genomic and proteomics

Upload: niko-hizkia-simatupang

Post on 06-Jul-2018

217 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/17/2019 Genomic and Proteomics

    1/12

    21st Century Directions in Biology 

    Since the first recognition of microorganisms,scientists have devised classification schemes with thegoal of systematically identifying species in an evolutionary 

    or phylogenetic context (Clarke 1985). This has consistently 

    proved more challenging for bacteria than for macroorgan-isms. Bacteria are asexual, so the classic definition of a speciesas a group of organisms that can interbreed and produce

    fertile offspring is difficult to apply. Furthermore, because of 

    their small size, bacteria have a limited range of morpholog-

    ical attributes. They do exhibit enormous biochemical diversity in both their metabolism and cell structure; this has proved

    to be a useful cue for the taxonomy of some groups, but by 

    no means all of them. It is important, then, that the molec-

    ular revolution that has transformed all of biology has had asgreat an impact on the taxonomy and systematics of bacteria

    as in any other area of biology. In the 1970s, on the basis of 

    molecular comparisons of evolutionarily conserved riboso-

    mal genes, Carl Woese proposed the then-heretical notion thatthe bacteria actually made up two separate domains, theBacteria and the Archaea, each as distinct from one another

    as they are from the Eukaryotes, the third domain that com-

    prises all “higher forms” of life (Woese 1987). This classifi-

    cation, supported by reams of additional molecular data, isnow the standard view among microbiologists. This is not to

    say that bacterial systematics is now fully standardized; indeed,

    there is a vigorous ongoing debate about what constitutes a

    bacterial species (Gevers et al. 2005, Achtman and Wagner2008). Nonetheless, molecular systematics has provided the

    crucial framework for building bacterial classification schemes.

    Despite the lack of a coherent species definition, the timely 

    classification, characterization, and identification of bacteriacontinue to be critical in many areas, including public health,

    clinical diagnosis, environmental monitoring, food safety 

    monitoring, and identification of biological threat agents.In particular, the recent advances of modern molecular tech-niques in genomics and proteomics have offered attractive

    alternatives to conventional microbiological procedures for

    characterizing and identifying microorganisms. These new 

    methods can provide a rapid, multidimensional data outputwith taxonomically relevant molecular information on both

    individual strains and whole populations.

    This article is primarily concerned with the identification

    of individual strains of bacteria that can be grown as axenic

    David Emerson is a senior research scientist at Bigelow Laboratory for Ocean

    Sciences, West Boothby Harbor, Maine. He is an environmental microbiologist 

    who has worked on developing methods for the rapid identification of a

    variety of environmentally important bacteria and archaea. Liane Agulto

    is a biologist at the American Type Culture Collection (ATCC) in Manassas,

    Virginia; her major interest is identifying disease-related biomarkers using

     proteomics technology. Henry Liu, a graduate student at Georgetown University 

    School of Medicine in Washington, DC, interned at the ATCC, where he

    worked on developing a diagnostic kit for rapid antimicrobial susceptibility

    testing. Liping Liu (e-mail: [email protected]) was the head of proteomics at the

     ATCC and is now the director of research and development at Stealth Peptides,

    Inc., in Rockville, Maryland. Her major expertise is in identifying biomarkers

    using proteomics technologies and in developing biotherapeutics. © 2008

     American Institute of Biological Sciences.

    Identifying and Characterizing 

    Bacteria in an Era of Genomics

    and Proteomics

    DAVID EMERSON, LIANE AGULTO, HENRY LIU, AND LIPING LIU

    The advent of new molecular technologies in genomics and proteomics is shifting traditional techniques for bacterial classification, identification,and characterization in the 21st century toward methods based on the elucidation of specific gene sequences or molecular components of a cell. Wediscuss current genotypic and proteomics technologies for bacterial identification and characterization, and present an overview of how these new 

    technologies complement conventional approaches. The new methods can be rapid, offer high throughput, and produce unprecedented levels of discrimination among strains of bacteria and archaea. Remaining challenges include developing appropriate standards and methods for thesetechniques’ routine application and establishing integrated databases that can handle the large amounts of data that they generate. We conclude by discussing the impacts of rapid bacterial identification on the environment and public health, as well as directions for future development in this

     field.

    Keywords: bacterial identification, bacterial characterization, genotype, proteomics, bioinformatics

    www.biosciencemag.org November 2008 / Vol. 58 No. 10 • BioScience 925

    21st Century Directions in Biology 

  • 8/17/2019 Genomic and Proteomics

    2/12

    cultures in the laboratory. The discrimination and identifi-cation of bacteria within mixed natural populations is also a

    rapidly developing field that utilizes some of the same tech-

    niques, but it is an entirely separate subject (Liu and Stahl 2007,

    Logue et al. 2008). Methods of bacterial identification can bebroadly delimited into genotypic techniques based on profiling

    an organism’s genetic material (primarily its DNA) and phe-notypic techniques based on profiling either an organism’s

    metabolic attributes or some aspect of its chemical compo-sition. Genotypic techniques have the advantage over pheno-

    typic methods that they are independent of the physiological

    state of an organism; they are not influenced by the com-

    position of the growth medium or by the organism’s phaseof growth. Phenotypic techniques, however, can yield more

    direct functional information that reveals what metabolic

    activities are taking place to aid the survival, growth, and

    development of the organism. These may be embodied, forexample, in a microbe’s adaptive ability to grow on a certain

    substrate, or in the degree to which it is resistant to a cohort

    of antibiotics. Because genotypic and phenotypic approaches

    are complementary and use different techniques, this review is divided into two parts. However, this division is historical;

    we predict that as molecular-based identification matures,

    there will be more and more overlap in the information

    obtained using different methodologies.

    Genotypic methodsGenotypic microbial identification methods can be broken

    into two broad categories: (1) pattern- or fingerprint-basedtechniques and (2) sequence-based techniques. Pattern-based

    techniques typically use a systematic method to produce a

    series of fragments from an organism’s chromosomal DNA.These fragments are then separated by size to generate a pro-file, or fingerprint, that is unique to that organism and its very 

    close relatives. With enough of this information, researchers

    can create a library, or database, of fingerprints from known

    organisms, to which test organisms can be compared. Whenthe profiles of two organisms match, they can be considered

    very closely related, usually at the strain or species level.

    Sequence-based techniques rely on determining the

    sequence of a specific stretch of DNA, usually, but not always,associated with a specific gene. In general, the approach is the

    same as for genotyping: a database of specific DNA sequences

    is generated, and then a test sequence is compared with it.

    The degree of similarity, or match, between the two sequencesis a measurement of how closely related the two organismsare to one another. A number of computer algorithms have

    been created that can compare multiple sequences to one

    another and build a phylogenetic tree based on the results

    (Ludwig and Klenk 2001). The example cited above of usingsequence comparisons of the ribosomal RNA (rRNA) gene

    to distinguish bacteria and archaea demonstrates how this

    information can be applied to identify relationships among

    microorganisms.Both fingerprinting techniques and sequence-based meth-

    ods have strengths and weaknesses. Traditionally, sequence-

    based methods, such as analysis of the 16S rRNA gene, haveproved effective in establishing broader phylogenetic rela-

    tionships among bacteria at the genus, family, order, and

    phylum levels, whereas fingerprinting-based methods are

    good at distinguishing strain- or species-level relationships butare less reliable for establishing relatedness above the species

    or genus level (Vandamme et al. 1996). When these methodsare coupled with other phenotypic tests, this creates a polypha-

    sic approach that is the standard for describing new bacter-ial species (Gillis et al. 2001).

     Specific genotyping methodologiesCurrent protocols for the identification of bacteria may uti-lize a variety of different fingerprinting- or sequence-based

    methods, either alone or, more often, in combination. These

    techniques are constantly evolving to embrace new method-

    ologies that provide both greater accuracy for identificationand higher sample throughput. Examples of some of the

    most widely used techniques are provided below.

    Fingerprinting-based methodologies. At present, fingerprintingtechniques are the most commonly used genotypic methods

    for bacterial identification. The most widely used of these

    methods are shown in table 1. Repetitive element PCR (rep-

    PCR), amplified fragment length polymorphism (AFLP),and random amplification of polymorphic DNA all utilize

    PCR to amplify multiple copies of short DNA fragments

    using defined sets of primers (Versalovic et al. 1994, Coccon-

    celli et al. 1995, Vos et al. 1995, Lin et al. 1996). These meth-ods are designed to take advantage of DNA polymorphisms

    in related organisms that may accrue as a result of a variety 

    of evolutionary mechanisms. Figure 1 provides an illustrationof the type of data obtained using rep-PCR. Multiplex PCR uses unique PCR primer sets for more than one organism;

    these sets can be separated on the basis of amplicon size as a

    way of rapidly identifying more than one microbe at a time

    in a mixed sample (Settanni and Corsetti 2007).Riboprinting does not use PCR, but instead utilizes a sen-

    sitive probing method to detect differences in gene patterns

    between strains and species (Bruce 1996). DuPont’s Ribo-

    Printer system (www2.dupont.com/Qualicon/en_US/ ) andthe DiversiLab system for rep-PCR (http://biomerieux-usa.

    com/diversilab) have both been developed as commercial

    products for bacterial identification. All of the methods de-

    scribed here have been used to identify bacteria in a multitudeof different ways, many of which can be found in the scien-tific literature. These applications include source tracking

    (Meays et al. 2004), authentication of isolates for archival

    purposes (Cleland et al. 2008), taxonomy and systematics

    (Vandamme et al. 1996, Gevers et al. 2005), and determina-tion of microbial population structures and community 

    studies (Savenlkoul et al. 1999), to name but a few.

     Sequence-based methodologies. The most widely usedsequence-based methods are also shown in table 1. Multilocus

    sequencing is one of the newest and, to date, one of the most

    21st Century Directions in Biology 

    926 BioScience •  November 2008 / Vol. 58 No. 10 www.biosciencemag.org 

    21st Century Directions in Biology 

  • 8/17/2019 Genomic and Proteomics

    3/12

    powerful methods developed to identify microbial species. In

    principle, this technique is akin to 16S rRNA gene sequence

    comparisons, except that, instead of one gene, the fragments

    of multiple “housekeeping” genes are each sequenced, andthe combined sequences are put together, or concatenated,

    into one long sequence that can be compared with other se-

    quences. Housekeeping genes are generally defined as en-

    coding for proteins that carry out essential cellular processes.A few examples include the gyrase B subunit ( gyrB); the

    alpha and beta subunits of RNA polymerase (rpoA and rpoB);

    and recA, a gene encoding for an enzyme important in DNA

    repair; there are a host of others (Zeigler 2003). Housekeep-ing-gene loci are present in most cells and tend to be conservedamong different organisms. As a result, general-purpose

    primers can be designed that will work using PCR to amplify 

    the same genes across multiple genera.

    In practice, the story is a bit more complicated; in mostcases, truly universal primer sets are not possible, so primers

    need to be designed for specific families or orders of bacteria.

    Two multilocus sequencing strategies are currently used:

    multilocus sequence typing (MLST) and multilocus sequenceanalysis (MLSA). MLST is a well-defined approach that uses

    a suite of 6 to 10 genetic loci, with appropriate primers for each

    locus to allow PCR amplification and sequencing of the

    products (usually 400 to 600 base pairs) (Maiden et al. 1998).

    The resulting concatenated sequences can then be compared

    with a curated database of sequences for the same organism.The result provides a high-resolution identification of an in-

    dividual strain that may reveal close evolutionary relationships

    among individual strains. This technique has proved useful

    in epidemiological studies, making it possible to track the out-break of virulent bacterial pathogens (Cooper and Feil 2004).

    Thus far, MLST, and the robust databases that have been

    created for it, has been applied only to a relatively small num-

    ber of common pathogens, using highly prescribed conditionsfor each organism, both for PCR primers and for databaseanalysis.

    MLSA also involves sequencing of multiple fragments of 

    conserved protein encoding genes, but it uses a more ad hoc

    approach to choosing the genes for comparative analysis. Asmaller subset (≤ 6) of genes or loci is typically used in MLSA

    than is used in MLST (Gevers et al. 2005). MLSA is typically 

    used to identify organisms in the broader context of probing

    species relationships within genera of families, rather thantracking the history of individual strains. As typically ap-

    plied, it does not have the analytical capacity to detect the very 

    21st Century Directions in Biology 

    www.biosciencemag.org November 2008 / Vol. 58 No. 10 • BioScience 927

    21st Century Directions in Biology 

    Table 1. Genotypic methods used in identifying bacteria.

    Method Technology Application Database

    Common fingerprinting methods

    Repetitive element polymerase Polymerase chain reaction (PCR) Identification at the species and User created; commercial individualchain reaction (rep-PCR) primers target specific repetitive strain levels database available

    elements randomly distributed inthe chromosomes of bacteria andarchaea

    Amplified fragment length Restriction digestion of chromosomal Identification at the species and User createdpolymorphism DNA is followed by PCR using adapters strain levels; can be used for both

    coupled to the restriction sites bacteria and archaea

    Riboprinting Restriction digest of chromosomal DNA Identification at the species and User created; commercial universalis followed by probing for ribosomal strain levels; often used in quality database availablegenes control and assurance

    Random amplification of A set of arbitrary short primers are Comparison of strains of known User createdpolymorphic DNA used to randomly amplify short species

    stretches of chromosomal DNA

    Pulsed-field gel electrophoresis Chromosomal DNA is cut into large Typing of pathogenic bacteria Public universal database adminis-fragments with rare-cutting restriction tered by the Centers for Diseaseenzymes, and fragment is determined Control and Prevention (www.cdc.gov/ 

    pulsenet)

    Multiplex PCR Multiple PCR primers are used for Identif ication of multiple species in User created

    diagnostic genes mixed samples such as food andclinical specimens

    Common DNA sequencing methods

    Small-subunit ribosomal gene Conserved primers are used to amplify The current gold standard in bacterial Public universal databases available,sequencing (SSU rDNA) and then sequence the SSU rDNA identification and determination of including the Ribosomal Database

    gene; sequences are then compared evolutionary relationships; may not Project (http://rdp.cme.msu.edu) andwith a database distinguish strains or species within greengenes (http://greengenes.lbl.gov )

    a genus

    Multilocus sequence typing DNA sequencing of a specific subset Typing of pathogenic bacteria; Public universal database available(MLST) of conserved and semiconserved epidemiology (administered by www.mlst.net)

    genes for a given species is followedby a comparison of concatenatedsequences

    Multilocus sequence analysis DNA sequencing of a specific subset Provides more robust identification User created; based on BLAST (Basicof conserved genes is followed by at the species level than traditional Local Alignment Search Tool) searchesa comparison of concatenated SSU rRNA gene sequencing against GenBanksequences; this method typicallyuses fewer genes than MLST does(only two or three)

  • 8/17/2019 Genomic and Proteomics

    4/12

    minor changes in sequence patterns that are useful in epi-

    demiologic studies. At present, MLSA is limited by a lack of standardization, and no central databases are available. For ex-ample, an analysis of eight recent papers that used MLSA to

    identify a wide range of bacterial phyla found that anywhere

    from two to six genes were used in the different individual

    studies (Devulder et al. 2005, Lodders et al. 2005, Naser et al.2005, Paradis et al. 2005, Thompson et al. 2005, Richter et al.

    2006, Chelo et al. 2007, Richert et al. 2007). Furthermore, no

    single gene was common to all studies, and most studies

    used completely different sets of genes. Although the techniqueproved useful for each individual study, the lack of cohesive-

    ness makes comprehensive comparative analyses impossible.

     The genomic future. The genomes of approximately 2000

    strains of bacteria and archaea have now been sequenced orare in the process of being sequenced. This has led to theadvent of using whole-genome comparisons between related

    species to determine the average nucleotide identity between

    two genomes (Goris et al. 2007). This technique currently de-

    fines a species at the genomic level as having 95% averagenucleotide identity between two strains. This corresponds to

    an estimate of at least 70% reassociation for DNA-DNA

    hybridization, which has been the traditional standard for

    defining bacterial species (Vandamme et al. 1996). Completegenome comparisons have proved to be more accurate than

    DNA-DNA hybridization, which requires very stringent

    928 BioScience •  November 2008 / Vol. 58 No. 10 www.biosciencemag.org 

    21st Century Directions in Biology 

    Figure 1. An example of genotyping archaea from extreme environments using repetitive element poly-

    merase chain reaction. The dendrogram compares the barcodes derived from four genera of extremely thermophilic archaea ( Methanocaldococcus, Methanotorris, Sulfolobus , and Thermococcus ) associ-

    ated with high-temperature environments like the hydrothermal vent shown in the photograph on the 

    left, and four genera of halophilic archaea ( Halobacterium, Haloferax, Haloarcula,and Halorubrum )

    associated with high-salt environments like the solar saltern shown on the right. This analysis providesa highly specific method for strain identification of these unique organisms, but it does not provide in-

     formation regarding their phylogenetic relatedness. Photographs: David Clelann.

  • 8/17/2019 Genomic and Proteomics

    5/12

    protocols and is often difficult to reproduce precisely be-

    tween laboratories. The rapid advent of the next generation

    of sequencing technologies is likely to make sequence-based

    methods more cost-effective and more readily available for use

    at all levels of bacterial classification and identification.

    This raises the question of whether it will soon be possi-

    ble to simply sequence the genome of an isolate to determinewhat it is and what it does. Can this be done for roughly the

    same cost as a standard battery of biochemical identification

    tests and a genotype analysis? Can it be done with the same

    or better speed and efficiency as current methods? These are

    the challenges faced by researchers who are developing and

    using genomics-based identification methods. It is difficult to

    predict how soon, if ever, whole-genome sequencing will be

    used as a routine means of bacterial identification; however,

    it is certain that the multilocus sequencing approaches de-

    scribed above will expand and mature rapidly. While we ap-

    preciate the technological challenges of DNA sequencing per

    se, perhaps an even greater challenge will be the establishmentof large, integrated databases that allow for the rapid assem-

    bly of sequence data to help researchers make robust com-

    parisons among sequences and predict identifications between

    bacteria with a high degree of confidence. The lack of stan-

    dardization for MLSA analysis needs to be addressed so that

    standards can be developed for comparisons of multiple

    taxa. Once these are in place, it will become progressively

    easier to develop MLSA- or MLST-type sequence-based

    strategies that accurately target multiple genes and can be used

    to provide a full range of genotypic information for all

    bacteria and archaea.

    Microarrays are another technology that shows promise asa means of simultaneously identifying specific microbes and

    providing ecological context for the population structure

    and functional structure of a given microbial community. Mi-

    croarrays work on the general principle of spotting probes for

    hundreds or thousands of genes onto a substrate (e.g., a glass

    slide) and then hybridizing sample DNA or RNA to it. The

    sample DNA or RNA is labeled with a fluorescent reporter

    molecule so that samples that hybridize with probes on the

    microarray can be detected rapidly. In terms of bacterial

    identification, several iterations of a “phylochip” that utilizes

    the small-subunit ribosomal gene as a target have been de-

    veloped, both for specific and for very broad groups of envi-ronmental bacteria (Liu et al. 2001, Wilson et al. 2002).

    Another example is the geochip, which has been developed

    to identify microbes involved in essential biogeochemical

    processes such as metal transformations, contaminant degra-

    dation, and primary carbon cycling (He et al. 2007). In the clin-

    ical realm, the use of microarrays is moving forward rapidly,

    both for diagnostic purposes and for understanding the fun-

    damentals of disease pathology (Frye et al. 2006, Richter et al.

    2006). However, because of their inherent complexity and rel-

    ative expense, microarrays have yet to be used as standard

    methods in microbial identification.

    Proteomics technologies in bacterialidentification and characterizationAlthough genotypic information is valuable in identifying an

    organism and determining how it is related to others, meth-

    ods that probe an organism’s phenotypic properties remaincritical for understanding the physiological and functional

    activities of an organism at the protein level. Phenotypicmethods that determine the activity of specific enzymes,

    such as catalase or oxidase, or metabolic functions, such as theability to degrade lactose, have long been a mainstay of bac-

    terial identification. The advent of new proteomics tools that

    are based primarily on mass spectrometry and allow rapid in-

    terrogation of biomolecules produced by an organism offersan excellent complement to classical microbiological and

    genomics-based techniques for bacterial classification, iden-

    tification, and phenotypic characterization. What is also in-

    teresting is that some of these techniques are integratinggenotypic and proteomic data to provide more complete

    information. The predominant proteomic technologies that

    have been explored for bacterial identification and charac-

    terization include matrix-assisted laser desorption/ionizationtime-of-flight mass spectrometry (MALDI-TOF-MS); elec-

    trospray ionization mass spectrometry (ESI-MS); surface-

     enhanced laser desorption/ionization (SELDI) mass

    spectrometry; one- or two-dimensional sodium dodecylsulfate–polyacrylamide gel electrophoresis (SDS-PAGE); or

    the combination of mass spectrometry, gel electrophoresis, and

    bioinformatics. (See figure 2 for a general integrated proteo-

    mics flowchart.) In addition to the above-mentioned classi-cal proteomics approaches, Fourier-transform infrared

    spectroscopy (FT-IR) has been used to classify and identify 

    bacterial samples (see, e.g., Al-Qadiri et al. 2006).

    Mass spectrometry–based bacterial characterization and

    identification. Mass spectrometry is a powerful analytical

    technique that has been used to identify unknown com-

    pounds, quantify known compounds, and elucidate the struc-ture and chemical properties of molecules. The development

    of mass spectrometry can be traced back to the late 19th

    century, when it was first used by J. J. Thomson (1899) to

    measure the mass-to-charge ratio of electrons. With the re-finement of this technology throughout the 20th century,

    mass-spectrometry applications have been expanded to

    include physical measurement, chemical characterization,

    and biological identification.One of the major breakthroughs in mass spectrometry 

    for the analysis of biological molecules was the soft ionization

    method (i.e., MALDI-TOF-MS and ESI-MS; see figure 3 for

    a simplified schematic representation). Until the develop-

    ment of the soft ionization method, the application of massspectrometry to biological materials was limited by the re-

    quirement that the sample be in vapor phase before ioniza-

    tion. Soft ionization has made it possible to study larger

    biological molecules and perform analyte sampling and ion-ization directly from native samples, including whole cells,

    using mass spectrometry (Fenn et al. 1989). Since its initial

    www.biosciencemag.org November 2008 / Vol. 58 No. 10 • BioScience 929

    21st Century Directions in Biology 

  • 8/17/2019 Genomic and Proteomics

    6/12

  • 8/17/2019 Genomic and Proteomics

    7/12

    MALDI-TOF-MS is rapidly becoming an accepted tech-

    nology for bacterial identification, so we will cite only one

    example of its use. Staphylococcus aureus, a bacterium com-

    monly found on human skin, causes infection during timesof uncontrolled growth. Improper use of antibiotics has

    rendered S. aureus resistant to the methicillin class of anti-

    biotics. The first outbreak of methicillin-resistant S. aureus(MRSA) was recorded in a European hospital in the early 1960s. Since then, the threat of MRSA has spread from

    hospitals and clinical settings to schools and public commu-

    nities, thus necessitating the use of techniques that can rapidly 

    identify and discriminate MRSA from methicillin-sensitive S.aureus (MSSA). Edward-Jones and colleagues (2000) developeda MALDI-TOF-MS method for the identification, typing,

    and discrimination of MRSA and MSSA. In this method, a

    sample is taken from a single bacterial colony and smeared

    onto a sample slide. The appropriate matrix is applied to thesample, which is then analyzed using MALDI-TOF-MS.

    MALDI-TOF-MS analysis shows that MRSA and MSSA yield

    distinct spectral peaks that allow for rapid distinction between

    the two and therefore, hypothetically, for appropriate treat-ment of S. aureus infections with respect to their resistance

    to antibiotics.

    To serve market needs, severalinstrument systems based on

    MALDI-TOF-MS have been

    developed for bacterial identi-

    fication and characterization.For instance, Bruker Daltonics’

    MALDI BioTyper is a systembased on the measurement of 

    high-abundance proteins, includ-ing many ribosomal proteins in

    a microorganism (Mellmann et

    al. 2008). The system is equipped

    with bioinformatics tools (clus-tering and phylogenetic dendro-

    gram construction) that allow 

    for the rapid identification and

    characterization of a known orunknown bacterial culture on the

    basis of proteomics signatures.

    Electrospray ionization mass

    spectrometry. ESI-MS also has

    the potential to play an impor-

    tant role in bacterial character-

    ization, especially for the analysisof cellular components. Proteins

    expressed by the bacteria can be

    extracted from the lysed cells

    and analyzed using ESI-MS.This technique allows for the

    analysis of both intracellular and

    extracellular proteins, carbo-hydrates, and lipids. A major ad-

    vantage of ESI-MS is its ability to perform tandem mass

    spectrometry, in which the protein of interest can be frag-

    mented for a second mass analysis that provides protein frag-

    ment sequence information, or a peptide fragmentationfingerprint, that can then be applied to a database search to

    identify that specific protein. This has significantly increased

    the accuracy of protein identification compared with identi-

    fication using only molecular weight information from asingle MALDI-TOF-MS analysis. A study by Krishnamurthy 

    and Ross (1996) reported that the total analysis time leading

    to unambiguous bacterial identification in samples is less

    than 10 minutes, with reproducible results. A system recently developed by Ibis Biosciences (the T5000 Biosensor System)can identify and characterize bacterial strains rapidly and

    effectively using a combination of PCR and ESI-MS tech-

    nology (Sampath et al. 2007). Another example of the use of 

    mass spectrometry to analyze nucleic acids for bacterial iden-tification is the combination of MALDI-TOF-MS with MLST

    analysis using Neisseria meningitidis (Honisch et al. 2007).

    These efforts exemplify how the integrated genotypic and pro-

    teomics technologies provide an even more powerful tool forbacterial identification. Additional descriptions of the ESI-MS

    technique and its applications for bacterial characterization

    www.biosciencemag.org November 2008 / Vol. 58 No. 10 • BioScience 931

    21st Century Directions in Biology 

    Figure 3. Schematic representation of soft ionization techniques used in mass spectrome-

    try. (a) Matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass

    spectrometry. The sample to be analyzed (the analyte) is mixed with organic matrices

    and deposited on the sample plate in the form of a small spot. The mixture is ionizedby the laser beam. The resulting ions move toward the mass analyzer, and the mass is

    detected to obtain the mass spectrum. (b) Electrospray ionization mass spectrometry

    (ESI-MS). The analyte is mixed with a solvent and sprayed from a narrow tube.

    Positively charged droplets in the spray move toward the mass-spectrometersampling orifice under the influence of electrostatic forces and pressure differentials.

     As the droplets move to the orifice, the solvent evaporates, causing the analyte

    ions to move toward the analyzer for mass analysis.

  • 8/17/2019 Genomic and Proteomics

    8/12

    may be found in a review article by Bons and colleagues(2005).

    Another advantage of ESI-MS is its ability to identify

    target bacteria in mixed samples. The resolution of ESI-MS

    is such that specific intracellular biomarkers for individual mi-croorganisms can be distinguished with enough confidence

    that they can be identified in unknown samples. For the pro-tein analysis, comparing the experimentally obtained protein

    profile of an unknown bacterial species with the profile in-formation found in a proteomics database will allow for the

    identification and characterization of the unknown species.

    Identifying the virulence factors of pathogenic bacteria is

    one of the major applications of liquid chromatography withtandem mass spectrometry (LC/MS/MS) (Chao et al. 2007).

    Once the putative virulence factors are identified, their func-

    tions and mechanism can be further characterized by pheno-

    typic analyses such as mutagenesis, conventional biochemicalmethods, and structural biology.

     Surface-enhanced laser desorption/ionization. SELDI is a

    relatively new technology, designed to perform mass spec-trometric analysis of protein mixtures retained on chemically 

    (e.g., cationic, ionic, hydrophobic) or biologically (e.g.,

    antibody, ligand) modified chromatographic chip surfaces.

    These varied chemical and biochemical surfaces allow dif-ferential capture of proteins based on the intrinsic properties

    of the proteins themselves. The SELDI mass spectrometer

    produces spectra of complex protein mixtures based on the

    mass-to-charge ratio of the proteins in the mixture and theirbinding affinity to the chip surface. Differentially expressed

    proteins may then be determined from these protein profilesby comparing peak intensity. Figure 4 illustrates the general

    procedure.

    SELDI technology has been applied extensively to bio-

    marker and protein profiling studies in the field of oncology (Yip and Lomas 2002). By contrast, only a limited number of 

    reports have investigated the applicability of SELDI for de-tecting and identifying bacterial pathogens (Seo et al. 2004)

    and virulence factors. However, these limited study resultsdemonstrate that SELDI technology offers an alternative

    approach to the other techniques for exploring bacterial pro-

    teomes, ultimately permitting bacterial identification based

    on a comparison of protein profiles and patterns. An exam-ple of how SELDI technology has been applied is its use in dis-

    tinguishing between four subspecies of Francisella tularensis,the causative agent of tularemia in humans. Of the four sub-

    species of F. tularensis, tularensis is the most infectious andthe only subspecies found in North America. Lundquist and

    colleagues (2005) showed that SELDI time-of-flight mass

    spectrometry is capable of generating unique and repro-

    ducible protein profiles for each subspecies, allowing thesubspecies to be distinguished from one another.

    Although the use of mass spectrometry has great potential

    for identifying bacteria by their spectral profile, many factors

    affect the reproducibility of bacterial spectra. Sample pre-paration, matrix selection, and differences in instrument

    quality and performance can all have an impact on the re-

    producibility of protein profiles (Wunschel et al. 2005). Just

    as important, the physiological state of the cell may also in-fluence the results of mass spectral analysis, and thus both the

    growth medium and the growth

    stage of the cells must be takeninto account. Using MALDI-TOF-MS technology to analyze

    and discriminate foodborne mi-

    croorganisms, Mazzeo and col-

    leagues (2006) concluded thatthe growth time did not affect

    the bacterial protein profile, but

    that different growth media did

    affect the mass spectra of Es-

    cherichia coli. Similarly, Walker

    and associates (2002) showed

    that culture medium—especially 

    with the addition of blood, as inColumbia blood agar—will causevariation in mass spectra profiles.

    With so many variables, many 

    scientists have introduced stan-

    dardized techniques for MALDI-TOF-MS of whole cells. Liu and

    colleagues (2007) developed a

    universal sample preparation

    method for characterizing gram-positive and gram-negative bac-

    teria using MALDI-TOF-MS.

    932 BioScience •  November 2008 / Vol. 58 No. 10 www.biosciencemag.org 

    21st Century Directions in Biology 

    Figure 4. Schematic representation of the surface-enhanced laser desorption/ionization

    technique. This technique utilizes aluminum-based chips, engineered with chemically

    or biologically modified surfaces. These varied surfaces allow the differential captureof proteins based on the intrinsic properties of the proteins themselves. Bacterial lysates

    are applied directly to the surfaces, where proteins with affinities to the surface will bind.

    Following a series of washes to remove nonspecifically bound proteins, the bound proteins

    are profiled using the integrated mass analyzer to generate a mass spectrum for further analysis. Abbreviations: MS, mass spectrometry; m/z, mass-to-charge ratio.

  • 8/17/2019 Genomic and Proteomics

    9/12

    These results emphasize the need to develop standardized tech-

    niques for preparing samples to use in the creation of mass

    spectra databases for bacterial identification.

    Gel-based bacterial characterization and identification. Bac-

    teria may also be differentiated on the basis of their cellular

    protein contents. The most established technique for exam-ining cellular protein content is to lyse cells and separate

    their entire protein complement using SDS-PAGE. This results

    in a migration pattern of the protein bands that is character-

    istic for a given bacterial strain (Vandamme et al. 1996). Re-

    searchers can identify bacteria by comparing their migration

    patterns with reference gel patterns in an established database.

    However, because SDS-PAGE analysis is slow and labor-

     intensive, and because the application necessitates precise

    culture conditions that yield fairly large amounts of sample

    material, it is not particularly useful for rapid identification

    of bacteria, particularly for field and point-of-care applications.

    Two-dimensional gel electrophoresis (2DE)—the combi-nation of isoelectric focusing (IEF) and SDS-PAGE—affords

    a high-resolution separation of up to several thousand

    spots in a single gel analysis. In 1975, O’Farrell (1975) intro-

    duced 2DE as a method for separating complex mixtures of 

    cellular proteins. In 2DE, proteins are separated by IEF elec-

    trophoresis in a pH gradient according to each protein’s

    isoelectric point in the first dimension, followed by the

    second-dimension SDS-PAGE separation according to the

    relative molecular weight of each protein. After the second-

    dimension separation, the gel can be stained with standard or

    sensitive staining solutions so that protein spots can be visu-

    alized and analyzed. Protein gel patterns or 2DE maps from

    known bacteria can be further scanned, analyzed, and stored

    in a reference database. To identify an unknown species, a 2DE

    map from the unknown sample is generated by running a 2DE

    gel and then comparing it with 2DE maps in the reference

    database for identification.

    When used as a stand-alone technique, 2DE is most often

    used for analyzing protein mixtures, isolating proteins of in-

    terest for identification, and comparing differential expression

    patterns of different types of samples. For more complex 

    proteomics analysis, 2DE is greatly enhanced when com-

    bined with mass spectrometry. For example, Redmond and

    associates (2004) analyzed the exosporium of Bacillus anthracis

    spores by isolating the proteins from the outer casing of thespore using SDS-PAGE and analyzing the isolated protein

    using delayed-extraction MALDI-TOF-MS. The team iden-

    tified several proteins associated with the exosporium of

    B. anthracis. Using these same methods, the whole proteome

    or subproteome (or both) has also been made available for

    many other bacteria, including E. coli, Bacillus subtilis, S.

    aureus, Pseudomonas aeruginosa, and Helicobacter pylori

    (Nouwens et al. 2000, Hecker et al. 2003, Peng et al. 2005,

    Pieper et al. 2006). A collective proteomics database with

    complete 2DE maps and mass spectra of known bacteria

    will allow investigators to compare and identify unknown bac-

    teria with great efficiency. However, building such a database

    will not be an easy undertaking.

     The database challenge. One thing modern genomic and pro-

    teomic approaches share is the generation of large data sets

    for any individual sample that is analyzed. These present a real

    challenge in terms of archiving the data, processing and in-tegrating data from many samples so that it can be used

    for comparative purposes in the broadest way possible, and

    developing robust quality control and quality assurance

    practices. All this should be done in an environment that isaccessible and easy to use for a broad group of scientists,

    many of whom may have little experience with a particular

    method of analysis. At present there are no comprehensive ge-

    nomic or proteomic databases for bacterial identification.There are, however, a great number of databases and tools

    available for both genomic and proteomic analysis that are

    essential for providing integrated data for specific types of 

    analysis. Two examples of databases that provide excellentanalysis for identification based on 16S rRNA gene sequence

    analysis are the Ribosomal Database Project (Cole et al. 2007)

    and greengenes (DeSantis et al. 2006). On the proteomics side,

    LC/MS/MS data analysis has improved by several means andcontinues to complement 2DE gel analysis, which is still

    limited by the inability of the pattern recognition software to

    resolve overlapping protein spots (Palagi et al. 2006). Protein

    identification and characterization has been carried out withmore depth since the development of predictive tools such as

    GlycoMod and databases such as PhosphoSite, which can

    reveal possible posttranslational modifications not other-

    wise accounted for in databases consisting simply of theoretical

    spectra (Barrett et al. 2005, Witze et al. 2007).More important, algorithms are evolving to adapt to actual

    experimental occurrences and parameters. One example of 

    such adaptation is the development of a recent algorithm thatpredicts missed cleavage sites as they typically occur during

    protein digests, in order to ease the return of, and render

    greater confidence to, mass spectra probability matches

    (Siepen et al. 2007). The goal of the ProDB platform proposedby Wilke and colleagues (2003) is not only to integrate data

    derived from various databases to enrich a protein profile but

    also to archive experimental conditions and parameters, such

    as growth and culture conditions as they might apply to bac-teria, to account for the subsequent effects on mass-spectra

    profile generation. Archiving and integrating specific exper-imental conditions as part of the proteomic bioinformatics

    may alleviate the need for stringent culturing standards whenattempting to identify proteins that aid the characterization

    and identification of clinically important bacteria. For a more

    comprehensive review on proteomic data analysis, see Lisacek 

    and colleagues (2006).

    ConclusionsAdvanced genomics and proteomics technologies will continueto play a critical role in bacterial identification and charac-

    terization in the 21st century. Bacterial characterization has

    www.biosciencemag.org November 2008 / Vol. 58 No. 10 • BioScience 933

    21st Century Directions in Biology 

  • 8/17/2019 Genomic and Proteomics

    10/12

    a number of practical applications, aside from being funda-

    mental to questions of bacterial systematics, taxonomy,

    and evolution. Rapid identification and discrimination of 

    pathogenic microbes has a major impact on public health in

    terms of correct diagnosis and timely disease treatment.

    The ability to identify specific indicator organisms is also

    important for determining water quality, and an enhancedunderstanding of the population structure of these organisms

    can allow researchers to identify the source of a particular

    contaminant. For example, methods are being developed

    to determine whether fecal bacteria found in public water

    supplies are from humans, mammals, or birds. This kind of

    information has a significant impact for treatment options.

    As researchers learn more about the community fabric of 

    microbial ecosystems, it is likely that we will come to recog-

    nize sentinel microbes that will tell us, by their presence (or

    absence) and abundance, important information about

    the state of that ecosystem. For example, the identification of 

    microbes that carry out specific transformations of nitrogenor phosphorus might indicate the status of these important

    nutrients in aquatic or soil ecosystems. Likewise, the presence

    of microorganisms with certain biodegradative capacities

    could be an indicator of specific pollutants in an environment.

    The ability to rapidly identify these individual organisms

    within populations of thousands of different species is

    essential for understanding how they will affect our eco-

    systems. Bacterial characterization will also assist in elucidating

    the mechanisms that govern microbial pathogenesis, and

    allow for the discovery of important protein targets essential

    to the development of vaccines, diagnostic kits, and thera-

    peutics for infectious diseases. It is these kinds of applicationsthat make the continued development of techniques for bac-

    terial identification important both for basic science and for

    the maintenance of human and environmental health.

     Acknowledgments

    The authors would like to thank Raymond Cypess (chief

    executive officer, American Type Culture Collection [ATCC])

    and Cohava Gelber (chief scientific and technology officer,

    ATCC) for their unconditional support in preparing this

    manuscript. We thank Scott Jenkins for his help in editing

    the manuscript and David Cleland for his help in preparing

    figure 1. We also acknowledge that many excellent papers,

    particularly those providing examples of the use of the tech-

    nologies described herein, could not be cited because of space

    limitations.

    References citedAchtman M, Wagner M. 2008. Microbial diversity and the genetic nature of 

    microbial species. Nature Reviews Microbiology 6: 431–440.

    Al-Qadiri HM, Lin M, Cavinato AG, Rasco BA. 2006. Fourier transform

    infrared spectroscopy, detection and identification of Escherichia coli

    O157:H7 and  Alicyclobacillus strains in apple juice. International

    Journal of Food Microbiology 111: 73–80.

    Barrett J, Brophy PM, Hamilton JV. 2005. Analysing proteomic data.

    International Journal of Parasitology 35: 543–553.

    Bons JA, Wodzig WK, van Dieijen-Visser MP. 2005. Protein profiling as a

    diagnostic tool in clinical chemistry: A review. Clinical and Chemical

    Laboratory Medicine 43: 1281–1290.

    Bruce J. 1996. Automated system rapidly identifies and characterizes micro-

    organisms in food. Food Technology 50: 77–81.

    Chao CC, Chelius D, Zhang T, Mutumanje E, Ching WM. 2007. Insight

    into the virulence of Rickettsia prowazekii by proteomic analysis and

    comparison with an avirulent strain. Biochimica Biophysica Acta 1774:

    373–381.

    Chelo IM, Ze-Ze L, Tenreiro R. 2007. Congruence of evolutionary relation-

    ships inside the Leuconostoc–Oenococcus–Weissella clade assessed by 

    phylogenetic analysis of the 16S rRNA gene, dnaA, gyrB, rpoC and dnaK.

    International Journal of Systematic and Evolutionary Microbiology 57:

    276–286.

    Clarke PH. 1985. The scientific study of bacteria. Pages 1–37 in Leadbetter

    ER, Poindexter JS, eds. Bacteria in Nature, vol. 1. New York: Plenum Press.

    Cleland D, Krader P, Emerson D. 2008. Use of the DiversiLab repetitive

    sequence-based PCR system for genotyping and identification of archaea.

    Journal of Microbiological Methods 73: 172–178.

    Cocconcelli PS, Porro D, Galandini S, Senini L. 1995. Development of RAPD

    protocol for typing of strains of lactic-acid bacteria and enterococci.

    Letters in Applied Microbiology 21: 376–379.

    Cole JR, Chai B, Farris RJ, Wang Q, Kulam-Syed-Mohideen AS, McGarrell

    DM, Bandela AM, Cardenas E, Garrity GM, Tiedje JM. 2007. The

    ribosomal database project (RDP-II): Introducing myRDP  space and

    quality controlled public data. Nucleic Acids Research 35: D169–D172.

    Cooper JE, Feil EJ. 2004. Multilocus sequence typing—what is resolved?

    Trends in Microbiology 12: 373–377.

    Dare D. 2006. Rapid bacterial characterization and identification by

    MALDI-TOF mass spectrometry. Pages 117–133 in Tang Y-W, Stratton

    CW, eds. Advanced Techniques in Diagnostic Microbiology. New York:

    Springer.

    DeSantis TZ, Hugenholtz P, Larsen N, Rojas M, Brodie EL, Keller K, Huber

    T, Dalevi D, Hu P, Andersen GL. 2006. Greengenes, a chimera-checked

    16S rRNA gene database and workbench compatible with ARB.

    Applied and Environmental Microbiology 72: 5069–5072.

    Devulder G, de Montclos P, Flandrois JP. 2005. A multigene approach to

    phylogenetic analysis using the genus  Mycobacterium as a model.International Journal of Systematic and Evolutionary Microbiology 55:

    293–302.

    Edwards-Jones V, Claydon MA, Evason DJ, Walker J, Fox AJ, Gordon DB. 2000.

    Rapid discrimination between methicillin-sensitive and methicillin-

     resistant Staphylococcus aureus by intact cell mass spectrometry. Journal

    of Medical Microbiology 49: 295–300.

    Fenn JB, Mann M, Meng CK, Wong SF, Whitehouse CM. 1989. Electrospray 

    ionization for mass spectrometry of large biomolecules. Science 246:

    64–71.

    Frye JG, Jesse T, Long F, Rondeau G, Porwollik S, McClelland M, Jackson CR,

    Englen M, Fedorka-Cray PJ. 2006. DNA microarray detection of

    antimicrobial resistance genes in diverse bacteria. International Journal

    of Antimicrobial Agents 27: 138–151.

    Gevers D, et al. 2005. Re-evaluating prokaryotic species. Nature Reviews

    Microbiology 3: 733–739.Gillis M, Vandamme P, De Vos P, Swings J, Kersters K. 2001. Polyphasic

    taxonomy. Pages 43–48 in Boone DR, Castenholtz RW, eds. Bergey’s

    Manual of Systematic Bacteriology, vol. 1. New York: Springer.

    Goris J, Konstantinidis KT, Klappenbach JA, Coenye T, Vandamme P, Tiedje

    JM. 2007. DNA-DNA hybridization values and their relationship to

    whole-genome sequence similarities. International Journal of Systematic

    and Evolutionary Microbiology 57: 81–91.

    He Z, et al. 2007. GeoChip: A comprehensive microarray for investigating

    biogeochemical, ecological and environmental processes. ISME Journal:

    Multidisciplinary Journal of Microbial Ecology 1: 67–77.

    Hecker M, Engelmann S, Cordwell SJ. 2003. Proteomics of Staphylococcus

    aureus— current state and future challenges. Journal of Chromatography 

    B: Analytical Technologies in the Biomedical and Life Sciences 787:

    179–195.

    934 BioScience •  November 2008 / Vol. 58 No. 10 www.biosciencemag.org 

    21st Century Directions in Biology 

  • 8/17/2019 Genomic and Proteomics

    11/12

    Holland R, Wilkes JG, Rafii F, Sutherland JB, Persons CC, Voorhees KJ, Lay 

    JO Jr. 1996. Rapid identification of intact whole bacteria based on

    spectral patterns using matrix-assisted laser desorption/ionization with

    time-of-flight mass spectrometry. Rapid Communications in Mass

    Spectrometry 10: 1227–1232.

    Honisch C, Chen Y, Mortimer C, Arnold C, Schmidt O, van den Boom D,

    Cantor CR, Shah HN, Gharbia SE. 2007. Automated comparative sequence

    analysis by base-specific cleavage and mass spectrometry for nucleic

    acid-based microbial typing. Proceedings of the National Academy of

    Sciences 104: 10649–10654.

    Krader P, Emerson D. 2004. Characterization of Archaea and some

    extremophilic bacteria using matrix-assisted laser desorption/ionization

    time-of-flight (MALDI-TOF) mass spectrometry. Extremophiles 8:

    259–268.

    Krishnamurthy T, Ross PL. 1996. Rapid identification of bacteria by direct

    matrix-assisted laser desorption/ionization mass spectrometric analysis

    of whole cells. Rapid Communications in Mass Spectrometry 10:

    1992–1996.

    Lay JO Jr. 2001. MALDI-TOF mass spectrometry of bacteria. Mass

    Spectrometry Reviews 20: 172–194.

    Lin JJ, Kuo J, Ma J. 1996. A PCR-based DNA fingerprinting technique: AFLP

    for molecular typing of bacteria. Nucleic Acids Research 24: 3649–3650.

    Lisacek F, Cohen-Boulakia S, Appel RD. 2006. Proteome informatics II:Bioinformatics for comparative proteomics. Proteomics 6: 5445–5466.

    Liu H, Du Z, Wang J, Yang R. 2007. Universal sample preparation method

    for characterization of bacteria by matrix-assisted laser desorption

    ionization–time of flight mass spectrometry. Applied Environmental

    Microbiology 73: 1899–1907.

    Liu WT, Stahl DA. 2007. Molecular approaches for the measurement of 

    density, diversity, and phylogeny. Pages 139–156 in Hurst CJ, Crawford

    RL, Garland JL, Lipson DA, Mills AL, Stetzenbach LD, eds. Manual of

    Environmental Microbiology. 3rd ed. Washington (DC): ASM Press.

    Liu WT, Mirzabekov AD, Stahl DA. 2001. Optimization of an oligonucleotide

    microchip for microbial identification studies: A non-equilibrium

    dissociation approach. Environmental Microbiology 3: 619–629.

    Lodders N, Stackebrandt E, Nubel U. 2005. Frequent genetic recombination

    in natural populations of the marine cyanobacterium  Microcoleus

    chthonoplastes. Environmental Microbiology 7: 434–442.Logue JB, Bürgmann H, Robinson CT. 2008. Progress in the ecological

    genetics and biodiversity of freshwater bacteria. BioScience 58: 103–113.

    Ludwig W, Klenk H-P. 2001. Overview: A phylogenetic backbone and

    taxonomic framework for prokaryotic systematics. Pages 49–65 in Boone

    DR, Castenholtz RW, eds. Bergey’s Manual of Systematic Bacteriology.

    Berlin: Springer.

    Lundquist M, Caspersen MB, Wikström P, Forsman M. 2005. Discrimina-

    tion of Francisella tularensis subspecies using surface enhanced laser

    desorption ionization mass spectrometry and multivariate data analy-

    sis. FEMS Microbiology Letters 243: 303–310.

    Maiden MCJ, et al. 1998. Multilocus sequence typing: A portable approach

    to the identification of clones within populations of pathogenic

    microorganisms. Proceedings of the National Academy of Sciences 95:

    3140–3145.

    Mazzeo MF, Sorrentino A, Gaita M, Cacace G, Di Stasio M, Facchiano A,

    Comi G, Malorni A, Siciliano RA. 2006. Matrix-assisted laser desorption

    ionization–time of flight mass spectrometry for the discrimination of 

    food-borne microorganisms. Applied and Environmental Microbiology 

    72: 1180–1189.

    Meays CL, Broersma K, Nordin R, Mazunder A. 2004. Source tracking fecal

    bacteria in water: A critical review of current methods. Journal of

    Environmental Management 73: 71–79.

    Mellmann A, et al. 2008. Evaluation of matrix-assisted laser desorption/

    ionization time-of-flight mass spectrometry (MALDI-TOF MS) in

    comparison to 16S rRNA gene sequencing for species identification of 

    nonfermenting bacteria. Journal of Clinical Microbiology 46: 1946–1954.

    Naser SM, Thompson FL, Hoste B, Gevers D, Dawyndt P, Vancanneyt M,

    Swings J. 2005. Application of multilocus sequence analysis (MLSA) for

    rapid identification of Enterococcus species based on rpoA and  pheS

    genes. Microbiology 151: 2141–2150.

    Nouwens AS, Cordwell SJ, Larsen MR, Molloy MP, Gillings M, Willcox MD,

    Walsh BJ. 2000. Complementing genomics with proteomics: The

    membrane subproteome of Pseudomonas aeruginosa PAO1. Electro-

    phoresis 21: 3797–3809.

    O’Farrell PH. 1975. High resolution two-dimensional electrophoresis of 

    proteins. Journal of Biological Chemistry 250: 4007–4021.

    Palagi PM, Hernandez P, Walther D, Appel RD. 2006. Proteome informatics

    I: Bioinformatics tools for processing experimental data. Proteomics

    6: 5435–5444.

    Paradis S, Boissinot M, Paquette N, Belanger SD, Martel EA, Boudreau DK,

    Picard FJ, Ouellette M, Roy PH, Bergeron MG. 2005. Phylogeny of the

    Enterobacteriaceae based on genes encoding elongation factor Tu and

    F-ATPase µ-subunit. International Journal of Systematic and Evo-

    lutionary Microbiology 55: 2013–2025.

    Peng X, Xu C, Ren H, Lin X, Wu L, Wang S. 2005. Proteomic analysis of the

    sarcosine-insoluble outer membrane fraction of Pseudomonas aeruginosa

    responding to ampicilin, kanamycin, and tetracycline resistance. Journal

    of Proteome Research 4: 2257–2265.

    Pieper R, Gatlin-Bunai CL, Mongodin EF, Parmar PP, Huang ST, Clark DJ,

    Fleischmann RD, Gill SR, Peterson SN. 2006. Comparative proteomic

    analysis of Staphylococcus aureus strains with differences in resistance to

    the cell wall-targeting antibiotic vancomycin. Proteomics 6: 4246–4258.

    Pignone M, Greth K, Cooper J, Emerson D, Tang J. 2006. Identification of

    mycobacteria by matrix-assisted laser desorption/ionization time-

    of-flight mass spectrometry. Journal of Clinical Microbiology 44:

    1963–1970.

    Redmond C, Baillie LW, Hibbs S, Moir AJ, Moir A. 2004. Identification of

    proteins in the exosporium of Bacillus anthracis. Microbiology 150:

    355–363.

    Richert K, Brambilla E, Stackebrandt E. 2007. The phylogenetic significance

    of peptidoglycan types: Molecular analysis of the genera Microbacterium

    and Aureobacterium based upon sequence comparison of gyrB, rpoB, recA

    and  ppk and 16SrRNA genes. Systematic and Applied Microbiology

    30: 102–108.

    Richter D, Postic D, Sertour N, Livey I, Matuschka FR, Baranton G. 2006.

    Delineation ofBorrelia burgdorferi

    sensu lato species by multilocussequence analysis and confirmation of the delineation of Borrelia

    spielmanii sp. nov. International Journal of Systematic and Evolutionary 

    Microbiology 56: 873–881.

    Sampath R, Hall TA, Massire C, Li F, Blyn LB, Eshoo MW, Hofstadler SA, Ecker

    DJ. 2007. Rapid identification of emerging infectious agents using PCR 

    and electrospray ionization mass spectrometry. Annals of the New York 

    Academy of Sciences 1102: 109–120.

    Savenlkoul PHM, Aarts HJM, De Haas J, Dijkshoorn L, Duim B, Otsen M,

    Rademaker JLW, Schouls L, Lenstra JA. 1999. Amplified-fragment

    length poly morphism analysis: The state of an art. Journal of Clinical

    Microbiology 37: 3083–3091.

    Seo GM, Kim SJ, Chai YG. 2004. Rapid profiling of the infection of Bacillus

    anthracis on human macrophages using SELDI-TOF mass spectroscopy.

    Biochemical and Biophysical Research Communications 325: 1236–1239.

    Settanni L, Corsetti A. 2007. The use of multiplex PCR to detect anddifferentiate food- and beverage-associated microorganisms: A review.

    Journal of Microbiological Methods 69: 1–22.

    Siepen JA, Keevil EJ, Knight D, Hubbard SJ. 2007. Prediction of missed

    cleavage sites in tryptic peptides aids protein identification in

    proteomics. Journal of Proteome Research 6: 399–408.

    Thompson FL, Gevers D, Thompson CC, Dawyndt P, Naser S, Hoste B,

    Munn CB, Swings J. 2005. Phylogeny and molecular identification of

    vibrios on the basis of multilocus sequence analysis. Applied and

    Environmental Microbiology 71: 5107–5115.

    Thomson J. 1899. On the masses of the ions in gases at low pressures.

    Philosophical Magazine 48: 547–567.

    Vandamme B, Pot B, Gillis M, de Vos P, Kersters K, Swings J. 1996. Polyphasic

    taxonomy, a consensus approach to bacterial systematics. Microbiological

    Reviews 60: 407–438.

    www.biosciencemag.org November 2008 / Vol. 58 No. 10 • BioScience 935

    21st Century Directions in Biology 

  • 8/17/2019 Genomic and Proteomics

    12/12

    Versalovic J, Schneider M, de Bruijn FJ, Lupski JR. 1994. Genomic finger-

    printing of bacteria using repetitive sequence based PCR (rep-PCR).

    Methods in Cellular and Molecular Biology 5: 25–40.

    Vos P, et al. 1995. AFLP: A new technique for DNA fingerprinting. Nucleic

    Acids Research 23: 4407–4414.

    Walker J, Fox AJ, Edwards-Jones V, Gordon DB. 2002. Intact cell mass

    spectrometry (ICMS) used to type methicillin-resistant Staphylococcus

    aureus: Media effects and inter-laboratory reproducibility. Journal of

    Microbiological Methods 48: 117–126.

    Wilke A, et al. 2003. Bioinformatics support for high-throughput

    proteomics. Journal of Biotechnology 106: 147–156.

    Wilson KH, Wilson WJ, Radosevich JL, DeSantis TZ, Viswanathan VS,

    Kuczmarski TA, Andersen GL. 2002. High-density microarray of small-

    subunit ribosomal DNA probes. Applied and Environmental Micro-

    biology 68: 2535–2541.

    Witze ES, Old WM, Resing KA, Ahn NG. 2007. Mapping protein post-

     translational modifications with mass spectrometry. Nature Methods

    4: 798–806.

    Woese CR. 1987. Bacterial evolution. Microbiological Reviews 51: 221–271.

    Wunschel SC, Jarman KH, Petersen CE, Valentine NB, Wahl KL, Schauki D,

    Jackman J, Nelson CP, White VE. 2005. Bacterial analysis by MALDI-TOF

    mass spectrometry: An inter-laboratory comparison. Journal of the

    American Society of Mass Spectrometry 16: 456–462.

    Yip TT, Lomas L. 2002. SELDI ProteinChip array in oncoproteomic

    research. Technology of Cancer Research Treatment 1: 273–280.

    Zeigler DR. 2003. Gene sequences useful for predicting relatedness of whole

    genomes in bacteria. International Journal of Systematic and Evo-

    lutionary Microbiology 53: 1893–1900.

    doi:10.1641/B581006Include this information when citing this material.

    936 BioScience •  November 2008 / Vol. 58 No. 10 www.biosciencemag.org 

    21st Century Directions in Biology