used as a positive control for necrosis, and unchallenged HCAEC were used as a negative control.

Results

Microarray Summary

The purpose of this microarray expression analysis was to provide a broad overview of host/pathogen interactions and pathways that are altered in response to these interactions to look at specific pathways that are associated with endothelial activation/dysfunction and to generate potential leads for future studies. A comparison of transcriptional profiles between uninfected HCAEC and S. mutans OMZ175 infected

HCAEC at 1 and 5 h was used to determine the overall transcriptional responses to

S. mutans challenge. RNA from confluent monolayers was isolated, labeled, and hybridized onto microarrays, each containing 54,675 probes representing more than

38,500 well characterized genes and UniGenes. A low level analysis was completed, using Robust Multi-Array Average (RMA), to create an expression matrix from background corrected, log2 transformed, and quantile normalized signal intensity data.

Signal intensity data for 54,675 probe sets were included in the unsupervised clustering

101

analysis and supervised class prediction analysis as these probe sets passed the initial filtering criteria. The unsupervised hierarchial clustering showed that biological replicates clustered together (data not shown). A time series analysis using data from all three conditions identified 7550 probe sets that were differentially expressed with a false discovery rate (FDR) of p<0.05. When comparing each of the treatments, the major separation node occurred between the 5 h OMZ175 treatment group and the uninfected control and 1 h OMZ175 treated groups (Figure 3-1), indicating that the cluster of up- regulated genes between the control and 1 h OMZ175 treatments were most similar.

Additionally, pairwise comparisons were completed as described above using different iterations of the three test conditions. For this analysis, signal intensities were normalized between groups and supervised analyses were completed. The leave-one- out cross-validation method was used to compute a misclassification rate, which indicated that the classifiers correctly predicted the treatment groups. Significance was set to p< 0.001, and the number of probe sets that were differentially expressed are reported in Table 3-2.

Ontology Analysis

Ontology analysis tools (BRB-ArrayTools Version 4.1.0 and Bioconductor annotation package Version 2.4.5) were used to group genes into biologically relevant metabolic pathways. The signal intensity data was separated into 3 classes based on treatment and 54,675 probe sets were included in the analysis as they passed the filtering criteria. The probe sets were organized into gene sets based on Biocarta

Pathway definitions. The total number of gene sets investigated was 309. The univariate test used was the F-test. The random variance model was not used because the distribution assumptions were not satisfied for this model. 120 out of 309 gene sets

102

exceeded the significance threshold of p<0.005. The LS/KS permutation tests identified

117 significant gene-sets; this test was designed to find gene sets that have more differentially expressed genes among the classes than expected by chance. Efron-

Tibshirani's test identified 12 significant gene-sets under 200 permutations; this test uses 'maxmean' statistics to find differentially expressed gene sets. Table 3-3 is an abridged list of the Biocarta Pathways in endothelial cells that were impacted over time when challenged with S. mutans OMZ175.

RT-PCR Validation of Microarray

RT-PCR was performed using primers to transcriptional factors (cFOS, JUN,

EGR1, EGR2, EGR3) that were shown to be differentially regulated during 5 h invasion with S. mutans OMZ175 compared to an uninfected control. These transcriptional factors are inducers of cell motility and apoptosis. These experiments were designed to validate the microarray experiment results, so the RNA used was isolated from a different set of invasion experiments than the microarray. The internal transcriptional control was beta-actin. Using microarray analysis, cFOS, JUN, EGR1, EGR2, EGR3 were found to have 62.5, 2.1, 45.5, 5.9, and 37 fold increases in transcriptional expression, respectively. The upregulation of these genes in HCAEC infected with S. mutans OMZ175 for 5 h was verified using RT-PCR, though the fold changes (in parentheses) were different than those observed with microarray: cFOS (9.1), JUN

(17.1), EGR1 (25.7), EGR2 (8.0), and EGR3 (4.2) (Figure 3-2).

Cytokines

Cytokines are a group of small proteins that include growth factors, interleukins, interferons, colony stimulating factors, and chemokines. Assessing the changes in the expression of these molecules serve a dual purpose 1) to validate that the cytokine

103

expression profile generated from the microarray analysis in this study is consistent with published data that shows that S. mutans invasion stimulate expression in endothelial cells 2) to determine the specific HCAEC chemokine response to S. mutans OMZ175 under in vitro culture conditions.

Changes in the HCAEC cytokine microarray profile were observed at both one and five h post infection with S. mutans OMZ175. However, changes were most significant 5 h post infection compared to the uninfected control (Tables 3-4, 3-5, 3-7). There are no published studies investigating the production of growth factors by endothelial cells in response to S. mutans infection. However, it would be expected that there would be an up-regulation of growth factors if S. mutans induced a “wounding” or hypoxic response

[279-281]. Specifically, vascular endothelial growth factor-A (VEGF-A), VEGF-C, hepatocyte growth factor (HGF), and heparin binding EGF-like growth factor (HBEGF) were found to have 3.2, 2.6, 2.9, 3.6 fold increases in transcriptional expression, respectively. Other growth factors that were impacted to a lesser, but significant, extent include transforming growth factor beta-1 (TGF-beta), TGF-beta-2, platelet derived growth factor (PDGF) A, PDGF-D, insulin-like growth factor, fibroblast growth factor 2

(FGF), FGF-16, FGF-18 (Table 3-4).

In addition to microarray analysis of mRNAs, the relative amount of protein expressed was measured by ELISA assays. To this end, assays were performed to measure VEGF and PDGF in conditioned media and cell lysates collected from HCAEC cultured in RPMI media supplemented with 0.1% FBS and 1% BSA infected with

S. mutans OMZ175. The results at 24 h post infection under continuous antibiotic demonstrated no differences between treatment and control groups (Figure 3-3). These

104

results indicate that any change in growth factor transcriptional induction at early time points does not appear to translate to increased growth factor production under the conditions assayed.

In addition to growth factor expression changes, we observed global changes in the expression of chemokines, cytokines, and some of their receptors using microarray analysis (Table 3-5). Changes were most significant five h post infection compared to the uninfected control. Cytokines, cytokine receptors, and chemokines that had greater than a two-fold increase (indicated in parenthesis) in expression include: CCL2 (2.5),

CCL20 (19.2), CD69 (13.9), CX3CL1 (2.2), CXCL1 (2.9), CXCL2 (24.4), CXCL3 (28.6),

CXCL5 (2.4), CXCL6 (2.1), CXCL8 (10), CXCL12 (2.2), IL6 (6.7), IL1RL1 (2), and IL11

(5.9). None of the factors in this class were shown to have a decreased change in expression greater than two-fold.

ELISAs were also performed to measure the levels of inflammatory cytokines

IL1A, IL1B, IL6, and IL8 in HCAEC conditioned media 5 h post-infection with S. mutans

OMZ175. The 5 h infection group included HCAEC cultured in EBM-2 media complete without antibiotic treatment. The 24 h infection group was treated with continuous antibiotics 5 h post infection. There was no difference in IL1A or IL1B levels when comparing the uninfected control to the S. mutans OMZ175 treated group at both 5 and

24 h post-infection (Figure 3-4 A & B). In contrast, both IL6 and IL8 levels increased significantly in the S. mutans OMZ175 infection group compared to the uninfected control at both 5 and 24 h post-infection (p<0.001) (Figure 3-4 C & D).

Cytoskeletal Elements and Adhesion Molecules

Cytoskeletal elements are important for cell motility, morphology, vesicle formation, proliferation, and survival. Changes in HCAEC cytoskeletal gene expression

105

were observed using microarray analysis at both 1 and 5 h post infection with S. mutans

OMZ175, with the most significant changes occurring at 5 h post infection (Table 3-6).

Those with greater than two fold induction include Rho-Related GTP-Binding Protein

Rho 6 (RND1; 5.6), Rho GTPase-activating protein 5 (ARHGAP5; 2.4), Ras Protein-

Specific Guanine Nucleotide-Releasing Factor 2 (RASGRF2; 2), and Ras-related

Protein Rab-23 (RAB23; 2.3). These proteins are classified as GTPase or GTPase regulatory proteins that appear to be involved in cytoskeleton organization. RND1 and

ARHGAP5 are members of the Rho family, which mediate the regulation of the cytoskeleton pathway. Overexpression of RND1 has been shown to lead to cell rounding in fibroblasts [282]. Furthermore, ARHGAP5 interaction with RND1 is critical to the cellular effects elicited by the expression of RND1 in fibroblasts [283]. They function as antagonists of RhoA GTPase [283], causing the disassembly of actin stress fibers in response to extracellular growth factors. RASGRF2, Ras-specific guanine nucleotide- releasing factor 2, is an activator of Ras and Rac1, both shown to be important for

S. pyogenes invasion of human cells [167, 284]. RAB23 is a member of the Rab

GTPase family and has been demonstrated to be required for the formation of Group A streptococci containing autophagosome-like vacuoles and eventual fusion to lysosomes during S. pyogenes infection [285]. Therefore, it is possible that S. mutans uses mechanisms of entry similar to S. pyogenes, and should be investigated further in future studies.

Endothelial cell adhesion molecules are cell surface proteins important for homing immune cells from the lumen to the interior of the artery. Changes in the HCAEC adhesion molecule expression were observed using microarray analysis at both one

106

and five h post infection with S. mutans OMZ175 with the most significant changes

occurring at five h post infection (Table 3-7). Specifically, intercellular adhesion

molecule (ICAM) -1, ICAM-4, selectin E (SELE), and vascular cell adhesion molecule 1

(VCAM-1) were found to have 3.3, 2.6, 35.7, 10.9 fold increases in transcriptional expression, respectively.

Previously published studies have shown that S. mutans infection of endothelial

cells impact expression of ICAM-1, VCAM-1, and SELE, therefore, we sought to identify other adhesion molecules that may be impacted by S. mutans infection using Human

RT2 profiler™ PCR array analysis. Targeted RT-PCR can be more sensitive than microarray profiling in identifying changes in gene expression. To ensure no additional changes were introduced, RNA samples that were collected and used for microarray analysis were pooled (no infection control and 5 h post infection), and the two sample sets were analyzed using RT-PCR. This data set (Table 3-8) confirmed an increase in the expression of ICAM-1, VCAM-1, SELE, with fold increases of 3.1, 31.0, and 81.8,

respectively. Additionally, this analysis identified matrix metallopeptidase (MMP)-1 and -

10 to be upregulated by 2.6 and 9.5 fold, respectively. Other MMPs that were shown to

be notably downregulated include MMP-2 and MMP-14 by -1.9 and -2.2 fold,

respectively.

Apoptosis

Changes in HCAEC gene expression of the apoptotic pathway were observed

using microarray analysis at both one and five h post infection with S. mutans OMZ175

with most significant changes occurring at five h post infection (Table 3-9). BIRC3, a

member of the apoptotic inhibition pathway mediators, demonstrated a 12.04 increase

in expression. TNFSF10, a member of the tumor necrosis factor superfamily that

107

functions to induce apoptosis, demonstrated a -2.82, -3.43, and -3.57 fold reduction in expression reflected by three microarray probe sets. The remaining genes involved in this pathway showed less than two fold changes including Caspase1, 2, 3, 6, 7, 8, 9, 10.

Caspase proteins are inducers of apoptosis. With the exception of caspase 3, all other caspase proteins were down regulated. Enzymatic assays were performed to validate these observations.

Caspase 3/7 activity was measured using HCAEC lysates collected 5 h post- infection with S. mutans OMZ175 (Figure 3-5), both with and without co-incubation with the broad-spectrum caspase inhibitor, z-VAD-FMK. Caspase activity was normalized to the activity of the uninfected control. In wells that were treated with caspase inhibitor z-

VAD-FMK, caspase activity was significantly reduced. Results show there was a statistically significant increase (p <0.001) in caspase activity in HCAEC infected with

OMZ175 and OMZ175:cnm; however, a 20 % induction in caspase activity is not expected to be biologically relevant. The results also indicate that the observed caspase induction was not stimulated by invasion, as the invasion deficient cnm mutant demonstrates similar levels of caspase activity to the invasive strain. Previous experiments indicated that S. mutans had no intrinsic caspase activity (data not shown), thus contribution to caspase activity by the bacteria was ruled out.

Considering the data generated from the microarray and biochemical assays were not conclusive, both flow cytometry and microscopy were performed to determine the population of apoptotic HCAEC in response to S. mutans infection. For these assays, camptothecin (10 µM) was used as an inducer of apoptosis and hydrogen peroxide (2 mM) was used to trigger necrosis. Flow cytometry was used to measure HCAEC fate

108

during infection with S. mutans OMZ175 over time (1.25, 2.5, and 5 h). Cells were stained with FITC labeled Annexin V and Ethidium homodimer III cell impermeable DNA stain. The results demonstrate that HCAEC live cell populations decreased over time with approximately 66% healthy cells remaining 5 h post-infection (Figure 3-6, 3-7).

Uninfected HCAEC control cells had approximately 84% healthy cells, thus increased

HCAEC death was attributed to bacterial infection. Flow cytometry was also used to measure HCAEC fate during an infection with S. mutans OMZ175 at MOI of 1 and 100

(Figure3-8, 3-9). The cell population distributions were similar between these two groups 4 h post-infection and correlated with the previous flow cytometry assay.

Analysis of population distribution indicated that both apoptosis and necrosis occurred, but at low levels. Camptothecin and hydrogen peroxide, agents selected as the apoptotic and necrotic positive controls, did not perform as expected based on flow cytometry results as a significant proportion of the population formed aggregates and were thus not reflected in the flow cytometry cell counts. Contrary to the flow cytometry cell count data, the cells from the positive control groups used in flow cytometry assays, monitored by phase-contrast microscopy prior to detachment and staining, were observed to have cell morphologies consistent with greater that 50% of the population undergoing apoptosis and necrosis. In retrospect, flow cytometry is designed to assess the state of cell population for suspension cell types. It is not amenable to adherent cell types because they form cell aggregates making it impossible to generate accurate population data. Therefore, microscopy was used to further investigate cell fate.

Consequently, fluorescent microscopic analysis was used to differentiate between apoptosis, necrosis, and healthy HCAEC populations 5 h post-infection with S. mutans.

109

The DNA stain, Hoechst 33342, was used to detect all nuclei; ethidium homodimer III was used to stain necrotic cells; FITC labeled Annexin V was used to stain apoptotic cells (Figure 3-10). Image J software was used to subtract background fluorescence, merge the images, and to count cells in each of the three categories. The average number of cells counted per treatment was 1031 ± 378. Camptothecin and hydrogen peroxide, agents selected as the apoptotic and necrotic positive controls, performed as expected, detecting 47.1% apoptotic cells and 94.8% necrotic cells in the respective treatment group. The results demonstrate that HCAEC live cell populations decrease

5 h post-infection by approximately 10 % (Figure 3-11), and the numbers were independent of S. mutans strain or viability. At 5 h post infection, S. mutans does not appear to induce a cell death response in HCAEC at levels that would suggest biological relevance.

Discussion

The purpose of this study was to investigate the gene expression changes of

HCAEC during attachment and invasion over time by S. mutans OMZ175 in comparison to uninfected HCAEC using microarray analysis. We hypothesized that S. mutans

OMZ175 invasion of HCAEC would result in endothelial dysfunction, a pathological state of the endothelium associated with the increased expression and production of adhesion molecules and inflammatory markers (chemokines and other cytokines). We also expected the up regulation of genes involved in the innate immune response at early time points to heighten over time when infected with S. mutans OMZ175, particularly genes involved in pathogen associated molecular pattern recognition and signaling, autophagy, and apoptosis based on previous studies.

110

The transcriptional profiling data presented in this study provides a global view of the HCAEC response to S. mutans OMZ175 infection. In this analysis, we focused on infection induced expression of cytokines, cytoskeletal elements, adhesion molecules, and factors linked to the apoptotic response. S. mutans is considered the primary etiologic agent of dental caries, affecting 60 to 90% of the population. S. mutans gains access to the bloodstream through procedures such as tooth brushing, tooth extraction, endodontic treatment, and periodontal surgery and is often the causative agent of infectious endocarditis. A variety of data suggest S. mutans may contribute to the pathology of cardiovascular diseases such as atherosclerosis [152, 154, 207]. The presence of S. mutans has also been recently linked to the pathology of hemorrhagic stroke [150]. Previously, S. mutans strains expressing a collagen binding protein (Cnm) have been shown to invade human coronary artery endothelial cells [127]. Serotype f strain S. mutans OMZ175 invades HCAEC in a Cnm, collagen binding protein, dependant manner and has induced an accelerated induction of atherosclerosis in an

ApoE null mouse model of disease [31, 126].

The inflammation response of endothelial cells to oral streptococci including

S. mutans has been the subject of several studies [139, 262, 286-289], and the cytokine profile generated during the study presented here agrees with previously published data

(e.g. CCL2, IL6 and CXCL8). CCL2 plays a crucial role in initiating coronary artery disease by recruiting monocytes/macrophages to the vessel wall, which leads to the formation of atherosclerotic lesions and vulnerability of the plaque [242]. Another important study has reported that macrophages are converted to atherosclerotic foam cells when infected with S. mutans [290]. Other factors, such as CXCL8, have been

111

reported to stimulate neutrophilic respiratory burst, degranulation, and adherence to endothelial luminal surfaces, and activate monocytes, directing their recruitment to atherosclerotic lesions [291]. Glucosyltransferases (excreted enzymes produced by

S. mutans) have been reported to stimulate the production of oxygen reactive species

(hydrogen peroxide) and TNF-α in macrophages [292] and induce the production of IL-6 in T cells [293].

While the role of individual chemokines or cytokines in the development or exacerbation of atherosclerosis is under investigation by many laboratories, epidemiological studies, as well as animal studies, have linked certain chemokines and cytokines to a pro-atherogenic or atheroprotective phenotype. For instance, circulating

CCL20 has been shown to be higher in asymptomatic patients with hypercholesterolemia, a disease that results in the deposition of cholesterol in vascular tissues, and to be overexpressed in atherosclerotic lesions from coronary artery patients

[294]. Furthermore, in a study using CCL20 receptor knockout mice (CCR6(-/-)ApoE (-/-)), it was reported that these mice had 40 % less atherosclerotic lesions and 44 % less macrophage infiltrates when compared to CCR6(+/+)ApoE (-/-) mice [295]. In contrast,

CXCL2 induction has been reported to be atheroprotective and has been shown to remain uninduced in patients with coronary artery atherosclerosis [296, 297]. However,

CXCL2 is a proinflammatory cytokine that is induced during bacterial infections and important for bacterial clearance by neutrophils, leukocytes that contributes to the development of atherosclerosis at various stages [298-300]. CXCL3 is specifically induced in macrophages in response to oxidized low-density lipoprotein of patients with hypercholesterolemia, and like CXCL2, it is reported to be involved in the endothelial

112

recruitment of neutrophils and monocytes [301]. In this study, we report a 19.2, 24.4, and 28.6 fold induction of CCL20, CXCL2, and CXCL3 in HCAEC 5 h post infection

(Table 3-5). Overall, the study reported here found that several chemokines were induced, as would be expected during the early response period after bacterial infection; among them, 10 were induced > 2 fold. An interesting observation was the induction of chemokines involved in recruiting monocytes and immature dendritic cells (e.g., CCL2,

CCL5, CCL8, CCL20, and CXCL12). A recent study has reported that S. mutans stimulated differentiation of monocytes to dendritic cells and has been hypothesized as the possible mechanism by which S. mutans is able to persist in the bloodstream [139].

Such a mechanism could facilitate S. mutans ability to colonize downstream sites such as cardiac lesions.

In addition we found significant induction of expression of growth factors by endothelial cells in response to S. mutans infection that have not been previously reported (p<0.001). For example, growth factors important for endothelial proliferation, migration, and angiogenesis were significantly up-regulated (i.e. VEGF, PDGF, HGF, and HBEGF). VEGF and PDGF protein levels in conditioned media and lysates of

HCAEC cultured in growth factor depleted media after S. mutans infection were assayed using ELISAs. We found no significant changes of VEGF or PDGF levels relative to the control. Unlike the microarray assay, these biochemical assays were performed under continuous antibiotic treatment to measure the endothelial response to intracellular bacteria. It is possible that the continuous antibiotic treatment interfered with growth factor production. For example, under these conditions bacteria which cycle

113

out of the cell are exposed to antibiotics and are unable to grow or secrete metabolic byproducts (e.g., lactic acid).

With respect to S. mutans infection, we hypothesize that growth factor induction may be the result of lactic acid secreted by S. mutans in culture media. Previous studies have shown that lactic acid is a small molecule product of human metabolism, mediated by a hypoxic environment in healthy tissue, and has been demonstrated to have a regulatory role in cell migration and is believed to be important in tissue growth and repair [302-304]. Furthermore, studies have shown that the presence of lactic acid impact tumor metastasis and angiogenesis, in a manner independent of oxygen concentration [305, 306]. Considering that S. mutans primary metabolic byproduct is

L-lactic acid, we hypothesize that S. mutans infection would have significant impact on pre-existing disease pathologies and could contribute to atherosclerosis through this mechanism. Preliminary data generated using a wound scratch model of HCAEC cultured in growth factor depleted media showed increased migration and/or cell proliferation when infected with S. mutans OMZ175 over a 72 h period. The results were only observed in cultures with intermediate pulse antibiotic treatments but not with continuous antibiotic treatment (data not shown). Furthermore, factors that are associated with cytoskeletal modifications would be expected to occur in conjunction with factors that regulate cell proliferation and migration and bacterial entry and trafficking. Those factors that were differentially expressed (Table 3-6) were induced or repressed in response to S. mutans 5 h post infection by a factor < 2 fold, except for

RND1 (5.6 fold; disassembly of actin stress fibers) and ARHGAP5 (2.4 fold; positive regulator of cell migration). Thus, we hypothesize that cell migration, rather than cell

114

proliferation, is induced upon infection with S. mutans. These results are interesting in

that other oral streptococci have also been linked to cardiovascular diseases, thus this

mechanism of streptococci pathogenesis would theoretically be invasion independent.

This previously unstudied phenomenon will be the focus of future studies.

During bacterial infection, the host cell inflammatory response functions to call

immune cells to the site of infection, while the expression of surface adhesion molecules

are up-regulated and function to capture leukocytes and facilitate diapedesis. For

example, upon stimulation with S. mutans OMZ175 purified PAc adhesin protein endothelial cells have been reported to result in increased expression of adhesion molecules and promotion of the transendothelial migration of neutrophils [254] through

specific binding of PAc to α5β1 integrin [140]. This binding event results in a signal

cascade involving phospholipase Cγ (PLCγ), focal adhesion kinase (FAK) and paxillin

phosphorlyation with downstream mitogen activating protein kinase (MAPK) and protein

kinase C (PKC) which in turn causes an increase in production of CXCL8 [189, 307].

The work presented here demonstrated an up-regulation in expression of adhesion

molecules ICAM-1, ICAM-4, VCAM-1, and SELE using both microarray and RT-PCR

analyses. ICAM-1 is constitutively expressed in resting endothelial cells at a low levels

[308, 309]. Once activated by cytokines, endothelial cells increase expression of

ICAM-1 and VCAM-1 [310]. P- and E- selectins are produced by endothelial cells in

acute, as well as, in active atheromatous plaques and serve as rolling molecules for

monocytes, neutrophils, effector T cells, B cells, and natural killer cells [310, 311].

E-selectin is not constitutively expressed; however, it is found on the surface of

activated endothelial cells [312]. P-selectin has been reported to be sequestered in

115

Weibel-Palade bodies (an endothelial cell organelle that contains biologically active

compounds that modulates local and systemic processes like inflammation) that

undergo exocytosis in activated endothelial cells depositing P-selectin to the membrane

[313]. We did not observe an increase in P-selectin expression at the transcriptional

level. Future studies will include measuring VCAM-1, E- and P-selectin protein levels after S. mutans infection to confirm endothelial activation.

Microarray analysis indicated inhibition of apoptotic pathways 1 and 5 h post

infection of HCAEC with S. mutans OMZ175. Subsequent analyses, measuring caspase

3/7 activity and induction of apoptosis and necrosis in HCAEC 5 h post infection with

S. mutans OMZ175, we found an induction of apoptosis; however the observed levels of

induction do not indicate a biologically relevant apoptotic response. Data presented

herein also indicated that a noninvasive strain OMZ175:cnm and nonviable heat killed

OMZ175 induced cell death at 5 h to a similar degree, indicating that invasion was not required for this response. Furthermore, induction of the autophagic pathway was not

detected by microarray analysis, except for a 2.3 fold induction of RAB23 reported to be

involved in autophagic vacuole assembly, suggesting an absence of autophagy driven

apoptosis. Another interesting observation derived from both microarray and RT-PCR

analyses were the induction of JUN, c-FOS and EGR-1. These factors have been shown to be key in induction of Ca+2 regulated growth inhibition and cell death [314]. In

addition to the transient modifications occurring upon entry, intracellular bacteria induce

drastic changes in the pattern of transcription and translation of infected cells, which

may undergo programmed cell death (e.g., apoptosis) or necrosis, taking over the fate

of their host cell with significant implications in the progression of diseases. Microarray

116

and RT-PCR analyses capture a moment in time and do not always reflect the final outcome of cell fate. In future studies, time course studies using microscopy will investigate the role of intracellular S. mutans in HCAEC fate. Unfortunately, methods available to investigate the mechanisms of apoptosis and necrosis are limited as

S. mutans grows robustly in culture media and endothelial cells are an adherent cell type.

In summary, while we observed an induction of cytokines and adhesion molecules which by in large agreed with published reports typical of a streptococci infection, we found changes in gene expression due to S. mutans infection that were unexpected or contrary to our expectations. We expected genes involved in the autophagic pathway to be induced, since S. mutans was observed in double membrane vacuoles of infected

HCAEC using TEM; however, none of these genes were changed 5 h post infection.

Other unexpected results include an anti-apoptotic response and the induction of growth factors post infection as cell death and growth arrest were expected responses to sepsis causing bacterial infections. Specifically, the induction of VEGF and repression of genes involved in apoptosis 5 h post infection with S. mutans is reminiscent of a vascular endothelial response during Bartonella and Chlamydia infections, two successful intracellular pathogens [315, 316]. Interestingly, infections with Bartonella and Chlamydia have been shown to induce host cell proliferation, while we currently hypothesize that S. mutans is inducing a cell migration response.

In conclusion, this study indicates that S. mutans can be an important bacterium in the pathology of human systemic disease that may or may not require invasion.

Although the data collected to date using cell culture models does not suggest that cnm

117

is important for the host cell response and fate of endothelial cells, our data indicates that invasion and bacterial trafficking is affected by cnm (Chapter 2), thus cnm appears to be important to bacterial fate and would be expected to impact bacterial persistence and development of chronic infection. Consequently, in vivo studies may be required to investigate the host response to S. mutans infection with respect to cnm.

118

Table 3-1. List of primers used in this study Gene Primer sequence cFOS_F TAGTTAGTAGCATGTTGAGCCAGG cFOS_R ACCACCTCAACAATGCATGA cJUN_F GGCTGGTGTTTCGGGAGTGT3 cJUN_R CGCCGCCTTCTGGTCTTTAC EGR1_F CTTCAACCCTCAGGCGGACA EGR1_R GGAAAAGCGGCCAGTATAGGT EGR2_F ACGTCGGTGACCATCTTTCCCAAT EGR2_R TGCCCATGTAAGTGAAGGTCTGGT EGR3_F GCTTTGTTCAGTTCGGATCGCCTT EGR3_R AAACAATGAGGTGTTTGGGTCGGG

Table 3-2. Supervised analysis of pairwise comparisons Condition A/Condition B Significant probe sets t-test p<0.001 OMZ175 1 h Uninfected 1507 OMZ175 5 h Uninfected 6600 OMZ175 1 h OMZ175 5 h 950

119

Table 3-3. Ontology analysis of endothelial cell pathways impacted by infection with S. mutans OMZ175 over time Impacted signaling p<0.01* Description pathway** MAPK 89/228 Regulates genes that are important to cell cycle regulation Toll-like receptor 26/62 Stimulation of the innate immune system and inflammation Phosphatidylinositol 30/66 Calcium released to the cytosol and PKC activation Cell cycle 27/58 Cell cycle regulation Wnt 28/70 Regulation of gene transcription, cytoskeleton, and intracellular calcium Adhesion and diapedesis 16/34 Lymphocyte rolling and diapedesis of lymphocytes Adhesion and diapedesis 14/29 Granulocyte rolling and diapedesis of granulocytes Cytokine-cytokine 15/48 Inflammatory response receptor Protein translation 28/51 Genes involved in protein production Apoptosis death pathway 45/86 Extrinsic cell programmed death Apoptosis caspase 29/62 Effectors of apoptosis pathway TNF stress related 30/57 Regulation of apoptotic pathways, NF-kB pathway activation, and stress-activated protein kinases IGF-1 signaling pathway 24/63 AKT signaling pathway activator EGF signaling pathway 33/85 Regulates growth, differentiation, migration, adhesion and cell survival PDGF signaling pathway 36/84 Regulates growth and angiogenesis, potent mitogen of smooth muscle cells VEGF, hypoxia, and 39/79 Regulates physiological and pathological angiogenesis angiogenesis such as tumor growth and ischemic diseases Hypoxia-inducible factor 26/48 Regulates oxygen homeostasis Low density lipid pathway 10/17 Involved in the production of "bad cholesterol" Rac1 cell motility 28/59 Stimulates the formation of filipodia and signaling pathway lamellopodia and regulates cell motility *Number of regulated genes in the pathway/Number of total genes defined for this pathway **BioCarta pathways database (http://www.biocarta.com/genes/index.asp)

120

Table 3-4. Gene expression changes of growth factor receptors and ligands 5 h post infection with S. mutans OMZ175 compared to uninfected control (p< 0.001) Fold change Gene symbol Gene name 2.6 VEGFC Vascular endothelial growth factor C 3.2 VEGFA Vascular endothelial growth factor A 1.2 TGFBI Transforming growth factor, beta-induced, 68kDa 1.4 TGFB2 Transforming growth factor, beta 2 1.5 PDGFA Platelet-derived growth factor alpha polypeptide 1.3 PDGFA Platelet-derived growth factor alpha polypeptide 1.5 PDGFD Platelet derived growth factor D 1.5 PDGFD Platelet derived growth factor D 1.2 IGF1 Insulin-like growth factor 1 (somatomedin C) 1.3 HDGFRP3 Hepatoma-derived growth factor, related protein 3 -1.2 HGS Hepatocyte growth factor-regulated tyrosine kinase substrate 2.9 HGF Hepatocyte growth factor 3.6 HBEGF Heparin-binding EGF-like growth factor 1.3 FGF2 Fibroblast growth factor 2 (basic) 1.4 FGF18 Fibroblast growth factor 18 1.3 FGF16 Fibroblast growth factor 16 -1.6 EGFR Epidermal growth factor receptor

121

Table 3-5. Microarray gene expression changes of chemokines and cytokines 5 h post infection with S. mutans OMZ175 compared to uninfected control (p< 0.001) Fold Gene change symbol Common name and function 1.3 CCL11 Eotaxin-1 selectively recruits eosinophils by inducing their chemotaxis 2.5 CCL2 MCP-1 recruits monocytes, memory T cells, and dendritic cells to the sites of inflammation produced by either tissue injury or infection 19.2 CCL20 Macrophage inflammatory protein-3α chemotactic for dendritic cells 1.6 CCL5 RANTES is chemotactic for T cells, eosinophils, and basophils 1.4 CCL8 MCP-2 is chemotactic for and activates mast cells, eosinophils and basophils, and monocytes, T cells, and NK cells 13.9 CD69 Involved in lymphocyte proliferation and functions as a signal-transmitting receptor in lymphocytes, including natural killer (NK) cells, and platelets 2.2 CX3CL1 Fractalkine in the soluble form recruits T-cells and monocytes; Promotes adhesion of leukocytes through CX3CR1 2.9 CXCL1 Neutrophil-activating protein is a secreted growth factor that binds to CXCR2 and is chemotactic for neutrophils 2.2 CXCL12 Stromal cell-derived factor 1 activates CXCR4 to induce the rapid uptake of intracellular calcium ions and to recruit T- cells and monocytes; Activates beta-arrestin pathway through binding CXCR7 24.4 CXCL2 Macrophage inflammatory protein 2-α regulates the production of VEGF and considered to be atheroprotective; Recruits neutrophils 28.6 CXCL3 GROγ targets neutrophils, fibroblasts, and melanoma cells by CXCR2 2.4 CXCL5 ENA-78 recruits neutrophils and endothelial cells by CXCR2; Produced in response to either IL1 or TNFα 2.1 CXCL6 Granulocyte chemotactic protein 2 targets neutrophils and endothelial cells 10 CXCL8 Neutrophil chemotactic factor or IL-8 induces chemotaxis and phagocytosis in neutrophils but also other granulocytes; Potent promoter of angiogenesis

122

Table 3-5. Continued Fold Gene Description change symbol 1.6 IL1A Induces inflammation, as well as, fever and sepsis

2 IL1RL1 Induced by proinflammatory stimuli, and may be involved in the function of helper T cells 6.7 IL6 Important mediator of fever and of the acute phase response 1.4 IL6ST The receptor systems for IL6, LIF, OSM, CNTF, IL11, CTF1 and BSF3 can utilize gp130 for initiating signal transmission; Binds to IL6/IL6R (alpha chain) complex -1.6 IL7 Induces inflammation via PI3K/AKT-dependent and - independent activation of NF-κB, recruits monocytes/macrophages, and has an active role in atherogenesis 1.5 IL7R Receptor (CD127) for interleukin-7

-1.2 IL10RB Accessory chain essential for the active interleukin 10 receptor complex; Co-expression of this and IL10RA proteins has been shown to be required for IL10-induced signal transduction 5.9 IL11 Key regulator of hematopoiesis, including the stimulation of megakaryocyte maturation 1.4 IL13RA2 Binds IL13 with high affinity; Regulates the effects of both IL-13 and IL-4 1.2 IL13RA1 A subunit of the interleukin 13 receptor that forms a receptor complex with IL4 receptor alpha, a subunit shared by IL13 and IL4 receptors; Bind to tyrosine kinase TYK2, and may be involved in JAK1, STAT3 and STAT6 activation -1.3 IL15RA High-affinity receptor for interleukin-15 which regulates T and natural killer cell activation and proliferation 1.4 IL18R1 Receptor for IL-18 which differentially regulates apoptosis mediated by TNFα and Fas 1.3 IL27RA Receptor for IL27 which is involved in the regulation of Th1- type immune responses and innate defense mechanisms -1.9 IL33 Induces T helper cells, mast cells, eosinophils and basophils to produce type 2 cytokines This table was generated from data collected at the The GeneCards Human Gene Database [317].

123

Table 3-6. Differential expression of genes involved in actin cytoskeleton organization, vesicle transport, cell proliferation, migration, and survival of HCAEC 5 h post infection with S. mutans OMZ175 compared to uninfected control (p<0.001) Fold Gene Change Name Description* 5.6 RND1 Disassembly of actin stress fibers 1.4 RND3 Disassembly of actin stress fibers 1.3 RHOBTB3 Required for endosome to Golgi transport; Specifically binds Rab9, but not other Rab proteins -1.3 ARHGEF18 Colocalizes with actin stress fibers; Positive regulation of apoptosis 2.4 ARHGAP5 Positive regulation of cell migration 1.3 ARHGAP31 Required for cell spreading, polarized lamellipodia formation and cell migration; Activates RAC1 and CDC42 1.4 ARHGAP29 Essential role in blood vessel tubulogenesis; Suppresses RhoA signaling and reduces ROCK and MYH9 activity -1.3 ARHGAP27 May be involved in clathrin-mediated endocytosis/receptor mediated endocytosis -1.6 ARHGAP26 Activates RhoA and CDC42 1.4 ARHGAP24 Filamin-A associated. Filamin-A is involved in ciliogenesis and early to late endosome transport; Suppresses RAC1 and CDC42 activity -1.9 ARHGAP22 Regulates endothelial cell capillary tube formation during angiogenesis 1.3 ARHGAP21 Interacts with GTP-bound ARF1 and ARF6; Organelle transport along microtubules -1.3 ARHGDIA Inhibits cell migration and invasion by indirectly inhibiting RAC1; Negative regulator of apoptosis and cell adhesion -1.3 RRAGB Activation of the TOR signaling cascade by amino acids 1.3 RRAGC Activation of the TOR signaling cascade by amino acids 1.9 RASL10A Potent inhibitor of cellular proliferation 1.3 RIT1 Activation of both EPHB2 and MAPK14 signaling pathways 1.7 RASD1 Involved in nitric oxide signaling 2 RASGRF2 Positive regulation of apoptosis -1.4 RASAL2 Inhibitory regulator of the Ras-cyclic AMP pathway -1.5 RASA3 Inhibitory regulator of the Ras-cyclic AMP pathway 1.3 RASA2 Inhibitory regulator of the Ras-cyclic AMP pathway -1.2 RASIP1 Required for the proper formation of vascular structures

124

Table 3-6. Continued Fold Gene Change Name Description* 1.2 RAB11FIP1 Endosomal recycling -1.3 RAB11FIP5 Transport from apical endosomes to apical plasma membrane 1.4 RAB14 Endocytic recycling 1.2 RAB18 Plays a role in apical endocytosis/recycling 1.2 RAB21 Regulates integrin internalization and recycling 2.3 RAB23 Involved in autophagic vacuole assembly and defense against pathogens 1.3 RAB2B Positive regulation of exocytosis 1.3 RAB31 Involved in the acidification of phagosomes containing pathogens -1.3 RAB31P May activate RAB8A and RAB8B -1.8 RAB33B Acts as a modulator of autophagosome formation -1.2 RAB35 Essential rate-limiting regulator of a recycling pathway to the plasma membrane 1.3 RAB38 Involved in the acidification of phagosomes containing pathogens -1.3 RAB3D Probably involved in regulated exocytosis 1.2 RAB40B Involved in substrate recognition and ubitquitination 1.3 RAB5A Required for the fusion of plasma membranes and early endosomes and involved in the regulation of filopodia extension 1.3 RAB7A Regulator in endo-lysosomal trafficking -1.4 RAB8B May be involved in vesicular trafficking -1.2 RABEP2 Role in membrane trafficking and early endosome fusion 1.2 RABGGTB Rab geranylgeranyltransferase activity -1.2 RAP2B May play a role in cytoskeletal rearrangements and regulate cell spreading 1.2 RAPGEF1 PDGF receptor signaling pathway in cell-cell adhesion -1.3 RAPGEF3 Positive regulation of angiogenesis 1.6 RAPGEF4 Involved in cAMP-dependent, PKA-independent exocytosis through interaction with RIMS2 1.8 RAPH1 Colocalizes at the tips of lamellipodia and filipodia and may colocalize with pathogens 1.6 RASGRP3 MAPK cascade

125

Table 3-6. Continued Fold Gene Change Name Description* 1.3 RASSF1 Required for death receptor-dependent apoptosis and induced in response to DNA damage -1.3 RASSF2 May promote apoptosis and cell cycle arrest 1.3 RHEB Activates the protein kinase activity of mTORC1, a protein that regulates cell growth and survival 1.3 RHOB Up-regulated by DNA damaging agents -1.2 RHOD Involved in endosome dynamics -1.2 RHOJ Regulation of cell morphology -1.3 RIN2 Downstream effector for RAB5B in endocytic pathway * This table was generated from data collected at UniProt Knowledgebase [310, 318].

126

Table 3-7. Microarray gene expression changes of leukocyte adhesion molecules 5 h post infection with S. mutans OMZ175 compared to uninfected control Gene Fold Description symbol Change ICAM4 2.6 Intercellular adhesion molecule 4 ICAM3 -1.3 intercellular adhesion molecule 3 ICAM2 -1.3 Intercellular adhesion molecule 2 ICAM1 3.3 Intercellular adhesion molecule 1 SELPLG 1.2 Selectin P ligand SELE 35.7 Selectin E VCAM1 10.9 Vascular cell adhesion molecule 1

Table 3-8. Human RT2 Profiler™ PCR array of adhesion molecule gene expression using pooled RNA previously isolated for microarray analysis 5 h post infection with S. mutans OMZ175 compared to uninfected control Gene Fold Description symbol regulation COL8A1 -2.5 Collagen, type VIII, alpha 1 ICAM1 3.1 Intercellular adhesion molecule 1 ITGA3 -2.1 Integrin, alpha 3 (CD49C, subunit of VLA-3 receptor) ITGB5 -1.8 Integrin, beta 5 MMP1 2.6 Matrix metallopeptidase 1 (interstitial collagenase) MMP10 9.5 Matrix metallopeptidase 10 (stromelysin 2) MMP14 -2.2 Matrix metallopeptidase 14 (membrane-inserted) MMP2 -1.9 Matrix metallopeptidase 2 (type IV collagenase) SELE 81.8 Selectin E SPG7 -3.1 Spastic paraplegia 7 THBS3 -1.9 Thrombospondin 3 VCAM1 31.0 Vascular cell adhesion molecule 1

127

Table 3-9. Microarray gene expression changes of genes involved in apoptotic pathways 5 h post infection with S. mutans OMZ175 compared to uninfected control. Fold Gene Name Description Change 1.24 APAF1 Apoptotic peptidase activating factor 1 1.29 BCL2 B-cell CLL/lymphoma 2 -1.22 BID BH3 interacting domain death agonist; Cleaved by Caspase 8 to yield t-Bid 12.04 BIRC3 Baculoviral IAP repeat-containing 3 -1.26 CASP10 Caspase 10, apoptosis-related cysteine peptidase 1.14 CASP3 Caspase 3, apoptosis-related cysteine peptidase -1.29 CASP6 Caspase 6, apoptosis-related cysteine peptidase -1.18 CASP7 Caspase 7, apoptosis-related cysteine peptidase -1.27 CASP8 Caspase 8, apoptosis-related cysteine peptidase -1.37 CASP9 Caspase 9, apoptosis-related cysteine peptidase -2.2 CCAR1 Cell division cycle and apoptosis regulator 1 1.39 CFLAR FLIP; Blocks activation of caspase 8 1.18 CHUK Conserved helix-loop-helix ubiquitous kinase -1.3 CIAPIN1 Cytokine induced apoptosis inhibitor 1; Dependent on growth factor stimulation -1.28 DFFB DNA fragmentation factor, 40kDa, beta polypeptide (caspase-activated DNase) -1.17 FADD Fas (TNFRSF6)-associated via death domain 1.28 LMNA Lamin A/C -1.06 MAP3K14 Mitogen-activated protein kinase kinase kinase 14 1.60 NFKB1 Nuclear factor of kappa light polypeptide gene enhancer in B-cells 1 -1.2 PAWR Pro-apoptotic protein by Fas pathway; downregulates BCL2 by ts interactions with WT1 -1.19 RIPK1 Receptor (TNFRSF)-interacting serine-threonine kinase 1 -1.26 SPTAN1 Spectrin, alpha, non-erythrocytic 1 (alpha-fodrin) -2.82 TNFSF10 Tumor necrosis factor (ligand) superfamily, member 10 -3.43 TNFSF10 Tumor necrosis factor (ligand) superfamily, member 10 -3.57 TNFSF10 Tumor necrosis factor (ligand) superfamily, member 10 1.09 XIAP X-linked inhibitor of apoptosis; Blocks activation of caspases 3 and 9

128

Figure 3-1. Hierarchical clustering of variance-normalized gene expression data. The expression pattern of the cRNA generated for each condition tested by microarray analysis was determined by a supervised analysis of variance- normalized data set of differentially expressed genes (FDR; (p< 0.05). In this heat map, each column represents an individual gene element on the array, and each row represents the gene expression state for each of the conditions tested. Each expression data point represents the relative fluorescence intensity of the cRNA from S. mutans OMZ175 infected HCAEC at 1 and 5 h to the fluorescence intensity of the cRNA of uninfected HCAEC. The distance matrix used to measure the relatedness of samples through gene expression space was 1 – Pearson’s correlation coefficient. The cluster is subdivided into two groups. Repressed genes are blue, and induced genes are red. Genes that did not change are white. The variation in the gene expression for a given gene is expressed as the distance from the mean observation for that gene according to the color scale below the heat map. Gene annotation enrichment analysis determined that 135 gene clusters were significantly impacted (p<0.001) and that 28 out of 135 annotated gene clusters were relevant to endothelial activation/dysfunction (eg. anti-apoptosis, apoptosis, immune response, and cell proliferation).

129

Figure 3-2. RT-PCR validation of microarray results. Single strand cDNA was generated from RNA samples using iScript™ cDNA synthesis kit (Bio-Rad) as per manufacturer instructions. The RT-PCR reactions were prepared using IQ SYBR Green RT-PCR kit (Bio-Rad) as per manufacturer instructions with custom primer pairs to transcription factors that were observed to be up- regulated in the microarray. RT-PCR data was analyzed by the comparative Ct method.

130

A)

B)

Figure 3-3. Enzyme linked immunosorbant assays to measure the levels of VEGF A) and PDGF B) in HCAEC lysates and conditioned media 24 h post infection with S. mutans. Infection was performed in RPMI media supplemented with 0.1% FBS and 1% BSA with continuous antibiotic treatment. There is no statistical difference in the presence of growth factor (VEGF or PDGF) in the lysates or supernatants of uninfected compared to HCAEC 24 h post infection. The error bars indicate standard error (n=6). Student’s t-test statistics to compare VEGF or PDGF mean levels detected in lysates and supernatants (conditioned media) between treatments and the uninfected control.

131

Figure 3-4. Enzyme linked immunosorbant assays to measure the levels of A) IL1A, B) IL1B; C) IL6; and D) IL8 in culture conditioned media 5 and 24 h post infection with S. mutans. Infection was performed in EBM2 media complete: 5 h time point without antibiotic treatment; 24 h time point without antibiotic for 5 h and 19 h with continuous antibiotic treatment. The error bars indicate standard deviation (n=4). Student’s t-test statistics to compare means between treatments at a certain time. (***, p<0.001; ns, not significant)

132

Figure 3-5. Caspase 3/7 activity to determine activation of the apoptotic pathway presented as a percent of the uninfected HCAEC control. HCAEC extracts were collected 5 h post infection and analyzed for caspase 3/7 activity using a colorimetric caspase. Caspase 3/7 protease activity was induced above basal levels independent of invasion, suggesting that innate surveillance by toll-like receptors may be activated in response to pathogen associated molecular patterns, but this result may not be biologically relevant in vivo. CI = broad- spectrum caspase inhibitor z-VAD-FMK 100µM

133

Figure 3-6. Flow cytometric analysis of HCAEC fate during infection with S. mutans OMZ175 over time. These are representative dot plots with FITC labeled Annexin V (FITC-A) versus Ethidium homodimer III cell impermeable DNA stain (PI-A). The top 3 dot plots were of the control treatment groups. Camptothecin is an alkaloid which inhibits the DNA enzyme topoisomerase I and induces apoptosis, while hydrogen peroxide induces necrosis. (n=3; 3839 ± 286 cells counted per condition) Upper left quadrant of the dot plot indicates percentage of necrotic cells. Upper right quadrant of the dot plot indicates percentage of dead cells. Lower left quadrant of the dot plot indicates percentage of live cells. Lower right quadrant of the dot plot indicates percentage of apoptotic cells.

134

Figure 3-7. Percentage of HCAEC among the four fate groups as determined by flow cytometry after infection with S. mutans OMZ175 over time. Stacked column chart shows the percent population within the four fate groups. Necrotic column indicates HCAEC treated with 2 mM hydrogen peroxide (control). Apoptotic column indicates HCAEC treated with Camptothecin 10 uM (control). (n=3)

135

Figure 3-8. Flow cytometric analysis of HCAEC fate after infection with S. mutans OMZ175 at MOI = 1 and MOI = 100. These are representative dot plots with FITC labeled Annexin V (FITC-A) versus Ethidium homodimer III cell impermeable DNA stain (PI-A). The top 3 dot plots were of the control treatment groups. Camptothecin is an alkaloid which inhibits the DNA enzyme topoisomerase I and induces apoptosis, while hydrogen peroxide induces necrosis. Upper left quadrant of the dot plot indicates percentage of necrotic cells. Upper right quadrant of the dot plot indicates percentage of dead cells. Lower left quadrant of the dot plot indicates percentage of live cells. Lower right quadrant of the dot plot indicates percentage of apoptotic cells. (n=1; 5621 ± 224 cells counted per condition)

136

Figure 3-9. Percentage of HCAEC among the four fate groups as determined by flow cytometry after infection with S. mutans OMZ175 at MOI = 1 and MOI = 100. Stacked column chart shows the percent population within the four fate groups. Necrotic column indicates HCAEC treated with 8 mM hydrogen peroxide (control). Apoptotic column indicates HCAEC treated with Camptothecin 10 uM (control). (n=1)

137

Figure 3-10. Microscopic analysis of HCAEC to differentiate between apoptosis, necrosis, and healthy cells 5 h post infection with S. mutans OMZ175. Panels A-D are micrographs of HCAEC treated with apoptosis inducing agent camptothecin (10 µM). Panels E-H are micrographs of uninfected HCAEC. Panels I-L are micrographs of HCAEC infected with S. mutans OMZ175 for 5 h. Panels A, E, and I micrographs were obtained with a 350/461nm filter set to detect all nuclei with DNA stain Hoechst 33342. Panels B, F, J micrographs were obtained with a 528/617nm filter set to detect necrotic cells stained with cell impermeable DNA intercalating dye ethidium homodimer III. Panels C, G, and K micrographs were obtained with a 492/514nm filter set to detect apoptotic cells stained with FITC labeled Annexin V. Panels D, H, and L are composite micrographs generated using Image J software. In the composite micrographs, blue nuclei are “healthy”, red or purple nuclei are necrotic, and green cells are apoptotic.

138

Figure 3-11. Percentage of HCAEC among the fate groups as determined by microscopy after 5 h infection with S. mutans. Stacked column chart shows the percent population within the fate groups. Necrotic column indicates HCAEC treated with 2 mM hydrogen peroxide. Apoptotic column indicates HCAEC treated with Camptothecin 10 µM. Dead OMZ175 group indicates that the bacteria were heat killed prior to infection. Results suggest that apoptosis is induced in a subset of infected cells independent of invasion.

139

CHAPTER 4 DISCUSSION AND FUTURE DIRECTIONS

General Summary and Discussion

This dissertation work challenges the current paradigm that S. mutans is a noninvasive pathogen, restricted to dental caries and infectious endocarditis. Invasive strains of S. mutans expressing the collagen binding protein, Cnm, have been isolated from patients with infectious endocarditis shown to induce hemorrhagic stroke and promote atherogenesis in mice [31, 150]. Non-invasive strains of S. mutans are the most common strains in the oral cavity and are the best studied oral organisms whereas relatively little is known about the invasive strains.

Route from Oral Cavity to Cardiovascular Tissues

In order to investigate S. mutans ability to transition from an oral site of infection to a cardiovascular tissue infection, antibiotic protection assays and transmission electron microscopy were used to determine the tropism of S. mutans to gingival and arterial tissues and to determine the role of Cnm in attachment and invasion of these cell types.

These studies are the first to show that S. mutans invades oral keratinocytes, gingival fibroblasts, and coronary artery smooth muscle cells in a cnm dependent manner with similar efficacy to its ability to attach to and invade HCAEC. The ability of cnm- expressing strains of S. mutans to invade cells associated with the gingiva may provide them an environment for a persistent infection as well as increased access to the bloodstream. This ability to invade oral keratinocytes provides an environmental niche that protects these strains from host defenses (oral healthcare, saliva, antibodies, defensins, bacteriocins, etc.). Thus, invasive strains of S. mutans strains would have a selective advantage by having the ability to use oral epithelium as a reservoir allowing

140

this opportunistic pathogen access to deeper tissues in the gingiva and the circulatory system particularly during incidences of gingival inflammation or injury. The reservoir model hypothesis (Figure 2-14) is supported by epidemiological studies that S. mutans can colonize the oral cavities of children before tooth eruption [228, 229, 319]. Once

S. mutans enters the circulatory system, strains capable of invading cardiovascular tissues would have a selective advantage by being removed from innate immunity factors (e.g., complement and antibodies) by entering the vascular endothelium. In such a situation this opportunistic pathogen may lead to the development of infectious endocarditis and could contribute to arterial inflammation and exacerbate cardiovascular disease development. The hypothesis that S. mutans contributes to other vascular disease besides endocarditis is supported by clinical data that suggest that specific strains of S. mutans may play a role in infections of cardiovascular tissues and that individuals harboring these strains may be at a higher risk for cardiovascular involved diseases [320]. S. mutans has been reported to be the predominant bacterial species isolated from diseased heart valve tissues and in atheromatous plaques [152, 153].

Furthermore, systemic infection with S. mutans strains expressing Cnm increased the risk of cerebral hemorrhagic stroke [150] and accelerated atherosclerosis in mice[31], where the endothelial cells lining blood vessels were damaged leading to exposure of the underlying extracellular matrix. Studies investigating the role of other collagen binding S. mutans adhesins (e.g. PAc, WapA) have shown that these molecules do not contribute to the invasive phenotype in a manner similar to Cnm [126]. Others have found that a S. mutans serotype c strain (GS-5), which we have subsequently

141

established as a noninvasive cnm negative strain, did not promote atherosclerosis in an

ApoE null mouse model [321].

Mechanisms of Dissemination

In this study, we investigated mechanism of dissemination using multiple derivations of the standard antibiotic protection assay (e.g., intracellular replication, intracellular growth curves, cell to cell transfer, exiting and re-infection) in conjunction with live cell counts, RT-PCR, and fluorescent microscopy. This work did not find evidence of S. mutans intracellular replication within primary endothelial cells; however, intracellular S. mutans were found to exit host endothelial cells, and they remained invasive. This data provides support that S. mutans has the potential to traverse the endothelium and to enter cells in the intima, such as smooth muscle cells. The ability of cnm-expressing strains of S. mutans to invade coronary artery smooth muscle cells may further support a role for S. mutans infection/invasion in atherosclerosis and provide information as to a possible mechanism.

Endothelial Response to S. mutans Infection

This work investigated the gene pattern changes of primary endothelial cells when infected with an invasive strain compared to a cognate mutant strain using microarrays.

The transcriptional profiles generated indicated that genes involved in cytoskeletal regulation and rearrangements were impacted in a Cnm-dependent manner. The specific mechanisms of S. mutans entry into endothelial cells (i.e. caveolin-mediated and/or clathrin-mediated endocytosis) is the topic of an NIH R21 awarded to the

Progulske-Fox laboratory, and these microarray results will be used to support that project. Furthermore, this work investigated the changes in the endothelial transcription response to S. mutans infection over time. It was known through previous studies that

142

S. mutans invasion of endothelial cells elicited changes in adhesion molecule expression, cytokine production, and outside-in signaling through binding to α5β1 integrins. However, this work is the first to report a global view of the impact of

S. mutans on endothelial cells.

We found an increase in the expression of genes whose enhanced expression leads to dysregulated tissue repair (e.g., transforming growth factor β2, TGFβ2) [322], genes important for the development of atherosclerotic lesions (e.g., low density lipoprotein receptor, LDLR [323, 324], and modified LDL scavenger receptor CD36

[325]), as well as genes involved in initiating morphological changes characteristic of atherosclerosis (e.g., platelet derived growth factor, PDGF [326]). Overall, the dysregulation of these genes are linked to net-positive growth stimulation and development of chronic inflammation indicative of atherosclerosis progression/ exacerbation [327]. Therefore, we hypothesize that endothelial injury and subsequent inflammation as a result of S. mutans invasion leads to endothelial activation and eventual dysfunction, the initiating event in cardiovascular disease. Of interest was the up-regulation of growth factors including VEGF and PDGF. These growth factors have been shown to be differentially expressed in the presence of lactate. This finding may open a new avenue of research focusing on the effect of bacterial lactic acid production on host cells of the cardiovascular system. VEGF is also an important factor in angiogenesis. Interestingly, the induction of VEGF and repression of genes involved in apoptosis in S. mutans infected HCAEC has also been reported for Bartonella and

Chlamydia vascular endothelial infections, and it is hypothesized that angiogenesis, cell proliferation, and repression of apoptosis are key components of their virulence.

143

Future Work

Introduction

S. mutans has long been recognized as a major pathogen of dental caries, the most common infectious disease in humans [38, 86, 87]. What is much less appreciated is its role in extra-oral diseases [88-97]. S. mutans is an opportunistic pathogen frequently identified in human bacteremias and is an etiologic agent of infectious endocarditis [207]. Infectious endocarditis, a valvular vegetation of the endothelial layer, is a serious cardiovascular disease with often fatal outcomes directly related to dental procedures [105, 133]. It is estimated that oral streptococci are responsible for 35 –

45% of infectious endocarditis cases each year in the U.S.A. and the incidence is increasing [148]. There is now also strong evidence that S. mutans is involved in hemorrhagic stroke, which occurs when a blood vessel ruptures in the brain [150].

Hemorrhagic stroke accounts for 10% of all strokes and has a 40% fatality rate, resulting in 24,000 deaths per year in the USA [149]. In addition, epidemiological and

DNA sequencing evidence supports a contributing role of S. mutans to atherosclerosis

[152, 153]. Atherosclerosis is a complex inflammatory disease afflicting medium and large sized arteries and is the leading cause of death in the USA [328]. The commonality of these diseases is that they are diseases of the cardiovascular system.

Intracellular pathogens have evolved multiple pathways and mechanisms to enter, traffic and survive within eukaryotic cells [329]. The process of internalization, coincident or subsequent to adherence is critical to the cell signaling and trafficking within host cells. Thus the mode of entry plays a major role in determining bacterial fate. There are no reports about the cellular events that govern S. mutans entry into cells. It is likely that strains of S. mutans expressing Cnm, such as OMZ175, are able to adhere to, enter,

144

and persist in HCAECs and that the mechanism of entry is critical to the trafficking and survival of S. mutans intracellularly. It is also likely that invasive S. mutans enters endothelial cells using specific mechanisms and that Cnm plays a central role in this entry. In addition, cnm has now been found in strains of S. sanguinis (Abranches, personal communication), S. suis and S. rattus. Therefore future studies investigating the role of cnm in endothelial cell invasion will have significance beyond S. mutans as well as cardiovascular disease pathology.

Entry and Intracellular Trafficking

Two important pathways for bacterial internalization into eukaryotic cells are the caveolae/lipid raft-mediated endocytosis pathway and the receptor-mediated (clathrin- dependent) endocytosis pathway. Entry via caveolae and/or lipid rafts has been reported for several Gram-positive bacteria including streptococci. Scanning electron microscopy (SEM) can be used to determine if S. mutans enters through a trigger mechanism (membrane-ruffling pattern), a zipper-like mechanism, both mechanisms of receptor mediated entry, and/or through caveolae entry in the host cell membrane.

Furthermore, protein markers (e.g. transferrin receptor), specific inhibitors, and gene- knockdowns (e.g. caveolin-1, clathrin) can be used to better define the entry process.

Methodology has recently been developed to isolate specific classes of endosomes using antibody conjugated magnetic nanoparticles [330, 331]. Isolation and characterization of S. mutans containing endosomes using magnetic nanoparticle methodology and Multidimensional Protein Identification Technology (MudPIT) could be used by modifying this methodology to isolate endocytic vesicles containing S. mutans

OMZ175. This analysis would provide new data concerning protein markers in the

S. mutans containing vesicles. For example, vesicles isolated as part of caveolae-

145

mediated entry would contain caveolin 1, while vesicles isolated as part of clathrin driven receptor-mediated entry would contain clathrin and the cargo receptor. In the event of the later situation, a host receptor that binds to S. mutans may be identified. It is possible that both modes of entry are used by S. mutans as described for

S. suis [175].

Role of Cnm in S. mutans Entry

The S. mutans surface protein adhesin, PAc, has been reported to bind to host proteins i.e. fibronectin and collagen [186]. Purified PAc protein has been shown to elicit a cytokine response and induced expression of leukocyte homing receptors (I-CAM, V-

CAM, and E-selectin) in endothelial cells [253, 254]. PAc has also been shown to be an important molecule in host cell adherence as it directly binds to the alpha 5 beta 1 integrin [140, 189]. However, studies investigating the role of S. mutans adhesion surface molecules such as PAc have not shown that these molecules contribute to the invasive phenotype in a manner similar to Cnm. Given that Cnm is required for invasion of endothelial cells [126], studies designed to elucidate the role of Cnm in invasion could be completed using purified Cnm coupled to latex beads to determine if Cnm is sufficient for entry. GAS invasins including a fibronectin binding protein have been functionally expressed in other bacteria [167]. Similarly, full length cnm could be expressed in Lactococcus lactis as previously described[167]. If Cnm is sufficient for entry of S. mutans OMZ175, then the heterologous expressing strains should behave as strain OMZ175. If Cnm is not sufficient, or induces a different kind of entry than the whole bacterium, as reported for a GAS protein [233], then there will either be minimal intracellular bacteria (not expected) or the bacteria may enter by a different mechanism.

146

It is expected that the coated beads and heterologous expression strains would

provide similar results. However if the heterologous strain results are consistent with

results from OMZ175 but the coated bead results are different, such as no

internalization, then an additional factor supplied by the live bacteria is likely required. It

may be that the coated beads as well as the heterologous strains will attach to the cells

but not be internalized, in which case it would concluded that Cnm is not sufficient for

entry. Whatever the outcome, these experiments would obtain additional understanding

of a group of important but minimally studied S. mutans strains.

Summary

The future work proposed would begin to fill a critical gap in our knowledge of

S. mutans pathogenesis. Follow up studies would include the transcellular passage of

S. mutans through the endothelium to deeper tissues and cell responses to invasion by

S. mutans. Data presented in this dissertation and that of others indicate that invasive strains of S. mutans also invade oral cells in vitro; furthermore, S. mutans can be

detected in predentate children, supporting this claim. Therefore these studies may

have significance beyond cardiovascular disease and endothelial cells. Finally, these

future studies would contribute information concerning the possible future application of

cnm as a biomarker for screening patients at risk for S. mutans non-oral infections.

147

APPENDIX MICROARRAY EXPRESSION DATA AND KEGG PATHWAYS

Table A-1. Microarray gene expression changes of genes with fold changes greater than 2 fold 5 h post infection with S. mutans OMZ175 compared to uninfected control. Parametric Fold- p-value Control * OMZ175 6h* change Gene symbol Gene name < 1e-07 1182.4 5556.65 4.8 ADAMTS1 ADAM metallopeptidase with thrombospondin type 1 motif, 1 < 1e-07 1470.92 4513.4 3 AKAP12 A kinase (PRKA) anchor protein 12 < 1e-07 85.18 225.19 2.6 ALCAM activated leukocyte cell adhesion molecule < 1e-07 2048 4451.27 2.2 ANGPT2 angiopoietin 2 < 1e-07 299.73 1110.89 3.7 ANKRD55 ankyrin repeat domain 55 1.50E-06 300.25 688.59 2.3 ANP32E acidic (leucine-rich) nuclear phosphoprotein 32 family, member E < 1e-07 73.39 604.67 8.3 AREG amphiregulin < 1e-07 58.89 143.26 2.4 ARHGAP5 Rho GTPase activating protein 5 < 1e-07 434.29 1034.7 2.4 ARID5B AT rich interactive domain 5B (MRF1-like) < 1e-07 135.06 5104.31 38.5 ATF3 activating transcription factor 3 < 1e-07 128.89 590.18 4.5 ATOH8 atonal homolog 8 (Drosophila) < 1e-07 2094.66 4576.41 2.2 ATP13A3 ATPase type 13A3 < 1e-07 776.05 1833.01 2.4 ATP2B1 ATPase, Ca++ transporting, plasma membrane 1 < 1e-07 54.66 113.38 2.1 BCL2A1 BCL2-related protein A1 < 1e-07 1637.75 3281.18 2 BCL6 B-cell CLL/lymphoma 6 < 1e-07 1649.14 4999.26 3 BHLHE40 basic helix-loop-helix family, member e40 < 1e-07 81.29 978.89 12 BIRC3 baculoviral IAP repeat-containing 3 < 1e-07 496.28 2642.16 5.3 BMP2 bone morphogenetic protein 2 1.00E-07 694.58 1598.5 2.3 CALCRL calcitonin receptor-like < 1e-07 142.52 324.6 2.3 CAMTA1 calmodulin binding transcription activator 1 < 1e-07 118.19 346.69 2.9 CBLN2 cerebellin 2 precursor < 1e-07 236.39 699.41 2.9 CCDC68 coiled-coil domain containing 68 < 1e-07 6144.16 15393.14 2.5 CCL2 chemokine (C-C motif) ligand 2 < 1e-07 124.72 2410.31 19.2 CCL20 chemokine (C-C motif) ligand 20

148

Table A-1. Continued. Parametric Fold- p-value Control * OMZ175 6h* change Gene symbol Gene name < 1e-07 1746.2 4262.55 2.4 CD274 CD274 molecule < 1e-07 22.55 311.91 13.9 CD69 CD69 molecule 1.00E-07 66.49 147.54 2.2 CDC14A CDC14 cell division cycle 14 homolog A (S. cerevisiae) 3.82E-05 195.36 380.04 2 CDC23 cell division cycle 23 homolog (S. cerevisiae) < 1e-07 688.59 1420.81 2.1 CDK6 cyclin-dependent kinase 6 < 1e-07 335.46 849.22 2.5 CEBPD CCAAT/enhancer binding protein (C/EBP), delta < 1e-07 189.03 507.58 2.7 CHSY3 chondroitin sulfate synthase 3 < 1e-07 306.55 916.51 3 CLDN14 claudin 14 3.10E-06 207.22 408.02 2 CLK4 CDC-like kinase 4 < 1e-07 282.58 1029.34 3.7 CNKSR3 CNKSR family member 3 < 1e-07 331.99 725.33 2.2 CREB5 cAMP responsive element binding protein 5 < 1e-07 117.17 512 4.3 CSF2 colony stimulating factor 2 (granulocyte-macrophage) < 1e-07 255.11 657.11 2.6 CSRNP1 cysteine-serine-rich nuclear protein 1 < 1e-07 158.41 350.31 2.2 CX3CL1 chemokine (C-X3-C motif) ligand 1 chemokine (C-X-C motif) ligand 1 (melanoma growth stimulating < 1e-07 3875.05 11405.96 2.9 CXCL1 activity, alpha) 1.70E-06 247.28 543.07 2.2 CXCL12 chemokine (C-X-C motif) ligand 12 < 1e-07 354.59 8569.53 24.4 CXCL2 chemokine (C-X-C motif) ligand 2 < 1e-07 154.88 4474.47 28.6 CXCL3 chemokine (C-X-C motif) ligand 3 < 1e-07 38.59 93.7 2.4 CXCL5 chemokine (C-X-C motif) ligand 5 chemokine (C-X-C motif) ligand 6 (granulocyte chemotactic protein 3.80E-06 1878.02 3942.79 2.1 CXCL6 2) < 1e-07 471.95 1470.92 3.1 CXCR7 chemokine (C-X-C motif) receptor 7 3.40E-06 186.43 420.95 2.3 DIRAS3 DIRAS family, GTP-binding RAS-like 3 < 1e-07 18.47 94.68 5 DKK2 dickkopf homolog 2 (Xenopus laevis) < 1e-07 328.56 700.63 2.1 DOK5 docking protein 5 < 1e-07 3432.4 13331.02 3.8 DUSP1 dual specificity phosphatase 1 < 1e-07 118.81 325.16 2.7 DUSP10 dual specificity phosphatase 10 < 1e-07 428.31 1807.78 4.2 DUSP5 dual specificity phosphatase 5

149

Table A-1. Continued. Parametric Fold- p-value Control * OMZ175 6h* change Gene symbol Gene name < 1e-07 193.01 376.76 2 DUSP8 dual specificity phosphatase 8 < 1e-07 955.43 2252.79 2.4 E2F7 E2F transcription factor 7 2.30E-06 60.03 146.02 2.4 EGOT eosinophil granule ontogeny transcript (non-protein coding) < 1e-07 132.51 5965.8 45.5 EGR1 early growth response 1 < 1e-07 81.15 471.14 5.9 EGR2 early growth response 2 < 1e-07 23.79 874.61 37 EGR3 early growth response 3 < 1e-07 73.9 145.76 2 EGR4 early growth response 4 4.00E-07 73.01 145.51 2 EIF4E eukaryotic translation initiation factor 4E < 1e-07 1293.89 2911.41 2.3 ELL2 elongation factor, RNA polymerase II, 2 < 1e-07 1680.88 5451.74 3.2 ERRFI1 ERBB receptor feedback inhibitor 1 < 1e-07 88.8 882.22 10 F3 coagulation factor III (thromboplastin, tissue factor) < 1e-07 182.28 365.82 2 FBXO32 F-box protein 32 3.00E-07 123.43 400.32 3.2 FGFR3 fibroblast growth factor receptor 3 fms-related tyrosine kinase 1 (vascular endothelial growth < 1e-07 1930.82 5792.62 3 FLT1 factor/vascular permeability factor receptor) < 1e-07 43.56 2787.97 62.5 FOS FBJ murine osteosarcoma viral oncogene homolog < 1e-07 121.31 2418.67 20 FOSB FBJ murine osteosarcoma viral oncogene homolog B < 1e-07 1612.41 3321.23 2 FOSL2 FOS-like antigen 2 < 1e-07 67.53 199.12 2.9 FREM3 FRAS1 related extracellular matrix 3 < 1e-07 367.09 2288.2 6.3 FST follistatin < 1e-07 67.53 1243.34 18.5 GEM GTP binding protein overexpressed in skeletal muscle < 1e-07 68.83 492 7.1 GPR137C G protein-coupled receptor 137C < 1e-07 22.94 142.77 6.3 GREM1 gremlin 1 < 1e-07 1573.76 3492.39 2.2 GULP1 GULP, engulfment adaptor PTB domain containing 1 < 1e-07 259.12 545.9 2.1 HABP4 hyaluronan binding protein 4 < 1e-07 1823.51 6080.61 3.3 HBEGF heparin-binding EGF-like growth factor < 1e-07 160.62 1049.15 6.7 HDAC9 histone deacetylase 9 3.72E-05 238.44 614.17 2.6 HEY1 hairy/enhancer-of-split related with YRPW motif 1 < 1e-07 25.77 75.98 2.9 HGF hepatocyte growth factor (hepapoietin A; scatter factor)

150

Table A-1. Continued. Parametric Fold- p-value Control * OMZ175 6h* change Gene symbol Gene name < 1e-07 189.36 368.37 2 HIVEP2 human immunodeficiency virus type I enhancer binding protein 2 9.00E-07 220.56 603.62 2.7 HLX H2.0-like homeobox < 1e-07 1047.33 2127.58 2 HMGCR 3-hydroxy-3-methylglutaryl-CoA reductase < 1e-07 355.82 1166.12 3.2 HMGCS1 3-hydroxy-3-methylglutaryl-CoA synthase 1 (soluble) < 1e-07 705.5 2320.15 3.3 ICAM1 intercellular adhesion molecule 1 intercellular adhesion molecule 4 (Landsteiner-Wiener blood 4.00E-07 25.24 66.72 2.6 ICAM4 group) < 1e-07 2058.67 4138.81 2 IDI1 isopentenyl-diphosphate delta isomerase 1 < 1e-07 64.45 388.7 5.9 IL11 interleukin 11 7.00E-07 1377.18 2792.81 2 IL1RL1 interleukin 1 receptor-like 1 < 1e-07 715.35 4746.01 6.7 IL6 interleukin 6 (interferon, beta 2) < 1e-07 3647.01 16130.46 4.3 IL8 interleukin 8 < 1e-07 2091.03 6483.24 3.1 INSIG1 insulin induced gene 1 < 1e-07 405.2 1006.41 2.5 IRF6 interferon regulatory factor 6 < 1e-07 1126.4 2921.51 2.6 ITGB8 integrin, beta 8 < 1e-07 1425.74 2998.45 2.1 JUN jun proto-oncogene < 1e-07 188.38 897.64 4.8 JUNB jun B proto-oncogene < 1e-07 3565.77 8192 2.3 JUND jun D proto-oncogene potassium intermediate/small conductance calcium-activated 1.10E-06 31.45 71.88 2.3 KCNN2 channel, subfamily N, member 2 < 1e-07 603.62 1190.62 2 KDM6B lysine (K)-specific demethylase 6B 2.00E-07 17.85 40.02 2.2 KIAA0146 KIAA0146 < 1e-07 2646.74 7434.4 2.8 KITLG KIT ligand < 1e-07 151.95 2977.74 19.6 KLF4 Kruppel-like factor 4 (gut) < 1e-07 149.86 438.82 2.9 KLF5 Kruppel-like factor 5 (intestinal) < 1e-07 6494.49 13517.12 2.1 KLF6 Kruppel-like factor 6 1.80E-06 90.51 185.46 2 LBH limb bud and heart development homolog (mouse) < 1e-07 1826.67 6059.57 3.3 LDLR low density lipoprotein receptor < 1e-07 101.48 789.61 7.7 LIF leukemia inhibitory factor (cholinergic differentiation factor)

151

Table A-1. Continued. Parametric Fold- p-value Control * OMZ175 6h * change Gene symbol Gene name < 1e-07 309.22 638.04 2.1 LONRF1 LON peptidase N-terminal domain and ring finger 1 < 1e-07 1719.17 3344.33 2 MAFF v-maf musculoaponeurotic fibrosarcoma oncogene homolog F < 1e-07 1465.83 3029.79 2.1 MALL mal, T-cell differentiation protein-like < 1e-07 2735.33 5556.65 2 MCL1 myeloid cell leukemia sequence 1 (BCL2-related) 2.90E-06 86.22 278.2 3.2 MGP matrix Gla protein < 1e-07 321.24 765.36 2.4 MMP1 matrix metallopeptidase 1 (interstitial collagenase) < 1e-07 305.49 1579.22 5.3 MMP10 matrix metallopeptidase 10 (stromelysin 2) < 1e-07 6677.07 19997.05 3 MT1E metallothionein 1E < 1e-07 1408.55 6112.3 4.3 MT1F metallothionein 1F < 1e-07 119.22 4762.49 40 MT1M metallothionein 1M < 1e-07 3019.3 12633.79 4.2 MT1X metallothionein 1X < 1e-07 7525.13 16469.39 2.2 MYADM myeloid-associated differentiation marker < 1e-07 623.83 1389.16 2.2 MYO1E myosin IE < 1e-07 182.59 465.46 2.6 NAMPT nicotinamide phosphoribosyltransferase < 1e-07 880.7 1967.98 2.2 NAV3 neuron navigator 3 < 1e-07 1770.57 4787.31 2.7 NCOA7 nuclear receptor coactivator 7 neural precursor cell expressed, developmentally down-regulated < 1e-07 672.09 1525.43 2.3 NEDD4L 4-like neural precursor cell expressed, developmentally down-regulated < 1e-07 3414.6 7525.13 2.2 NEDD9 9 < 1e-07 103.43 234.35 2.3 NEK3 NIMA (never in mitosis gene a)-related kinase 3 < 1e-07 930.91 1900.94 2 NEXN nexilin (F actin binding protein) nuclear factor of activated T-cells, cytoplasmic, calcineurin- < 1e-07 423.14 947.18 2.2 NFATC1 dependent 1 nuclear factor of activated T-cells, cytoplasmic, calcineurin- < 1e-07 255.11 645.83 2.5 NFATC2 dependent 2 < 1e-07 438.06 1154.06 2.6 NFIL3 nuclear factor, interleukin 3 regulated nuclear factor of kappa light polypeptide gene enhancer in B-cells < 1e-07 105.42 292.04 2.8 NFKB2 2 (p49/p100) nuclear factor of kappa light polypeptide gene enhancer in B-cells < 1e-07 2373.01 6316.89 2.6 NFKBIA inhibitor, alpha

152

Table A-1. Continued. Parametric Fold- p-value Control* OMZ175 6h* change Gene symbol Gene name nuclear factor of kappa light polypeptide gene enhancer in B-cells < 1e-07 673.25 3666.02 5.6 NFKBIZ inhibitor, zeta 3.00E-07 72.88 221.32 3 NPTX1 neuronal pentraxin I < 1e-07 291.53 734.19 2.5 NR4A1 nuclear receptor subfamily 4, group A, member 1 < 1e-07 30.17 554.48 18.5 NR4A2 nuclear receptor subfamily 4, group A, member 2 < 1e-07 73.9 235.16 3.2 NR4A3 nuclear receptor subfamily 4, group A, member 3 < 1e-07 332.57 714.11 2.1 PCDH17 protocadherin 17 < 1e-07 71.01 206.86 2.9 PDLIM3 PDZ and LIM domain 3 < 1e-07 79.2 224.8 2.9 PDLIM3 PDZ and LIM domain 3 < 1e-07 238.44 509.35 2.1 PDLIM4 PDZ and LIM domain 4 < 1e-07 1080.51 2198.8 2 PDLIM5 PDZ and LIM domain 5 < 1e-07 470.32 948.83 2 PICALM phosphatidylinositol binding clathrin assembly protein < 1e-07 65.57 127.78 2 PID1 phosphotyrosine interaction domain containing 1 < 1e-07 962.07 3801.89 4 PLA2G4A phospholipase A2, group IVA (cytosolic, calcium-dependent) < 1e-07 504.95 1047.33 2.1 PPP1R10 protein phosphatase 1, regulatory (inhibitor) subunit 10 < 1e-07 581.04 1230.48 2.1 PPP1R15A protein phosphatase 1, regulatory (inhibitor) subunit 15A < 1e-07 13.57 62.03 4.5 PRR16 proline rich 16 prostaglandin-endoperoxide synthase 2 (prostaglandin G/H < 1e-07 373.51 10903.47 29.4 PTGS2 synthase and cyclooxygenase) < 1e-07 67.3 323.47 4.8 PTHLH parathyroid hormone-like hormone < 1e-07 268.26 606.77 2.3 RAB23 RAB23, member RAS oncogene family < 1e-07 72.13 171.85 2.4 RAI2 retinoic acid induced 2 < 1e-07 221.71 443.41 2 RASGRF2 Ras protein-specific guanine nucleotide-releasing factor 2 < 1e-07 391.4 903.89 2.3 RBM24 RNA binding motif protein 24 < 1e-07 1232.61 4211.15 3.4 RCAN1 regulator of calcineurin 1 < 1e-07 378.07 828.87 2.2 RIMKLB ribosomal modification protein rimK-like family member B < 1e-07 321.8 709.18 2.2 RIPK2 receptor-interacting serine-threonine kinase 2

153

Table A-1. Continued. Parametric Fold- p-value Control * OMZ175 6h * change Gene symbol Gene name < 1e-07 183.23 1013.41 5.6 RND1 Rho family GTPase 1 < 1e-07 111.24 559.31 5 RRAD Ras-related associated with diabetes < 1e-07 45.73 231.52 5 RRAD Ras-related associated with diabetes < 1e-07 204.36 465.46 2.3 RUNDC3B RUN domain containing 3B < 1e-07 5175.56 10423.12 2 SAT1 spermidine/spermine N1-acetyltransferase 1 < 1e-07 152.75 5527.84 35.7 SELE selectin E sema domain, transmembrane domain (TM), and cytoplasmic 8.00E-07 202.6 426.82 2.1 SEMA6D domain, (semaphorin) 6D < 1e-07 205.79 3219.23 15.6 SERPINB2 serpin peptidase inhibitor, clade B (ovalbumin), member 2 1.20E-06 505.83 990.83 2 SLC2A3 solute carrier family 2 (facilitated glucose transporter), member 3 < 1e-07 83.43 225.58 2.7 SLC35F5 solute carrier family 35, member F5 solute carrier family 4, sodium bicarbonate cotransporter, member 3.03E-05 38.85 75.45 2 SLC4A7 7 solute carrier family 7 (cationic amino acid transporter, y+ system), < 1e-07 563.2 2846.55 5 SLC7A2 member 2 4.00E-07 92.09 204.72 2.2 SMC4 structural maintenance of 4 1.13E-05 73.52 148.83 2 SOCS1 suppressor of cytokine signaling 1 < 1e-07 258.23 1215.64 4.8 SOCS3 suppressor of cytokine signaling 3 < 1e-07 313 612.05 2 SPAG9 sperm associated antigen 9 < 1e-07 770.69 1541.37 2 SPRY1 sprouty homolog 1, antagonist of FGF signaling (Drosophila) < 1e-07 970.44 3929.15 4 SPRY2 sprouty homolog 2 (Drosophila) < 1e-07 632.53 1804.65 2.9 SPRY4 sprouty homolog 4 (Drosophila) < 1e-07 435.04 958.74 2.2 SQSTM1 sequestosome 1 < 1e-07 519.15 1084.26 2.1 ST3GAL6 ST3 beta-galactoside alpha-2,3-sialyltransferase 6 1.00E-07 276.76 601.53 2.2 ST8SIA4 ST8 alpha-N-acetyl-neuraminide alpha-2,8-sialyltransferase 4 < 1e-07 522.76 1316.51 2.5 STK38L serine/threonine kinase 38 like < 1e-07 147.03 358.29 2.4 TBX3 T-box 3 1.40E-06 982.29 2503.97 2.6 TFPI2 tissue factor pathway inhibitor 2 < 1e-07 336.63 792.35 2.4 TMEM158 transmembrane protein 158 (gene/pseudogene) 2.00E-07 80.73 219.79 2.7 TMEM200A transmembrane protein 200A

154

Table A-1. Continued. Parametric Fold- p-value Control* OMZ175 6h * change Gene symbol Gene name < 1e-07 513.78 3685.13 7.1 TNFAIP3 tumor necrosis factor, alpha-induced protein 3 < 1e-07 506.7 1325.66 2.6 TNFAIP8 tumor necrosis factor, alpha-induced protein 8 < 1e-07 163.71 512 3.1 TRAF1 TNF receptor-associated factor 1 < 1e-07 1353.52 5113.16 3.8 TRIB1 tribbles homolog 1 (Drosophila) < 1e-07 27.67 110.47 4 TSLP thymic stromal lymphopoietin < 1e-07 404.5 4397.6 10.9 VCAM1 vascular cell adhesion molecule 1 < 1e-07 512 1643.44 3.2 VEGFA vascular endothelial growth factor A < 1e-07 1465.83 3821.7 2.6 VEGFC vascular endothelial growth factor C 3.90E-06 145.76 340.14 2.3 VIM vimentin < 1e-07 127.78 265.03 2.1 ZBTB16 zinc finger and BTB domain containing 16 < 1e-07 285.53 4060.66 14.3 ZFP36 zinc finger protein 36, C3H type, homolog (mouse) < 1e-07 45.57 108.57 2.4 ZNF93 zinc finger protein 93 *Background corrected, normalized signal intensity readout.

155

Table A-2. Microarray gene expression changes of genes with fold reductions less than -2 fold 5 h post infection with S. mutans OMZ175 compared to uninfected control. Parametric p-value Control OMZ175 6h Fold-change Gene symbol Description < 1e-07 567.12 245.57 -2.3 AIM1 absent in melanoma 1 < 1e-07 3432.4 1307.41 -2.6 ANGPTL4 angiopoietin-like 4 4.00E-07 116.36 53.91 -2.2 ANO2 anoctamin 2 < 1e-07 3723.65 1365.3 -2.7 BMP4 bone morphogenetic protein 4 < 1e-07 1761.39 514.67 -3.4 CABLES1 Cdk5 and Abl enzyme substrate 1 4.80E-06 107.82 48.59 -2.2 CCAR1 cell division cycle and apoptosis regulator 1 2.10E-06 104.33 52.71 -2 CCDC76 coiled-coil domain containing 76 < 1e-07 1995.45 888.36 -2.3 CGNL1 cingulin-like 1 < 1e-07 1280.51 491.14 -2.6 CHST1 carbohydrate (keratan sulfate Gal-6) sulfotransferase 1 carbohydrate (N-acetylgalactosamine 4-sulfate 6-O) < 1e-07 1423.28 558.34 -2.6 CHST15 sulfotransferase 15 < 1e-07 3834.97 1509.65 -2.5 CLDN5 claudin 5 < 1e-07 158.68 63.12 -2.5 CLMN calmin (calponin-like, transmembrane) < 1e-07 512 258.68 -2 CNTNAP3B similar to cell recognition molecule CASPR3 < 1e-07 4600.26 2176.06 -2.1 DDIT4 DNA-damage-inducible transcript 4 1.98E-05 645.83 320.68 -2 DUSP4 dual specificity phosphatase 4 < 1e-07 1761.39 844.82 -2.1 EFNA1 ephrin-A1 < 1e-07 8436.92 2344.4 -3.6 EIF5 eukaryotic translation initiation factor 5 < 1e-07 543.07 210.11 -2.6 ENC1 ectodermal-neural cortex 1 (with BTB-like domain) < 1e-07 212.31 85.63 -2.5 EPHX4 epoxide hydrolase 4 < 1e-07 1549.41 602.58 -2.6 FAM124B family with sequence similarity 124B < 1e-07 635.83 170.07 -3.7 FAM13B family with sequence similarity 13, member B < 1e-07 6793.79 3208.09 -2.1 FAM43A family with sequence similarity 43, member A < 1e-07 1130.31 456.67 -2.5 FAM84B family with sequence similarity 84, member B < 1e-07 934.14 445.72 -2.1 FAM89A family with sequence similarity 89, member A < 1e-07 355.82 139.34 -2.6 FILIP1 filamin A interacting protein 1

156

Table A-2. Continued. Parametric p-value Control OMZ175 6h Fold-change Gene symbol Description < 1e-07 536.52 244.72 -2.2 FLRT2 fibronectin leucine rich transmembrane protein 2 < 1e-07 91.14 41.36 -2.2 FSTL5 follistatin-like 5 < 1e-07 242.19 116.16 -2.1 GATA3 GATA binding protein 3 < 1e-07 905.46 312.45 -2.9 GIMAP1 GTPase, IMAP family member 1 < 1e-07 1629.26 740.57 -2.2 GIMAP2 GTPase, IMAP family member 2 1.00E-07 1761.39 892.99 -2 GIMAP4 GTPase, IMAP family member 4 < 1e-07 3578.15 1549.41 -2.3 GIMAP6 GTPase, IMAP family member 6 < 1e-07 1606.83 524.57 -3.1 GIMAP7 GTPase, IMAP family member 7 < 1e-07 2040.91 924.48 -2.2 GIMAP8 GTPase, IMAP family member 8 3.30E-06 32.67 15.73 -2.1 GOPC golgi-associated PDZ and coiled-coil motif containing < 1e-07 179.77 60.03 -3 GPR146 G protein-coupled receptor 146 heterogeneous nuclear ribonucleoprotein D (AU-rich element RNA 2.60E-06 453.51 218.27 -2.1 HNRNPD binding protein 1, 37kDa) < 1e-07 413 196.72 -2.1 HOXA3 homeobox A3 < 1e-07 268.73 130.24 -2.1 HOXA9 homeobox A9 < 1e-07 1344.17 688.59 -2 HOXD1 homeobox D1 7.00E-07 517.35 244.72 -2.1 HRCT1 histidine rich carboxyl terminus 1 < 1e-07 226.36 86.37 -2.6 HSD17B2 hydroxysteroid (17-beta) dehydrogenase 2 < 1e-07 548.75 251.17 -2.2 INO80C INO80 complex subunit C < 1e-07 703.06 331.42 -2.1 INPP5D inositol polyphosphate-5-phosphatase, 145kDa 2.40E-06 371.57 190.02 -2 KIAA1274 KIAA1274 < 1e-07 1071.19 549.7 -2 KIAA1370 KIAA1370 < 1e-07 1164.1 237.21 -4.9 KIT v-kit Hardy-Zuckerman 4 feline sarcoma viral oncogene homolog < 1e-07 396.86 172.45 -2.3 LFNG LFNG O-fucosylpeptide 3-beta-N-acetylglucosaminyltransferase 4.00E-07 532.82 251.6 -2.1 LHX6 LIM homeobox 6 < 1e-07 1302.89 629.25 -2.1 LOC158376 hypothetical LOC158376 < 1e-07 193.01 70.4 -2.7 MBD5 methyl-CpG binding domain protein 5 < 1e-07 919.69 454.3 -2 MERTK c-mer proto-oncogene tyrosine kinase < 1e-07 166.57 83.14 -2 MGC16121 hypothetical protein MGC16121

157

Table A-2. Continued. Parametric p-value Control OMZ175 6h Fold-change Gene symbol Description 2.00E-07 88.95 41.43 -2.2 MMP28 matrix metallopeptidase 28 < 1e-07 437.31 175.16 -2.5 MN1 meningioma (disrupted in balanced translocation) 1 2.00E-07 2206.43 1126.4 -2 MTUS1 microtubule associated tumor suppressor 1 < 1e-07 782.8 326.29 -2.4 MYLIP myosin regulatory light chain interacting protein 3.90E-06 492.85 241.77 -2 N4BP2L2 NEDD4 binding protein 2-like 2 < 1e-07 101.48 52.07 -2 NAV2 neuron navigator 2 < 1e-07 211.94 108.2 -2 NBPF1 neuroblastoma breakpoint family, member 1 < 1e-07 461.44 220.94 -2.1 NFAT5 nuclear factor of activated T-cells 5, tonicity-responsive < 1e-07 1278.29 638.04 -2 NR1D2 nuclear receptor subfamily 1, group D, member 2 < 1e-07 2019.8 1024 -2 NR2F1 nuclear receptor subfamily 2, group F, member 1 < 1e-07 1981.67 1018.69 -2 NRARP NOTCH-regulated ankyrin repeat protein 6.00E-07 190.02 83.72 -2.3 OAS1 2',5'-oligoadenylate synthetase 1, 40/46kDa 3.30E-06 121.1 34.54 -3.5 PDK4 pyruvate dehydrogenase kinase, isozyme 4 < 1e-07 403.8 170.96 -2.4 PLLP plasmolipin < 1e-07 376.76 164.85 -2.3 PNMAL1 PNMA-like 1 2.00E-07 987.41 487.75 -2 PPP1R16B protein phosphatase 1, regulatory (inhibitor) subunit 16B < 1e-07 296.63 137.66 -2.2 PPP1R3C protein phosphatase 1, regulatory (inhibitor) subunit 3C < 1e-07 2711.73 1084.26 -2.5 PRICKLE1 prickle homolog 1 (Drosophila) 2.10E-06 58.79 27.47 -2.1 PRKAA1 protein kinase, AMP-activated, alpha 1 catalytic subunit 8.00E-06 94.35 45.81 -2.1 REV3L REV3-like, catalytic subunit of DNA polymerase zeta (yeast) 1.80E-06 98.53 34.84 -2.8 RGS7BP regulator of G-protein signaling 7 binding protein 1.00E-07 667.44 316.82 -2.1 RPS24 ribosomal protein S24 2.70E-06 53.82 26.72 -2 SFRS18 splicing factor, arginine/serine-rich 18 < 1e-07 3152.98 1573.76 -2 SLC39A10 solute carrier family 39 (zinc transporter), member 10 < 1e-07 418.04 131.83 -3.2 SNAI2 snail homolog 2 (Drosophila) < 1e-07 476.06 225.58 -2.1 SPSB1 splA/ryanodine receptor domain and SOCS box containing 1 < 1e-07 4812.26 2414.49 -2 TFRC transferrin receptor (p90, CD71) < 1e-07 924.48 472.77 -2 TGFBRAP1 transforming growth factor, beta receptor associated protein 1

158

Table A-2. Continued. Parametric p-value Control OMZ175 6h Fold-change Gene symbol Description < 1e-07 244.3 120.68 -2 TIGD2 tigger transposable element derived 2 < 1e-07 830.31 232.73 -3.6 TNFSF10 tumor necrosis factor (ligand) superfamily, member 10 < 1e-07 172.74 59.51 -2.9 TRA2A transformer 2 alpha homolog (Drosophila) < 1e-07 666.29 259.57 -2.6 TRIB2 tribbles homolog 2 (Drosophila) 2.36E-05 167.15 85.92 -2 VASH1 vasohibin 1 4.50E-06 558.34 267.33 -2.1 XAF1 XIAP associated factor 1 7.50E-06 191.34 92.89 -2.1 XPO1 exportin 1 (CRM1 homolog, yeast) < 1e-07 1190.62 554.48 -2.2 ZBTB1 zinc finger and BTB domain containing 1 < 1e-07 103.97 41.14 -2.5 ZBTB24 zinc finger and BTB domain containing 24 6.40E-06 241.35 119.43 -2 ZCCHC18 zinc finger, CCHC domain containing 18 9.00E-07 334.88 129.11 -2.6 ZNF207 zinc finger protein 207 < 1e-07 132.51 51.54 -2.6 ZNF345 zinc finger protein 345 < 1e-07 2931.66 1337.2 -2.2 ZNF521 zinc finger protein 521 < 1e-07 116.77 56.89 -2.1 ZNF658 zinc finger protein 658 < 1e-07 371.57 111.24 -3.3 ZNF792 zinc finger protein 792 2.00E-07 176.68 87.58 -2 ZSCAN16 zinc finger and SCAN domain containing 16

159

Generate using Kyoto Encyclopedia of Genes and Genomes (KEGG) Pathway Database (http://www.genome.jp/dbget- bin/www_bget?map04210). Each box may represent more than one gene or probe set, hence the “purple” designation.

160

Generate using Kyoto Encyclopedia of Genes and Genomes (KEGG) Pathway Database (http://www.genome.jp/dbget- bin/www_bget?map04350). Each box may represent more than one gene or probe set, hence the “purple” designation.

161

Generate using Kyoto Encyclopedia of Genes and Genomes (KEGG) Pathway Database (http://www.genome.jp/dbget- bin/www_bget?map04010). Each box may represent more than one gene or probe set, hence the “purple” designation.

162

Generate using Kyoto Encyclopedia of Genes and Genomes (KEGG) Pathway Database (http://www.genome.jp/dbget- bin/www_bget?map04620). Each box may represent more than one gene or probe set, hence the “purple” designation.

163

Generate using Kyoto Encyclopedia of Genes and Genomes (KEGG) Pathway Database (http://www.genome.jp/dbget- bin/www_bget?map04620). Each box may represent more than one gene or probe set, hence the “purple” designation.

164

Generate using Kyoto Encyclopedia of Genes and Genomes (KEGG) Pathway Database (http://www.genome.jp/dbget- bin/www_bget?map04370). Each box may represent more than one gene or probe set, hence the “purple” designation.

165

LIST OF REFERENCES

1. Breslow JL: Cardiovascular disease burden increases, NIH funding decreases. Nat Med 1997, 3(6):600-601.

2. Braunwald E: Shattuck lecture--Cardiovascular medicine at the turn of the millennium: triumphs, concerns, and opportunities. N Engl J Med 1997, 337(19):1360-1369.

3. Berenson GS, Srinivasan SR, Bao W, Newman WP, 3rd, Tracy RE, Wattigney WA: Association between multiple cardiovascular risk factors and atherosclerosis in children and young adults. The Bogalusa Heart Study. N Engl J Med 1998, 338(23):1650-1656.

4. Glantz SA, Parmley WW: Passive smoking and heart disease. Mechanisms and risk. J Am Med Assoc 1995, 273(13):1047-1053.

5. Kannel WB: Blood pressure as a cardiovascular risk factor: prevention and treatment. J Am Med Assoc 1996, 275(20):1571-1576.

6. Leaverton PE, Sorlie PD, Kleinman JC, Dannenberg AL, Ingster-Moore L, Kannel WB, Cornoni-Huntley JC: Representativeness of the Framingham risk model for coronary heart disease mortality: a comparison with a national cohort study. J Chronic Dis 1987, 40(8):775-784.

7. Mehta JL, Saldeen TG, Rand K: Interactive role of infection, inflammation and traditional risk factors in atherosclerosis and coronary artery disease. J Am Coll Cardiol 1998, 31(6):1217-1225.

8. Rothenbacher D, Brenner H, Hoffmeister A, Mertens T, Persson K, Koenig W: Relationship between infectious burden, systemic inflammatory response, and risk of stable coronary artery disease: role of confounding and reference group. Atherosclerosis 2003, 170(2):339-345.

9. Haraszthy VI, Zambon JJ, Trevisan M, Zeid M, Genco RJ: Identification of periodontal pathogens in atheromatous plaques. J Periodontol 2000, 71(10):1554-1560.

10. Kozarov E, Sweier D, Shelburne C, Progulske-Fox A, Lopatin D: Detection of bacterial DNA in atheromatous plaques by quantitative PCR. Microbes Infect 2006, 8(3):687-693.

11. Kozarov EV, Dorn BR, Shelburne CE, Dunn WA, Jr., Progulske-Fox A: Human atherosclerotic plaque contains viable invasive Actinobacillus actinomycetemcomitans and Porphyromonas gingivalis. Arterioscler Thromb Vasc Biol 2005, 25(3):e17-18.

166

12. Lockhart PB, Bolger AF, Papapanou PN, Osinbowale O, Trevisan M, Levison ME, Taubert KA, Newburger JW, Gornik HL, Gewitz MH et al: Periodontal disease and atherosclerotic vascular disease: does the evidence support an independent association?: A scientific statement from the American Heart Association. Circulation 2012, 125(20):2520-2544.

13. Senba T, Kobayashi Y, Inoue K, Kaneto C, Inoue M, Toyokawa S, Suyama Y, Suzuki T, Miyano Y, Miyoshi Y: The association between self-reported periodontitis and coronary heart disease--from MY Health Up Study. J Occup Health 2008, 50(3):283-287.

14. Ylostalo PV, Jarvelin MR, Laitinen J, Knuuttila ML: Gingivitis, dental caries and tooth loss: risk factors for cardiovascular diseases or indicators of elevated health risks. J Clin Periodontol 2006, 33(2):92-101.

15. Beck JD, Eke P, Heiss G, Madianos P, Couper D, Lin D, Moss K, Elter J, Offenbacher S: Periodontal disease and coronary heart disease: a reappraisal of the exposure. Circulation 2005, 112(1):19-24.

16. Elter JR, Champagne CM, Offenbacher S, Beck JD: Relationship of periodontal disease and tooth loss to prevalence of coronary heart disease. J Periodontol 2004, 75(6):782-790.

17. Persson RE, Hollender LG, Powell VL, MacEntee M, Wyatt CC, Kiyak HA, Persson GR: Assessment of periodontal conditions and systemic disease in older subjects. II. Focus on cardiovascular diseases. J Clin Periodontol 2002, 29(9):803-810.

18. Dorn JM, Genco RJ, Grossi SG, Falkner KL, Hovey KM, Iacoviello L, Trevisan M: Periodontal disease and recurrent cardiovascular events in survivors of myocardial infarction (MI): the Western New York Acute MI Study. J Periodontol 2010, 81(4):502-511.

19. Dietrich T, Jimenez M, Krall Kaye EA, Vokonas PS, Garcia RI: Age-dependent associations between chronic periodontitis/edentulism and risk of coronary heart disease. Circulation 2008, 117(13):1668-1674.

20. Hung HC, Joshipura KJ, Colditz G, Manson JE, Rimm EB, Speizer FE, Willett WC: The association between tooth loss and coronary heart disease in men and women. J Public Health Dent 2004, 64(4):209-215.

21. Joshipura KJ, Wand HC, Merchant AT, Rimm EB: Periodontal disease and biomarkers related to cardiovascular disease. J Dent Res 2004, 83(2):151- 155.

22. Meurman JH, Janket SJ, Qvarnstrom M, Nuutinen P: Dental infections and serum inflammatory markers in patients with and without severe heart

167

disease. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 2003, 96(6):695- 700.

23. Tonetti MS, D'Aiuto F, Nibali L, Donald A, Storry C, Parkar M, Suvan J, Hingorani AD, Vallance P, Deanfield J: Treatment of periodontitis and endothelial function. N Engl J Med 2007, 356(9):911-920.

24. Ide M, Jagdev D, Coward PY, Crook M, Barclay GR, Wilson RF: The short-term effects of treatment of chronic periodontitis on circulating levels of endotoxin, C-reactive protein, tumor necrosis factor-alpha, and interleukin-6. J Periodontol 2004, 75(3):420-428.

25. Beck JD, Offenbacher S: Systemic effects of periodontitis: epidemiology of periodontal disease and cardiovascular disease. J Periodontol 2005, 76(11 Suppl):2089-2100.

26. Chiu B: Multiple infections in carotid atherosclerotic plaques. Am Heart J 1999, 138(5 Pt 2):S534-536.

27. Rafferty B, Jonsson D, Kalachikov S, Demmer RT, Nowygrod R, Elkind MS, Bush H, Jr., Kozarov E: Impact of monocytic cells on recovery of uncultivable bacteria from atherosclerotic lesions. J Intern Med 2011, 270(3):273-280.

28. Gibson FC, 3rd, Hong C, Chou HH, Yumoto H, Chen J, Lien E, Wong J, Genco CA: Innate immune recognition of invasive bacteria accelerates atherosclerosis in apolipoprotein E-deficient mice. Circulation 2004, 109(22):2801-2806.

29. Lalla E, Lamster IB, Hofmann MA, Bucciarelli L, Jerud AP, Tucker S, Lu Y, Papapanou PN, Schmidt AM: Oral infection with a periodontal pathogen accelerates early atherosclerosis in apolipoprotein E-null mice. Arterioscler Thromb Vasc Biol 2003, 23(8):1405-1411.

30. Li L, Messas E, Batista EL, Jr., Levine RA, Amar S: Porphyromonas gingivalis infection accelerates the progression of atherosclerosis in a heterozygous apolipoprotein E-deficient murine model. Circulation 2002, 105(7):861-867.

31. Kesavalu L, Lucas AR, Verma RK, Liu L, Dai E, Sampson E, Progulske-Fox A: Increased atherogenesis during Streptococcus mutans infection in ApoE- null mice. J Dent Res 2012, 91(3):255-260.

32. Paster BJ, Boches SK, Galvin JL, Ericson RE, Lau CN, Levanos VA, Sahasrabudhe A, Dewhirst FE: Bacterial diversity in human subgingival plaque. J Bacteriol 2001, 183(12):3770-3783.

33. Dewhirst FE, Chen T, Izard J, Paster BJ, Tanner AC, Yu WH, Lakshmanan A, Wade WG: The human oral microbiome. J Bacteriol 2010, 192(19):5002-5017.

168

34. Vartoukian SR, Palmer RM, Wade WG: Strategies for culture of 'unculturable' bacteria. FEMS Microbiol Lett 2010, 309(1):1-7.

35. Kolbert CP, Persing DH: Ribosomal DNA sequencing as a tool for identification of bacterial pathogens. Curr Opin Microbiol 1999, 2(3):299-305.

36. Russell RRB: Pathogenesis of oral streptococci. In: Gram-Positve Pathogens. 2 edn. Edited by Fischetti VA, Novick, R.P., Ferretti, J.J., Portnoy D.A., Rood, J.I. . Washington D.C.: ASM Press; 2006: 332-339.

37. Marsh M, Martin, M.V.: Acquisition, adherence, distribution and metabolism of the oral microflora. In: Oral Microbiol. 4 edn. Woburn, MA: Wright; 1999: 34- 57.

38. Loesche WJ: Role of Streptococcus mutans in human dental decay. Microbiol Rev 1986, 50:353-380.

39. Li J, Helmerhorst EJ, Leone CW, Troxler RF, Yaskell T, Haffajee AD, Socransky SS, Oppenheim FG: Identification of early microbial colonizers in human dental biofilm. J Appl Microbiol 2004, 97(6):1311-1318.

40. Kolenbrander PE, London J: Adhere Today, Here Tomorrow - Oral Bacterial Adherence. J Bacteriol 1993, 175(11):3247-3252.

41. Kolenbrander PE, Andersen RN, Moore LVH: Intrageneric Coaggregation among Strains of Human Oral Bacteria - Potential Role in Primary Colonization of the Tooth Surface. Appl Environ Microbiol 1990, 56(12):3890- 3894.

42. De Vos P: Bergey's Manual of Systematic Bacteriology, vol. 3, 2nd edn. Dordrecht ; New York Springer; 2009.

43. Kreth J, Merritt J, Qi F: Bacterial and host interactions of oral streptococci. DNA Cell Biol 2009, 28(8):397-403.

44. Nobbs AH, Lamont RJ, Jenkinson HF: Streptococcus adherence and colonization. Microbiol Mol Biol Rev 2009, 73(3):407-450, Table of Contents.

45. Hamilton IR, Buckley ND: Adaptation by Streptococcus mutans to acid tolerance. Oral Microbiol Immunol 1991, 6(2):65-71.

46. Li YH, Lau PC, Lee JH, Ellen RP, Cvitkovitch DG: Natural genetic transformation of Streptococcus mutans growing in biofilms. J Bacteriol 2001, 183(3):897-908.

47. Banas JA: Virulence properties of Streptococcus mutans. Front Biosci 2004, 9:1267-1277.

169

48. Lemos JA, Burne RA: A model of efficiency: stress tolerance by Streptococcus mutans. Microbiology 2008, 154(Pt 11):3247-3255.

49. Senadheera D, Cvitkovitch DG: Quorum sensing and biofilm formation by Streptococcus mutans. Adv Exp Med Biol 2008, 631:178-188.

50. Ajdic D, McShan WM, McLaughlin RE, Savic G, Chang J, Carson MB, Primeaux C, Tian R, Kenton S, Jia H et al: Genome sequence of Streptococcus mutans UA159, a cariogenic dental pathogen. Proc Natl Acad Sci USA2002, 99(22):14434-14439.

51. Yamada T, Takahashi-Abbe S, Abbe K: Effects of oxygen on pyruvate formate-lyase in situ and sugar metabolism of Streptococcus mutans and Streptococcus sanguis. Infect Immun 1985, 47(1):129-134.

52. Rawlings ND, Tolle DP, Barrett AJ: MEROPS: the peptidase database. Nucleic Acids Res 2004, 32(Database issue):D160-164.

53. MEROPS Peptidase Database [http://merops.sanger.ac.uk]

54. Hall-Stoodley L, Stoodley P: Evolving concepts in biofilm infections. Cell Microbiol 2009, 11(7):1034-1043.

55. Flemming HC, Neu TR, Wozniak DJ: The EPS matrix: the "house of biofilm cells". J Bacteriol 2007, 189(22):7945-7947.

56. Sutherland I: Biofilm exopolysaccharides: a strong and sticky framework. Microbiology 2001, 147(Pt 1):3-9.

57. Stoodley P, Sauer K, Davies DG, Costerton JW: Biofilms as complex differentiated communities. Ann Rev Microbiol 2002, 56:187-209.

58. Horswill AR, Stoodley P, Stewart PS, Parsek MR: The effect of the chemical, biological, and physical environment on quorum sensing in structured microbial communities. Anal Bioanal Chem 2007, 387(2):371-380.

59. Donlan RM, Costerton JW: Biofilms: survival mechanisms of clinically relevant microorganisms. Clin Microbiol Rev 2002, 15(2):167-193.

60. Lewis K: Persister cells, dormancy and infectious disease. Nat Rev Microbiol 2007, 5(1):48-56.

61. Brooun A, Liu S, Lewis K: A dose-response study of antibiotic resistance in Pseudomonas aeruginosa biofilms. Antimicrob Agents Chemother 2000, 44(3):640-646.

62. Guiot E, Georges P, Brun A, Fontaine-Aupart MP, Bellon-Fontaine MN, Briandet R: Heterogeneity of diffusion inside microbial biofilms determined by

170

fluorescence correlation spectroscopy under two-photon excitation. Photochem Photobiol 2002, 75(6):570-578.

63. Banas JA, Vickerman MM: Glucan-binding proteins of the oral streptococci. Crit Rev Oral Biol Med 2003, 14(2):89-99.

64. Sciotti MA, Yamodo I, Klein JP, Ogier JA: The N-terminal half part of the oral streptococcal antigen I/IIf contains two distinct binding domains. FEMS Microbiol Lett 1997, 153(2):439-445.

65. Demuth DR, Davis CA, Corner AM, Lamont RJ, Leboy PS, Malamud D: Cloning and expression of a Streptococcus sanguis surface antigen that interacts with a human salivary agglutinin. Infect Immun 1988, 56(9):2484-2490.

66. Brady LJ, Maddocks SE, Larson MR, Forsgren N, Persson K, Deivanayagam CC, Jenkinson HF: The changing faces of Streptococcus antigen I/II polypeptide family adhesins. Mol Microbiol 2010, 77(2):276-286.

67. Nakano K, Nomura R, Taniguchi N, Lapirattanakul J, Kojima A, Naka S, Senawongse P, Srisatjaluk R, Gronroos L, Alaluusua S et al: Molecular characterization of Streptococcus mutans strains containing the cnm gene encoding a collagen-binding adhesin. Arch Oral Biol 2010, 55(1):34-39.

68. Nomura R, Nakano K, Naka S, Nemoto H, Masuda K, Lapirattanakul J, Alaluusua S, Matsumoto M, Kawabata S, Ooshima T: Identification and characterization of a collagen-binding protein, Cbm, in Streptococcus mutans. Mol Oral Microbiol 2012, 27(4):308-323.

69. Gibbons RJ, van Houte J: Bacterial adherence and the formation of dental plaque. In: Bacterial Adherence. edn. Edited by Beachy EH. London: Chapman and Hall; 1980.

70. Loesche WJ: Microbiology of dental decay and periodontal disease. In: Medical Microbiology. 4th edn. Edited by Baron S. Galveston (TX); 1996.

71. Kolenbrander PE, Palmer RJ, Jr., Rickard AH, Jakubovics NS, Chalmers NI, Diaz PI: Bacterial interactions and successions during plaque development. Periodontol 2000 2006, 42:47-79.

72. Suchett-Kaye G, Morrier JJ, Barsotti O: Interactions between non-immune host cells and the immune system during periodontal disease: Role of the gingival keratinocyte. Crit Rev Oral Biol M 1998, 9(3):292-305.

73. Tonetti MS, Imboden MA, Lang NP: Neutrophil migration into the gingival sulcus is associated with transepithelial gradients of interleukin-8 and ICAM-1. J Periodontol 1998, 69(10):1139-1147.

171

74. Sugiyama A, Uehara A, Iki K, Matsushita K, Nakamura R, Ogawa T, Sugawara S, Takada H: Activation of human gingival epithelial cells by cell-surface components of black-pigmented bacteria: augmentation of production of interleukin-8, granulocyte colony-stimulating factor and granulocyte- macrophage colony-stimulating factor and expression of intercellular adhesion molecule 1. J Med Microbiol 2002, 51(1):27-33.

75. Wilson M, Reddi K, Henderson B: Cytokine-inducing components of periodontopathogenic bacteria. J Periodontal Res 1996, 31(6):393-407.

76. Dye BA, Li X, Beltran-Aguilar ED: Selected oral health indicators in the United States, 2005-2008. NCHS data brief 2012(96):1-8.

77. Hamada S, Slade HD: Biology, immunology, and cariogenicity of Streptococcus mutans. Microbiol Rev 1980, 44(2):331-384.

78. Preston AJ, Edgar WM: Developments in dental plaque pH modelling. Journal of dentistry 2005, 33(3):209-222.

79. Stephan RM, Miller BF: A quantitative method for evaluating physical and chemical agents which modify production of acids in bacterial plaques on human teeth. J Dent Res 1943, 22:45–51.

80. Stephan RM: Changes in hydrogen ion concentration on tooth surfaces and in carious lesions. J Am Dent Assoc 1940, 27:718-723.

81. Chassy BM, Beall JR, Bielawski RM, Porter EV, Donkersloot JA: Occurrence and distribution of sucrose-metabolizing enzymes in oral streptococci. Infect Immun 1976, 14(2):408-415.

82. Dashper SG, Reynolds EC: Lactic acid excretion by Streptococcus mutans. Microbiol-Uk 1996, 142:33-39.

83. Shapiro-Shelef M, Calame K: Regulation of plasma-cell development. Nat Rev Immunol 2005, 5(3):230-242.

84. Brewer JW, Hendershot LM: Building an antibody factory: a job for the unfolded protein response. Nat Immunol 2005, 6(1):23-29.

85. Hahn CL, Best AM, Tew JG: Cytokine induction by Streptococcus mutans and pulpal pathogenesis. Infect Immun 2000, 68(12):6785-6789.

86. Petersen PE: Priorities for research for oral health in the 21st century--the approach of the WHO Global Oral Health Programme. Community Dent Health 2005, 22(2):71-74.

172

87. Petersen PE, Yamamoto T: Improving the oral health of older people: the approach of the WHO Global Oral Health Programme. Community Dent Oral Epidemiol 2005, 33(2):81-92.

88. Malicka M, Zielinski R, Piotrowska V, Andrzejewski J, Zakrzewska A: Can dental problems have influence on difficulties in treating paranasal sinusitis in children?. Przeglad lekarski 2011, 68(1):68-72.

89. Zeledon JI, McKelvey RL, Servilla KS, Hofinger D, Konstantinov KN, Kellie S, Sun Y, Massie LW, Hartshorne MF, Tzamaloukas AH: Glomerulonephritis causing acute renal failure during the course of bacterial infections. Histological varieties, potential pathogenetic pathways and treatment. Int Urol Nephrol 2008, 40(2):461-470.

90. Ullman RF, Strampfer MJ, Cunha BA: Streptococcus mutans vertebral osteomyelitis. Heart Lung 1988, 17(3):319-321.

91. Rajpal RS, Leibsohn JA, Liekweg WG, Gross CM, Olinger GN, Rose HD, Bamrah VS: Infected left atrial myxoma with bacteremia simulating infective endocarditis. Arch Intern Med 1979, 139(10):1176-1178.

92. Sattler FR, Ruskin J: Empyema due to Streptococcus mutans. Chest 1977, 71(2):229-231.

93. Durand R, Gunselman EL, Hodges JS, Diangelis AJ, Michalowicz BS: A pilot study of the association between cariogenic oral bacteria and preterm birth. Oral Dis 2009, 15(6):400-406.

94. Romero R, Schaudinn C, Kusanovic JP, Gorur A, Gotsch F, Webster P, Nhan- Chang CL, Erez O, Kim CJ, Espinoza J et al: Detection of a microbial biofilm in intraamniotic infection. Am J Obstet Gynecol 2008, 198(1):135 e131-135.

95. Rabe LK, Winterscheid KK, Hillier SL: Association of viridans group streptococci from pregnant women with bacterial vaginosis and upper genital tract infection. J Clin Microbiol 1988, 26(6):1156-1160.

96. Reeder JC, Westwell AJ, Hutchinson DN: Indifferent streptococci in normal and purulent eyes of neonates. J Clin Pathol 1985, 38(8):942-945.

97. Parker MT, Ball LC: Streptococci and aerococci associated with systemic infection in man. J Med Microbiol 1976, 9(3):275-302.

98. Morency AM, Rallu F, Laferriere C, Bujoldg E: Eradication of intra-amniotic Streptococcus mutans in a woman with a short cervix. J Obstet Gynaecol Can 2006, 28(10):898-902.

173

99. Ioannidis O, Kakoutis E, Katsifa H, Rafail S, Chatzopoulos S, Kotronis A, Makrantonakis N: Streptococcus mutans: a rare cause of retroperitoneal abscess. Adv Med Sci 2011, 56(1):113-118.

100. Ferrieri P, Gewitz MH, Gerber MA, Newburger JW, Dajani AS, Shulman ST, Wilson W, Bolger AF, Bayer A, Levison ME et al: Unique features of infective endocarditis in childhood. Circulation 2002, 105(17):2115-2126.

101. Dekkers P, Elbers HR, Morshuis WJ, Jaarsma W: Infected left atrial myxoma. J Am Soc Echocardiogr 2001, 14(6):644-645.

102. Vogt PR, Jenni R, Turina MI: Infected left atrial myxoma with concomitant mitral valve endocarditis. Eur J Cardiothorac Surg 1996, 10(1):71-73.

103. Nomura R, Hamada M, Nakano K, Nemoto H, Fujimoto K, Ooshima T: Repeated bacteraemia caused by Streptococcus mutans in a patient with Sjogren's syndrome. J Med Microbiol 2007, 56(Pt 7):988-992.

104. Siudikiene J, Machiulskiene V, Nyvad B, Tenovuo J, Nedzelskiene I: Dental caries increments and related factors in children with type 1 diabetes mellitus. Caries Res 2008, 42(5):354-362.

105. Li X, Kolltveit KM, Tronstad L, Olsen I: Systemic diseases caused by oral infection. Clin Microbiol Rev 2000, 13(4):547-558.

106. Forner L, Larsen T, Kilian M, Holmstrup P: Incidence of bacteremia after chewing, tooth brushing and scaling in individuals with periodontal inflammation. J Clin Periodontol 2006, 33(6):401-407.

107. Kinane DF, Riggio MP, Walker KF, MacKenzie D, Shearer B: Bacteraemia following periodontal procedures. J Clin Periodontol 2005, 32(7):708-713.

108. Lofthus JE, Waki MY, Jolkovsky DL, Otomo-Corgel J, Newman MG, Flemmig T, Nachnani S: Bacteremia following subgingival irrigation and scaling and root planing. J Periodontol 1991, 62(10):602-607.

109. Carroll GC, Sebor RJ: Dental flossing and its relationship to transient bacteremia. J Periodontol 1980, 51(12):691-692.

110. Daly C, Mitchell D, Grossberg D, Highfield J, Stewart D: Bacteraemia caused by periodontal probing. Aust Dent J 1997, 42(2):77-80.

111. Rosenfeld ME, Campbell LA: Pathogens and atherosclerosis: update on the potential contribution of multiple infectious organisms to the pathogenesis of atherosclerosis. Thromb Haemost 2011, 106(5):858-867.

112. Nakano K, Nomura R, Ooshima T: Streptococcus mutans and cardiovascular diseases. Jpn Dent Sci Rev 2008, 44(1):29-37.

174

113. Meyer DH, Fives-Taylor PM: Oral pathogens: from dental plaque to cardiac disease. Curr Opin Microbiol 1998, 1:88-95.

114. Nakano K, Ooshima T: Serotype classification of Streptococcus mutans and its detection outside the oral cavity. Future Microbiol 2009, 4(7):891-902.

115. Linzer R, M. S. Reddy, M. J. Levine: Molecular Microbiology and Immunology of Streptococcus mutans. Amsterdam, The Netherlands: Elsevier Science Publishers; 1986.

116. Tsukioka Y, Yamashita Y, Oho T, Nakano Y, Koga T: Biological function of the dTDP-rhamnose synthesis pathway in Streptococcus mutans. J Bacteriol 1997, 179(4):1126-1134.

117. Tsukioka Y, Yamashita Y, Nakano Y, Oho T, Koga T: Identification of a fourth gene involved in dTDP-rhamnose synthesis in Streptococcus mutans. J Bacteriol 1997, 179(13):4411-4414.

118. Yamashita Y, Tsukioka Y, Nakano Y, Tomihisa K, Oho T, Koga T: Biological functions of UDP-glucose synthesis in Streptococcus mutans. Microbiology 1998, 144 ( Pt 5):1235-1245.

119. Yamashita Y, Tsukioka Y, Tomihisa K, Nakano Y, Koga T: Genes involved in cell wall localization and side chain formation of rhamnose-glucose polysaccharide in Streptococcus mutans. J Bacteriol 1998, 180(21):5803- 5807.

120. Yamashita Y, Shibata Y, Nakano Y, Tsuda H, Kido N, Ohta M, Koga T: A novel gene required for rhamnose-glucose polysaccharide synthesis in Streptococcus mutans. J Bacteriol 1999, 181(20):6556-6559.

121. Shibata Y, Ozaki K, Seki M, Kawato T, Tanaka H, Nakano Y, Yamashita Y: Analysis of loci required for determination of serotype antigenicity in Streptococcus mutans and its clinical utilization. J Clin Microbiol 2003, 41(9):4107-4112.

122. Benabdelmoumene S, Dumont S, Petit C, Poindron P, Wachsmann D, Klein JP: Activation of human monocytes by Streptococcus mutans serotype f polysaccharide: immunoglobulin G Fc receptor expression and tumor necrosis factor and interleukin-1 production. Infect Immun 1991, 59(9):3261- 3266.

123. Soell M, Lett E, Holveck F, Scholler M, Wachsmann D, Klein JP: Activation of human monocytes by streptococcal rhamnose glucose polymers is mediated by CD14 antigen, and mannan binding protein inhibits TNF-alpha release. J Immunol 1995, 154(2):851-860.

175

124. Nakano K, Nomura R, Matsumoto M, Ooshima T: Roles of oral bacteria in cardiovascular diseases--from molecular mechanisms to clinical cases: Cell-surface structures of novel serotype k Streptococcus mutans strains and their correlation to virulence. J Pharmacol Sci 2010, 113(2):120-125.

125. Nomura R, Naka S, Nakano K, Taniguchi N, Matsumoto M, Ooshima T: Detection of oral streptococci with collagen-binding properties in saliva specimens from mothers and their children. Int J Paediatr Dent 2010, 20(4):254-260.

126. Abranches J, Miller JH, Martinez AR, Simpson-Haidaris PJ, Burne RA, Lemos JA: The collagen-binding protein Cnm is required for Streptococcus mutans adherence to and intracellular invasion of human coronary artery endothelial cells. Infect Immun 2011, 79(6):2277-2284.

127. Abranches J, Zeng L, Belanger M, Rodrigues PH, Simpson-Haidaris PJ, Akin D, Dunn WA, Jr., Progulske-Fox A, Burne RA: Invasion of human coronary artery endothelial cells by Streptococcus mutans OMZ175. Oral Microbiol Immunol 2009, 24(2):141-145.

128. Guha S, Padh H: Cathepsins: fundamental effectors of endolysosomal proteolysis. Indian J Biochem Biophys 2008, 45(2):75-90.

129. Maxfield FR, Yamashiro DJ: Endosome acidification and the pathways of receptor-mediated endocytosis. Adv Exp Med Biol 1987, 225:189-198.

130. Huotari J, Helenius A: Endosome maturation. EMBO J 2011, 30(17):3481- 3500.

131. Matsui R, Cvitkovitch D: Acid tolerance mechanisms utilized by Streptococcus mutans. Future Microbiol 2010, 5(3):403-417.

132. Diment S, Stahl P: Macrophage endosomes contain proteases which degrade endocytosed protein ligands. J Biol Chem 1985, 260(28):15311- 15317.

133. Drangsholt MT: A new causal model of dental diseases associated with endocarditis. Ann Periodontol 1998, 3(1):184-196.

134. Giglio JA, Rowland RW, Dalton HP, Laskin DM: Suture removal-induced bacteremia: a possible endocarditis risk. J Am Dent Assoc 1992, 123(8):65- 66, 69-70.

135. Herzberg MC: Platelet-streptococcal interactions in endocarditis. Crit Rev Oral Biol Med 1996, 7(3):222-236.

176

136. Plummer C, Wu H, Kerrigan SW, Meade G, Cox D, Ian Douglas CW: A serine- rich glycoprotein of Streptococcus sanguis mediates adhesion to platelets via GPIb. Br J Haematol 2005, 129(1):101-109.

137. Nakano K, Tsuji M, Nishimura K, Nomura R, Ooshima T: Contribution of cell surface protein antigen PAc of Streptococcus mutans to bacteremia. Microbes Infect 2006, 8(1):114-121.

138. Nakano K, Nomura R, Nemoto H, Lapirattanakul J, Taniguchi N, Gronroos L, Alaluusua S, Ooshima T: Protein antigen in serotype k Streptococcus mutans clinical isolates. J Dent Res 2008, 87(10):964-968.

139. Hahn CL, Schenkein HA, Tew JG: Endocarditis-associated oral streptococci promote rapid differentiation of monocytes into mature dendritic cells. Infect Immun 2005, 73(8):5015-5021.

140. Engels-Deutsch M, Rizk S, Haikel Y: Streptococcus mutans antigen I/II binds to alpha5beta1 integrins via its A-domain and increases beta1 integrins expression on periodontal ligament fibroblast cells. Arch Oral Biol 2011, 56(1):22-28.

141. Qian H, Dao ML: Inactivation of the Streptococcus mutans wall-associated protein A gene (wapA) results in a decrease in sucrose-dependent adherence and aggregation. Infect Immun 1993, 61(12):5021-5028.

142. Love RM, McMillan MD, Jenkinson HF: Invasion of dentinal tubules by oral streptococci is associated with collagen recognition mediated by the antigen I/II family of polypeptides. Infect Immun 1997, 65(12):5157-5164.

143. Nomura R, Nakano K, Taniguchi N, Lapirattanakul J, Nemoto H, Gronroos L, Alaluusua S, Ooshima T: Molecular and clinical analyses of the gene encoding the collagen-binding adhesin of Streptococcus mutans. J Med Microbiol 2009, 58(Pt 4):469-475.

144. Engels-Deutsch M, Pini A, Yamashita Y, Shibata Y, Haikel Y, Scholler-Guinard M, Klein JP: Insertional inactivation of pac and rmlB genes reduces the release of tumor necrosis factor alpha, interleukin-6, and interleukin-8 induced by Streptococcus mutans in monocytic, dental pulp, and periodontal ligament cells. Infect Immun 2003, 71(9):5169-5177.

145. Matsumoto-Nakano M, Tsuji M, Inagaki S, Fujita K, Nagayama K, Nomura R, Ooshima T: Contribution of cell surface protein antigen c of Streptococcus mutans to platelet aggregation. Oral Microbiol Immunol 2009, 24(5):427-430.

146. Taniguchi N, Nakano K, Nomura R, Naka S, Kojima A, Matsumoto M, Ooshima T: Defect of glucosyltransferases reduces platelet aggregation activity of Streptococcus mutans: analysis of clinical strains isolated from oral cavities. Arch Oral Biol 2010, 55(6):410-416.

177

147. Chen Y-Y, Peng B, Yang Q, Glew MD, Veith PD, Cross KJ, Goldie KN, Chen D, O'Brien-Simpson N, Dashper SG et al: The outer membrane protein LptO is essential for the O-deacylation of LPS and the co-ordinated secretion and attachment of A-LPS and CTD proteins in Porphyromonas gingivalis. Mol Microbiol 2011, 79(5):1380-1401.

148. Hill EE, Herijgers P, Herregods MC, Peetermans WE: Evolving trends in infective endocarditis. Clin Microbiol Infect 2006, 12(1):5-12.

149. Roger VL, Go AS, Lloyd-Jones DM, Benjamin EJ, Berry JD, Borden WB, Bravata DM, Dai S, Ford ES, Fox CS et al: Heart disease and stroke statistics-2012 update: A report from the American Heart Association. Circulation 2012, 125(1):E2-E220.

150. Nakano K, Hokamura K, Taniguchi N, Wada K, Kudo C, Nomura R, Kojima A, Naka S, Muranaka Y, Thura M et al: The collagen-binding protein of Streptococcus mutans is involved in haemorrhagic stroke. Nat Commun 2011, 2:485.

151. Offermanns S: Activation of platelet function through G protein-coupled receptors. Circ Res 2006, 99(12):1293-1304.

152. Nakano K, Nemoto H, Nomura R, Inaba H, Yoshioka H, Taniguchi K, Amano A, Ooshima T: Detection of oral bacteria in cardiovascular specimens. Oral Microbiol Immunol 2009, 24(1):64-68.

153. Nakano K, Inaba H, Nomura R, Nemoto H, Takeda M, Yoshioka H, Matsue H, Takahashi T, Taniguchi K, Amano A et al: Detection of cariogenic Streptococcus mutans in extirpated heart valve and atheromatous plaque specimens. J Clin Microbiol 2006, 44(9):3313-3317.

154. Nakano K, Nemoto H, Nomura R, Homma H, Yoshioka H, Shudo Y, Hata H, Toda K, Taniguchi K, Amano A et al: Serotype distribution of Streptococcus mutans a pathogen of dental caries in cardiovascular specimens from Japanese patients. J Med Microbiol 2007, 56(Pt 4):551-556.

155. Roger VL, Go AS, Lloyd-Jones DM, Benjamin EJ, Berry JD, Borden WB, Bravata DM, Dai S, Ford ES, Fox CS et al: Heart disease and stroke statistics-2012 update: A report from the American Heart Association. Circulation 2012, 125(22):E1002-E1002.

156. Endemann DH, Schiffrin EL: Endothelial dysfunction. J Am Soc Nephrol 2004, 15(8):1983-1992.

157. Taddei S, Virdis A, Mattei P, Ghiadoni L, Sudano I, Salvetti A: Defective L- arginine-nitric oxide pathway in offspring of essential hypertensive patients. Circulation 1996, 94(6):1298-1303.

178

158. Celermajer DS, Sorensen KE, Gooch VM, Spiegelhalter DJ, Miller OI, Sullivan ID, Lloyd JK, Deanfield JE: Non-invasive detection of endothelial dysfunction in children and adults at risk of atherosclerosis. Lancet 1992, 340(8828):1111-1115.

159. Singh KV, Nallapareddy SR, Sillanpaa J, Murray BE: Importance of the collagen adhesin ace in pathogenesis and protection against Enterococcus faecalis experimental endocarditis. PLoS Pathog 2010, 6(1):e1000716.

160. Gibson FC, 3rd, Ukai T, Genco CA: Engagement of specific innate immune signaling pathways during Porphyromonas gingivalis induced chronic inflammation and atherosclerosis. Front Biosci 2008, 13:2041-2059.

161. Gibson FC, 3rd, Yumoto H, Takahashi Y, Chou HH, Genco CA: Innate immune signaling and Porphyromonas gingivalis-accelerated atherosclerosis. J Dent Res 2006, 85(2):106-121.

162. Madan M, Bishayi B, Hoge M, Messas E, Amar S: Doxycycline affects diet- and bacteria-associated atherosclerosis in an ApoE heterozygote murine model: cytokine profiling implications. Atherosclerosis 2007, 190(1):62-72.

163. Madan M, Bishayi B, Hoge M, Amar S: Atheroprotective role of interleukin-6 in diet- and/or pathogen-associated atherosclerosis using an ApoE heterozygote murine model. Atherosclerosis 2008, 197(2):504-514.

164. Madan M, Amar S: Toll-like receptor-2 mediates diet and/or pathogen associated atherosclerosis: proteomic findings. PLoS One 2008, 3(9):e3204.

165. Piroth L, Que YA, Widmer E, Panchaud A, Piu S, Entenza JM, Moreillon P: The fibrinogen- and fibronectin-binding domains of Staphylococcus aureus fibronectin-binding protein A synergistically promote endothelial invasion and experimental endocarditis. Infect Immun 2008, 76(8):3824-3831.

166. Rohde M, Muller E, Chhatwal GS, Talay SR: Host cell caveolae act as an entry-port for group A streptococci. Cell Microbiol 2003, 5(5):323-342.

167. Amelung S, Nerlich A, Rohde M, Spellerberg B, Cole JN, Nizet V, Chhatwal GS, Talay SR: The FbaB-type fibronectin-binding protein of Streptococcus pyogenes promotes specific invasion into endothelial cells. Cell Microbiol 2011.

168. Rohde M, Graham RM, Branitzki-Heinemann K, Borchers P, Preuss C, Schleicher I, Zahner D, Talay SR, Fulde M, Dinkla K et al: Differences in the aromatic domain of homologous streptococcal fibronectin-binding proteins trigger different cell invasion mechanisms and survival rates. Cell Microbiol 2011, 13(3):450-468.

179

169. Jenkinson HF, Lamont RJ: Oral microbial communities in sickness and in health. Trends Microbiol 2005, 13(12):589-595.

170. Tamilselvam B, Almeida RA, Dunlap JR, Oliver SP: Streptococcus uberis internalizes and persists in bovine mammary epithelial cells. Microb Pathog 2006, 40(6):279-285.

171. Courtney HS, Hasty DL, Dale JB: Molecular mechanisms of adhesion, colonization, and invasion of group A streptococci. Ann Med 2002, 34(2):77- 87.

172. Nitsche-Schmitz DP, Rohde M, Chhatwal GS: Invasion mechanisms of Gram- positive pathogenic cocci. Thromb Haemost 2007, 98(3):488-496.

173. Burnham CA, Shokoples SE, Tyrrell GJ: Invasion of HeLa cells by group B Streptococcus requires the phosphoinositide-3-kinase signalling pathway and modulates phosphorylation of host-cell Akt and glycogen synthase kinase-3. Microbiology 2007, 153(Pt 12):4240-4252.

174. Ring A, Tuomanen E: Host cell invasion by Streptococcus pneumoniae. Subcell Biochem 2000, 33:125-135.

175. Benga L, Goethe R, Rohde M, Valentin-Weigand P: Non-encapsulated strains reveal novel insights in invasion and survival of Streptococcus suis in epithelial cells. Cell Microbiol 2004, 6(9):867-881.

176. Nobbs AH, Shearer BH, Drobni M, Jepson MA, Jenkinson HF: Adherence and internalization of Streptococcus gordonii by epithelial cells involves beta1 integrin recognition by SspA and SspB (antigen I/II family) polypeptides. Cell Microbiol 2007, 9(1):65-83.

177. Stinson MW, Alder S, Kumar S: Invasion and killing of human endothelial cells by viridans group streptococci. Infect Immun 2003, 71(5):2365-2372.

178. Stinson MW, Barua PK, Bergey EJ, Nisengard RJ, Neiders ME, Albini B: Binding of Streptococcus mutans antigens to heart and kidney basement membranes. Infect Immun 1984, 46(1):145-151.

179. Tortora G. J. DBH: Principles of Anatomy and Physiology, 7 edn. New York, NY: HarperCollins; 1993.

180. Rostand KS, Esko JD: Microbial adherence to and invasion through proteoglycans. Infect Immun 1997, 65(1):1-8.

181. Reitsma S, Slaaf DW, Vink H, van Zandvoort MA, oude Egbrink MG: The endothelial glycocalyx: composition, functions, and visualization. Eur J Physiol 2007, 454(3):345-359.

180

182. Law DA, Nannizzi-Alaimo L, Phillips DR: Outside-in integrin signal transduction. Alpha IIb beta 3-(GP IIb IIIa) tyrosine phosphorylation induced by platelet aggregation. J Biol Chem 1996, 271(18):10811-10815.

183. Scibelli A, Roperto S, Manna L, Pavone LM, Tafuri S, Della Morte R, Staiano N: Engagement of integrins as a cellular route of invasion by bacterial pathogens. Vet J 2007, 173(3):482-491.

184. Conforti G, Dominguez-Jimenez C, Zanetti A, Gimbrone MA, Jr., Cremona O, Marchisio PC, Dejana E: Human endothelial cells express integrin receptors on the luminal aspect of their membrane. Blood 1992, 80(2):437-446.

185. Yang JJ, Chen YM, Kurokawa T, Gong JP, Onodera S, Yasuda K: Gene expression, glycocalyx assay, and surface properties of human endothelial cells cultured on hydrogel matrix with sulfonic moiety: Effect of elasticity of hydrogel. J Biomed Mater Res 2010, 95(2):531-542.

186. Petersen FC, Assev S, van der Mei HC, Busscher HJ, Scheie AA: Functional variation of the antigen I/II surface protein in Streptococcus mutans and Streptococcus intermedius. Infect Immun 2002, 70(1):249-256.

187. Ridker PM: On evolutionary biology, inflammation, infection, and the causes of atherosclerosis. Circulation 2002, 105(1):2-4.

188. Bartruff JB, Yukna RA, Layman DL: Outer membrane vesicles from Porphyromonas gingivalis affect the growth and function of cultured human gingival fibroblasts and umbilical vein endothelial cells. J Periodontol 2005, 76(6):972-979.

189. Al-Okla S, Chatenay-Rivauday C, Klein JP, Wachsmann D: Involvement of alpha5beta1 integrins in interleukin 8 production induced by oral viridans streptococcal protein I/IIf in cultured endothelial cells. Cell Microbiol 1999, 1(2):157-168.

190. Nomura R, Naka S, Nemoto H, Inagaki S, Taniguchi K, Ooshima T, Nakano K: Potential involvement of collagen-binding proteins of Streptococcus mutans in infective endocarditis. Oral Dis 2013, 19(4):387-393.

191. Almeida RA, Oliver SP: Trafficking of Streptococcus uberis in bovine mammary epithelial cells. Microb Pathog 2006, 41(2-3):80-89.

192. Almeida RA, Dunlap JR, Oliver SP: Binding of Host Factors Influences Internalization and Intracellular Trafficking of Streptococcus uberis in Bovine Mammary Epithelial Cells. Vet Med Int 2010, 2010:319192.

193. Nakagawa I, Amano A, Mizushima N, Yamamoto A, Yamaguchi H, Kamimoto T, Nara A, Funao J, Nakata M, Tsuda K et al: Autophagy defends cells against invading group A Streptococcus. Science 2004, 306(5698):1037-1040.

181

194. Huang YT, Teng LJ, Ho SW, Hsueh PR: Streptococcus suis infection. J Microbiol Immunol Infect 2005, 38(5):306-313.

195. Gottschalk M, Segura M, Xu J: Streptococcus suis infections in humans: the Chinese experience and the situation in North America. Anim Health Res Rev 2007, 8(1):29-45.

196. Fisher K, Phillips C: The ecology, epidemiology and virulence of Enterococcus. Microbiology 2009, 155(Pt 6):1749-1757.

197. Koch S, Hufnagel M, Theilacker C, Huebner J: Enterococcal infections: host response, therapeutic, and prophylactic possibilities. Vaccine 2004, 22(7):822-830.

198. Nallapareddy SR, Singh KV, Duh RW, Weinstock GM, Murray BE: Diversity of ace, a gene encoding a microbial surface component recognizing adhesive matrix molecules, from different strains of Enterococcus faecalis and evidence for production of ace during human infections. Infect Immun 2000, 68(9):5210-5217.

199. Ryding U, Flock JI, Flock M, Soderquist B, Christensson B: Expression of collagen-binding protein and types 5 and 8 capsular polysaccharide in clinical isolates of Staphylococcus aureus. J Infect Dis 1997, 176(4):1096- 1099.

200. Hienz SA, Schennings T, Heimdahl A, Flock JI: Collagen binding of Staphylococcus aureus is a virulence factor in experimental endocarditis. J Infect Dis 1996, 174(1):83-88.

201. Heart Disease and Stroke Prevention: Addressing the Nation's Leading Killers: At A Glance 2011 [http://www.cdc.gov/chronicdisease/resources/ publications/aag/dhdsp.htm]

202. Centers for Disease Control and Prevention: Million hearts: strategies to reduce the prevalence of leading cardiovascular disease risk factors-- United States, 2011. MMWR 2011, 60(36):1248-1251.

203. Eke PI, Dye BA, Wei L, Thornton-Evans GO, Genco RJ, Cdc Periodontal Disease Surveillance workgroup: James Beck GDRP: Prevalence of periodontitis in adults in the United States: 2009 and 2010. J Dent Res 2012, 91(10):914-920.

204. Parahitiyawa NB, Jin LJ, Leung WK, Yam WC, Samaranayake LP: Microbiology of odontogenic bacteremia: beyond endocarditis. Clin Microbiol Rev 2009, 22(1):46-64.

182

205. Lockhart PB, Brennan MT, Sasser HC, Fox PC, Paster BJ, Bahrani-Mougeot FK: Bacteremia associated with toothbrushing and dental extraction. Circulation 2008, 117(24):3118-3125.

206. Berlutti F, Catizone A, Ricci G, Frioni A, Natalizi T, Valenti P, Polimeni A: Streptococcus mutans and Streptococcus sobrinus are able to adhere and invade human gingival fibroblast cell line. Int J Immunopathol Pharmacol 2010, 23(4):1253-1260.

207. Nakano K, Nomura R, Nemoto H, Mukai T, Yoshioka H, Shudo Y, Hata H, Toda K, Taniguchi K, Amano A et al: Detection of novel serotype k Streptococcus mutans in infective endocarditis patients. J Med Microbiol 2007, 56(Pt 10):1413-1415.

208. Nakano K, Lapirattanakul J, Nomura R, Nemoto H, Alaluusua S, Gronroos L, Vaara M, Hamada S, Ooshima T, Nakagawa I: Streptococcus mutans clonal variation revealed by multilocus sequence typing. J Clin Microbiol 2007, 45(8):2616-2625.

209. Panagia M, Winston B, Click JW, Sabatine MS, Resnic FS: A rare complication of infective endocarditis. Circulation 2012, 125(10):1316-1317.

210. Gdoura R, Pereyre S, Frikha L, Hammami N, Clerc M, Sahnoun Y, Bebear C, Daoud M, de Barbeyrac B, Hammami A: Culture-negative endocarditis due to Chlamydia pneumoniae. J Clin Microbiol 2002, 40(2):718-720.

211. Gaydos CA, Summersgill JT, Sahney NN, Ramirez JA, Quinn TC: Replication of Chlamydia pneumoniae in vitro in human macrophages, endothelial cells, and aortic artery smooth muscle cells. Infect Immun 1996, 64(5):1614-1620.

212. Rosenshine I, Ruschkowski S, Finlay BB: Inhibitors of cytoskeletal function and signal transduction to study bacterial invasion. Methods Enzymol 1994, 236:467-476.

213. Vosbeck K, James PR, Zimmermann W: Antibiotic action on phagocytosed bacteria measured by a new method for determining viable bacteria. Antimicrob Agents Ch 1984, 25(6):735-741.

214. Tang P, Foubister V, Pucciarelli MG, Finlay BB: Methods to study bacterial invasion. J Microbiol Meth 1993, 18(3):227-240.

215. Wang ZY, Wang JQ, Zhou Y, Zhao D, Xiao B: Quantitative detection of Streptococcus mutans and bacteria of dental caries and no caries groups in permanent teeth from a north China population. Chin Med J 2012, 125(21):3880-3884.

183

216. Hasegawa Y, Mans JJ, Mao S, Lopez MC, Baker HV, Handfield M, Lamont RJ: Gingival epithelial cell transcriptional responses to commensal and opportunistic oral microbial species. Infect Immun 2007, 75(5):2540-2547.

217. Eisen MB, Spellman PT, Brown PO, Botstein D: Cluster analysis and display of genome-wide expression patterns. Proc Natl Acad Sci U S A 1998, 95(25):14863-14868.

218. Tonetti MS: Molecular factors associated with compartmentalization of gingival immune responses and transepithelial neutrophil migration. J Periodontal Res 1997, 32(1 Pt 2):104-109.

219. Uitto VJ: Degradation of basement membrane collagen by proteinases from human gingiva, leukocytes and bacterial plaque. J Periodontol 1983, 54(12):740-745.

220. Peng TK, Nisengard RJ, Levine MJ: The alteration in gingival basement membrane antigens in chronic periodontitis. J Periodontol 1986, 57(1):20-24.

221. Sugiyama E, Baehni P, Cimasoni G: An in vitro study of polymorphonuclear leucocyte-mediated injury to human gingival keratinocytes by periodontopathic bacterial extracts. Arch Oral Biol 1992, 37(12):1007-1012.

222. Krasse B: The proportional distribution of different types of streptococci in saliva and plaque material. Odontol Revy 1953, 4(4):304-312.

223. Hamada S, Masuda N, Ooshima T, Sobue S, Kotani S: Epidemiological survey of Streptococcus mutans among Japanese children. Identification and serological typing of the isolated strains. Jpn J Microbiol 1976, 20(1):33-44.

224. Malicka E, Sitko R, Zawisza B, Heimann J, Kajewski D, Kita A: Nondestructive analysis of single crystals of selenide spinels by X-ray spectrometry techniques. Anal Bioanal Chem 2011, 399(9):3285-3292.

225. Rudney JD, Chen R: The vital status of human buccal epithelial cells and the bacteria associated with them. Arch Oral Biol 2006, 51(4):291-298.

226. Rudney JD, Chen R, Zhang G: Streptococci dominate the diverse flora within buccal cells. J Dent Res 2005, 84(12):1165-1171.

227. Liljemark WF, Gibbons RJ: Proportional distribution and relative adherence of Streptococcus miteor (mitis) on various surfaces in the human oral cavity. Infect Immun 1972, 6(5):852-859.

228. Milgrom P, Riedy CA, Weinstein P, Tanner AC, Manibusan L, Bruss J: Dental caries and its relationship to bacterial infection, hypoplasia, diet, and oral hygiene in 6- to 36-month-old children. Commun Dent Oral Epidemiol 2000, 28(4):295-306.

184

229. Wan AK, Seow WK, Purdie DM, Bird PS, Walsh LJ, Tudehope DI: Oral colonization of Streptococcus mutans in six-month-old predentate infants. J Dent Res 2001, 80(12):2060-2065.

230. Tankkunnasombut S, Youcharoen K, Wisuttisak W, Vichayanrat S, Tiranathanagul S: Early colonization of mutans streptococci in 2- to 36- month-old Thai children. Pediatr Dent 2009, 31(1):47-51.

231. Reyes L, Herrera D, Kozarov E, Rolda S, Progulske-Fox A: Periodontal bacterial invasion and infection: contribution to atherosclerotic pathology. J Periodontol 2013, 84(4 Suppl):S30-50.

232. Molinari G, Talay SR, Valentin-Weigand P, Rohde M, Chhatwal GS: The fibronectin-binding protein of Streptococcus pyogenes, SfbI, is involved in the internalization of group A streptococci by epithelial cells. Infect Immun 1997, 65(4):1357-1363.

233. Kaur SJ, Nerlich A, Bergmann S, Rohde M, Fulde M, Zahner D, Hanski E, Zinkernagel A, Nizet V, Chhatwal GS et al: The CXC chemokine-degrading protease SpyCep of Streptococcus pyogenes promotes its uptake into endothelial cells. J Biol Chem 2010, 285(36):27798-27805.

234. Amelung S, Nerlich A, Rohde M, Spellerberg B, Cole JN, Nizet V, Chhatwal GS, Talay SR: The FbaB-type fibronectin-binding protein of Streptococcus pyogenes promotes specific invasion into endothelial cells. Cell Microbiol 2011, 13(8):1200-1211.

235. Schmittgen TD, Livak KJ: Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc 2008, 3(6):1101-1108.

236. Vita JA: Endothelial function. Circulation 2011, 124(25):e906-912.

237. Deanfield JE, Halcox JP, Rabelink TJ: Endothelial function and dysfunction: testing and clinical relevance. Circulation 2007, 115(10):1285-1295.

238. Lerman A, Zeiher AM: Endothelial function: Cardiac events. Circulation 2005, 111(3):363-368.

239. Ross R: Atherosclerosis-an inflammatory disease. New Engl J Med 1999, 340(2):115-126.

240. Glass CK, Witztum JL: Atherosclerosis. The road ahead. Cell 2001, 104(4):503-516.

241. Barlic J, Murphy PM: Chemokine regulation of atherosclerosis. J Leukoc Biol 2007, 82(2):226-236.

185

242. Kraaijeveld AO, de Jager SC, van Berkel TJ, Biessen EA, Jukema JW: Chemokines and atherosclerotic plaque progression: towards therapeutic targeting? Curr Pharm Des 2007, 13(10):1039-1052.

243. Tedgui A, Mallat Z: Cytokines in atherosclerosis: pathogenic and regulatory pathways. Physiol Rev 2006, 86(2):515-581.

244. Braunersreuther V, Mach F: Leukocyte recruitment in atherosclerosis: potential targets for therapeutic approaches? Cell Mol Life Sci 2006, 63(18):2079-2088.

245. Rao RM, Yang L, Garcia-Cardena G, Luscinskas FW: Endothelial-dependent mechanisms of leukocyte recruitment to the vascular wall. Circ Res 2007, 101(3):234-247.

246. Blankenberg S, Barbaux S, Tiret L: Adhesion molecules and atherosclerosis. Atherosclerosis 2003, 170(2):191-203.

247. Hansson GK, Libby P: The immune response in atherosclerosis: A double- edged sword. Nat Rev Immunol 2006, 6(7):508-519.

248. Abiko Y: Passive immunization against dental caries and periodontal disease: Development of recombinant and human monoclonal antibodies. Crit Rev Oral Biol Med 2000, 11(2):140-158.

249. Hoge M, Amar S: Role of interleukin-1 in bacterial atherogenesis. Drugs Today 2006, 42(10):683-688.

250. Keller TT, Mairuhu AT, de Kruif MD, Klein SK, Gerdes VE, ten Cate H, Brandjes DP, Levi M, van Gorp EC: Infections and endothelial cells. Cardiovasc Res 2003, 60(1):40-48.

251. Parent C, Eichacker PQ: Neutrophil and endothelial cell interactions in sepsis. The role of adhesion molecules. Infect Dis Clin North Am 1999, 13(2):427-447, x.

252. Vallet B, Wiel E: Endothelial cell dysfunction and coagulation. Crit Care Med 2001, 29(7 Suppl):S36-41.

253. Vernier A, Diab M, Soell M, Haan-Archipoff G, Beretz A, Wachsmann D, Klein JP: Cytokine production by human epithelial and endothelial cells following exposure to oral viridans streptococci involves lectin interactions between bacteria and cell surface receptors. Infect Immun 1996, 64(8):3016-3022.

254. Vernier-Georgenthum A, al-Okla S, Gourieux B, Klein JP, Wachsmann D: Protein I/II of oral viridans streptococci increases expression of adhesion molecules on endothelial cells and promotes transendothelial migration of neutrophils in vitro. Cell Immunol 1998, 187(2):145-150.

186

255. Hajishengallis G, Sharma A, Russell MW, Genco RJ: Interactions of oral pathogens with toll-like receptors: possible role in atherosclerosis. Ann Periodontol 2002, 7(1):72-78.

256. Soell M, Diab M, Haan-Archipoff G, Beretz A, Herbelin C, Poutrel B, Klein JP: Capsular polysaccharide types 5 and 8 of Staphylococcus aureus bind specifically to human epithelial (KB) cells, endothelial cells, and monocytes and induce release of cytokines. Infect Immun 1995, 63(4):1380-1386.

257. Soell M, Holveck F, Scholler M, Wachsmann RD, Klein JP: Binding of Streptococcus mutans SR protein to human monocytes: production of tumor necrosis factor, interleukin 1, and interleukin 6. Infect Immun 1994, 62(5):1805-1812.

258. Katsargyris A, Theocharis SE, Tsiodras S, Giaginis K, Bastounis E, Klonaris C: Enhanced TLR4 endothelial cell immunohistochemical expression in symptomatic carotid atherosclerotic plaques. Expert Opin Ther Targets 2010, 14(1):1-10.

259. Schoneveld AH, Hoefer I, Sluijter JP, Laman JD, de Kleijn DP, Pasterkamp G: Atherosclerotic lesion development and toll like receptor 2 and 4 responsiveness. Atherosclerosis 2008, 197(1):95-104.

260. Edfeldt K, Swedenborg J, Hansson GK, Yan ZQ: Expression of toll-like receptors in human atherosclerotic lesions: a possible pathway for plaque activation. Circulation 2002, 105(10):1158-1161.

261. den Dekker WK, Cheng C, Pasterkamp G, Duckers HJ: Toll like receptor 4 in atherosclerosis and plaque destabilization. Atherosclerosis 2010, 209(2):314- 320.

262. Nagata E, de Toledo A, Oho T: Invasion of human aortic endothelial cells by oral viridans group streptococci and induction of inflammatory cytokine production. Mol Oral Microbiol 2011, 26(1):78-88.

263. Tieu BC, Lee C, Sun H, Lejeune W, Recinos A, 3rd, Ju X, Spratt H, Guo DC, Milewicz D, Tilton RG et al: An adventitial IL-6/MCP1 amplification loop accelerates macrophage-mediated vascular inflammation leading to aortic dissection in mice. J Clin Invest 2009, 119(12):3637-3651.

264. Best PJ, Hasdai D, Sangiorgi G, Schwartz RS, Holmes DR, Jr., Simari RD, Lerman A: Apoptosis : Basic concepts and implications in coronary artery disease. Arterioscler Thromb Vasc Biol 1999, 19(1):14-22.

265. Geng YJ, Libby P: Evidence for apoptosis in advanced human atheroma. Colocalization with interleukin-1 beta-converting enzyme. Am J Pathol 1995, 147(2):251-266.

187

266. Han DK, Haudenschild CC, Hong MK, Tinkle BT, Leon MB, Liau G: Evidence for apoptosis in human atherogenesis and in a rat vascular injury model. Am J Pathol 1995, 147(2):267-277.

267. Gao L, Abu Kwaik Y: Hijacking of apoptotic pathwaysby bacterial pathogens. Microbes Infect 2000, 2(14):1705-1719.

268. Handfield M, Mans JJ, Zheng G, Lopez MC, Mao S, Progulske-Fox A, Narasimhan G, Baker HV, Lamont RJ: Distinct transcriptional profiles characterize oral epithelium-microbiota interactions. Cell Microbiol 2005, 7(6):811-823.

269. Klenk M, Nakata M, Podbielski A, Skupin B, Schroten H, Kreikemeyer B: Streptococcus pyogenes serotype-dependent and independent changes in infected HEp-2 epithelial cells. Isme J 2007, 1(8):678-692.

270. Nakhjiri SF, Park Y, Yilmaz O, Chung WO, Watanabe K, El-Sabaeny A, Park K, Lamont RJ: Inhibition of epithelial cell apoptosis by Porphyromonas gingivalis. FEMS Microbiol Lett 2001, 200(2):145-149.

271. DeLeo FR: Modulation of phagocyte apoptosis by bacterial pathogens. Apoptosis 2004, 9(4):399-413.

272. Weinrauch Y, Zychlinsky A: The induction of apoptosis by bacterial pathogens. Annu Rev Microbiol 1999, 53:155-187.

273. Pollanen MT, Salonen JI, Grenier D, Uitto VJ: Epithelial cell response to challenge of bacterial lipoteichoic acids and lipopolysaccharides in vitro. J Med Microbiol 2000, 49(3):245-252.

274. Wang PL, Shirasu S, Daito M, Ohura K: Streptococcus mutans lipoteichoic acid-induced apoptosis in cultured dental pulp cells from human deciduous teeth. Biochem Biophys Res Commun 2001, 281(4):957-961.

275. RT² Profiler™ PCR Array Data Analysis version 3.5 [http://www.sabiosciences.com/pcrarraydataanalysis.php]

276. Vermes I, Haanen C, Steffens-Nakken H, Reutelingsperger C: A novel assay for apoptosis. Flow cytometric detection of phosphatidylserine expression on early apoptotic cells using fluorescein labelled annexin V. J Immunol Methods 1995, 184(1):39-51.

277. Schneider CA, Rasband WS, Eliceiri KW: NIH Image to ImageJ: 25 years of image analysis. Nat Methods 2012, 9(7):671-675.

278. ImageJ [http://imagej.nih.gov/ij/]

188

279. Grando Mattuella L, Westphalen Bento L, de Figueiredo JA, Nor JE, de Araujo FB, Fossati AC: Vascular endothelial growth factor and its relationship with the dental pulp. J Endod 2007, 33(5):524-530.

280. Roberts-Clark DJ, Smith AJ: Angiogenic growth factors in human dentine matrix. Arch Oral Biol 2000, 45(11):1013-1016.

281. Telles PD, Hanks CT, Machado MA, Nor JE: Lipoteichoic acid up-regulates VEGF expression in macrophages and pulp cells. J Dent Res 2003, 82(6):466-470.

282. Nobes CD, Lauritzen I, Mattei MG, Paris S, Hall A, Chardin P: A new member of the Rho family, Rnd1, promotes disassembly of actin filament structures and loss of cell adhesion. J Cell Biol 1998, 141(1):187-197.

283. Wennerberg K, Forget MA, Ellerbroek SM, Arthur WT, Burridge K, Settleman J, Der CJ, Hansen SH: Rnd proteins function as RhoA antagonists by activating p190 RhoGAP. Curr Biol 2003, 13(13):1106-1115.

284. Purushothaman SS, Wang B, Cleary PP: M1 protein triggers a phosphoinositide cascade for group A Streptococcus invasion of epithelial cells. Infect Immun 2003, 71(10):5823-5830.

285. Nozawa T, Aikawa C, Goda A, Maruyama F, Hamada S, Nakagawa I: The small Rab9A and Rab23 function at distinct steps in autophagy during Group A Streptococcus infection. Cell Microbiol 2012, 14(8):1149-1165.

286. de Toledo A, Nagata E, Yoshida Y, Oho T: Streptococcus oralis coaggregation receptor polysaccharides induce inflammatory responses in human aortic endothelial cells. Mol Oral Microbiol 2012, 27(4):295-307.

287. Jiang Y, Russell TR, Schilder H, Graves DT: Endodontic pathogens stimulate monocyte chemoattractant protein-1 and interleukin-8 in mononuclear cells. J Endod 1998, 24(2):86-90.

288. Jiang Y, Magli L, Russo M: Bacterium-dependent induction of cytokines in mononuclear cells and their pathologic consequences in vivo. Infect Immun 1999, 67(5):2125-2130.

289. Jiang Y, Schilder H: An optimal host response to a bacterium may require the interaction of leukocytes and resident host cells. J Endod 2002, 28(4):279-282.

290. Qi M, Miyakawa H, Kuramitsu HK: Porphyromonas gingivalis induces murine macrophage foam cell formation. Microb Pathog 2003, 35(6):259-267.

291. Charo IF, Taubman MB: Chemokines in the pathogenesis of vascular disease. Circ Res 2004, 95(9):858-866.

189

292. Choi I, Jo G, Kim S, Jung C, Kim Y, Shin K: Stimulation of various functions in murine peritoneal macrophages by glucans produced by glucosyltransferases from Streptococcus mutans. Biosci Biotechnol Biochem 2005, 69(9):1693-1699.

293. Chia JS, Lien HT, Hsueh PR, Chen PM, Sun A, Chen JY: Induction of cytokines by glucosyltransferases of Streptococcus mutans. Clin Diagn Lab Immunol 2002, 9(4):892-897.

294. Calvayrac O, Rodriguez-Calvo R, Alonso J, Orbe J, Martin-Ventura JL, Guadall A, Gentile M, Juan-Babot O, Egido J, Beloqui O et al: CCL20 is increased in hypercholesterolemic subjects and is upregulated by LDL in vascular smooth muscle cells: role of NF-kappaB. Arterioscler Thromb Vasc Biol 2011, 31(11):2733-2741.

295. Wan W, Lim JK, Lionakis MS, Rivollier A, McDermott DH, Kelsall BL, Farber JM, Murphy PM: Genetic deletion of chemokine receptor Ccr6 decreases atherogenesis in ApoE-deficient mice. Circ Res 2011, 109(4):374-381.

296. Castillo L, Rohatgi A, Ayers CR, Owens AW, Das SR, Khera A, McGuire DK, de Lemos JA: Associations of four circulating chemokines with multiple atherosclerosis phenotypes in a large population-based sample: results from the Dallas Heart Study. J Interferon Cytokine Res 2010, 30(5):339-347.

297. McPherson R, Davies RW: Inflammation and coronary artery disease: insights from genetic studies. Can J Cardiol 2012, 28(6):662-666.

298. Munoz N, Van Maele L, Marques JM, Rial A, Sirard JC, Chabalgoity JA: Mucosal administration of flagellin protects mice from Streptococcus pneumoniae lung infection. Infect Immun 2010, 78(10):4226-4233.

299. Massena S, Christoffersson G, Hjertstrom E, Zcharia E, Vlodavsky I, Ausmees N, Rolny C, Li JP, Phillipson M: A chemotactic gradient sequestered on endothelial heparan sulfate induces directional intraluminal crawling of neutrophils. Blood 2010, 116(11):1924-1931.

300. Soehnlein O: Multiple roles for neutrophils in atherosclerosis. Circ Res 2012, 110(6):875-888.

301. Martin-Fuentes P, Civeira F, Solanas-Barca M, Garcia-Otin AL, Jarauta E, Cenarro A: Overexpression of the CXCL3 gene in response to oxidized low- density lipoprotein is associated with the presence of tendon xanthomas in familial hypercholesterolemia. Biochem Cell Biol 2009, 87(3):493-498.

302. Beckert S, Farrahi F, Aslam RS, Scheuenstuhl H, Konigsrainer A, Hussain MZ, Hunt TK: Lactate stimulates endothelial cell migration. Wound Repair Regen 2006, 14(3):321-324.

190

303. Lamalice L, Le Boeuf F, Huot J: Endothelial cell migration during angiogenesis. Circ Res 2007, 100(6):782-794.

304. Hunt TK, Aslam RS, Beckert S, Wagner S, Ghani QP, Hussain MZ, Roy S, Sen CK: Aerobically derived lactate stimulates revascularization and tissue repair via redox mechanisms. Antioxid Redox Signal 2007, 9(8):1115-1124.

305. Walenta S, Wetterling M, Lehrke M, Schwickert G, Sundfor K, Rofstad EK, Mueller-Klieser W: High lactate levels predict likelihood of metastases, tumor recurrence, and restricted patient survival in human cervical cancers. Cancer Res 2000, 60(4):916-921.

306. Bonuccelli G, Tsirigos A, Whitaker-Menezes D, Pavlides S, Pestell RG, Chiavarina B, Frank PG, Flomenberg N, Howell A, Martinez-Outschoorn UE et al: Ketones and lactate "fuel" tumor growth and metastasis: Evidence that epithelial cancer cells use oxidative mitochondrial metabolism. Cell Cycle 2010, 9(17):3506-3514.

307. Zeisel MB, Druet VA, Sibilia J, Klein JP, Quesniaux V, Wachsmann D: Cross talk between MyD88 and focal adhesion kinase pathways. J Immunol 2005, 174(11):7393-7397.

308. Attar MA, Bailie MB, Christensen PJ, Brock TG, Wilcoxen SE, Paine R, 3rd: Induction of ICAM-1 expression on alveolar epithelial cells during lung development in rats and humans. Exp Lung Res 1999, 25(3):245-259.

309. Huang GT, Eckmann L, Savidge TC, Kagnoff MF: Infection of human intestinal epithelial cells with invasive bacteria upregulates apical intercellular adhesion molecule-1 (ICAM)-1) expression and neutrophil adhesion. J Clin Invest 1996, 98(2):572-583.

310. Consortium TU: Reorganizing the protein space at the Universal Protein Resource (UniProt). Nucleic Acids Res 2012, 40(D1):D71-D75.

311. McEver RP: Selectins: lectins that initiate cell adhesion under flow. Curr Opin Cell Biol 2002, 14(5):581-586.

312. Millan MT, Geczy C, Stuhlmeier KM, Goodman DJ, Ferran C, Bach FH: Human monocytes activate porcine endothelial cells, resulting in increased E- selectin, interleukin-8, monocyte chemotactic protein-1, and plasminogen activator inhibitor-type-1 expression. Transplantation 1997, 63(3):421-429.

313. Metcalf DJ, Nightingale TD, Zenner HL, Lui-Roberts WW, Cutler DF: Formation and function of Weibel-Palade bodies. J Cell Sci 2008, 121(Pt 1):19-27.

314. Chen L, Wang S, Zhou Y, Wu X, Entin I, Epstein J, Yaccoby S, Xiong W, Barlogie B, Shaughnessy JD, Jr. et al: Identification of early growth response

191

protein 1 (EGR-1) as a novel target for JUN-induced apoptosis in multiple myeloma. Blood 2010, 115(1):61-70.

315. Resto-Ruiz SI, Schmiederer M, Sweger D, Newton C, Klein TW, Friedman H, Anderson BE: Induction of a potential paracrine angiogenic loop between human THP-1 macrophages and human microvascular endothelial cells during Bartonella henselae infection. Infect Immun 2002, 70(8):4564-4570.

316. Carratelli CR, Paolillo R, Rizzo A: Chlamydia pneumoniae stimulates the proliferation of HUVEC through the induction of VEGF by THP-1. Int Immunopharmacol 2007, 7(3):287-294.

317. The GeneCards Human Gene Database [http://www.genecards.org/]

318. UniProt Knowledgebase [http://www.uniprot.org/]

319. Edwardsson S, Mejare B: Streptococcus milleri (Guthof) and Streptococcus mutans in the mouths of infants before and after tooth eruption. Arch Oral Biol 1978, 23(9):811-814.

320. Nakano K, Nomura R, Nemoto H, al. e: Detection of novel serotype k Streptococcus mutans in infective endocarditis patients. J Med Microbiol 2007(56):1413-1415.

321. Zhang T, Kurita-Ochiai T, Hashizume T, Du Y, Oguchi S, Yamamoto M: Aggregatibacter actinomycetemcomitans accelerates atherosclerosis with an increase in atherogenic factors in spontaneously hyperlipidemic mice. FEMS Immunol Med Microbiol 2010, 59(2):143-151.

322. Seki E, De Minicis S, Osterreicher CH, Kluwe J, Osawa Y, Brenner DA, Schwabe RF: TLR4 enhances TGF-beta signaling and hepatic fibrosis. Nat Med 2007, 13(11):1324-1332.

323. Linton MF, Babaev VR, Gleaves LA, Fazio S: A direct role for the macrophage low density lipoprotein receptor in atherosclerotic lesion formation. J Biol Chem 1999, 274(27):19204-19210.

324. Chen Y, Ruan XZ, Li Q, Huang A, Moorhead JF, Powis SH, Varghese Z: Inflammatory cytokines disrupt LDL-receptor feedback regulation and cause statin resistance: a comparative study in human hepatic cells and mesangial cells. Am J Physiol-Renal 2007, 293(3):F680-687.

325. Greaves DR, Gordon S: The macrophage scavenger receptor at 30 years of age: current knowledge and future challenges. J Lipid Res 2009, 50 Suppl:S282-286.

326. Vanhoutte PM: Endothelial dysfunction and atherosclerosis. Eur Heart J 1997, 18 Suppl E:E19-29.

192

327. Libby P: Changing concepts of atherogenesis. J Intern Med 2000, 247(3):349-358.

328. Go AS, Mozaffarian D, Roger VL, Benjamin EJ, Berry JD, Borden WB, Bravata DM, Dai S, Ford ES, Fox CS, Franco S, Fullerton HJ, Gillespie C; et al. Heart disease and stroke statistics-2013 update: A report from the American Heart Association. Circulation 2013, 127(1):E6-E245.

329. Casadevall A: Evolution of intracellular pathogens. Annu Rev Microbiol 2008, 62:19-33.

330. Wittrup A, Zhang SH, Svensson KJ, Kucharzewska P, Johansson MC, Morgelin M, Belting M: Magnetic nanoparticle-based isolation of endocytic vesicles reveals a role of the heat shock protein GRP75 in macromolecular delivery. Proc Natl Acad Sci U S A 2010, 107(30):13342-13347.

331. Wittrup A, Zhang SH, ten Dam GB, van Kuppevelt TH, Bengtson P, Johansson M, Welch J, Morgelin M, Belting M: ScFv antibody-induced translocation of cell-surface heparan sulfate proteoglycan to endocytic vesicles: evidence for heparan sulfate epitope specificity and role of both syndecan and glypican. J Biol Chem 2009, 284(47):32959-32967.

193

BIOGRAPHICAL SKETCH

Edith M. Sampson is a United States Army Signal Corps veteran with training in communications and information systems. After her military service, Edith graduated with an Associate of Arts (A. A.) degree summa cum laude from Daytona Beach

Community College 1996. After receiving an A. A. degree, she transferred to the

University of Florida and completed a Bachelor of Science degree through the

Microbiology and Cell Science (MCS) Department graduating cum laude. In 2004, Edith received a M. S. degree in MCS in the College of Agriculture at UF under the mentorship of Thomas Bobik. Her graduate project was to complete a genetic characterization of the twenty one genes of the propanediol utilization (pdu) operon in

Salmonella enterica. To complete this assignment, she was trained in bacterial genetics, molecular biology, chromatography, and transmission electron microscopy.

While working on this project, discoveries were made that led to a better understanding of Salmonella microcompartment protein composition and function and cobalamin

(vitamin B12) reduction and activation during 1,2-propanediol metabolism.

After leaving the microbiology department, Edith accepted a senior biological scientist position to pursue medical science research with Dr. Gregory Schultz, director and professor of the Wound Institute in the Department of Obstetrics and Gynecology at

UF. While in this lab, she was cross trained in a variety of methodologies including eukaryotic cell culture (integument and eye tissues), monoclonal antibody production, gene therapies (antisense oligos, siRNA and ribozymes), enzyme linked immunosorbant assays, collagenase assays, zymography, eukaryotic molecular biology, microarray technology, and immunohistochemistry (IHC). The duties of this position required experimental support of many collaborative projects including NEI and 194

industry funded research. When the opportunity came, she accepted a position as research manager in 2006 with Patrick Antonelli, Professor and Chairman of the

Department of Otolaryngology (ENT).

As research manager for ENT, Edith was responsible for designing and directing hypothesis driven research while ensuring timely completion of experimental objectives to accomplish overall project goals. As a researcher, she developed methods for growing and assessing staphylococcal, pseudomonal, and streptococcal biofilms on medical devices such as tympanosotomy tubes, cochlear implants, and other materials.

As a part of this objective, she was trained to process samples and operate the scanning electron microscope. She also received electrocochleography training to determine the effects of various medical treatments on hearing using animal models.

This position both broadened her skills and experience in medical research and cemented her desire to conduct independent research.

In 2009, Edith was accepted into the Interdisciplinary Program in Medical Sciences in the Immunology and Microbiology Concentration. Under the mentorship of Dr. Ann

Progulske-Fox in the College of Dentistry’s Department of Oral Biology, Edith completed her dissertation work presented in this document.

195