biofilm models and methods of

Upload: verghese-george-m

Post on 04-Jun-2018

220 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/13/2019 Biofilm Models and Methods Of

    1/21

    Biofilm models and methods of

    biofilm assessmentANIL KISHEN & MARKUS HAAPASALO

    Endodontic microbes dwell within the infected root canal system as surface-adherent biofilm structures. In order to

    simulate thisin vivosituation, a variety ofin vitrobiofilm models are currently used in Endodontics for different

    microbiological experiments. Unfortunately a cogent selectionof the best models to be used for a particular research

    application has not been obtained. This article outlines various factors to be considered while developing biofilm

    models in Endodontics. Different in vitroendodontic biofilm models, devices used to generate biofilms, and biofilm

    assays used to analyze these biofilm structures qualitatively and quantitatively are also presented in this article.

    Received 9 December 2011; accepted 3 March 2012.

    Introduction: changing paradigm inendodontic infection

    Biofilm is a mode of microbial growth where dynamic

    communities of interacting sessile cells are irreversibly

    attached to a solid substratum, as well as to each other,

    and are embedded in a matrix of extracellular poly-

    meric substances (EPS) (1). While endodontic micro-

    bial flora is established to be less diverse compared tothe oral microbial flora, the task of disinfecting a root

    canal system is one of the most prominent challenges

    in Dentistry. Endodontic microbes dwell within the

    entire root canal anatomy as surface-adherent biofilm

    (intraradicular biofilm). The endodontic bacterial

    activities that are usually confined to the intracanal

    spaces may, under certain conditions, form biofilm on

    locations beyond the apical foramen (extraradicular

    biofilm) (24). The geometrical and anatomical com-

    plexity in the root canal system tends to shelter the

    bacterial biofilm from root canal disinfectants andinstrumentation procedures (2). Furthermore, the

    progression of endodontic infection alters the nutri-

    tional and environmental status of the root canal

    system, apparently rendering it more anaerobic and

    depleting it of nutrients. This yields a tough ecological

    niche for the surviving microorganisms (5). The

    biofilm mode of growth allows the resident bacteria to

    survive unfavorable environmental and nutritional

    conditions (6,7). On the above basis, it is vital to

    consider bacterial biofilm models as essential models

    for in vitro microbiological investigations and the

    assessment of different disinfectants and disinfection

    strategies in Endodontics.

    Biofilm model systems: factors tobe considered

    Bacterial biofilms are developed in order to study themicrobial interactions within the root canal space or

    between bacteria and host immune cells (8,9). Cur-

    rently, they are more commonly grown to test the

    efficacy of different irrigants/medicaments and irriga-

    tion procedures in Endodontics. It should be noted

    that the antimicrobial resistance observed in biofilm

    bacteria is not generally due to classic genetic mech-

    anisms; instead, this arises due to certain peculiarities

    of biofilm growth. The types of resident bacterial

    species, nature of bacterial adherence to substrate,

    physico-chemical characteristics of the substrate, thick-ness of the biofilm, bacterial cell density, amount of

    EPS, and phenotypical/genotypical modification of

    the resident bacteria are all factors that could contrib-

    ute to antimicrobial resistance in biofilm bacteria

    (6,10,11). Typically, different antimicrobial resistance

    mechanisms may act concurrently or synergistically in

    a biofilm structure, and understanding some of these

    mechanisms is the key to developing biofilm model

    systems for different application in Endodontics.

    bs_bs_banner

    Endodontic Topics 2012, 22, 5878

    All rights reserved

    2012 John Wiley & Sons A/S

    ENDODONTIC TOPICS 20121601-1538

    58

  • 8/13/2019 Biofilm Models and Methods Of

    2/21

    Broadly, factors associated with bacterial adherence,

    bacteriasubstrate interaction, and biofilm ultrastruc-

    ture should be standardized to develop useful biofilm

    models forin vitroexperiments (6). Currently, there is

    no universally acceptedin vitromodel that reproduces

    biofilm infection in Endodontics.

    Bacterial adherence andbacteriasubstrate interaction

    The earliest stage in the formation of most oral bio-

    films involves the adsorption of macromolecules from

    tissue fluids such as saliva onto a biomaterial (natural

    or synthetic) surface, leading to the formation of a

    conditioning layer. The conditioning fluid will form a

    layer of adsorbed inorganic and organic molecules on

    the solid surface, and alters the physical/chemical

    properties of the surface. The conditioning layer,formed prior to the influx of microorganisms, will

    selectively promote the adhesion of microbial cells to

    the surface. It may also serve as a source of nutrition

    for adherent bacteria. Bacteria can generally form bio-

    films on any surface that is conditioned with such

    conditioning fluids (11). The next step in the devel-

    opment of a biofilm is the adhesion of microbial cells

    to the substrate surface. The adhesive potential of

    microbes to natural (e.g. dentin) or synthetic (e.g.

    restorative/endodontic material) biomaterials is

    considered to be a vital ecologic and pathogenicdeterminant in biofilm-mediated infection. Bacterial

    adherence to a surface is influenced by (i) the environ-

    mental conditions such as pH, temperature, fluid flow

    rate, nutrient availability, etc.; (ii) bacteria-associated

    factors such as type of bacteria (species/strain),

    growth phase of bacteria (log or stationary phase),

    type and charge of the surface molecules, etc.; and (iii)

    substrate-associated factors such as physical and

    chemical characteristics of the substrate. It is crucial to

    standardize these parameters in order to develop

    clinically realistic biofilm model systems for in vitroexperiments (12,13).

    The initial phase of the bacteriasubstrate interaction

    is determined by the physical and chemical properties

    (e.g. surface energy and charge density) of the bacteria

    and substrate (PHASE 1 of bacterial adherence: trans-

    port of microbe to substrate surface). This reversible

    interaction is followed by the molecular-level non-

    specific interactions between the bacterial surface

    structures and the substrate. This phase of microbial

    adherence to a substrate is mediated by the bacterial

    surface structures such as fimbrae, pili, flagella, and

    EPS (PHASE 2: initial non-specific microbialsubstrate

    adherence phase). The bacterial surface structures form

    bridges between the bacteria and the conditioning film

    (11). Porphyromonas gingivalis, Streptococcus mitis,

    Streptococcus salivarius, Prevotella intermedia, Prevo-tella nigrescens, Streptococcus mutans, andActinomyces

    naeslundii are some of the oral bacteria possessing

    surface structures (11,12). The bridges formed

    between bacteria and substrate are a combination

    of electrostatic attraction and covalent/hydrogen

    bonding. Initially the bonds between bacteria and sub-

    strate may not be strong. However, with time these

    bonds gain in strength, making the bacterial attach-

    ment irreversible. In the final stage, a more specific

    bacterial adhesion to the substrate is established via

    polysaccharide adhesin or ligand formation (PHASE 3:specific microbialsubstrate adherence phase). In this

    phase, adhesin or ligand molecules on the bacterial cell

    surface will bind to receptors on the substrate. Specific

    bacterial adhesion is less affected by environmental

    factors (13,14). It is critical to realize that these phases

    involved in bacterial adherence to a substrate are a

    dynamic process which occurs as a function of time.

    The reversible and irreversible steps in phase 1 of the

    bacterial adherence occur in a few seconds to minutes,

    while phase 2 and 3 interaction take a few hours to

    days to occur, depending upon the bacteria and theenvironment conditions (Fig. 1). Therefore, while

    developingin vitromodels, it is important to provide

    sufficient bacteriasubstrate interaction time and

    optimum environmental conditions. The development

    and maturation of biofilm structure occurs subsequent

    to bacterial adherence.

    Biofilm ultrastructure

    During the development of a biofilm, the resident

    bacterial cells proliferate, leading to expansion of thebiofilm structure. In this stage, the mono-layer of

    microbes (primary colonizers) attracts the secondary

    colonizers, forming micro-colonies, and the collection

    of micro-colonies gives rise to the final structure of the

    biofilm. Ultrastructurally, a biofilm consists of a popu-

    lation of bacterial cells attached irreversibly to a sub-

    strate and encased in a hydrated, polyanionic matrix of

    EPS, proteins, polysaccharides, and nucleic acids

    (15,16). Bacteria themselves account for a variable

    Biofilm models and methods of biofilm assessment

    59

  • 8/13/2019 Biofilm Models and Methods Of

    3/21

    fraction of the total biofilm volume (typically 535%)

    (17). A mature biofilm will be a metabolically active

    community of microorganisms where individuals share

    duties and benefits (16). For instance, some microor-ganisms help in adhering to the solid support while

    some others (such asFusobacterium nucleatum) create

    bridges between different species. This signifies

    the relevance of a polymicrobial biofilm over a

    mono-species biofilm. The physiological characteris-

    tics of the resident microorganisms in a biofilm also

    offer an inherent resistance to antimicrobial agents

    (17,18). Bacterial species such as staphylococci,

    enterococci,Klebsiella pneumoniae,Pseudomonasspp.,

    etc., are inherently resistant to many antimicrobials

    (1719).Interestingly, it has been reported that the biofilms

    formed in pure cultures of bacteria under laboratory

    conditions and the mixed-species biofilms formed in

    natural ecosystems show a similar basic organization in

    which cells grow in matrix-enclosed micro-colonies

    separated by a network of open-water channels (19).

    The thickness of the EPS will influence the biofilm

    permeability and consequently provide a certain degree

    of protection or barrier effect against physical and

    chemical threats. Each step in the development of

    biofilm, from the adherence of bacteria to a substrate tothe final formation of a matured biofilm structure, as

    well as protein expression/slime production, is modu-

    lated by a large number of variables, including type of

    bacterial species, environmental conditions, and age of

    the biofilm. Previous studies on endodontic biofilm

    models have shown that mature biofilms and biofilms

    with limited nutrient supply are more resistant to irri-

    gants such as chlorhexidine and Light Activated Dis-

    infection than early (immature) biofilms under normal

    nutrient conditions. Studies have emphasized that the

    age or degree of maturation, nutritional condition of

    the biofilm, and interactions between resident bacterial

    species are some of the major confounding factors indesigningin vitrobiofilm models to test the efficacy of

    various endodontic disinfectants (2026). On the

    above basis, it is important to use relevant bacteria

    (primary colonizers), and provide ideal environmental

    conditions (substrate, fluid conditioning, nutritional

    conditions, and temperature) and optimum bacteria

    substrate interaction time (matured biofilm) in order

    to achieve a standardized endodontic biofilm model for

    in vitroapplications.

    Two types of microbial interactions occur at the

    cellular level during the formation of biofilm. One is theprocess of recognition between a suspended cell and a

    cell already attached to substratum. This type of inter-

    action is termed co-adhesion. In the second type of

    interaction, genetically distinct cells in suspension rec-

    ognize each other and clump together. This type of

    interaction is called co-aggregation. This association is

    highly specific and occurs between co-aggregating

    partners only. Interestingly, most oral bacteria recog-

    nize each other as co-aggregating partners. Fuso-

    bacterium nucleatum, a Gram-negative filamentous

    anaerobe, can co-aggregate with all oral bacteria tested,and can act as a bridging bacterium that binds together

    even non-aggregating bacteria (6). The association

    of long-filamentous bacteria and surface-adsorbed

    spherical-shaped cocci produce the characteristic

    corncob structure of oral biofilms (24). The attachment

    of cocci to filamentous bacteria is said to be mediated

    via fimbriae of the oral streptococci. Although the

    genetic makeup of the bacteria is the main determinant

    of co-aggregation, the physico-chemical characteristics

    Fig. 1. Schematic diagram showing the stages in the development of a biofilm.

    Kishen & Haapasalo

    60

  • 8/13/2019 Biofilm Models and Methods Of

    4/21

    of the environment also play a crucial role (24,26). It

    was observed that bacteria in a co-aggregated suspen-

    sion were significantly more resistant to antimicrobials

    when compared to planktonic suspension, while bacte-

    ria in a biofilm mode showed the most resistance to

    antimicrobials (25,26). Consequently, antimicrobial

    agents selected on the basis of traditional susceptibilitymethods (such as broth-based Minimal Inhibitory

    Concentration [MIC]) may not be very appropriate to

    eliminate co-aggregated or biofilm bacteria.

    Bacterial biofilm models:in vitrodevelopment

    Laboratory models are conventionally used to mimic

    natural biofilms for different experimental purposes.

    Thesein vitrobiofilms are easy to control and useful in

    obtaining standardized biofilm models with predict-able structure and behavior. Conventional biofilm

    models range from monocultures in static growth con-

    ditions to diverse mixed cultures in dynamic growth

    conditions. The static biofilm models use different sub-

    strates (e.g. glass, polycarbonate, silicon, hydroxyl

    apatite, nitrocellulose, enamel, dentin) to grow bio-

    films while the dynamic biofilm models use reactor or

    fermenting systems to grow biofilms on a particular

    substrate. Both aerobic and anaerobic environments

    can be employed for in vitro biofilm development.

    Figures 26 show differentin vitrobiofilms grown ondifferent substrates. Given that thein vivoconditions

    are commonly dynamic, studies evaluating biofilm for-

    mation under static conditions might be somewhat

    misleading depending upon the research question.

    These in vitrobacterial biofilm models are routinely

    applied to: (i) examine the adherence of specific bac-

    terial species to any biomaterial surface (27); (ii) study

    the nature and pattern of early microbial biofilm for-

    mation on a particular substrate (28); (iii) study the

    interaction between different biofilm bacteria and host

    immune cells (29); and (iv) test the efficacy of antimi-crobial agents or antimicrobial treatment strategies

    (30,31). Currently in Endodontics, most in vitro

    biofilms are developed and utilized for testing anti-

    microbials and irrigation strategies (Table 1). Pres-

    ently, published results on the activity of disinfectants

    show noticeable discrepancies between experiments,

    and this may be attributed to the diversity of the

    microbial growth phase, biofilm models, and

    procedures/assays utilized for the analysis. Obviously,

    Fig. 2. (a) Photograph of a biofilm grown in vitro forthree weeks on a collagen-coated hydroxyapatite disc.The biofilm is coated with a palladiumgold mixture forSEM. (b) SEM image of mixed bacterial (multi-species)biofilm grown anaerobically for seven days in BHI brothon collagen-coated hydroxyapatite disc. Low magnifica-tion. (c) SEM image of six-month-old biofilm grownanaerobically in BHI broth on collagen-coated hydrox-

    yapatite disc. Several bacterial morphotypes includingcoiled spirochetes can be seen.

    Biofilm models and methods of biofilm assessment

    61

  • 8/13/2019 Biofilm Models and Methods Of

    5/21

    the number of parameters needs to be considered in

    the design of a representative biofilm model for appli-

    cations in Endodontics (Fig. 7).

    In recent years, it has become evident that a number

    of parameters may be important when performing

    adherence assays, including the microbial concentra-tion in the inoculum, incubation time, growth condi-

    tions, and substrate properties. If laboratory strains are

    used for adherence assays, then it is important that

    they are representative of clinical isolates. In addition,

    assays that do not take into account the presence of

    saliva may be unsuitable for the study of adhesion and

    early biofilm formation (27). Findings from experi-

    ments with planktonic and biofilm bacteria (50) have

    revealed large differences in the dynamics of killing

    Fig. 3. (a) SEM of three-day-old biofilm grown anaero-bically in BHI broth on a nitrocellulose filter. Cocci and

    long rod-shaped bacteria dominate in the specimen. Lowmagnification. (b) Three-day-old biofilm grown anaero-bically in BHI broth on a nitrocellulose filter. Cocci andlong rod-shaped bacteria dominate in the specimen.High magnification.

    Fig. 4. (a) SEM image of six-week-old biofilm grownaerobically in BHI broth on a dentin disc. (b) Lowmagnification SEM image of the previous sample. Cocciand long rods dominate in the sample. (c) Mixture ofbacteria grown aerobically for one month on dentin hasfailed to grow a mature biofilm. Smeared dentin can beseen between the bacteria.

    Kishen & Haapasalo

    62

  • 8/13/2019 Biofilm Models and Methods Of

    6/21

    between planktonic and biofilm bacteria. There is nodoubt that the results from planktonic killing studies

    must be interpreted with caution and direct extrap-

    olation as to the efficacy of the agent in complex

    in vivo systems is not possible. A comparison of the

    planktonic and biofilm tests in a study indicated that

    planktonic killing tests may be useful for preliminary

    screening of new disinfectants before proceeding onto

    more complex biofilm designs (50).

    Biofilm devices: flow cells and fermentorsThere are several in vitro devices that are used to

    develop biofilms. Some of them produce biofilms irri-

    gated with fresh culture medium; in this case, the

    biofilms experience a continuous flow of medium

    supplemented with fresh nutrients. These in vitro

    devices are used to grow dynamic biofilm models. The

    flow cell system is one of the most utilized dynamic

    models. It consists of a transparent chamber of fixed

    depth through which the growth medium flows. The

    inlet tubing supplies growth medium and the outlettubing drains the medium to a waste reservoir. The

    growth medium is passed through the cell with the aid

    of a peristaltic pump, which controls the flow rate of

    the medium. Pre-fabricated flow cell systems are avail-

    able commercially or they can be custom-made based

    on any particular application. In conjunction with a

    microscope, charge-coupled-device (CCD) camera, or

    Confocal Laser Scanning Microscopy (CLSM), this

    method can be used to observe the early events in

    biofilm formation in real time (55).

    Chemostats are also used to grow dynamic biofilmsof microbes on experimental substrates submerged

    within the chemostat. One of the most important

    features of chemostats is that microbial biofilms can be

    grown at a constant rate and under constant culture

    conditions (temperature, pH). Similar to chemostat,

    there is another category of reactors in which biofilms

    are formed on thin filter membranes in a physiological

    steady state. These systems permit evaluation of

    growth rate dependence and cell-cycle specificity of

    Fig. 5. (a) Mixture of bacteria grown anaerobically for one month on dentin. (b) Mixed bacterial biofilm grownanaerobically in BHI broth for one month on dentin. (c) Mixed bacteria biofilm grown anaerobically for one monthin BHI on root canal wall dentin. (d) High magnification image of the previous SEM picture.

    Biofilm models and methods of biofilm assessment

    63

  • 8/13/2019 Biofilm Models and Methods Of

    7/21

    antibacterial agents. Finally, there are constant depth

    reactors in which surface growth is periodically

    removed to maintain a constant geometry of thebiofilm. In these reactors, microorganisms can be

    grown in a physiological steady state with all culture

    parameters constant. The system can generate large

    numbers of biofilms with comparable and repro-

    ducible data (55).

    The static biofilm system generates biofilms that

    have exhausted important nutrient components at

    the end of an overnight incubation. The key features

    of this system are that numerous biofilms can be

    handled at any given time, and it does not require

    time-consuming sterilization and set-up procedures,

    allowing it to be used as a high-throughput systemfor biofilm analysis (31). This system provides a basis

    for the rapid screening of biofilm mutants (56),

    biomass development, and biofilm-forming capacity

    (57), as well as extracellular matrix composition (58).

    Essentially, detection of any microbial phenotype that

    can be processed by a microtiter plate/reader can be

    used for the approach. However, this system is

    incompatible with CLSM, which is the preferred

    methodology for studying the structure of biofilms.

    Fig. 6. (a) Multi-species biofilm grown anaerobically for three weeks on a glass coverslip conditioned with media for24 hrs. The biofilm was sparse and not uniform. (b) High magnification image of the previous SEM picture showingmixed bacterial flora. (c) The biofilms were approximately 20 microns thick at certain areas. (d) High magnificationimage of biofilm with abundant extracellular matrix.

    Kishen & Haapasalo

    64

  • 8/13/2019 Biofilm Models and Methods Of

    8/21

    Table 1: List of literature of in vitrobiofilm models for different endodontic applications

    Authors Type of Model Purpose Preparation of Biofilm

    Shabahang &

    Torabinejad,

    2003 (32)

    In vitro

    E. faecalis

    The antimicrobial effect of MTAD: anin

    vitroinvestigation

    Extracted human teeth contaminated with

    E. faecalisfor 4 weeks

    Dentinal shavings and CFU-based

    method was used for the analysis

    Duggan &

    Sedgley, 2007

    (33)

    In vitro

    E. faecalisstrains

    recovered from root

    canals, oral cavity,

    and non-oral

    sources

    Biofilm formation of oral and endodontic

    E. faecalis

    96-well plates for 24 hours

    Crystal violet assay used for assessment

    (optical density at 570 nm)

    George &

    Kishen, 2007

    (34)

    In vitro

    E. faecalis

    (Gram-positive),

    Actinomycetes

    actinomycetemcomitans

    (Gram-negative)

    Methylene blue dissolved in different

    formulations: water, 70% glycerol, 70%

    polyethylene glycol, and a mixture of

    glycerol:ethanol:water (30:20:50) was

    tested

    Two-day-old biofilms in multi-well plates

    (polystyrene)

    Four-day-old biofilms in human teeth

    CFU-based method

    George &

    Kishen, 2008

    (35)

    In vitro

    E. faecalis

    This study aimed to investigate the effect

    of including an oxidizer and oxygen

    carrier in photosensitization

    formulation to disinfect a mature

    endodontic biofilm by light activated

    disinfection

    Human teeth (10-week-old biofilm)

    CFU-based method

    McGill et al.,

    2008 (36)

    In vitro

    E. faecalis

    The efficacy of dynamic irrigation using a

    commercially available system

    (RinsEndo)

    A collagen-based bio-molecular film

    formed on extracted human teeth

    Digital image analysis of the canal surfaces

    (ipWin4)

    Sainsbury et al.,

    2009 (37)

    Ex vivo DIAGNOdent laser fluorescence

    assessment of endodontic infection

    Extracted teeth with endodontic

    pathology

    Fluorescence emissions in thenear-infrared range were measured

    Shen et al.,

    2009 (38)

    In vitro

    Multi-species

    Evaluation of the effect of two

    chlorhexidine preparations on biofilm

    bacteria in vitro: a three-dimensional

    quantitative analysis

    Collagen-coated hydroxyapatite (CHA)

    and uncoated hydroxyapatite (HA)

    discs

    Confocal laser scanning microscopy was

    used for the analysis of dead versus

    viable cells

    Williamson

    et al., 2009

    (39)

    In vitro

    E. faecalis

    (clinical isolate)

    Antimicrobial susceptibility of

    monoculture biofilms to 6% NaOCl,

    2% CHX,

  • 8/13/2019 Biofilm Models and Methods Of

    9/21

    Table 1: Continued

    Authors Type of Model Purpose Preparation of Biofilm

    Kishen et al.,

    2010 (41)

    In vitro

    E. faecalis

    Efflux pump inhibitor potentiates

    antimicrobial photodynamic

    inactivation ofEnterococcus faecalis

    biofilm

    Microwell plates

    CFU-based method

    Confocal laser scanning microscopy

    Hiraishi et al.,

    2010 (42)

    In vitro

    E. faecalis

    Antimicrobial efficacy of 3.8% silver

    diamine fluoride

    Membrane filters

    CFU-based method

    Shrestha et al.,

    2010 (43)

    In vitro

    E. faecalis

    Nanoparticulates for anti-biofilm

    treatment and effect of aging on its

    antibacterial activity

    Microwell plates/saliva

    CFU-based method

    Confocal laser scanning microscopy

    Liu et al., 2010

    (44)

    In vitro

    E. faecalis

    Biofilm formation capability of

    Enterococcus faecaliscells in starvation

    phase and its susceptibility to sodium

    hypochlorite

    Human dentin and polystyrene blocks

    CFU-based method

    SEM

    Chavez de Paz

    et al., 2010

    (45)

    In vitro(clinical

    isolates)

    E. faecalis,L. paracasei,

    S. anginosus,

    S. gordonii

    The effects of antimicrobials on

    endodontic biofilm bacteria

    24-hour biofilm within a mini-flow cell

    system

    Confocal microscopy and image analysis

    Su et al., 2010

    (46)

    In vivo This study explored the effect of surgical

    endodontic treatment of refractory

    periapical periodontitis with

    extraradicular biofilm

    Resected root-end samples

    Soares et al.,

    2010 (47)

    In vitro

    E. faecalis

    Effectiveness of chemomechanical

    preparation with alternating use of

    sodium hypochlorite and EDTA in

    eliminating intracanalEnterococcus

    faecalisbiofilm

    Human teeth (21-day-old biofilm)

    SEM

    CFU-based method

    Bhuva et al.,

    2010 (48)

    In vitro

    E. faecalis

    The effectiveness of passive ultrasonic

    irrigation on intraradicularEnterococcus

    faecalisbiofilms in extracted

    single-rooted human teeth

    Human teeth

    SEM-based image analysis

    Shen et al.,

    2010 (49)

    In vitro

    Multi-species biofilm

    (subgingivial

    plaque)

    The synergistic antimicrobial effect by

    mechanical agitation and two

    chlorhexidine preparations

    Collagen-coated hydroxyapatite (CHA)

    discs

    (3 weeks old)

    Confocal laser scanning microscopy

    Pappen et al.,

    2010 (50)

    In vitro

    Multi-species biofilm

    (subgingivial

    plaque)

    To investigate the antibacterial effect

    of Tetraclean, MTAD, and five

    experimental irrigants using both

    direct exposure test with planktonic

    cultures and mixed-species in vitro

    biofilm model

    Collagen-coated hydroxyapatite (CHA)

    discs

    (2 weeks old)

    Confocal laser scanning microscopy

    Shen et al.,

    2010 (51)

    In vitro

    Multi-species biofilm

    (subgingivial

    plaque)

    The aim of this study was to enumerate

    viable bacteria at different growth

    stages of a multi-species oral biofilm

    and to compare results obtained with

    the LIVE/DEAD BacLight Kit with

    those from culturing and plate

    counting

    Collagen-coated hydroxyapatite (CHA)

    discs

    Confocal laser scanning microscopy

    CFU-based method

    Kishen & Haapasalo

    66

  • 8/13/2019 Biofilm Models and Methods Of

    10/21

    Table 1: Continued

    Authors Type of Model Purpose Preparation of Biofilm

    Lundstrom

    et al., 2010

    (52)

    In vitro(multi-species)

    Streptococcus sanguinis,

    Actinomyces viscosus,

    Fusobacterium

    nucleatum,Peptostreptococcus

    micros,and

    Prevotella nigrescens

    Bactericidal activity of stabilized chlorine

    dioxide as an endodontic irrigant in a

    polymicrobial biofilm tooth model

    system

    Permanent bovine incisors coated with

    mucin and inoculated with standardized

    suspensions of bacteria (anaerobically

    for 14 days)

    CFU-based method

    Hope et al.,

    2010 (53)

    In vitro

    E. faecalis

    A direct comparison between extracted

    tooth and filter-membrane biofilm

    models of endodontic irrigation

    Human teeth

    CFU-based method

    Upadya &

    Kishen, 2010

    (9)

    In vitro

    E. faecalisand

    P. aeruginosa

    To evaluate the efficacy of light-activated

    disinfection (LAD) using Methylene

    blue (33) and a non-coherent light

    source on Gram-positive and

    Gram-negative bacteria in different

    growth modes. The influence ofdifferent photosensitizer (PS)

    formulations in the MB-mediated

    LAD of biofilms was also evaluated.

    Mono-species biofilms in 24-well

    polystyrene plates (4 days)

    CFU-based method

    Confocal laser scanning microscopy

    George et al.,

    2010 (28)

    In vitro

    E. faecalis

    This study examined the biofilm-forming

    capacity ofE. faecalison gutta-percha

    points under different nutrient status

    and surface conditioning with saliva

    and serum

    Gutta-percha

    Conditioned with saliva or serum (2, 4,

    and 12-weeks)

    Biofilm growth for 2 weeks

    CFU-based method

    SEM

    Badr et al.,

    2011 (54)

    In vitro

    E. faecalis

    A laboratory evaluation of the

    antibacterial and cytotoxic effect of

    liquorice when used as root canalmedicament

    Grown on cellulose nitrate membrane

    filters

    CFU-based method

    Shen et al.,

    2011 (21)

    In vitro

    Multi-species biofilm

    (subgingivial

    plaque)

    The aim of this study was to examine the

    susceptibility of multi-species biofilms

    at different phases of growth to root

    canal irrigants (2% chlorhexidine

    [CHX] or CHX-Plus)

    Collagen-coated hydroxyapatite (CHA)

    discs (2 days to months)

    Confocal laser scanning microscopy

    CFU-based method

    Fig. 7. Schematic diagram showing different factors that would influence the structure and development ofin vitrobiofilms.

    Biofilm models and methods of biofilm assessment

    67

  • 8/13/2019 Biofilm Models and Methods Of

    11/21

    Structural evaluation of biofilm requires the use of

    irrigated biofilm systems. An irrigated and flow-

    through cell system allows for the study of the devel-

    opment of biofilm over time. These analyses include

    non-destructive evaluation of temporal and spatial

    expression of selected genes and the complete life-

    cycle of biofilm formation and dispersal. Several find-

    ings on the unique behavioral responses of biofilm

    cells that cannot be obtained using static microtiter-

    based systems were observed with the aid of irrigated

    biofilm flow systems (59).

    Biofilm assays

    Biofilm assays are used to characterize factors such

    as (i) number and type of microorganism, (ii) vitality

    (dead/living cells) of the resident microbial popula-

    tion, (iii) age, (iv) thickness (mono-layered or multi-

    layered), (v) structure (homogeneous, irregular,dense, porous), and (vi) surface topography (peaks

    and valleys) of biofilms. Different techniques such as

    (i) microbiological culture techniques, (ii) colori-

    metric techniques, (iii) microscopic techniques, (iv)

    physical methods, (v) biochemical methods, and (vi)

    molecular methods are applied to biofilm assays. The

    basic outline of the experimental methods to assess

    antibacterial or antibiofilm efficacy of irrigants or irri-

    gation strategies is shown in Figure 8.

    Microbiological culture techniques

    The biofilm formed on a substrate can be quantified by

    directly enumerating the Colony Forming Units

    (CFU) of the bacteria adhering to the surface. The

    CFU measurement will provide information on the

    amount of viable bacteria adherent to the substrate or

    growing within the biofilm structure. However, the

    CFU may only detect bacteria that are able to initiatecell division at a sufficient rate to form colonies and

    whose growth requirements are supported by the

    culture medium used. Several protocols recommend

    the removal of biofilm bacteria from the substrate by a

    sonication or centrifugation process. In such cases, the

    CFU is usually determined from the supernatant

    obtained after the sonication/centrifugation proce-

    dure. The recovery of microorganisms after treatment

    with disinfectants remains an important point in these

    experiments. The bacteria can be sensitive to these

    procedures and changing growth conditions and, inthat case, there may be a 24-hour to more than a week

    lag phase in the bacterial response. A recent study

    showed that in older, starved biofilms the bacteria are

    viable based on the green staining pattern as observed

    by CLSM, but over 99% of these bacteria could not be

    grown when removed from the biofilm and grown in

    a culture media (49). Ultrasonic vibrations and

    enzymes are used to remove bacterial biofilm before

    quantification. However, it is imperative to use an

    Fig. 8. Basic outline of the experimental methods for assessing antimicrobial efficacy using in vitrobiofilm.

    Kishen & Haapasalo

    68

  • 8/13/2019 Biofilm Models and Methods Of

    12/21

  • 8/13/2019 Biofilm Models and Methods Of

    13/21

    without using fluorescent probes by using plasmid-

    encoded green fluorescent protein (GFP). These

    transformedE. coliO157:H7 have been used to study

    their attachment onto a surface (62,63). The viability

    of these cells may be determined by staining the trans-

    formed cells with membrane-impermeable fluorescent

    dye (63).

    Scanning Electron Microscopy (SEM) and Transmis-

    sion Electron Microscopy (TEM) have been effective

    workhorses in biofilm analyses for many years. High-resolution electron microscopy has been employed for

    the morphological and structural characterization of

    microbial biofilms. The main disadvantage with these

    techniques is the need for extensive sample preparation

    steps such as fixation, dehydration, freeze- or critical

    point-drying, and sputtering. These treatments can

    deeply affect the original biofilm morphology

    (Fig. 12). Environmental SEM (ESEM) is a relatively

    new technique that represents a powerful alternative to

    conventional SEM (high vacuum) as it allows the

    imaging of biological samples in their original hydratedcondition at relatively high resolution (64). Structural

    modifications in microbial biofilm architecture, par-

    ticularly an overall loss of matrix volume, were appre-

    ciable when comparing conventional high-vacuum

    SEM to ESEM images. Sutton et al. (65) compared

    different dehydration techniques and showed that

    freeze-dried samples presented significant detachment

    of microbial biofilm from the substrate, while more

    complex dehydration procedures such as critical point

    Fig. 11. Fluorescent microscope image showingE. faecaliscells adhering to type 1 collagen (100 , oil immersion).(a) Control. (b) After EDTA treatment.

    Fig. 12. SEM images of (a) E. faecalis cells adhering toroot canal dentin and (b) multi-layered mono-speciesbiofilm ofE. faecalison root canal dentin.

    Kishen & Haapasalo

    70

  • 8/13/2019 Biofilm Models and Methods Of

    14/21

    drying caused an almost complete disappearance of the

    EPS matrix. Although proper fixation processes were

    applied, the collapse of the biofilm structure upon

    dehydration procedures is mainly due to the lack of a

    self-sustaining scaffold in the EPS matrix. Neverthe-

    less, bacteria cells maintained their shape and dimen-

    sions in vacuum after fixation and could clearly beidentified by SEM. In the ESEM mode, the semi-

    transparent appearance of the EPS and the low signal-

    to-noise ratio at high pressures results in a limited

    image resolution. In brief, ESEM represents an effec-

    tive technique for detecting highly hydrated bacterial

    biofilms by preserving the substantial EPS component.

    On the contrary, conventional high-vacuum SEM

    allows a detailed examination of the cellular compo-

    nents and favors the detection of the three-dimensional

    hollow structures, but fails to show the actual biofilm

    architecture consisting of a large volume of EPS matrixsurrounding the cells. The combined use of conven-

    tional SEM and ESEM techniques can therefore

    provide complementary information on different

    biofilm components, bacterial cells, and the extracellu-

    lar matrix (65).

    Epifluorescent microscopy has been used to study

    bacterial biofilm microstructure. Biofilms grown on

    biomaterial surfaces are usually stained with a fluores-

    cent dye and viewed under an epifluorescent micro-

    scope. In a study, binary species biofilms were stained

    using two different fluorescent probes for each organ-ism and observed under an epifluorescent microscope

    using two excitation wavelengths. Two different images

    and the background biofilm were captured with appro-

    priate wavelengths. The images were then combined to

    construct a new image that simultaneously showed

    both organisms (66). Epifluorescentmicroscopy is used

    to determine viable cells, biofilm cell arrangement,

    micro-colony formation, biofilm pH, and distribution

    of chemicals in a biofilm structure (67).

    CLSM is a particularly important biofilm analysis

    technique that is restricted to 50200-mm-thickbiofilm structures. CLSM has overcome some of the

    limitations exhibited by most of the earlier micro-

    scopic techniques such as epifluorescence, SEM, and

    TEM. Together with improvements in the molecular

    techniques for bacteria, CLSM has become an impor-

    tant tool for studying biofilms. Green fluorescent

    protein (GFP) tagging of certain bacterial strains such

    as Pseudomonas aeruginosa is utilized to study biofilm

    formation. This method uses a fluorescent imaging-

    based analysis or CLSM to quantify the biofilm struc-

    tures. The preferred method of tagging has been to

    construct chromosomal insertions in order to ensure a

    stable gene dosage of the tag sequence (68,69).

    Recently introduced, time-lapse CLSM together with

    thegfpreporter system has been used to study the role

    ofagrin biofilm formation and has given an interest-ing insight into gene regulation during the course of

    biofilm development (70). This technique is likely to

    be an important tool for future studies on the regula-

    tors and genes involved in biofilm development.

    CLSM creates a thin (~0.3 mm) plane of focus

    (optical sections) in which out-of-focus light will be

    blocked, either conventionally by optical barriers or

    by applying the physics of light absorption as

    equipped in multi-photon microscopy (71). These

    optical sections can then be stacked by software to

    generate a three-dimensional reconstructed image ofthe entire biofilm. The CLSM images can be used to

    determine the thickness and distribution of cells in a

    biofilm structure. CLSM can also be used to deter-

    mine the pH gradients in biofilms. The interior pH of

    biofilms is measured by a fluorescent lifetime imaging

    technique using fluorescein as a pH indicator. Cur-

    rently, the use of a fluorescent dye combination

    (LIVE/DEAD BAC light) with CLSM has become a

    routine practice for in vitro biofilm analysis. The

    LIVE/DEAD Bacterial Viability kit (Molecular

    Probes, Eugene, OR) contains separate vials of thetwo component dyes (SYTO 9 and propidium

    iodide). The dyes are used in a 1:1 mixture for stain-

    ing the biofilm bacteria following the manufacturers

    instructions. The dead cells emit red light and the

    viable cells emit green light under CLSM examina-

    tion (Fig. 13). In a recent in vitrostudy, it was shown

    that bacteria in the multi-species anaerobic biofilm

    grown under nutrient deprivation changed into the

    viable-but-non-cultural (VBNC) state but could be

    returned to the normal physiological state and cul-

    tured by re-establishing the supply of nutrients whilethey were still in the biofilm. The results from this

    study indicated that viability staining was a better

    reflection of the true viability of the biofilm bacte-

    ria than the culturing method during starvation. This

    finding needs to be taken into account when assessing

    results from cultural studies employed to determine

    in vivoroot canal biofilms (51).

    A fluorescence in situ hybridization (FISH) tech-

    nique using probes to target specific 16S rRNA

    Biofilm models and methods of biofilm assessment

    71

  • 8/13/2019 Biofilm Models and Methods Of

    15/21

    sequences in bacteria is applied for the simultaneous

    analysis of the spatial distribution of both Gram-

    positive and Gram-negative bacteria in biofilms.

    FISH is a recognized tool for the specific and sensi-

    tive identification of target organisms within complex

    microbial communities. Visualization of FISH-

    labeled cells in biofilms can be carried out by fluo-

    rescent microscopy and LSCM (68,69). However,

    CLSM is preferred in a biofilm analysis because it

    allows a three-dimensional non-invasive visualizationof cells and the computational reconstruction of

    mature biofilms without distortion of their structure

    (25,70).

    Physical methods: thickness, weight, area,and density measurements

    Basic physical parameters such as biofilm thickness,

    area, weight (wet and dry), and density estimates are

    used to quantify biofilm growth. A thickness measure-

    ment by light microscopy is usually effective in thinbiofilms but may not work with thick biofilms. In this

    method, the biofilm is placed on the stage of a micro-

    scope that has calibration scales on the fine control and

    the objective is lowered until the biofilm surface is in

    focus and the fine adjustment dial setting of the micro-

    scope is recorded (71). The microscope objective is

    then focused on the substrate surface, preferably in an

    area with no biofilm. The difference in the fine adjust-

    ment settings can be used to calculate the thickness. A

    simple manual-gauge needle method (72) and an elec-

    tronic probe to measure biofilm thickness (73) have

    also been described. A properly prepared SEM sample

    or cryosection enables the estimation of biofilm thick-

    ness and also reveals layering of embedded bacterial

    cells (74). Biofilm wet-weight is a useful measure of

    the biomass, especially on tared substrates. This is a

    very simple and quick procedure. The substrate can be

    weighed before biofilm growth (with the assumption

    that no substrate solubilization occurred duringbiofilm formation) and then cleaned, dried, and

    weighed again in order to record the dry biofilm

    weight. If both wet and dry weight measurements on

    the same biofilm sample are performed, the approxi-

    mate density may be determined by assuming that the

    volume of the biofilm sample is the same as the water

    volume estimated as the wet weight minus the dry

    weight. Routinely, for comparative purposes, physical

    parameters such as biofilm density and weight can be

    calculated per unit of substratum.

    Biochemical methods: biomass andextracellular matrix (ECM)

    Microbial biomass denotes the total number of

    microbes in a given area. The measurement of micro-

    bial biomass is considered to be a rapid method and

    includes measurements of the wet or dry weight of the

    entire biofilm, measurements of the cell contents,

    measurements of the cellular activities or viable cells,

    Fig. 13. Three-dimensional confocal laser scanning microscopy reconstruction of E. faecalis biofilm (inlet showssagittal section) (60 ). Left: the biofilm has received no treatment. Right: the biofilm has been subjected to Light

    Activated Disinfection with Methylene blue and laser (660 nm).

    Kishen & Haapasalo

    72

  • 8/13/2019 Biofilm Models and Methods Of

    16/21

    etc. One method that is used for the early detection of

    viable bacteria is based on metabolic activity. Adeno-

    sine triphosphate (ATP) bioluminescence is widely

    used to determine the metabolic activity of a bacterial

    population. This technique requires a cell lysis step to

    release ATP, which is determined by a luciferine

    luciferase reaction (75). However, it should be notedthat the rate of lysis and the ATP content vary depend-

    ing on the microorganism. Hence, the ATP assay

    cannot be correlated with the initial number of micro-

    bial cells. Test strains genetically modified (containing

    genes for bioluminescence) have also been developed

    and used for in vitro analysis (76). All of these

    methods present advantages and disadvantages.

    Except for the microscopy-based techniques, most

    other methods require a good number of viable cells

    or the ability of bacteria to multiply to a significant

    number in a normal period of time. This will facilitatethe detection of even a minimum microbial population

    level. Revival of bacteria is also a concern because it is

    unknown how to determine the time needed before

    physical or biochemical measurements are made (77

    81). Currently there is no standardized rapid method

    that can replace conventional biochemical assays.

    Further research is required before employing rapid

    methods more routinely for the detection of viable

    bacteria in biofilm assays.

    Molecular biological methods

    Molecular biological techniques have provided a great

    deal of genetic information on biofilm bacteria. The

    primary goal of most of these methods is to develop

    standard assays to study factors affecting bacterial

    adherence and biofilm formation. Microarray analysis

    and the use of defined regulatory mutants have been

    important tools for studying biofilm development. In

    addition, cloning and expression of bacterial virulence

    factors in less pathogenic organisms is another impor-tant tool for assessing the role of bacterial factors in

    biofilm-mediated infections (78). It is important to

    realize that, when molecular-based analyses of bacterial

    adherence and biofilm formation are performed, the

    bacteria are grown under appropriate laboratory con-

    ditions. These conditions might not be a standard

    protocol or a clinically realistic condition for the resi-

    dent bacterial cells. Although such experiments can be

    used for a relative comparison between experimental

    groups, there is a possibility of data disparity between

    laboratories andin vivosituations.

    Enzyme-linked immunosorbent assay (ELISA) is a

    very sensitive method used to detect the presence of

    antigens or antibodies of interest in a sample. ELISA

    can be used for quantitative analysis when used in

    conjunction with standard curves. ELISA is typicallyperformed using one of two detection methods: the

    direct or indirect assay. In direct ELISA, an enzyme-

    linked (labeled) antibody is used to directly detect the

    captured antigen or antibody of interest. In the more

    common indirect ELISA, a detection or primary anti-

    body is bound to the sample antigen/antibody and

    then a secondary labeled antibody (antiglobulin) is

    used to detect the primary antibody. For any ELISA

    procedure, the sample antigen/antibodies of interest

    are concentrated and solublized in an appropriate

    buffer. ELISA has been used as an alternative methodto quantify biomass within biofilms and even protein

    production in biofilms (82,83). ELISA may be used to

    quantify the population of a particular bacterium in a

    mixed biofilm. An ELISA-based approach can circum-

    vent errors due to cell clumping and EPS production,

    which can lead to significant errors in bacterial quan-

    tification. The disadvantages of ELISA are similar to all

    antibody-based methods and are related to cross-

    reactivity and non-specific signal production. This

    method is also poorly suited for low concentrations of

    antigens (59).The detection of differential gene expression may

    also aid in capitalizing on the novel high-resolution

    and specific assays to understand differential gene

    expressions in biofilm communities. However, current

    assays may only depict the average signal or response

    from all of the cells in the biofilm. This measurement

    would not provide signals from the specific cell popu-

    lation in the biofilm that responded to specific

    environmental/treatment-mediated changes. These

    localized cells responses would be useful for the iden-

    tification and design of interfering therapeutic meas-ures. Furthermore, there are several factors in an

    in vivoenvironment that may influence bacterial adhe-

    sion and biofilm formation. The shear forces, salivary/

    plasma protein binding of biomaterials/device, and

    the immune response are some of the in vivo factors

    that are difficult to reproduce in vitro. Therefore, it is

    questionable whether the assessment of gene expres-

    sion in vitro is actually indicative of gene expression

    in vivo.

    Biofilm models and methods of biofilm assessment

    73

  • 8/13/2019 Biofilm Models and Methods Of

    17/21

    Polymerase chain reaction (PCR) is a method that

    allows exponential amplification of short DNAsequences. The method relies on thermal cycling and

    enzymatic replication of the DNA. Primers, which

    consist of short DNA fragments/sequences comple-

    mentary to the target region and a DNA polymerase,

    are key components to enable selective and repeated

    amplification. As PCR progresses, the DNA generated

    is itself used as a template for replication, setting up a

    chain reaction in which the DNA template is expo-

    nentially amplified. This method of analysis is mostly

    used as a qualitative tool for detecting the presence or

    absence of a particular bacterial DNA. A real-timepolymerase chain reaction, also called quantitative

    real-time polymerase chain reaction(Q-PCR) is based

    on PCR and is employed to amplify and simulta-

    neously quantify a targeted DNA molecule. RT-PCR

    enables both detection and quantification of one or

    more specific sequences in a DNA sample. The key

    feature in RT-PCR is that the amplified DNA is

    detected as the reaction progresses in real time(84).

    This is a new approach compared to standard PCR

    where the product of the reaction is detected at the

    end. Two common methods for the detection ofproducts in real-time PCR are (i) non-specific fluores-

    cent dyes that intercalate with any double-stranded

    DNA, and (ii) sequence-specific DNA probes consist-

    ing of oligonucleotides that are labeled with a fluo-

    rescent reporter, which permits detection only after

    hybridization of the probe with its complementary

    DNA target. RT-PCR can be used to estimate the

    number of copies of a target gene in a sample and is

    reported to be more sensitive than conventional quali-

    tative PCR. This method has been used to detect and

    quantify bacterial populations in a biofilm. Often, theRT-PCR is combined with reverse transcription to

    quantify messenger RNA and non-coding RNA in

    cells or tissues. Quantitative reverse transcriptase real-

    time PCR (qRT-PCR) can be used effectively to quan-

    tify the number of RNA transcripts of specific genes

    from bacteria growing in biofilms. qRT-PCR has a

    large dynamic range and may be used to verify gene

    expression data obtained from microarrays. In addi-

    tion, qRT-PCR is sensitive and therefore may be used

    to quantify gene expression from biofilm samples

    where only a small amount of biological material isavailable (8588).

    Miscellaneous advanced techniques

    Atomic force microscopy (AFM) has been applied

    recently to study the forces of interaction between

    bacteria cells and between bacteria cells and substrates

    (8992) (Fig. 14). In order to use AFM to determine

    bacteriasubstrate interaction, the bacteria cell or

    substrate particle is attached onto an AFM tip and the

    forces of interaction between bacterial cells andbetween the bacterial cell and substrate are deter-

    mined. Briefly, as the AFM tip approaches the sub-

    strate and the gap between the two interacting bodies

    closes to the nanometer range, the interacting forces

    developed are registered by the AFM tip (90). The

    AFM force cur ves can be used to estimate the duration

    of interaction and adhesion events in the interaction

    between the bacteria and the substrate. AFM has

    also become an accepted tool to measure interaction

    Fig. 14. Atomic force microscope images showing the details of bacterial cell surfaces in the nanometric range.

    Kishen & Haapasalo

    74

  • 8/13/2019 Biofilm Models and Methods Of

    18/21

    forces between bacteria and substrates (92). In this

    analysis, positively charged polymers, such as poly-

    ethyleneimine and poly-L-lysine, are necessary to

    securely attach bacteria onto the cantilever tips. The

    physical attachment of bacterial cells using positively

    charged polymers might promote structural rearrange-

    ments in bacterial cell surface structures, which in turnmay affect the value of the forces measured. Based on

    this concept, an investigation aimed to study the

    effects of endodontic irrigants on the adherence ofE.

    faecalis to dentin (93). The findings from this study

    highlighted that chemicals which altered the physico-

    chemical properties of dentin might influence the

    nature of bacterial adherence and adhesion forces to

    dentin that are factors in biofilm formation. Recently,

    mechanical tools such as micromanipulators have been

    used to sample individual cells or biofilm compart-

    ments. However, the sensitivity of such tools is too lowto allow any analysis of a population of cells (ideally

    less than 1,000 cells). Laser-based optical tweezers are

    non-invasive and non-contact tools that can probe the

    interaction between microscopic objects such as bac-

    teria and collagen with sub-pN sensitivity. The optical

    tweezers technique gives more quantitative informa-

    tion about the forces of interaction between bacteria

    and substrate (93).

    Fourier Transform Infra-Red (FTIR) spectroscopy

    has been applied to characterize the chemical compo-

    sition of mature biofilm structures. In an FTIR spec-troscopic analysis, infrared radiation is interacted with

    a test sample. During this interaction, some of the

    infrared radiation is absorbed by the sample and some

    of it is transmitted through the sample. The resulting

    spectrum represents the molecular level absorption

    and transmission, which is a molecular fingerprint of

    the sample. FTIR spectroscopy can be used for the

    qualitative and quantitative analysis of the chemical

    constituents on a biofilm structure (70). On a similar

    line, biophysical techniques such as solid-state nuclear

    magnetic resonance (NMR) are powerful analyticaltools and have been applied to study the constituents

    of bacterial biofilm. NMR spectroscopy techniques

    have been used as a non-invasive method to obtain

    metabolic information of viable prokaryotic cell sus-

    pensions, eukaryotic cells, and tissue samples (95).

    NMR spectroscopy techniques are also useful to

    obtain metabolic information in planktonic cells,

    adherent bacterial cells, and in situ biofilm bacteria

    (94,95).

    Conclusion

    A variety of biofilm models are used for different experi-

    mental purposes in Endodontics today. One of the main

    issues for researchers is making a rational choice regard-

    ing the best model to use for their particular research

    problem. Generally, systems that closely reproduce invivoconditions should be chosen when the aim is solely

    to reproduce natural biofilms under laboratory condi-

    tions. However, there is no single, ideal biofilm model

    for all applications. Direct, non-destructive visualiza-

    tion of biofilms is advantageous in monitoring changes

    in biofilm bacteria and structures. Recent advances in

    CLSM, flow cytometry, micromanipulator-assisted

    analysis, GFP tagging, and FISH have made biofilm

    characterization very comprehensive. In spite of the

    amount of work carried out and the multitude of

    available methods, the quantification of bacterialbiofilm and the evaluation of the disinfectant activity

    remain a major challenge in Endodontics. Efforts are

    warranted to standardize the type of biofilm models,

    test methods, parameters used in the analysis, sample

    collection, and analysis of results.

    References

    1. Costerton JW, Lewandowski Z, DeBeer D, Caldwell D,Korber D, James G. Biofilms, the customized micro-

    niche. J Bacteriol1994: 176: 21372142.2. Nair PNR, Sjgren U, Krey G, Kahnberg K-E, Sund-qvist G. Intraradicular bacteria and fungi in root-filled,asymptomatic human teeth with therapy-resistant peri-apical lesions: a long-term light and electron micro-scopic follow-up study. J Endod1990: 16: 580588.

    3. Nair PNR. Light and electron microscopic studies ofroot canal flora and periapical lesions.J Endod1987: 13:2939.

    4. Nair PNR. On the causes of persistent apical periodon-titis: a review. Int Endod J2006: 39: 249281.

    5. Sundqvist G, Figdor D. Life as an endodontic pathogen:ecological differences between the untreated and root-filled root canals.Endod Topics2003: 6: 328.

    6. Baumgartner JC, Siqueira JR JF, Sedgley CM, KishenA. Microbiology of endodontic disease. In: Ingle JI,Bakland LK, Baumgartner JC, eds. Ingles Endodontics,6th edn. Hamilton: BC Decker, 2008: 221222.

    7. Grenier D, Mayrand D. Nutritional relationshipsbetween oral bacteria. Infect Immun 1986: 53: 616620.

    8. Sundqvist G. Ecology of the root canal flora. J Endod1992: 18: 427430.

    9. Casadevall A, Pirofski LA. Hostpathogen interactions:redefining the basic concepts of virulence and pathoge-nicity. Infect Immun1999:67: 37033713.

    Biofilm models and methods of biofilm assessment

    75

  • 8/13/2019 Biofilm Models and Methods Of

    19/21

    10. Chavez de Paz LE. Redefining the persistent infectionin root canals: possible role of biofilm communities. JEndod2007:33: 652662.

    11. Miron J, Ben-Ghedalia D, Morrison M. Invited review:adhesion mechanisms of rumen cellulolytic bacteria. JDairy Sci2001:84: 1294309.

    12. Cowan, MM, Taylor KG, Doyle RJ. Energetics of the

    initial phase of adhesion of Streptococcus sanguis tohydroxyapatite. J Bacteriol1987: 169: 29953000.

    13. Costerton JW, Stewart PS, Greenberg EP. Bacterialbiofilm: a common cause of persistent infections.Science1999: 284: 13181322.

    14. Costerton JW, Lewandowski Z. The biofilm lifestyle.Adv Dent Res1997: 11: 192195.

    15. del Pozo JL, Patel R. The challenge of treating biofilm-associated bacterial infections. Clin Pharmacol Ther2007: 82: 204209.

    16. Huang R, Li M, Gregory RL. Bacterial interactions indental biofilm. Virulence2011: 2: 435444.

    17. Costerton JW, Stewart PS. Battling biofilms. Sci Am2001: 285: 7481.

    18. Sun D, Accavitti MA, Bryers JD. Inhibition of biofilmformation by monoclonal antibodies against Staphy-lococcus epidermidis RP62A accumulation-associatedprotein.Clin Diagn Lab Immunol2005: 12: 93100.

    19. Stoodley P, Sauer K, Davies DG, Costerton JW. Bio-films as complex differentiated communities. AnnualRev Microbiol2002:56:187209.

    20. Lim Z, Cheng JL, Lim TW, Teo EG, Wong J, George S,Kishen A. Light activated disinfection: an alternativeendodontic disinfection strategy.Aust Dent J2009:54:108114.

    21. Shen Y, Stojicic S, Haapasalo M. Antimicrobial efficacyof chlorhexidine against bacteria in biofilms at different

    stages of development.J Endod2011: 37: 657661.22. Kolenbrander PE, Parrish KD, Andersen RN, Green-berg EP. Intergeneric coaggregation of oral Treponemaspp. with Fusobacterium spp. and intrageneric coaggre-gation among Fusobacterium spp. Infect Immun1995:63: 45844588.

    23. Jones SJ. A special relationship between spherical andfilamentous microorganisms in mature human dentalplaque.Arch Oral Biol1972:17: 613616.

    24. Rosan B, Correeia FF, DiRienzo JM. Corncobs: amodel for oral microbial biofilms. In: Busscher HJ,Evans LV, eds.Oral Biofilms and Plaque Control: Con-cepts in Dental Plaque Formation. Amsterdam:Harwood Academic Publishers, 1998: 145162.

    25. Upadya MH, Kishen A. Influence of bacterial growthmodes on the susceptibility to light-activated disinfec-tion.Int Endod J2010: 43: 978987.

    26. McBain AJ. Chapter 4: In vitro biofilm models: anoverview. Adv Appl Microbiol2009:69: 99132.

    27. Kishen A, Sum CP, Mathew S, Lim CT. Influence ofirrigation regimens on the adherence of Enterococcusfaecalis to root canal dentin. J Endod2008: 34: 850854.

    28. George S, Basrani B, Kishen A. Possibilities ofgutta-percha-centered infection in endodontically

    treated teeth: an in vitro study. J Endod 2010: 36:12411244.

    29. Mathew S, Yaw-Chyn L, Kishen A. Immunogenicpotential of Enterococcus faecalis biofilm undersimulated growth conditions. J Endod2010: 36: 832836.

    30. Pratten J, Ready D. Use of biofilm model systems to

    study antimicrobial susceptibility. Methods Mol Biol2010: 642: 203215.

    31. Merritt JH, Kadouri DE, OToole GA. Growing andanalyzing static biofilms. Curr Protoc Microbiol 2011:22: 1B.1.11B.1.18.

    32. Shabahang S, Torabinejad M. Effect of MTAD onEnterococcus faecalis-contaminated root canals ofextracted human teeth. J Endod2003: 29: 576579.

    33. Duggan JM, Sedgley CM. Biofilm formation of oral andendodontic Enterococcus faecalis. J Endod 2007: 33:815818.

    34. George S, Kishen A. Photophysical, photochemical, andphotobiological characterization of methylene blue for-mulations for light-activated root canal disinfection. J

    Biomed Opt2007: 12: 034029.35. George S, Kishen A. Augmenting the antibiofilm effi-

    cacy of advanced noninvasive light activated disinfectionwith emulsified oxidizer and oxygen carrier. J Endod2008: 34: 11191123.

    36. McGill S, Gulabivala K, Mordan N, Ng YL. The efficacyof dynamic irrigation using a commercially availablesystem (RinsEndo) determined by removal of a collagenbio-molecular film from an ex vivo model. Int Endod J2008: 41: 602608.

    37. Sainsbury AL, Bird PS, Walsh LJ. DIAGNOdent laserfluorescence assessment of endodontic infection. JEndod2009: 35: 14041407.

    38. Shen Y, Qian W, Chung C, Olsen I, Haapasalo M.Evaluation of the effect of two chlorhexidine prepara-tions on biofilm bacteria in vitro: a three-dimensionalquantitative analysis. J Endod2009: 35: 981985.

    39. Williamson AE, Cardon JW, Drake DR. Antimicrobialsusceptibility of monoculture biofilms of a clinicalisolate of Enterococcus faecalis. J Endod 2009: 35:9597.

    40. Shahriari S, Mohammadi Z, Mokhtari MM, Yousefi R.Effect of hydrogen peroxide on the antibacterialsubstantivity of chlorhexidine. Int J Dent2010: 2010:946384.

    41. Kishen A, Upadya M, Tegos GP, Hamblin MR. Effluxpump inhibitor potentiates antimicrobial photodynamic

    inactivation of Enterococcus faecalisbiofilm. PhotochemPhotobiol2010: 86: 13431349.

    42. Hiraishi N, Yiu CK, King NM, Tagami J, Tay FR.Antimicrobial efficacy of 3.8% silver diamine fluorideand its effect on root dentin.J Endod2010:36: 10261029.

    43. Shrestha A, Shi Z, Neoh KG, Kishen A. Nanoparticu-lates for antibiofilm treatment and effect of aging on itsantibacterial activity. J Endod2010: 36: 10301035.

    44. Liu H, Wei X, Ling J, Wang W, Huang X. Biofilmformation capability of Enterococcus faecalis cells in

    Kishen & Haapasalo

    76

  • 8/13/2019 Biofilm Models and Methods Of

    20/21

    starvation phase and its susceptibility to sodiumhypochlorite.J Endod2010: 36: 630635.

    45. Chvez de Paz LE, Bergenholtz G, Svenster G. Theeffects of antimicrobials on endodontic biofilm bacteria.J Endod2010:36: 7077.

    46. Su L, Gao Y, Yu C, Wang H, Yu Q. Surgical endodontictreatment of refractory periapical periodontitis with

    extraradicular biofilm.Oral Surg Oral Med Oral PatholOral Radiol Endod2010: 110: e4044.

    47. Soares JA, Roque de Carvalho MA, Cunha Santos SM,Mendona RM, Ribeiro-Sobrinho AP, Brito-Jnior M,Magalhes PP, Santos MH, de Macdo Farias L. Effec-tiveness of chemomechanical preparation with alter-nating use of sodium hypochlorite and EDTA ineliminating intracanal Enterococcus faecalis biofilm.J Endod2010:36: 894898.

    48. Bhuva B, Patel S, Wilson R, Niazi S, Beighton D, Man-nocci F. The effectiveness of passive ultrasonic irrigationon intraradicular Enterococcus faecalis biofilms inextracted single-rooted human teeth.Int Endod J2010:43: 241250.

    49. Shen Y, Stojicic S, Qian W, Olsen I, Haapasalo M. Thesynergistic antimicrobial effect by mechanical agitationand two chlorhexidine preparations on biofilm bacteria.J Endod2010:36: 100104.

    50. Pappen FG, Shen Y, Qian W, Leonardo MR, GiardinoL, Haapasalo M. In vitroantibacterial action of Tetra-clean, MTAD and five experimental irrigation solutions.Int Endod J2010:43: 528535.

    51. Shen Y, Stojicic S, Haapasalo M. Bacterial viability instarved and revitalized biofilms: comparison of viabilitystaining and direct culture. J Endod 2010: 36: 18201823.

    52. Lundstrom JR, Williamson AE, Villhauer AL, Dawson

    DV, Drake DR. Bactericidal activity of stabilized chlo-rine dioxide as an endodontic irrigant in a polymicrobialbiofilm tooth model system. J Endod2010:36: 18741878.

    53. Hope CK, Garton SG, Wang Q, Burnside G, FarrellyPJ. A direct comparison between extracted tooth andfilter-membrane biofilm models of endodontic irriga-tion using Enterococcus faecalis. Arch Microbiol 2010:192: 775781.

    54. Badr AE, Omar N, Badria FA. A laboratory evaluationof the antibacterial and cytotoxic effect of Liquorice

    when used as root canal medicament.Int Endod J2011:44: 5158.

    55. Pavarina AC, Dovigo LN, Sanit PV, Machado AL,

    Giampaolo ET, Vergani CE. Dynamic models forin vitrobiofilm formation. In: Bailey WC, ed. Biofilms:Formation, Development and Properties. Hauppauge,NY: Nova Science Publishers, Inc., 2011: 125162.

    56. Kulasekara HD, Ventre I, Kulasekara BR, Lazdunski A,Filloux A, Lory S. A novel two-component system con-trols the expression ofPseudomonas aeruginosafimbrialcup genes. Mol Microbiol2005:55: 368380.

    57. Watnick PI, Kolter R. Steps in the development of aVibrio choleraeEl Tor biofilm. Mol Microbiol1999:34:586595.

    58. Friedman L, Kolter R. Genes involved in matrix forma-tion in Pseudomonas aeruginosa PA14 biofilms. MolMicrobiol2004: 51: 675690.

    59. Davey ME, Otoole GA. Microbial biofilms: fromecology to molecular genetics. Microbiol Mol Biol Rev2000: 64: 847867.

    60. Johansen C, Falholt P, Gram L. Enzymatic removal and

    disinfection of bacterial biofilms.Appl Environ Microbiol1997: 63: 37243728.

    61. Di Bonaventura G, Pompilio A, Picciani C, Iezzi M,DAntonio D, Piccolomini R. Biofilm formation by theemerging fungal pathogenTrichosporon asahii: develop-ment, architecture, and antifungal resistance. Anti-microb Agents Chemother2006:50: 32693276.

    62. Burnett SL, Chen J, Beuchat LR. Attachment ofEscherichia coliO157:H7 to the surfaces and internalstructures of apples as detected by confocal scanninglaser microscopy. Appl Environ Microbiol 2000: 66:46794687.

    63. Takeuchi K, Frank JF. Expression of red-shifted greenfluorescent protein by Escherichia coli O157:H7 as a

    marker for the detection of cells on fresh produce. JFood Prot2001:64: 298304.

    64. McKinlay KJ, Allison FJ, Scotchford CA, Grant DM,Oliver JM, King JR, Wood JV, Brown PD. Comparisonof environmental scanning electron microscopy withhigh vacuum scanning electron microscopy as applied tothe assessment of cell morphology. J Biomed Mater ResA2004: 69: 359366.

    65. Sutton NA, Hughes N, Handley PS. A comparison ofconventional SEM techniques, low temperature SEMand the electroscan wet scanning electron microscope tostudy the structure of a biofilm of Streptococcus cristaCR3. J Appl Bacteriol1994: 76: 448454.

    66. Trachoo N, Frank JF. Effectiveness of chemical sani-tizers againstCampylobacter jejuni-containing biofilms.J Food Prot2002:65: 11171121.

    67. Wolfaardt GM, Lawrence JR, Hendry MJ, Robarts RD,Caldwell DE. Development of steady-state diffusiongradients for the cultivation of degradative microbialconsortia. Appl Environ Microbiol 1993: 59: 23882396.

    68. Harraghy N, Seiler S, Jacobs K, Hannig M, MengerMD, Herrmann M. Advances in in vitro and in vivomodels for studying the staphylococcal factors involvedin implant infections. Int J Artif Organs 2006: 29:368378.

    69. Sheppard, CRJ, Shotton, DM.Confocal Laser Scanning

    Microscopy. Oxford, UK: BIOS Scientific PublishersLtd., 1997.

    70. Kishen A, George S, Kumar R. Enterococcus faecalis-mediated biomineralized biofilm formation on rootcanal dentinein vitro.J Biomed Mater Res A2006:77:406415.

    71. Bakke R, Olsson PQ. Biofilm thickness measurementsby light microscopy. J Microbiol Methods 1986: 5:9398.

    72. Peyton BM, Characklis WG. A statistical analysis of theeffect of substrate utilization and shear stress on the

    Biofilm models and methods of biofilm assessment

    77

  • 8/13/2019 Biofilm Models and Methods Of

    21/21

    kinetics of biofilm detachment.Biotechnol Bioeng1993:41: 728735.

    73. Main C, Geddes DA, McNee SG, Collins WJ, SmithDC, Weetman DA. Instrumentation for measurementof dental plaque thickness in situ.J Biomed Eng1984: 6:151154.

    74. Mattila-Sandholm T, Wirtanen G. Biofilm formation

    in the industry: a review. Food Rev Int 1992: 8:573603.

    75. Stewart GS.In vivobioluminescence: new potentials formicrobiology. Lett Appl Microbiol1990:10: 18.

    76. Walker AJ, Stewart GS, Sheppard F, Bloomfield SF,Holah JT, Denyer SP. Bioluminescence imaging as atool for studying biocide challenge upon planktonic andsurface attached bacteria. Bin Comp Microbiol1994:6:1617.

    77. Marshall KC, Stout R, Mitchell R. Mechanisms of theinitial events in the sorption of marine bacteria to solidsurfaces. J Gen Microbiol1971:68: 337348.

    78. Marshall KC. Colonization, adhesion and biofilms. In:Hurst CJ, Knudsen GR, McInerney MJ, Stetzenbach

    LD, eds. Manual of Environmental Microbiology.Washington, DC: American Society for MicrobiologyPress, 1997: 35865.

    79. DeBeer D, Stoodley P, Roe F, Lewandowski Z.Effects of biofilm structures on oxygen distribution andmass transport. Biotechnol Bioeng 1994: 43: 11311138.

    80. McAllister TA, Bae HD, Jones GA, Cheng KJ. Micro-bial attachment and feed digestion in the rumen. JAnim Sci1994: 72: 30043018.

    81. Millsap KW, Reid G, Van Der Mei HC, Busscher HJ.Adhesion of Lactobacillus species in urine and phos-phate buffer to silicone rubber and glass under flow.

    Biomaterials1997: 18: 8791.82. Lee HA, Wyatt GM, Bramham S, Morgan MR.Enzyme-linked immunosorbent assay for Salmonellatyphimurium in food: feasibility of 1-day Salmonelladetection. Appl Environ Microbiol 1990: 56: 15411546.

    83. Bauer-Kreisel P, Eisenbeis M, Scholz-Muramatsu H.Quantification ofDehalospirillum multivoransin mixed-culture biofilms with an enzyme-linked immunosorbentassay. Appl Environ Microbiol 1996: 62: 30503052.

    84. Prez-Osorio AC, Franklin MJ. qRT-PCR of microbialbiofilms.CSH Protoc2008:2008: pdb.prot5066.

    85. Kindaichi T, Kawano Y, Ito T, Satoh H, Okabe S.Population dynamics and in situ kinetics of nitrifyingbacteria in autotrophic nitrifying biofilms as determinedby real-time quantitative PCR. Biotechnol Bioeng2006:94: 11111121.

    86. Heydorn A, Ersbll B, Kato J, Hentzer M, Parsek MR,Tolker-Nielsen T, Givskov M, Molin S. Statistical analy-

    sis of Pseudomonas aeruginosa biofilm development:impact of mutations in genes involved in twitchingmotility, cell-to-cell signaling, and stationary-phasesigma factor expression. Appl Environ Microbiol2002:68: 20082017.

    87. Wimpenny J, Manz W, Szewzyk U. Heterogeneity inbiofilms.FEMS Microbiol Rev2000: 24: 661671.

    88. Thurnheer T, Gmr R, Guggenheim B. Multiplex FISHanalysis of a six-species bacterial biofilm. J MicrobiolMethods2004: 56: 3747.

    89. Postollec F, Norde W, de Vries J, Busscher HJ, van derMei HC. Interactive forces between co-aggregating andnon-co-aggregating oral bacterial pairs. J Dent Res2006: 85: 231234.

    90. Razatos A, Ong YL, Sharma MM, Georgiou G. Molecu-lar determinants of bacterial adhesion monitored byatomic force microscopy. Proc Natl Acad Sci USA1998: 95: 1105911064.

    91. Gaboriaud F, Dufrne YF. Atomic force microscopy ofmicrobial cells: application to nanomechanical proper-ties, surface forces and molecular recognition forces.Colloids Surf B Biointerfaces2007:54: 1019.

    92. Vadillo-Rodrguez V, Busscher HJ, Norde W, De VriesJ, Dijkstra RJ, Stokroos I, Van Der Mei HC. Compari-son of atomic force microscopy interaction forcesbetween bacteria and silicon nitride substrata for threecommonly used immobilization methods.Appl Environ

    Microbiol2004:70: 54415446.93. Sum C, Mohanty S, Gupta PK, Kishen A. Influence ofendodontic chemical treatment onEnterococcus faecalisadherence to collagen studied with laser scanning con-focal microscopy and optical tweezers: a preliminarystudy. J Biomed Opt2008:13: 044017.

    94. Grivet JP, Delort AM, Portais JC. NMR and micro-biology: from physiology to metabolomics. Biochimie2003: 85: 823840.

    95. Majors PD, McLean JS, Pinchuk GE, Fredrickson JK,Gorby YA, Minard KR, Wind RA. NMR methods forinsitu biofilm metabolism studies. J Microbiol Methods2005: 62: 337344.

    Kishen & Haapasalo