Take a look at the Recent articles

Diagnostic Molecular Microbiology and its applications: Current and Future Perspectives

Hemant Vira

Vivek Bhat

Preeti Chavan

DOI: 10.15761/CMID.1000105

Article
Article Info
Author Info
Figures & Data

Abstract

Infectious diseases are a major cause of morbidity and mortality throughout the world. Although the field of diagnostic microbiology has substantially evolvedover the past few decades, it is still heavily reliant on cultures and serology, which while being cost effective is largely time consuming and sometimes less sensitive and specific. Molecular biology on the other hand has developed with rapid and confirmatory diagnosis. However, the merger of molecular diagnostics in routine diagnostic microbiology labs has been slow paced mostly due to higher costs and lack of infrastructure.Molecular methods have progressed beyond identification to detect antibiotic marker genes, fastidious bacteria, and uncultivable microbes. It has found a scope in mycology and parasitology, in which the basic conventional techniquesmay sometimes be unable to make a definitive diagnosis of the concerned pathogens. In the field of virology where culturing may be impractical in some clinical settings, it has developed various multiplexing procedures which can detect and quantitate the viral copies present in the crude specimen. Besides the field offers different types of permutations and combinations like direct sequencing for finding the variants, whole genome sequencing, epidemiological testing like plasmid profiling, RFLP etc. which can point out the infection and different types of PCR customisation such as nested, multiplex and Real Time (RT) PCR. In the present review, we have described the various molecular typing techniques and their application in microbial testing with a briefing of the tests that have been already standardised with relevant sample acceptance and rejection criteria for commonly encountered pathogens and pointed towards future directions converging automated closed DNA, RNA extraction/amplification platforms, Next Generation Sequencing (NSG), Microarrays and Digital PCR (dPCR) into its arsenal.

Keywords

Molecular; Diagnostic; Medical; Microbiology

Introduction

Infectious disease is the main cause of mortality and morbidity worldwide. The different types of infection caused by various types of pathogens including bacteria, viruses, parasites and fungi are ever increasing [1]. Increasing age, diseases such as cancer, immunosuppression etc are contributing factors. Although the era of antibiotic, antiviral, antimycotic, and antiparasitic has led to alleviation from these infectious agents,the rampant use of these agents has led to the emergence of multi drug resistant (MDR) pathogens which if not diagnosed and contained early, could spread to large geographical areas [2].The role played by diagnostic systems is the ‘golden standard’ for enacting rapid treatment regimes. Routine clinical& microbiological procedures such as cultures, serology, and microscopy still remains the procedures of choice in terms of diagnosis and are also cost effective [3]. However the routine microbiological cultures are by themselves, not confirmatory tests for the accurate diagnosis of the pathogen. Even with cultivable bacteria, cultures fail to reveal an organism in many patients with sign and symptoms consistent with infectious disease. Due to the above reasons, there is a large scope for molecular biology procedures in the diagnostic clinical microbiology laboratory.

With the advent of PCR technology about 30 years ago came the era of molecular diagnostics. It is due molecular testing that the phenotypes exhibited by a pathogen can be genetically confirmed [4]. It ensures rapid diagnosis at a cost effective price, thus increasing the diagnostic arsenal for pathogen identification. This review enlists the various types of molecular typing methods which have been inculcated in molecular microbiology diagnostic labs. It also describes the need for more advanced molecular methods for increasing the sensitivity and specificity of diagnosis and also the routine protocols for various bacterial, viral, fungal, and parasites that have been already standardised at the genus and species levelswith a briefing on sample acceptance and rejection criteria for the enlisted pathogens.

Molecular typing techniques

The advent of nucleic acid amplification techniques has led to the advancement of molecular typing techniques [5]. It offers the advantage of rapid confirmatory diagnosis. Since the start of the field of medical microbiology, the main techniques have relied on the identification of phenotypic characters like biochemical characterisation, morphological view, and cultures. The inert nature of the DNA molecule makes it the most suitable marker for confirmatory diagnosis [6]. The various types of molecular techniques have been reviewed in the following sections.

Restriction analysis

The microbial DNA consists of various sites containing sequences which are repetitive in nature. These repeats are tandemly repeated after constant intervals [6]. Also restriction enzymes or endonuclease (R.E) a class of DNA–cleaving enzymes isolates from bacteria are used to cleave these DNA at a particular and specific sequence which results in the fragmentation of DNA molecule. This technique thus can detect the fragmentation pattern of the target and compare it with the in house pathogenic strains and handout results whether the isolated pathogens are similar or dissimilar in origins. Thus, in this technique the DNA is cut with the restriction enzymes [7].

Plasmid profiling

In these technique plasmids which are extrachromosomal DNA elements are cut with restriction enzymes (RE). Transfer of plasmids is very common between members of the family Enterobacteriaceae. It is mainly useful in epidemiological outbreaks in which the plasmids are cut with the same RE which may result in same restriction patterns from plasmids isolated from various pathogens in the outbreak area thus showing the profiles in outbreak associated strains [8]. It has been carried out in opportunistic multidrug resistant Pseudomonas aeruginosaisolated from wound infections, which carried a pattern of resistance to various antibiotics [9]. A similar study was used for screening of Neisseria gonorohoeae [10] and Shigella spp.[11] among others [12,13]. It has been also widely carried out to identify multidrug resistant isolates present in sewage associated with health care centres [14]. A study carried out used this technique for the study of the emergence of drug resistance in diarrheagenic E. coli in paediatric population in a developing country [15, 16].

Restriction Fragment Length Polymorphism (RFLP)

RFLP has been mainly used in multi drug resistant Mycobacterium tuberculosis outbreaks for simultaneous identification and differentiation in which polymorphisms present in DR locus which is characterised by repetitive sequence interspersed between non repetitive sequences has been hybridised by a probe known as spacer oligotyping or spoligotyping. It can differentiate M. bovisfrom M. tuberculosis, a distinction which is often difficult to make by traditional methods [17]. In parasitology PCR-RFLP has been used to diagnose and differentiate Old and New World Leishmania species [18] and also for differentiating Fasciola hepatica and Fasciola gigantic [19]. In a study carried out in outbreak in Japan characterization of enterohemorrhagic Escherichia coli O111 and O157 Strains were done by RFLP [20]. In case of the highly transmissible zoonotic infection Brucellosis, caused by the slow growing bacterium Brucellaspp.this technique has found importance [21].

Pulse Field Gel Electrophoresis (PFGE)

Withthe advent of RFLP, there arose a need to perform high resolution separation on agarose gel to look out for similarity in banding patterns obtained after RE digestion. In 1982, Schwartz introduced the concept that DNA molecules larger than 50kb can be separated by using two alternating electric fields.This led to the technique of PFGE. It is based on the digestion of bacterial DNA with RE that recognises few sites along the chromosome, generating large DNA fragments (30-800Kb). The basis for PFGE separation is the size-dependent time-associated reorientation of DNA migration. When visualised electronically bacterial isolates with identical or very similar band patterns are more likely to be related genetically than with more divergent band patterns.PFGE has been used for the detection of Listeria monocytogenesand Campylobacter spp. which are food borne pathogen difficult to culture [22,23]. It is also widely applied to the diagnosis of eukaryotic DNA like parasites which are dense and complex in nature [24].

PCR amplification of gene target

This method is the basis of all types of PCR technology. In this method designed primers which amplify a unique region of microorganisms are used. Thus, this method is target specific.

Multiplex PCR

Multiplexing is one of the most widely applied techniques in the field of diagnostic microbiology. It is a variant of PCR, which enables simultaneous amplification of many targets of interest in one reaction by using more than one pair of primers. Its main use is in the field of virology in which primers are designed to detect different types of viruses from a sample [25]. Different types of panels are available from brands such as Biomuriex, Roche, life technologies, Qaigenetcwhich are named syndromically e.g. Respiratory panel, Gastrointestinal panel, Meningitis panel etc. Multiplexing is also useful for differentiating at the species levels e.g. Multiplex PCR for identification of Campylobacter coli and Campylobacter jejunifrom pure cultures and stools [26], rapid detection of Methicillin Resistant Staphylococcus aureus (MRSA) from blood culture bottles [27], acquired carbapenemase resistance [28]; and in parasite detection, e.g. for differential diagnosis of helminths like Taeniasis and Cysticercosis [29].

Nested PCR

Involves two consecutive PCR reactions of 25 cycles. The first PCR uses primers external to the sequence of interest. The second PCR uses the product of the first PCR in conjunction with one or more nested primers to amplify the sequence within the region flanked by the initial set of primers. It has been designed for various bacterial [30-33], fungal [34, 35], and parasitic infections.

Real time PCR (RT-PCR)

It allows viewing the increase in the amount of DNA as it is amplified. All real time PCR systems rely upon the detection and quantitation of the fluorescent reporter, the signal of which increases in direct proportion of the amount of PCR product in a reaction. The most economical reporter is the double strand DNA specific dye SYBR green, which upon excitation emits light. However SYBR green has its limitations as it will bind to any double stranded DNA in a reaction [36]. Two most popular alternatives to SYBR green are TaqMan and molecular beacons. Both technologies depend on hybridisation probes relying on fluorescence resonance energy transfer (FRET) and quantization [37]. There are numerous laboratory-developed real time PCR tests, including assays for viruses like Dengue [38], Ebola [39] and bacteria like Pseudomonas aeruginosa [40].

Direct Sequencing

Direct sequencing is the most reliable method for the detection of sequences at the molecular level. The first whole genome sequenced was that of bacteriophage φX174. Given below is a briefing of some of the methods applied in microbial sequencing. It is noteworthy that sequencing has helped to differentiate fungi and parasites at the species level which is difficult to achieve by current morphological and phenotypic techniques [41].

Whole Genome Sequencing (WGS)

WGS has changed the landscape of molecular biology with its advent in 2005 by Roche. It is the most cost effective approach for solving many epidemiologic outbreaks and confirmation of pathogens after cultures [42,43]. The near market platforms available are Roche 454, Pacific Biosciences, Ion Torrent, and IlluminaMiSeq which can perform operations in 48hrs [44]. In terms of sample preparation for pathogens Oxford Nanopore is also very reliable [45].

Target Gene Sequencing

It is the most common method of sequencing used which relies on Sanger dideoxy chain terminator method. In this the primers complementary to the gene of interest are used to amplify the band containing the gene of interest and sequenced using an automated sequencer like the Applied Biosystems 8180xl genetic analyser. It is used for diagnosing multi drug resistance in microorganisms like presence of acquired carbapenamase resistance geneNDM-1 gene [28, 46].

Universal gene target

These are specific sequences used in microbial diagnostics which are highly conserved and also impart certain phylogenetic traits at the genus and species levels.

Ribosomal RNA (rRNA)

The bacterial pathogens which are unidentifiable with routine testing, slow growers, uncultivable can be identified by amplification of DNA encoding ribosomal RNA genes followed by DNA sequencing. In bacteria there are three genes that make up rRNA functionality, i.e. 5S, 16S, and 23S rRNA. The 16S has been employed for identification purposes due to it being highly conserved and having a moderate copy number depending on the genus. Besides 16S rRNA genes are found in all bacteria and accumulate mutations at a slow, constant rate over time, hence serving as “molecular clocks” [47]. The use of this technique has led to the discovery of novel clinical isolates and culture negative infections [48]. It has been widely used to identify tuberculosis and non-tuberculosis infections [49] and biothreat agents like Brucella spps [50]. Recently, there have been several reports regarding the use of the large subunit (23S rRNA) which is specific for bacterial species recognition. It has been used frequently for the detection of Stenotrophomonas maltophila from patients with cystic fibrosis [51].

Heat Shock proteins

The use of 16S rRNA although applied routinely bears some limitations. Strains with less than 97.5%16S rRNA gene sequence identity are unlikely to be related at the species level.However, there are a number of strains that share less than 50% DNA similarity by reassociation and therefore are classified as distinct species, but share 99% to 100% 16S rRNA gene sequence identity. For example, Mycobacterium chelonae and Mycobacterium abscessus have more than 99% 16S rRNAgene sequence identity, but their DNA similarity by reassociation is only 35% [52]. In such circumstance sequencing of essential genes such as the heat shock proteins (HSP; HSP60, HSP65, groEL, groER, etc.), have been shown to be useful [53,54]. The heat shock response is an important homeostatic response that enables the microbial cells survive the conditions of stress. This phenomenon is observed in all microorganisms from bacteria, fungi, to parasites [55]. A polymorphism in HSP 70 of Leishmania spps has been used for the differentiation of neotropical Leishmania species, as well as Trypanosoma cruzi [56].

Antibiotic resistance gene

Antibiotic resistance among microorganisms has become a worldwide issue. A review published by Fluit et al. has highlighted on the increasing numbers of multidrug resistant bacteria and its molecular detection [4]. Molecular detection by Xpert MTB/RIF automated system (GeneXpert, Cepheid) has found a wide scale acceptance for the detection of rifampicin resistant tuberculosis, which is on the rise in developing countries [57]. Also the detection of NDM-1 and its variant has been carried out by conventional PCR against the NDM-1 gene with members of the enterobactericeae family [58]. For detection of H pyroli a slow growing ulcerative pathogen resistance markers amplification strategies are used [59].In parasitology it has found applications for detection of chloroquine resistant Plasmodium falciparum by using nested PCR followed by mutation-specific, restriction-endonuclease digestion for detection of mutations in pfcrt and pfmdrgene for treatment regimens [60].

Molecular standardised diagnostic protocols

In this section we review some of the standardised protocols employed in a diagnostic molecular microbiology laboratory at the genus and species levels for various clinically important pathogens including bacteria, fungi, parasites and viruses. We also include the sample acceptance and rejection criteria as cited in the literature.

Bacteriology

Most medically important classes of pathogens belong to the class of bacteria and their molecular detection is the need of the hour for rapid confirmatory diagnosis [61]. Molecular standardised protocols have been cited in table 1. Whipple’s disease is a rare but fatal infection caused by Tropheryma whipplei the diagnosis of which until recently only relied on histopathology and electron microscopy, often from post morterm material. PCR now allows the diagnosis of neuro-Whipple’s disease and endocarditis by the detection of T. whipplei from non-invasive specimens [62]. Molecular diagnosis can also help to diagnose uncultivable pathogens like that of cat scratch disease Bartonella henselae, Q fever due to Coxiella burnetti, and male urethritis caused by Mycoplasma genitalium [63].

Table 1. Standardised molecular protocols in diagnostic bacteriology

Bacteria

Acceptable Specimen

Unacceptable Specimen

Limitations/Comments      

Primer Sequence/Probe and Method References

Bordetella pertussis

Nasopharyngeal aspirates, nasal swabs or throat swabs.Sputum, endotracheal tube aspirates,and BAL fluids are also occasionally processed

The potential for false B. pertussis detection due to B. Holmseii infection must also be kept in mind

-

[72-75]

Bordetella parapertussis

Nasopharyngeal aspirates, nasal swabs or throat swabs.Sputum, endotracheal tube aspirates, and BAL fluids are also occasionally processed

-

-

[72-75]

Brucellosis

Bacterial isolates, Blood (EDTA), Serum, Buffy coat, fresh tissue (liver, spleen,thyroid, bone, synovial tissue and fluid)

-

Assays were validated by testing historical culture confirmed B. suis and B. melitensis patient isolates as well as cattle derived B. abortus isolates

[76,77]

Burkholderia cepacia

Designed for the identification of suspected B. cepaciacomplex and B. gladioli cultures and has not yet been validated for the detection of these organisms in clinical samples.

-

Species specific assays are not currently available forall of the B. cepacia complex species (e.g. Burkholderia dolosa,

Burkholderia pyrrocinia)

[78-81]

Chlamydia

Acceptable specimens are urine, urethral swab, endocervical swab, dry swab (including throat, rectal and vaginal), tampon  cells collected in liquid based, cytology medium PreservCytTM (Hologic, Bedford, MA, USA) and SurePathTM (BD, Franklin Lakes, NJ, USA)

Not validated for other specimens

Most commercial assays, however, do not differentiate between LGV and other serovars of C. trachomatis

[82,83]

Chlamydophila

Respiratory material, including throat swabs, nose swabs, nasopharyngeal aspirates, bronchoalveolar lavage, whole blood (EDTA), urine and CSF, occasionally environmental/bird specimens

Wound swabs, drainage fluid, faeces

No international controls are available for C. psittaci due to restrictions on transport

of potential bioterrorism agents

[84-86]

Coxiellaburnetii

Blood, serum, bone marrow, biopsies

(Including those imbedded in paraffin) and cerebrospinal fluid.

-

-

[63]

Diarrheagenic Escherichia coli Pathotypes (DEP)

Including Enterohaemorrhagic (EHEC)/Shiga-toxin

E. coli (STEC)

Infectious colonies isolated after processing.

-

-

[87-90]

Haemophilus ducreyi

Impression smears or press slides, swabs, and biopsies only.

-

-

[91-93]

Klebsiella granulomatis.

------do-----------

-

Only K. granulomatis carries the restriction site specific to this organism

[92]

Haemophilus influenzae

– Capsulated and Non-typeable

H. influenza cultures only

Not  been validated for the detection of H. influenza in clinical samples

-

[94-96]

Mycobacterium tuberculosis Complex

Respiratory tract

Non-respiratory origin, the sensitivity and specificity of the assay has been estimated to be 69.8% and 95.6%, respectively.

Potential for environmental mycobacteria to contaminate samples

[94-96]

Mycobacteria Other Than Mycobacterium tuberculosis

Fresh tissue, cerebrospinal fluid, respiratory specimens, and body fluids, including uncoagulated blood in EDTA tubes

Formalin fixed and paraffin embedded tissue samples, clotted blood, serum, swabs,

urine and faeces

-

[97,98]

Mycoplasma genitalium

Urine and genital swabs

-

M. genitalium DNA may be detected in asymptomatic individuals

[99,100]

Mycoplasma pneumoniae

Sputum and Bronchoalveolar lavage (BAL) specimens, nasopharyngeal and throat swabs, nasopharyngeal aspirates, tracheal aspirates and pleural fluid specimens

Not been validated for any other specimen than those listed.

M. pneumonia may be detected for some time after acute infection following the resolution of clinical symptoms

[101-103]

Neisseria gonorrhoeae

Urine, urethral swab, endocervical swab, dry swab (including throat, rectal and vaginal)

Not been validated for any other specimen than those listed

PHLN guidelines for the use and interpretation of nucleic acid detection tests for Neisseria gonorrhoeae

[104-107]

Neisseria meningitidis

EDTA whole blood, CSF and aspirates from joints. Heparinised or citrated samples can be tested

Plasma and serum samples show a decreased recovery of N. meningitides DNA

The ctrAPCR assay is used as a diagnostic test for meningococcal disease

[108,109]

Pseudomonas  aeruginosa

P. aeruginosa isolates only

Used in sputum samples, but has not been validated

-

[110-113]

Rickettsia

Blood, biopsies, and cerebrospinal fluid

Clotted blood, serum, swabs and urine

-

[114]

Streptococcus pneumoniae

CSF and other sterile fluids.

Not appropriate to test respiratory specimens

ply gene is not specific for pneumococci and been detected in Streptococcus mitis and Streptococcus oralis isolates

[115]

Treponema pallidum

Swabs from skin lesions only

Not validated for other specimens

The gold standard for the detection of T. pallidum is probably rabbit inoculation

[116-118]

Virology

The field of virology has already adopted diagnosis by molecular methods and table 2 sites the medically important viruses encountered in clinical labs. Genotypic mutation analysis of the virus which correlates with phenotypic resistance has been designed for retroviruses like HIV in treatment naïve and for viral rebound in patients already on treatment, to establish if resistance has developed which requires a change in treatment [64,65].

Table 2.  Standardised molecular protocols in diagnostic virology

Virus

Acceptable Specimen

Unacceptable Specimen

Limitations/Comments      

Primer Sequence/Probe and Protocol References

Adenovirus

Clinical specimens (throat or nasal swabs or faecal samples)

Not been validated for any other specimen than those listed

Most common technique is to passage Adenovirus in A549 cells before genetic analysis

[119-121]

Cytomegalovirus (CMV)

New born Screening card, fresh placenta, fresh umbilical cord, amniotic fluid, urine, serum, plasma, buffy coats, formalin-fixed, paraffin-embedded tissues

Not been validated for any other specimen than those listed

Generally two different CMV PCR are performed on each sample to minimise false positivity

[122-126]

Dengue Virus

Serum

Not been validated for any other specimen than those listed

‘Not detected’ should not be considered as absence of virus.

[127-131]

Enterovirus

Faeces, CSF, nasopharyngeal aspirates, cell culture supernatant, throat, vesicle and

rectal swabs

Blood and tissue samples

The specimen should be kept at 4 degree C

[132-135]

Epstein Barr Virus

CSF, Blood, Biopsies

Not been validated for any other specimen than those listed

Stored at -20 degree C

[136,137]

Flavivirus

CSF, CNS tissues and whole blood

Not been validated for any other specimen than those listed

West Nile virus assay also detects Kunjin virus.

[137]

Hepatitis A Virus

Whole blood, serum, and EDTA tubes

Heparin tubes as it is a potent inhibitor of PCR

Haemolysed samples are causes PCR inhibition

[138]

Hepatitis B Virus

-----------do-----------

-------do--------

---------------do------------------

[138]

Hepatitis C Virus

-----------do-----------

-------do---------

---------------do-------------------

[139]

Hepatitis D Virus

-----------do-----------

-------do---------

---------------do-------------------

[140]

Hepatitis E Virus

-----------do-----------

-------do---------

---------------do-------------------

[141]

Herpes Simplex Virus Type 1 and 2

 CSF, brain biopsy, FFPE

Not been validated for any other specimen than those listed

PCR detection has become a confirmatory test for HSV encephalitis

[142-147]

Human Bocavirus

Respiratory secretions, nose/throat swabs, stool samples, whole blood

Not been validated for any other specimen than those listed

-

[148]

Human Coronaviruses

Respiratory specimens (Nasopharyngeal aspirates, nose and throat swabs and bronchial samples)

Not been validated for any other specimen than those listed

-

[149-151]

Human Herpes Viruses 6, 7 and 8

EDTA blood, bone marrow and body fluids (amniotic fluid, CSF), biopsies, newborn screening cards

Not been validated for any other specimen than those listed

-

[122,124,125]

Human Papillomavirus

Vaginal swab, tampon, FFPE, cells collected in liquid based cytology medium PreservCytTM (Hologic, Massachusetts, USA) and SurePathTM (BD, New Jersey, USA)

Not been validated for any other specimen than those listed.

For keratinised tissues, specific methods should be followed

[152,153]

Human Polyomaviruses – JCV and BKV

EDTA blood, CSF, body fluids, urine, biopsies

Not been validated for any other specimen than those listed

JCV and BKV may occur at the same time in same patients

[93,154-158]

Human Polyomaviruses – KIV and WUV

Nasopharyngeal aspirates, bronchial washes and other respiratory secretions, nose/throat swabs, cerebrospinal fluid, urine, whole blood, and stool samples

Not been validated for any other specimen than those listed

-

[159-161]

Human Rhinoviruses

Nasopharyngeal aspirates, nose and

throat swabs, nasal or bronchial washings, broncho alveolar lavage or middle ear fluids

Not been validated for any other specimen than those listed

The assay may miss some viral strains

[162]

Influenza Virus A H5N1 (“Avian Influenza”)

Throat swab, nasal swab or nasal wash

Not been validated for any other specimen than those listed

Prior testing with current strains is mandatory

[130]

Influenza Virus A H1N1 (2009) (“Human Swine Influenza”)

Nasopharyngeal aspirates, bronchial washings, throat swab, nose swab

Not been validated for any other specimen than those listed

-

[113,163,164]

Influenza Type C

Respiratory secretions, bronchial lavage and washings, nose/throat swabs

Not been validated for any other specimen than those listed.

-

[165]

Measles Virus

EDTA blood, Urine, nasopharyngeal aspirates, throat and nasopharyngeal

swab

Not been validated for any other specimen than those listed

No cross-reactivity identified between human and viral DNA in the cited protocols

[166,167]

Norovirus

Faeces

Not been validated for any other specimen than those listed

[168-171]

Respiratory Syncytial Virus Types A and B

Respiratory swabs, sputa or respiratory tissues, and paranasal aspirates

Not been validated for any other specimen than those listed

The coat protein gene of MS2 RNA coliphage and A and B (RSVA and

RSVB)

[172]

Rotavirus

Faeces

Not been validated for any other specimen than those listed

RNA should be completely free from residual stool inhibitors

[173-177]

Varicella Zoster virus

CSF and CNS tissue

Not been validated for any other specimen than those listed

-

[178-180]

Mycology and Parasitology

The molecular diagnosis of parasites and fungi has been a revolution as they are difficult to diagnose at the species level [24]. Although still in a developing phase in parasitology, the lack of culturing techniques has necessitated diagnosis by molecular techniques. The medically encountered parasites and fungi are elaborated in table 3. In mycology the most common testing employed is for molecular detection of Pneumocystis jiroveci (Pneumocystis carinii) an opportunistic fungus causing severe pneumonia in HIV-infected patients, which has made the microscopic and silver staining of tissue specimens a thing of the past [66].

Table 3.  Standardised molecular protocols in diagnostic mycology and parasitology

Fungi/Parasites

Acceptable Specimen

Unacceptable Specimen

Limitations/Comments      

Primer Sequence/Probe and Method References

Aspergillus Species

BAL, EDTA whole blood, serum, fresh and paraffin embedded tissue, vitreous fluid, ascitic fluid and CSF

Sputum and plasma

Penicillium spp. can also be amplified by the assay

[181-184]

Cryptosporidium

Fresh or frozen stool samples

-

PCR has a high sensitivity

[185-196]

Cyclospora

Fresh or frozen stool samples

-

The cited assay has a detection limit of 0.5 oocyst

[189]

Dientamoeba fragilis

Fresh or frozen stool samples

-

The cited assay amplifies 18s rDNA

[197-199]

Entamoeba histolytica

Fresh or frozen stool samples

The cited assay amplifies small subunit (18S) rRNA gene

[200-211]

Giardia

Fresh or frozen stool samples

-

-

[212-216]

Malaria (P. falciparum, P. vivax, P. malariae, P. ovale)

Whole blood samples

-

-

[155,217-219]

Microsporidia

Fresh or frozen stool samples

-

-

[220-227]

Pneumocystis jirovecii

Sputum, induced sputum, bronchial washings, bronchial lavages and lung biopsies

-

The cited assay can be used only as qualitative assay

[228,229]

Trichomonas vaginalis

Genital swabs and urine

-

The cited assay targets  the β-tubulin and 18S rRNA genes

[230]

Toxoplasma gondii

CSF, CNS tissue, and FFPE

CSF supernatant, plasma, and serum

Negative results should be confirmed with serology

[231-233]

Future applications:

The introduction of PCR technology by Nobel Prize winning scientist Dr KaryMullis in 1983 has revolutionised the field of medical diagnostics including microbiology.A number of automated extraction/detection systems are available in the market which will become essential in molecular labs in the future. This system, such as QiaSymphony (Qiagen) offers automated DNA extraction, PCR setup, including reagent preparation, dilution series and sample pipetting. Other automated systems such as the Abbott m1000 system, the ABI PRISM 6100 Nucleic Acid PrepStation, 6700 Automated Nucleic Acid Workstation, and the Corbett Robotics X-tractor Gene perform DNA extraction in a closed system.

In future a diverse array of next generation sequencing (NSG) platforms like Roche 454 will be routinely used as it can sequence ~500 bp approaching that of Sanger sequencing at lower cost [67]. For large bacterial sequences IlluminaHiSeq platform sequence analysers are the most useful, as it delivers ~300 Gb of raw data per eight‑lane flow cell in the form of a 100 bp reads and provide rapid sequencing [68]. The future is promising for bench top NGS analysers, like Ion PGM and the IlluminaMiSeq than the larger counterparts [43]. A new platform of NSG released in 2012 by UK startup Oxford Nanopore Technologies has led to landmark improvement by introducing two sequencing platforms (the GridION and MinION) capable of delivering high-throughput, ultra-long sequence reads at low cost. It offers chips that are configured to read 2,000 or 8,000 pores simultaneously and that reads can be up to tens of kilobases in length. Because it reads native DNA, the Oxford Nanopore technology is anticipated to work with fairly crude samples and low DNA concentrations this makes colony processing a thing of the past [45]. Another future application lies in Micorarrays which are designed to simultaneously monitor whole-genome, host and pathogen gene expression, providing a complete view of disease progression [69]. The most recent generation of microarrays performs sequence analysis often performing WHG in a single experiment. One such array the Gene Chip Customseq Arrays (Affymetrix, CA, US) can sequence up to 300 kb of a genome in 48hrs with minimal amplification of the genomic target thus enabling pathogen detection and identification [70].The limit of detection in Gene Chip is as low as 10 femtograms for pathogenic DNA much below the detection limits of existing technologies. Thus the flexibility and high-throughput nature of microarrays offers an unprecedented opportunity for infectious disease diagnosis. Also the platform which holds a huge scope is digital PCR (dPCR) which is still in its early stage of development. The sample is diluted and partitioned into hundreds or even millions of separate reaction chambers so that each contains one or no copies of the sequence of interest. By counting the number of ‘positive’ partitions (in which the sequence is detected) versus ‘negative’ partitions (in which it is not), scientists can determine exactly how many copies of a DNA molecule were in the original sample [71]. In one type of platform manufactured by Fluidigm and Life Technologies, reactions are created in within specially designed chips or plates. While in other platforms developed by BioRad and RainDance, reagents are sequestered into individual droplets i.e. Droplet Digital PCR (ddPCR).

Thus the future of diagnostic microbiology holds a tremendous scope offered by molecular diagnostics which has the potential to transform the precision, sensitivity and specificity of pathogen detection in a rapid and cost effective manner.

Competing interests

None declared

References

  1. Leeuwen WB Van (2009) Molecular Diagnostics in Clinical Microbiology. 1: 5–20.
  2. French GL (2010) The continuing crisis in antibiotic resistance. Int J Antimicrob Agents 36: S3–S7. [Crossref]
  3. N Khardori (2014) Future of diagnostic microbiology. Indian J Med Microbiol 32: 371–377. [Crossref]
  4. Fluit ADC, Visser MR, Schmitz F (2001) Molecular Detection of Antimicrobial Resistance. 14: 836–871. [Crossref]
  5. Speers DJ (2006) Clinical Applications of Molecular Biology for Infectious Diseases. 27: 39–51.
  6. Seeman NC(2003) DNA in a material world. Nature 421: 427–431.
  7. Hammoudi D, Ayoub Moubareck C, Karam Sarkis D (2014) How to detect carbapenemase producers? A literature review of phenotypic and molecular methods. J Microbiol Methods 107: 106–118. [Crossref]
  8. Mayer LW (1988) Use of Plasmid Profiles in Epidemiologic Surveillance of Disease Outbreaks and in Tracing the Transmission of Antibiotic Resistance. Clin Microbiol Rev 1: 228–243. [Crossref]
  9. Road E, Harcourt P, State R (2012) Plasmid Profile Analysis of Multidrug Resistant Pseudomonas aeruginosa Isolated from Wound Infections in South West, Nigeria. 20: 766–775.
  10. Ndip LM, Aroke G, Mbacham W, Ndip LM, Titanji VP (2014) Antibiogram and Plasmid Profiles of Neisseria gonorrhoeae Isolates from Cameroon : Useful Tools for Epidemiological Survey. Afr J Reprod Health 7: 100-105. [Crossref]
  11. Chun D, Cho DT, Seol SY, Suh MH, Lee YC (1984) R plasmids conferring multiple drug resistance from shigella isolated in Korea. J Hyg (Lond) 92: 53–160. [Crossref]
  12. Stunt RA, Amyes AK, Thomson CJ, Payne DJ, Amyes SG (1998) The production of a novel carbapenem- hydrolysing lactamase in. 835–839. [Crossref]
  13. Si S, Oo A, Odeigha P, Shodipo K, Ja A, et al. (2013) Plasmid profile of Escherichia coli 0157 : H7 from apparently healthy animals. 2: 322–324.
  14. Pijush S, Majharul IM, Kant OR, Gokul B (2013) profiling of multi-drug resistant isolates present in sewage. Int J Med Res 2: 923–930.
  15. Uma B, Prabhakar K, Rajendran S, Kavitha K, Sarayu YL (2009) Antibiotic Sensitivity and Plasmid Profiles of Escherichia coli Isolated from Pediatric Diarrhea. J Glob Infect Dis 1: 107-110. [Crossref]
  16. Malkawi HI, Youssef MT (1998) Antibiotic Susceptibility Testing and Plasmid Profiles of Escherichia coli Isolated from Diarrhoeal Patients. J Trop Pediatr 44: 128-132. [Crossref]
  17. Kamerbeek J, Schouls LEO, Kolk A, Agterveld MVAN, Soolingen DVAN, (1997) Simultaneous Detection and Strain Differentiation of Mycobacterium tuberculosis for Diagnosis and Epidemiology. J Clin Microbiol 35: 907–914. [Crossref]
  18. Marfurt J, Niederwieser I, Makia ND, Beck H, Felger I (2003) Diagnostic genotyping of Old and New World Leishmania species by PCR-RFLP. Diagn Microbiol Infect Dis 46: 115–124. [Crossref]
  19. Marcilla A, Bargues MD, Mas-Coma S (2002) A PCR-RFLP assay for the distinction between Fasciola hepatica and Fasciola gigantica. Mol Cell Probes 16: 327-333. [Crossref]
  20. Watahiki M, Isobe J, Kimata K, Shima T, Kanatani J, et al. (2014) Characterization of Enterohemorrhagic Escherichia coli O111 and O157 Strains Isolated from Outbreak Patients in Japan. 52: 2757–2763. [Crossref]
  21. Bricker BJ (2002) PCR as a diagnostic tool for brucellosis. Vet Microbiol 90: 435–446. [Crossref]
  22. Chou CH, Wang C (2006) Genetic relatedness between Listeria monocytogenes isolates from seafood and humans using PFGE and REP-PCR. Int J Food Microbiol 110: 135–148. [Crossref]
  23. Silva DT, Tejada TS, Blum-Menezes D, Dias PA, Timm CD (2016) Campylobacter species isolated from poultry and humans , and their analysis using PFGE in southern Brazil. Int J Food Microbiol 217: 189–194. [Crossref]
  24. Monis PT, Giglio S, Keegan AR, Thompson RCA (2005) Emerging technologies for the detection and genetic characterization of protozoan parasites. Trends Parasitol 21: 340-346. [Crossref]
  25. Elnifro EM, Ashshi AM, Cooper RJ (2000) Multiplex PCR : Optimization and Application in Diagnostic Virology. Clin Microbiol Rev 13: 559–570. [Crossref]
  26. Persson S, Olsen KEP (2016) Multiplex PCR for identification of Campylobacter coli and Campylobacter jejuni from pure cultures and directly on stool samples. J Med Microbiol 54: 1043–1047. [Crossref]
  27. Louie L, Goodfellow J, Mathieu P, Glatt A, Louie M, et al. (2002) Rapid Detection of Methicillin-Resistant Staphylococci from Blood Culture Bottles by Using a Multiplex PCR Assay. J Clin Microbiol 40: 2786–2790. [Crossref]
  28. Poirel L, Walsh TR, Cuvillier V, Nordmann P (2011) Multiplex PCR for detection of acquired carbapenemase genes. Diagn Microbiol Infect Dis 70: 119–123. [Crossref]
  29. Yamasaki H, Allan JC, Sato MO, Nakao M, Sako Y, et al. (2004) DNA Differential Diagnosis of Taeniasis and Cysticercosis by Multiplex PCR. J Clin Microbiol 42: 548–553. [Crossref]
  30. Calsamiglia M, Pijoan C, Trigo A (2016) Application of a nested polymerase chain reaction assay to detect Mycoplasma hyopneumoniae from nasal swabs. J Vet Diagn Invest 251: 246–251. [Crossref]
  31. Kim DM, Yun NR, Yang TY, Lee JH, Yang JT, et al. (2006) Usefulness of nested pcr for the diagnosis of scrub typhus in clinical practice : a prospective study. Am J Trop Med Hyg 75: 542–545. [Crossref]
  32. Carroll NM, Jaeger EE, Choudhury S, Dunlop AA, Matheson MM, et al. (2000) Detection of and Discrimination between Gram-Positive and Gram-Negative Bacteria in Intraocular Samples by Using Nested PCR. 38: 1753–1757. [Crossref]
  33. Song Q, Lange TÃ, Spahr AÃ, Adler G, Bode G (2016) Characteristic distribution pattern of Helicobacter pylori in dental plaque and saliva detected with nested PCR. J Med Microbiol 49: 49-353. [Crossref]
  34. Rappelli P, Are R, Casu G, Fiori PL, Cappuccinelli P (1998) Development of a Nested PCR for Detection of Cryptococcus neoformans in Cerebrospinal Fluid. J Clin Microbiol 36: 3438–3440. [Crossref]
  35. Oz HS, Hughes WT (2000) Search for Pneumocystis carinii DNA in upper and lower respiratory tract of humans. Diagn Microbiol Infect Dis 37: 161–164. [Crossref]
  36. Klein D (2002) Quantification using real-time PCR technology: applications and limitations. Trends Mol Med 8: 257–260. [Crossref]
  37. Espy MJ, Uhl JR, Sloan LM, Buckwalter SP, Jones MF, et al. (2006) Real-Time PCR in Clinical Microbiology : Applications for Routine Laboratory Testing. Clin Microbiol Rev 19: 165–256. [Crossref]
  38. Yean Y, Heng C, Chong T, Devi S (2006) Rapid detection, serotyping and quantitation of dengue viruses by TaqMan real-time one-step RT-PCR. J Virol Methods 138: 123–130. [Crossref]
  39. Leroy EM, Baize S, Lu CY, Mccormick JB, Georges AJ, et al. (2000) Diagnosis of Ebola Haemorrhagic Fever by RT-PCR in an Epidemic Setting. J Med Virol 467: 463–467. [Crossref]
  40. Savli H, Karadenizli A, Kolayli F, Gundes S, Ozbek U, et al. (2016) Expression stability of six housekeeping genes : a proposal for resistance gene quantification studies of Pseudomonas aeruginosa by real-time quantitative RT-PCR. J Med Microbiol 2003: 403–408. [Crossref]
  41. Balajee SA, Borman AM, Brandt ME, Cano J, Dannaoui E, et al. (2009) Species in the Clinical Mycology Laboratory : Where Are We and Where Should We Go from Here ?. J Clin Microbiol 47: 877–884. [Crossref]
  42. Didelot X, Bowden R, Wilson DJ, Peto TEA, Crook DW (2012) Transforming clinical microbiology with bacterial genome sequencing. Nat Publ Gr 13: 601–612. [Crossref]
  43. Hasman H, Saputra D, Sicheritz-ponten T, Lund O, Svendsen A, et al. (2014) Rapid Whole-Genome Sequencing for Detection and Characterization of Microorganisms Directly from Clinical Samples. J Clin Microbiol 52: 139–146. [Crossref]
  44. Loman NJ, Misra RV, Dallman TJ, Constantinidou C, Gharbia SE, et al. (2012) Performance comparison of benchtop high-throughput sequencing platforms. Nat Biotechnol 30: 434–439. [Crossref]
  45. Eisenstein M (2012) Oxford Nanopore announcement sets sequencing sector abuzz. Nat Biotechnol30: 295–296. [Crossref]
  46. Tada T, Miyoshi-Akiyama T, Shimada K, Kirikae T (2014) Biochemical analysis of metallo-β-lactamase NDM-3 from a multidrug-resistant escherichia coli strain isolated in japan. Antimicrob Agents Chemother 58: 3538–3540. [Crossref]
  47. Woese CR (1987) Bacterial evolution. Microbiol Rev 51: 221–271. [Crossref]
  48. Woo PCY, Lau SKP, Teng JLL, Tse H, Yuen K (2008) Then and now : use of 16S rDNA gene sequencing for bacterial identification and discovery of novel bacteria in clinical microbiology laboratories. Clin Microbiol Infect 14: 908–934. [Crossref]
  49. Toney NC, Toney SR, Butler WR (2010) Utility of high-performance liquid chromatography analysis of mycolic acids and partial 16S rRNA gene sequencing for routine identification of Mycobacterium spp . in a national reference laboratory. Diagn Microbiol Infect Dis 67: 143–152. [Crossref]
  50. Gee JE, De BK, Levett PN, Whitney AM, Novak RT, (2004) Use of 16S rRNA Gene Sequencing for Rapid Confirmatory Identification of Brucella Isolates. J Clin Microbiol 42: 3649–3654. [Crossref]
  51. Whitby PW, Carter KB, Burns JL, Royall JA, LiPuma JJ, et al. (2000) Identification and detection of Stenotrophomonas maltophilia by rRNA-directed PCR. J Clin Microbiol 38: 4305–4309. [Crossref]
  52. Patel JB (2001) 16S rRNA Gene Sequencing for Bacterial Pathogen Identification in the Clinical Laboratory. Mol Diagn 6: 313–322. [Crossref]
  53. Mcnabb A, Eisler D, Adie K, Amos M, Rodrigues M, et al. (2004) Assessment of Partial Sequencing of the 65-Kilodalton Heat Shock Protein Gene (hsp65) for Routine Identification of Mycobacterium Species Isolated from Clinical Sources. J Clin Microbiol 42: 3000–3011. [Crossref]
  54. Clarke L, Moore JE, Millar BC, Garske L, Xu J, et al. (2016) Development of a diagnostic PCR assay that targets a heat-shock protein gene (groES) for detection of Pseudomonas spp . in cystic fibrosis patients. J Med Microbiol 52: 759–763. [Crossref]
  55. Osterloh A, Breloer M (2008) Heat shock proteins : linking danger and pathogen recognition. 1–8.
  56. Garcia L, Kindt A, Bermudez H, Llanos-cuentas A, Doncker S De, et al. (2004) Culture-Independent Species Typing of Neotropical Leishmania for Clinical Validation of a PCR-Based Assay Targeting Heat Shock Protein 70 Genes. J Clin Microbiol 42: 2294–2297. [Crossref]
  57. Boehme CC, Nabeta P, Hillemann D, Nicol MP, Shenai S, et al. (2010) Rapid molecular detection of tuberculosis and rifampin resistance. N Engl J Med 363: 1005–1015. [Crossref]
  58. Yong D, Toleman MA, Giske CG, Cho HS, Sundman K, et al. (2009) Characterization of a New Metallo- ␤ -Lactamase Gene, bla NDM-1 , and a Novel Erythromycin Esterase Gene Carried on a Unique Genetic Structure in Klebsiella pneumoniae Sequence Type 14 from India. Antimicrob Agents Chemother 53: 5046–5054. [Crossref]
  59. Owen RJ(2002) Molecular testing for antibiotic resistance in Helicobacter pylori. Gut 50: 285-289. [Crossref]
  60. Djimoe A, Doumbo OK, Cortese JF, Kayentao K, Doumbo S, et al. (2001) A Molecular marker for chloroquine-resistant Falciparum malaria. N Engl J Med 344: 257–263. [Crossref]
  61. Versalovic J, Lupski JR, Versalovic J (2002) Molecular detection and genotyping : more accurate and of pathogens: rapid answers. 10: 15–21. [Crossref]
  62. Dutly F, Dutly F, Altwegg M, Altwegg M (2001) Whipple's disease and "Tropheryma whippelii". Clin Microbiol Rev 14: 561–583. [Crossref]
  63. Fenollar F, Raoult D (2004) Molecular genetic methods for the diagnosis of fastidious microorganisms. Apmis 112: 785–807. [Crossref]
  64. Hirsch MS, Günthard HF, Schapiro JM, Brun-Vézinet F, Clotet B, et al. (2008) Antiretroviral drug resistance testing in adult HIV-1 infection: 2008 recommendations of an International AIDS Society-USA panel. Clin Infect Dis 47: 266–285. [Crossref]
  65. MacArthur RD (2004) An updated guide to genotype interpretation. AIDS Read 14: 256–258. [Crossref]
  66. Kitada K, Oka S, Kimura S, Shimada K, Serikawa T, et al. (1991) Detection of Pneumocystis-Carinii Sequences by Polymerase Chain-Reaction - Animal-Models and Clinical-Application to Noninvasive Specimens. J Clin Microbiol 29: 1985–1990. [Crossref]
  67. Ellington MJ, Cartwright EJP, Gillespie SH, Ko CU, Brown NM, et al. (2012) Routine Use of Microbial Whole Genome Sequencing in Diagnostic and Public Health Microbiology. PLoS Pathog 8: e1002824. [Crossref]
  68. Harrison EM, Paterson GK, Holden MTG, Larsen J, Stegger M, et al. (2013) Whole genome sequencing identifies zoonotic transmission of MRSA isolates with the novel mecA homologue mecC. EMBO Mol Med 5: 509–515. [Crossref]
  69. Mcloughlin KS(2011) Microarrays for pathogen detection and analysis. Brief Funct Genomics 10: 342–353. [Crossref]
  70. Severgnini M, Cremonesi P, Consolandi C, De Bellis G, Castiglioni B (2011) Advances in DNA Microarray Technology for the Detection of Foodborne Pathogens. Food Bioprocess Technol 4: 936–953.
  71. Baker M (2012) Digital PCR hits its stride. Nat Methods 9: 541–544.
  72. Kösters K, Reischl U, Schmetz J, Riffelmann M, Wirsing von König CH (2002) Real-time LightCycler PCR for detection and discrimination of Bordetella pertussis and Bordetella parapertussis. J Clin Microbiol 40: 1719–1722. [Crossref]
  73. Register KB, Sanden GN (2006) Prevalence and sequence variants of IS481 in Bordetella bronchiseptica: Implications for IS481-based detection of Bordetella pertussis. J Clin Microbiol 44: 4577–4583. [Crossref]
  74. Templeton KE, Scheltinga SA, Zee A Van Der, Diederen BMW, Kruijssen AM, et al. (2003) Evaluation of Real-Time PCR for Detection of and Discrimination Bordetella holmesii for Clinical Diagnosis. J Clin Microbiol 41: 4121–4126. [Crossref]
  75. Yih WK, Silva EA, Ida J, Harrington N, Lett SM, et al. (1999) Bordetella holmesii-like organisms isolated from Massachusetts patients with pertussis-like symptoms. Emerg Infect Dis 5: 441–443. [Crossref]
  76. Redkar R, Rose S, Bricker B, DelVecchio V(2001) Real-time detection of Brucella abortus, Brucella melitensis and Brucella suis. Mol Cell Probes 15: 43–52. [Crossref]
  77. Sifuentes-Rincón AM, Revol A, Barrera-Saldaña HA (1997) Detection and differentiation of the six Brucella species by polymerase chain reaction. Mol Med 3: 734–739. [Crossref]
  78. Bauernfeind A, Schneider I, Jungwirth R, Roller C. (1999) Discrimination of Burkholderia multivorans and Burkholderia vietnamiensis from Burkholderia cepacia genomovars I, III, and IV by PCR.J Clin Microbiol 37: 1335–1339. [Crossref]
  79. Kidd TJ, Bell SC, Coulter C (2003) Genomovar diversity amongst Burkholderia cepacia complex isolates from an Australian adult cystic fibrosis unit. Eur J Clin Microbiol Infect Dis 22: 434–437. [Crossref]
  80. Mahenthiralingam E, Bischof J, Byrne SK, Radomski C, Davies JE, et al. (2000) DNA-Based Diagnostic Approaches for Identification of Burkholderia cepacia Genomovars I and III DNA-Based Diagnostic Approaches for Identification of Burkholderia cepacia Complex , Burkholderia vietnamiensis. J Clin Microbiol 38: 3165–3173. [Crossref]
  81. Vandamme P, Henry D, Coenye T, Nzula S, Vancanneyt M, LiPuma JJ, et al.(2002) Burkholderia anthina sp. nov. and Burkholderia pyrrocinia, two additional Burkholderia cepacia complex bacteria, may confound results of new molecular diagnostic tools. FEMS Immunol Med Microbiol 33: 143–149. [Crossref]
  82. Holland SM, Gaydos CA, Quinn TC (1990) Detection and differentiation of Chlamydia trachomatis, Chlamydia psittaci, and Chlamydia pneumoniae by DNA amplification. Infect Dis 162: 984–987. [Crossref]
  83. Morré SA, Spaargaren J, Fennema JS, de Vries HJ, Coutinho RA, et al. (2005) Real-time Polymerase Chain Reaction To Diagnose Lymphogranuloma Venereum. Emerg Infect Dis 11: 1311–1312. [Crossref]
  84. Madico G, Quinn TC, Boman J, Gaydos CA (2000) Touchdown enzyme time release-PCR for detection and identification of Chlamydia trachomatis, C. pneumoniae, and C. psittaci using the 16S and 16S- 23S spacer rRNA genes. J Clin Microbiol 38: 1085–1093. [Crossref]
  85. Ménard A, Clerc M, Subtil A, Mégraud F, Bébéar C, de Barbeyrac B(2006) Development of a real-time PCR for the detection of Chlamydia psittaci. J Med Microbiol 55: 471–473. [Crossref]
  86. Messmer TO, Skelton SK, Moroney JF, Daugharty H, Fields BS (1997) Application of a nested, multiplex PCR to psittacosis outbreaks. J Clin Microbiol 35: 2043–2046. [Crossref]
  87. Chapman TA, Wu XY, Barchia I, Bettelheim KA, Driesen S, et al. (2006) Comparison of virulence gene profiles of Escherichia coli strains isolated from healthy and diarrheic swine. Appl Environ Microbiol 72: 4782–4795. [Crossref]
  88. Coimbra RS, Grimont F, Grimont PA (1999) Identification of Shigella serotypes by restriction of amplified O- antigen gene cluster. Res Microbiol, 150: 543–553. [Crossref]
  89. Lehmacher A, Meier H, Aleksic S, Bockemühl J (1998) Detection of hemolysin variants of Shiga toxin-producing Escherichia coli by PCR and culture on vancomycin-cefixime-cefsulodin blood agar. Appl Environ Microbiol 64: 2449–2453. [Crossref]
  90. Wu XY, Chapman T, Trott DJ, Bettelheim K, Do TN, et al. (2007) Comparative analysis of virulence genes, genetic diversity, and phylogeny of commensal and enterotoxigenic Escherichia coli isolates from weaned pigs. Appl Environ Microbiol 73: 83–91. [Crossref]
  91. Mackay IM, Gardam T, Arden KE, McHardy S, Whiley DM, et al. (2003) Co-detection and discrimination of six human herpesviruses by multiplex PCR-ELAHA. J Clin Virol 28: 291–302. [Crossref]
  92. Mackay IM, Harnett G, Jeoffreys N, Bastian I, Sriprakash KS, Siebert D, et al. (2006) Detection and discrimination of herpes simplex viruses, Haemophilus ducreyi, Treponema pallidum, and Calymmatobacterium (Klebsiella) granulomatis from genital ulcers. Clin Infect Dis 42: 1431–1438. [Crossref]
  93. Whiley DM, Arden KE, Mackay IM, Syrmis MW, Sloots TP(2004) Simultaneous Detection and Differentiation of Human Polyomaviruses JC and BK by a Rapid and Sensitive PCR-ELAHA Assay and a Survey of the JCV Subtypes Within an Australian Population. J Med Virol 72: 467–472. [Crossref]
  94. Gonin P, Lorange M, Delage G (2000) Performance of a multiplex PCR for the determination of Haemophilus influenzae capsular types in the clinical microbiology laboratory. Diagn Microbiol Infect Dis 37: 1–4. [Crossref]
  95. Maaroufi Y, De Bruyne JM, Heymans C, Crokaert F (2007) Real-time PCR for determining capsular serotypes of Haemophilus influenzae. J Clin Microbiol 45: 2305–2308. [Crossref]
  96. Zhou J, Law DKS, Sill ML, Tsang RSW (2007) Nucleotide sequence diversity of the bexA gene in serotypeable Haemophilus influenzae strains recovered from invasive disease patients in canada. J Clin Microbiol 45:1996–1999. [Crossref]
  97. Boddinghaus B, Roga2021 Copyright OAT. All rights reserv1990) Detection and identification of mycobacteria by amplification of rRNA. J Clin Microbiol28: 1751–1759. [Crossref]
  98. McCallum a D, Watkin SW, Faccenda JF (2011) Non-tuberculous mycobacterial infections in the Scottish Borders: identification, management and treatment outcomes - a retrospective review. J R Coll Physicians Edinb 41: 294–303. [Crossref]
  99. Ross JDC, Jensen JS (2006) Mycoplasma genitalium as a sexually transmitted infection: implications for screening, testing, and treatment. Sex Transm Infect 82: 269–271. [Crossref]
  100. Jensen JS, Bjornelius E, Dohn B, Lidbrink P (2004) Use of TaqMan 5’nuclease real-time PCR for quantitative detection of Mycoplasma genitalium DNA in males with and without urethritis who were attendees at a sexually transmitted disease clinic. J Clin Microbiol 42: 683.
  101. Daxboeck F, Krause R, Wenisch C (2003) Laboratory diagnosis of Mycoplasma pneumoniae infection. Clin Microbiol Infect 9: 263–273. [Crossref]
  102. Gil JC, Cedillo RL, Mayagoitia BG, Paz MD (1993) Isolation of Mycoplasma pneumoniae from asthmatic patients. Ann Allergy 70: 23–25. [Crossref]
  103. Loens K, Beck T, Ursi D, Pattyn S, Goossens H, et al. (2006) Two quality control exercises involving nucleic acid amplification methods for detection of Mycoplasma pneumoniae and Chlamydophila pneumoniae and carried out 2 years apart (in 2002 and 2004). J Clin Microbiol 44: 899–908. [Crossref]
  104. Goire N, Nissen MD, LeCornec GM, Sloots TP, Whiley DM (2008) A duplex Neisseria gonorrhoeae real-time polymerase chain reaction assay targeting the gonococcal porA pseudogene and multicopy opa genes. Diagn Microbiol Infect Dis 61: 6–12. [Crossref]
  105. Smith DW, Tapsall JW, Lum G (2005) Guidelines for the use and interpretation of nucleic acid detection tests for Neisseria gonorrhoeae in Australia: a position paper on behalf of the Public Health Laboratory Network. Commun Dis Intell Q Rep 29: 358–365. [Crossref]
  106. Tabrizi SN, Chen S, Tapsall J, Garland SM(2005) Evaluation of opa-based real-time PCR for detection of Neisseria gonorrhoeae. Sex Transm Dis 32:199–202. [Crossref]
  107. Whiley DM, Sloots TP (2005) Comparison of three in-house multiplex PCR assays for the detection of Neisseria gonorrhoeae and Chlamydia trachomatis using real-time and conventional detection methodologies. Pathology 37: 364–370. [Crossref]
  108. Bryant PA, Li HY, Zaia A, Griffith J, Hogg G, et al. (2004) Prospective Study of a Real-Time PCR That Is Highly Sensitive , Specific , and Clinically Useful for Diagnosis of Meningococcal Disease in Children. Society 42: 2919–2925. [Crossref]
  109. Hackett SJ, Carrol ED, Guiver M, Marsh J, Sills JA, et al. (2002) Improved case confirmation in meningococcal disease with whole blood Taqman PCR. Arch Dis Child 86: 449–452. [Crossref]
  110. Høiby N, Frederiksen B, Pressler T (2005) Eradication of early Pseudomonas aeruginosa infection. J Cyst Fibros 4: 49–54. [Crossref]
  111. Jaffe RI, Lane JD, Bates CW (2001) Real-time identification of Pseudomonas aeruginosa direct from clinical samples using a rapid extraction method and polymerase chain reaction (PCR). J Clin Lab Anal 15: 131–137. [Crossref]
  112. Khan AA, Cerniglia CE (1994) Detection of Pseudomonas aeruginosa from clinical and environmental samples by amplification of the exotoxin A gene using PCR. Appl Environ Microbiol 60: 3739–3745. [Crossref]
  113. Whiley DM, Lambert SB, Bialasiewicz S, Goire N, Nissen MD, et al. (2008) False-negative results in nucleic acid amplification tests-do we need to routinely use two genetic targets in all assays to overcome problems caused by sequence variation? Crit Rev Microbiol 34: 71–76. [Crossref]
  114. Stenos J, Graves SR, Unsworth NB (2005) A highly sensitive and specific real-time PCR assay for the detection of spotted fever and typhus group Rickettsiae. Am J Trop Med Hyg 73: 1083–1085. [Crossref]
  115. Corless CE, Guiver M, Borrow R, Fox AJ, Kaczmarski EB (2001) Influenzae and Streptococcus pneumoniae in Suspected Cases of Meningitis and Septicemia Using Real-Time PCR. 39: 1553–1558. [Crossref]
  116. Larsen SA, Steiner BM, Rudolph AH (1995) Laboratory diagnosis and interpretation of tests for syphilis . These include : Laboratory Diagnosis and Interpretation of Tests for Syphilis. Clin Microbiol Rev 8: 1–21. [Crossref]
  117. Leslie DE, Azzato F, Karapanagiotidis T, Leydon J, Fyfe J (2007) Development of a real-time PCR assay to detect Treponema pallidum in clinical specimens and assessment of the assay’s performance by comparison with serological testing. J Clin Microbiol 45: 93–96. [Crossref]
  118. Orle KA, Gates CA, Martin DH, Body BA, Weiss JB (1996) Simultaneous PCR detection of Haemophilus ducreyi, Treponema pallidum, and herpes simplex virus types 1 and 2 from genital ulcers. J Clin Microbiol 34: 49–54. [Crossref]
  119. Gray GC, McCarthy T, Lebeck MG, Schnurr DP, Russell KL, et al. (2007) Genotype prevalence and risk factors for severe clinical adenovirus infection, United States 2004-2006. Clin Infect Dis 45: 1120–1131. [Crossref]
  120. Selvaraj G, Kirkwood C, Bines J, Buttery J (2006) Molecular epidemiology of adenovirus isolates from patients diagnosed with intussusception in Melbourne, Australia. J Clin Microbiol 44: 3371–3373. [Crossref]
  121. Xu W, Mcdonough MC, Erdman DD (2000) Species-Specific Identification of Human Adenoviruses by a Multiplex PCR Assay Species-Specific Identification of Human Adenoviruses by a Multiplex PCR Assay. J Clin Microbiol 38: 4114–4120. [Crossref]
  122. Barbi M, Binda S, Primache V, Caroppo S, Didò P, et al. (2000) Cytomegalovirus DNA detection in Guthrie cards: A powerful tool for diagnosing congenital infection. J Clin Viro17: 159–165. [Crossref]
  123. Barbi M, Binda S, Primache V, Luraschi C, Corbetta C, et al. (2006) Diagnosis of congenital cytomegalovirus infection by detection of viral DNA in dried blood spots. J Clin Virol 35: 206–209.
  124. Howard J, Hall B, Brennan LE, Arbuckle S, Craig ME, et al. (2009) Utility of newborn screening cards for detecting CMV infection in cases of stillbirth. J Clin Virol 44: 215–218. [Crossref]
  125. McIver CJ, Jacques CFH, Chow SSW, Munro SC, Scott GM, et al. (2005) Development of multiplex PCRs for detection of common viral pathogens and agents of congenital infections. J Clin Microbiol 43: 5102–5110. [Crossref]
  126. Utz U, Britt W, Vugler L, Mach M (1989) Identification of a neutralizing epitope on glycoprotein gp58 of human cytomegalovirus. J Virol 63: 1995–2001. [Crossref]
  127. Chien L, Liao T, Shu P, Gubler DJ, Chang GJ, et al. (2006) Development of Real-Time Reverse Transcriptase PCR Assays To Detect and Serotype Dengue Viruses. J Clin Microbiol 44: 1295–1304. [Crossref]
  128. Das S, Pingle MR, Muñoz-Jordán J, Rundell MS, Rondini S, et al. (2008) Detection and serotyping of dengue virus in serum samples by multiplex reverse transcriptase PCR-ligase detection reaction assay. J Clin Microbiol46: 3276–3284. [Crossref]
  129. Lanciotti RS, Calisher CH, Gubler DJ, Chang GJ, Vorndam AV (1992) Rapid detection and typing of dengue viruses from clinical samples by using reverse transcriptase-polymerase chain reaction. J Clin Microbiol 30: 545–551. [Crossref]
  130. Smith G, Smith I, Harrower B, Warrilow D, Bletchly C (2006) A simple method for preparing synthetic controls for conventional and real-time PCR for the identification of endemic and exotic disease agents. J Virol Methods 135: 229–234. [Crossref]
  131. Warrilow D, Northill JA, Pyke A, Smith GA (2002) Single rapid TaqMan fluorogenic probe based PRC assay that detects all four dengue serotypes. J Med Virol 66: 524–528. [Crossref]
  132. Fowlkes AL, Honarmand S, Glaser C, Yagi S, Schnurr D, et al. (2008) Enterovirus-associated encephalitis in the California encephalitis project, 1998-2005. J Infect Dis 198: 1685–1691. [Crossref]
  133. Oberste MS, Maher K, Williams AJ, Dybdahl-Sissoko N, Brown BA, et al. (2006) Species-specific RT-PCR amplification of human enterovirus: A tool for rapid species identification of uncharacterized enteroviruses. J Gen Virol 87: 119–128. [Crossref]
  134. Verstrepen WA, Kuhn S, Kockx MM, Van De Vyvere ME, Mertens AH (2001) Rapid detection of enterovirus RNA in cerebrospinal fluid specimens with a novel single-tube real-time reverse transcription-PCR assay. J Clin Microbiol 39: 4093–4096. [Crossref]
  135. Zoll GJ, Melchers WJG, Kopecka H, Jambroes G, Van der Poel HJA, et al. (1992) General primer-mediated polymerase chain reaction for detection of enteroviruses: Application for diagnostic routine and persistent infections. J Clin Microbiol 30: 160–165. [Crossref]
  136. Chambon M, Archimbaud C, Bailly JL, Henquell C, Regagnon C, et al. (2001) Semiquantitative PCR analysis of Epstein-Barr virus DNA in clinical samples of patients with EBV-associated diseases. J Med Virol 65: 348–357. [Crossref]
  137. Stanley KK, Szewczuk E (2005) Multiplexed tandem PCR: Gene profiling from small amounts of RNA using SYBR Green detection. Nucleic Acids Res 33: e180. [Crossref]
  138. Nainan O V, Xia G, Vaughan G, Margolis HS (2006) Diagnosis of Hepatitis A Virus Infection : a Molecular Approach. 19: 63–79. [Crossref]
  139. Simmonds P, Bukh J, Combet C, Deleage G, Enomoto N, et al. (2005) Consensus proposals for a unified system of nomenclature of hepatitis C virus genotypes. Hepatology 42: 962–973. [Crossref]
  140. Simpson LH, Battegay M, Hoofnagle JH, Waggoner JG, Di Bisceglie AM (1994) Hepatitis delta virus RNA in serum of patients with chronic delta hepatitis. Dig Dis Sci 39: 2650–2655. [Crossref]
  141. Erker JC, Desai SM, Mushahwar IK (1999) Rapid detection of Hepatitis E virus RNA by reverse transcription-polymerase chain reaction using universal oligonucleotide primers. J Virol Methods 81: 109–113. [Crossref]
  142. Aberle SW, Puchhammer-Stockl, E (2002) Diagnosis of herpesvirus infections of the central nervous system. J Clin Virol25: 79–85. [Crossref]
  143. Cinque P, Cleator GM, Weber T, Monteyne P, Sindic CJ, van Loon AM (1996) The role of laboratory investigation in the diagnosis and management of patients with suspected herpes simplex encephalitis: a consensus report. The EU Concerted Action on Virus Meningitis and Encephalitis. J Neurol Neurosurg Psychiatry 61: 339–345. [Crossref]
  144. Mitchell PS, Espy MJ, Smith TF, Toal DR, Rys PN, et al. (1997) Laboratory diagnosis of central nervous system infections with herpes simplex virus by PCR performed with cerebrospinal fluid specimens. J Clin Microbiol 35: 2873–2877. [Crossref]
  145. Tebas P, Nease RF, Storch GA (1998) Use of the polymerase chain reaction in the diagnosis of herpes simplex encephalitis: a decision analysis model. Am J Med 105: 287–295. [Crossref]
  146. Troendle-Atkins J, Demmler GJ, Buffone GJ (1993) Rapid diagnosis of herpes simplex virus encephalitis by using the polymerase chain reaction. J Pediatr 123: 376–380. [Crossref]
  147. Watzinger F, Suda M, Preuner S, Baumgartinger R, Baskova L, et al. (2004) Real-Time Quantitative PCR Assays for Detection and Monitoring of Pathogenic Human Viruses in Immunosuppressed Pediatric Patients. J Clin Microbiol 42: 5189–5198. [Crossref]
  148. Allander T, Tammi MT, Eriksson M, Bjerkner A, Tiveljung-Lindell A, et al. (2005) Cloning of a human parvovirus by molecular screening of respiratory tract samples. Proc Natl Acad Sci U S A 102: 12891–12896. [Crossref]
  149. Dare RK, Fry AM, Chittaganpitch M, Sawanpanyalert P, Olsen SJ, et al. (2007) Human Coronavirus Infections in Rural Thailand : A Comprehensive Study Using Real-Time Reverse- Transcription Polymerase Chain Reaction Assays. , 30333: 1321–1328. [Crossref]
  150. Gunson RN, Collins TC, Carman WF (2005) Real-time RT-PCR detection of 12 respiratory viral infections in four triplex reactions. J Clin Virol 33: 341–344. [Crossref]
  151. Elden LJ, Loon AM, Alphen F, Hendriksen KA, Hoepelman AI, et al. (2004) Frequent detection of human coronaviruses in clinical specimens from patients with respiratory tract infection by use of a novel real-time reverse-transcriptase polymerase chain reaction. J Infect Dis 189: 652-657. [Crossref]
  152. Gravitt PE, Peyton CL, Alessi TQ, Wheeler CM, Hildesheim a, Schiffman MH, et al. (2000) Improved Amplification of Genital Human Papillomaviruses Improved Amplification of Genital Human Papillomaviruses. 38: 357–361. [Crossref]
  153. Stevens MP, Rudland E, Garland SM, Tabrizi SN (2006) Assessment of MagNA pure LC extraction system for detection of human papillomavirus (HPV) DNA in PreservCyt samples by the Roche AMPLICOR and LINEAR ARRAY HPV tests. J Clin Microbiol 44: 2428–2433. [Crossref]
  154. Arthur RR, Dagostin S, Shah K V (1989) Detection of BK virus and JC virus in urine and brain tissue by the polymerase chain reaction. J Clin Microbiol 27: 1174–1179. [Crossref]
  155. Bialasiewicz S, Whiley DM, Nissen MD, Sloots TP (2007) Impact of competitive inhibition and sequence variation upon the sensitivity of malaria PCR. J Clin Microbiol 45: 1621–1623. [Crossref]
  156. Chesters PM, Heritage J, Mccance DJ, The S, Diseases I, et al. (1983) Persistence of DNA Sequences of BK Virus and JC Virus in Normal Human Tissues and in Diseased Tissues Persistence of DNA Sequences of BK Virus and JC Virus in Normal Human Tissues and in Diseased Tissues. J Infect Dis 147: 676–684. [Crossref]
  157. Heritage J, Chesters PM, McCance DJ (1981) The persistence of papovavirus BK DNA sequences in normal human renal tissue. J Med Virol 8: 143–150. [Crossref]
  158. Pal A, Sirota L, Maudru T, Peden K, Lewis AM (2006) Real-time, quantitative PCR assays for the detection of virus-specific DNA in samples with mixed populations of polyomaviruses. J Virol Methods 135: 32–42. [Crossref]
  159. Allander T, Andreasson K, Gupta S, Bjerkner A, Bogdanovic G, et al. (2007) Identification of a third human polyomavirus. J Virol81: 4130–4136. [Crossref]
  160. Bialasiewicz S, Whiley DM, Lambert SB, Gould A, Nissen MD, et al. (2007) Development and evaluation of real-time PCR assays for the detection of the newly identified KI and WU polyomaviruses. J Clin Virol 40: 9–14. [Crossref]
  161. Gaynor AM, Nissen MD, Whiley DM, Mackay IM, Lambert SB, Wu G, et al (2007) Identification of a novel polyomavirus from patients with acute respiratory tract infections. PLoS Pathog, 3: 0595–0604. [Crossref]
  162. Lu X, Holloway B, Dare RK, Kuypers J, Yagi S, et al. (2008) Real-time reverse transcription-PCR assay for comprehensive detection of human rhinoviruses. J Clin Microbiol 46: 533–539. [Crossref]
  163. Whiley DM, Sloots TP (2005) A 5-nuclease real-time reverse transcriptase-polymerase chain reaction assay for the detection of a broad range of influenza A subtypes, including H5N1. Diagn Microbiol Infect Dis 53: 335–337. [Crossref]
  164. Whiley DM, Bialasiewicz S, Bletchly C, Faux CE, Harrower B, Gould AR, et al. (2009) Detection of novel influenza A(H1N1) virus by real-time RT-PCR. J Clin Virol 45: 203–204. [Crossref]
  165. Matsuzaki Y, Sugawara K, Mizuta K, Tsuchiya E, Muraki Y, et al. (2002) Antigenic and genetic characterization of influenza C viruses which caused two outbreaks in Yamagata City, Japan, in 1996 and 1998. J Clin Microbiol 40: 422–429. [Crossref]
  166. Hübschen JM, Kremer JR, De Landtsheer S, Muller CP (2008) A multiplex TaqMan PCR assay for the detection of measles and rubella virus. J Virol Methods 149: 246–250. [Crossref]
  167. El Mubarak HS, De Swart RL, Osterhaus ADME, Schutten M (2005) Development of a semi-quantitative real-time RT-PCR for the detection of measles virus. J Clin Virol 32: 313–317. [Crossref]
  168. Bull RA, Tu ET, Mciver CJ, Rawlinson WD, White PA (2006) Emergence of a New Norovirus Genotype II . 4 Variant Associated with Global Outbreaks of Gastroenteritis Emergence of a New Norovirus Genotype II. 4 Variant Associated with Global Outbreaks of Gastroenteritis. Methods44: 327–333. [Crossref]
  169. Hansman GS, Katayama K, Maneekarn N, Peerakome S, Khamrin P, et al. (2004) Genetic Diversity of Norovirus and Sapovirus in Hospitalized Infants with Sporadic Cases of Acute Gastroenteritis in Chiang Mai, Thailand. J Clin Microbiol 42: 1305–1307. [Crossref]
  170. Jiang X, Huang PW, Zhong WM, Farkas T, Cubitt DW, Matson DO (1999) Design and evaluation of a primer pair that detects both Norwalk- and Sapporo-like caliciviruses by RT-PCR. J Virol Methods 83: 145–154. [Crossref]
  171. Kojima S, Kageyama T, Fukushi S, Hoshino FB, Shinohara M, et al. (2002) Genogroup-specific PCR primers for detection of Norwalk-like viruses. J Virol Methods 100: 107–114. [Crossref]
  172. Cubie HA, Inglis JM, Leslie EE, Edmunds AT, Totapally B (1992) Detection of respiratory syncytial virus in acute bronchiolitis in infants. J Med Virol, 38: 283–287. [Crossref]
  173. Elschner M, Prudlo J, Hotzel H, Otto P, Sachse K (2002) Nested reverse transcriptase-polymerase chain reaction for the detection of group A rotaviruses. J Vet Med B Infect Dis Vet Public Health 49: 77–81. [Crossref]
  174. Gentsch JR, Glass RI, Woods P, Gouvea V, Gorziglia M, et al. (1992) Identification of group A rotavirus gene 4 types by polymerase chain reaction. J Clin Microbiol 30: 1365–1373. [Crossref]
  175. Gouvea V, Glass RI, Woods P, Taniguchi K, Clark HF, Forrester B, et al. (1990) Polymerase chain reaction amplification and typing of rotavirus nucleic acid from stool Polymerase Chain Reaction Amplification and Typing of Rotavirus Nucleic Acid from Stool Specimens. J Clin Microbiol 28: 276–282. [Crossref]
  176. Simmonds MK, Armah G, Asmah R, Banerjee I, Damanka S, et al. (2008) New oligonucleotide primers for P-typing of rotavirus strains: Strategies for typing previously untypeable strains. J Clin Virol 42: 368–373. [Crossref]
  177. Zeng SQ, Halkosalo A, Salminen M, Szakal ED, Puustinen L, Vesikari T (2008) One-step quantitative RT-PCR for the detection of rotavirus in acute gastroenteritis. J Virol Methods 153: 238–240. [Crossref]
  178. Cooper C, Ratnamohan JWVM, Arbuckle S Congenital varicella syndrome diagnosed by polymerase chain reaction-scarring of the spinal cord , not the skin. J Paediatr Child Health 36: 186-188. [Crossref]
  179. Hausfater P, Fillet AM, Rozenberg F, Arthaud M, Trystram D, et al. (2004) Prevalence of Viral Infection Markers by Polymerase Chain Reaction Amplification and Interferon-Alpha Measurements among Patients Undergoing Lumbar Puncture in An Emergency Department. J Med Virol 73: 137–146. [Crossref]
  180. Ihekwaba UK, Kudesia G, McKendrick MW (2008) Clinical Features of Viral Meningitis in Adults: Significant Differences in Cerebrospinal Fluid Findings among Herpes Simplex Virus, Varicella Zoster Virus, and Enterovirus Infections. Clin Infect Dis 47: 783–789. [Crossref]
  181. Halliday C, Hoile R, Sorrell T, James G, Yadav S, et al. (2006) Role of prospective screening of blood for invasive aspergillosis by polymerase chain reaction in febrile neutropenic recipients of haematopoietic stem cell transplants and patients with acute leukaemia. Br J Haematol 132: 478–486. [Crossref]
  182. Halliday C, Wu QX, James G, Sorrell T (2005) Development of a Nested Qualitative Real-Time PCR Assay To Detect Aspergillus Species DNA in Clinical Specimens Development of a Nested Qualitative Real-Time PCR Assay To Detect Aspergillus Species DNA in Clinical Specimens. 43:13–16. [Crossref]
  183. Skladny H, Buchheidt D, Baust C, Seifarth W, Leib-mösch C, et al. (1999) Specific Detection of Aspergillus Species in Blood and Bronchoalveolar Lavage Samples of Immunocompromised Patients by Two-Step PCR Specific Detection of Aspergillus Species in Blood and Bronchoalveolar Lavage Samples of Immunocompromised Patients by two-step PCR. 37: 3865–3871. [Crossref]
  184. Williamson EC, Leeming JP, Palmer HM, Steward CG, Warnock D, Marks DI, et al.(2000) Diagnosis of invasive aspergillosis in bone marrow transplant recipients by polymerase chain reaction. Br J Haematol 108: 132–139. [Crossref]
  185. Abubakar I, Aliyu S, Arumugam C, Hunter P, Usman N (2012) Prevention and treatment of cryptosporidiosis in immunocompromised patients ( Review ). Cochrane Collab 5.
  186. Balatbat AB, Jordan GW, Tang YJ, Silva JJ (1996) Detection of Cryptosporidium parvum DNA in human feces by nested PCR. J Clin Microbiol 34: 1769–1772. [Crossref]
  187. Coupe S, Sarfati C, Hamane S, Derouin F, Coupe S, et al. (2005) Detection of Cryptosporidium and Identification to the Species Level by Nested PCR and Restriction Fragment Length Polymorphism. J Clincal Microbiol 43: 1017–1023.
  188. Fotedar R, Stark D, Beebe N, Marriott D, Ellis J, et al. (2007) PCR detection of Entamoeba histolytica, Entamoeba dispar, and Entamoeba moshkovskii in stool samples from Sydney, Australia. J Clin Microbiol 45: 1035–1037. [Crossref]
  189. Garcia LS. (2007) Diagnostic Medical Parasitology FAQ ’ s. ASM Press, Washington, DC.
  190. Higgins J, Fayer R, Trout J, Xiao L (2001) Real-time PCR for the detection of Cryptosporidium parvum. J Microbiol Methods 47: 323–337. [Crossref]
  191. Joachim A (2004) Human cryptosporidiosis: An update with special emphasis on the situation in Europe. J Vet Med Ser B Infect Dis Vet Public Heal 51: 251–259. [Crossref]
  192. Kaushik K, Khurana S, Wanchu A, Malla N (2008) Evaluation of staining techniques, antigen detection and nested PCR for the diagnosis of cryptosporidiosis in HIV seropositive and seronegative patients. Acta Trop 107: 1–7. [Crossref]
  193. Limor JR, Lal A a, Xiao L (2002) Detection and Differentiation of Cryptosporidium Parasites That Are Pathogenic for Humans by Real-Time PCR Detection and Differentiation of Cryptosporidium Parasites That Are Pathogenic for Humans by Real-Time PCR. 40: 2335–2338. [Crossref]
  194. Morgan U, Weber R, Xiao L, Sulaiman I, Thompson CA, et al. (2000) Molecular Characterization of Cryptosporidium Isolates Obtained from Human Immunodeficiency Virus-Infected Individuals Living in Switzerland, Kenya, and the United States Molecular Characterization of Cryptosporidium Isolates Obtained from Human Immunod. Society 38: 1180–1183.
  195. Morgan UM, Pallant L, Dwyer BW, Forbes DA, Rich G, et al. (1998) Comparison of PCR and microscopy for detection of Cryptosporidium parvum in human fecal specimens: Clinical trial. J Clin Microbiol 36: 995–998. [Crossref]
  196. Pedraza-Daz S, Amar CFL, McLauchlin J, Nichols GL, Cotton KM, et al. (2001) Cryptosporidium meleagridis from humans: Molecular analysis and description of affected patients. J Infect 42: 243–250.
  197. Peek R, Reedeker FR, Van Gool T (2004) Direct Amplification and Genotyping of Dientamoeba fragilis from Human Stool Specimens. J Clin Microbiol. 42: 631–635. [Crossref]
  198. Stark D, Beebe N, Marriott D, Ellis J, Harkness J (2005) Detection of Dientamoeba fragilis in fresh stool specimens using PCR. Int J Parasitol 35: 57–62. [Crossref]
  199. Stark DJ, Beebe N, Marriott D, Ellis JT, Harkness J (2006) Dientamoebiasis: Clinical importance and recent advances. Trends Parasitol 22: 92–96. [Crossref]
  200. Calore EE, Calore NM, Cavaliere MJ (2002) Salpingitis due to Entamoeba histolytica. Braz J Infect Dis 6: 97–99. [Crossref]
  201. Evangelopoulos A, Legakis N, Vakalis N(2001) Microscopy, PCR and ELISA applied to the epidemiology of amoebiasis in Greece. Parasitol Int 50: 185–189. [Crossref]
  202. Hamzah Z, Petmitr S, Mungthin M, Leelayoova S, Chavalitshewinkoon-Petmitr P (2006) Differential detection of Entamoeba histolytica, Entamoeba dispar, and Entamoeba moshkovskii by a single-round PCR assay. J Clin Microbiol 44: 3196–3200. [Crossref]
  203. Haque R, Mollah NU, Ali IKM, Alam K, Eubanks A, et al. (2000) Diagnosis of amebic liver abscess and intestinal infection with the TechLab Entamoeba histolytica II antigen detection and antibody tests. J Clin Microbiol 38: 3235–3239. [Crossref]
  204. Haque R, Neville LM, Hahn P, Petri WA (1995) Rapid diagnosis of Entamoeba infection by using Entamoeba and Entamoeba histolytica stool antigen detection kits. J Clin Microbiol 33: 2558–2561. [Crossref]
  205. Knappik M, B??rner U, Jelinek T (2005) Sensitivity and specificity of a new commercial enzyme-linked immunoassay kit for detecting Entamoeba histolytica IgG antibodies in serum samples. Eur J Clin Microbiol Infect Dis 24: 701–703. [Crossref]
  206. Qvarnstrom Y, James C, Xayavong M, Holloway BP, Visvesvara GS, et al (2005) Comparison of Real-Time PCR Protocols for Differential Laboratory Diagnosis of Amebiasis. J Clin Microbiol 43: 5491–5497. [Crossref]
  207. Sanchez-Guillen MC, Perez-Fuentes R, Salgado-Rosas H, Ruiz-Arguelles A, Ackers J, et al. (2002) Differentiation of Entamoeba histolytica/Entamoeba dispar by PCR and their correlation with humoral and cellular immunity in individuals with clinical variants of amoebiasis. Am J Trop Med Hyg 66: 731–737. [Crossref]
  208. Som I, Azam A, Bhattacharya A, Bhattacharya S (2000) Inter- and intra-strain variation in the 5.8S ribosomal RNA and internal transcribed spacer sequences of Entamoeba histolytica and comparison with Entamoeba dispar, Entamoeba moshkovskii and Entamoeba invadens. Int J Parasitol 30: 723–728. [Crossref]
  209. Stauffer W, Abd-Alla M, Ravdin JI (2006) Prevalence and incidence of Entamoeba histolytica infection in South Africa and Egypt. Arch Med Res 37: 266–269. [Crossref]
  210. Troll H, Marti H, Weiss N (1997) Simple differential detection of Entamoeba histolytica and Entamoeba dispar in fresh stool specimens by sodium acetate-acetic acid-formalin concentration and PCR. J Clin Microbiol 35: 1701–1705. [Crossref]
  211. van Hal SJ, Stark DJ, Fotedar R, Marriott D, Ellis JT, Harkness JL (2007) Amoebiasis: Current status in Australia. Med J Aust 186: 412–416. [Crossref]
  212. Ghosh S, Debnath A, Sil A, De S, Chattopadhyay DJ (2000) PCR detection of Giardia lamblia in stool: targeting intergenic spacer region of multicopy rRNA geneMol Cell Probes14: 181–189. [Crossref]
  213. Oster N, Gehrig-Feistel H, Jung H, Kammer J, McLean JE (2006) Evaluation of the immunochromatographic CORIS Giardia-Strip test for rapid diagnosis of Giardia lamblia. Eur J Clin Microbiol Infect Dis 25: 112–115. [Crossref]
  214. Savioli L, Smith H, Thompson A (2006) Giardia and Cryptosporidium join the “Neglected Diseases Initiative.” Trends Parasitol 22: 203–208. [Crossref]
  215. Verweij JJ, Schinkel J, Laeijendecker D, Van Rooyen MAA, Van Lieshout L, Polderman AM(2003) Real-time PCR for the detection of Giardia lamblia. Mol Cell Probes 17: 223–225. [Crossref]
  216. Weitzel T, Dittrich S, Möhl I, Adusu E, Jelinek T (2006) Evaluation of seven commercial antigen detection tests for Giardia and Cryptosporidium in stool samples. Clin Microbiol Infect 12: 656–659. [Crossref]
  217. Charles DM, Hart J, Davis WA, Sullivan E, Dowse GK (2005) Notifications of imported malaria in Western Australia, 1990-2001: Incidence, associated factors and chemoprophylaxis. Med J Aust 182: 164–167. [Crossref]
  218. Perandin F, Manca N (2004) Development of a real-time PCR assay for detection of Plasmodium falciparum, Plasmodium vivax, and Plasmodium ovale for routine clinical diagnosis. Journal of clinical 42: 1214–1219.
  219. Whiley DM, LeCornec GM, Baddeley A, Savill J, Syrmis MW, et al. (2004) Detection and differentiation of Plasmodium species by polymerase chain reaction and colorimetric detection in blood samples of patients with suspected malaria. Diagn Microbiol Infect Dis 49: 25–29. [Crossref]
  220. Brasil P, de Lima DB, de Paiva DD, Lobo MS, Sodré FC, et al. (2000) Clinical and diagnostic aspects of intestinal microsporidiosis in HIV-infected patients with chronic diarrhea in Rio de Janeiro, Brazil. Rev Inst Med Trop Sao Paulo, 42: 299–304. [Crossref]
  221. Breton J, Bart-Delabesse E, Biligui S, Carbone A, Seiller X, et al. (2007) New highly divergent rRNA sequence among biodiverse genotypes of Enterocytozoon bieneusi strains isolated from humans in Gabon and Cameroon. J Clin Microbiol 45: 2580–2589. [Crossref]
  222. Dowd SE, Gerba CP, Javier Enriquez F, Pepper IL (1998) PCR amplification and species determination of microsporidia in formalin-fixed feces after immunomagnetic separation. Appl Environ Microbiol 64: 333–336. [Crossref]
  223. Fedorko DP, Nelson N a, Cartwright CP (1995) Identification of microsporidia in stool specimens by using PCR and restriction Updated information and services can be found at : These include : Identification of Microsporidia in Stool Specimens by Using PCR and Restriction Endonucleases. 33: 1739–1741.
  224. Franzen C, Müller A, Hartmann P, Hegener P, Schrappe M, et al. (1998) Polymerase chain reaction for diagnosis and species differentiation of microsporidia. Folia Parasitol (Praha) 45: 140–148. [Crossref]
  225. Rinder H, Janitschke K, Aspöck H, Da Silva AJ, Deplazes P, et al. (1998) Blinded, externally controlled multicenter evaluation of light microscopy and PCR for detection of microsporidia in stool specimens. The Diagnostic Multicenter Study Group on Microsporidia. J Clin Microbiol 36: 1814–1818. [Crossref]
  226. Stark D, Van Hal S, Barratt J, Ellis J, Marriott D, Harkness J (2009) Limited genetic diversity among genotypes of Enterocytozoon bieneusi strains isolated from HIV-infected patients from Sydney, Australia. J Med Microbiol 58: 355–357. [Crossref]
  227. Verweij JJ, ten Hove R, Brienen EAT, van Lieshout L (2007) Multiplex detection of Enterocytozoon bieneusi and Encephalitozoon spp. in fecal samples using real-time PCR. Diagn Microbiol Infect Dis 57: 163–167. [Crossref]
  228. Morris A, Wei K, Afshar K, Huang L (2008) Epidemiology and clinical significance of pneumocystis colonization. J Infect Dis 197: 10–17. [Crossref]
  229. Palladino S, Kay I, Fonte R, Flexman J (2001) Use of real-time PCR and the LightCycler system for the rapid detection of Pneumocystis carinii in respiratory specimens. Diagn Microbiol Infect Dis 39: 233–236. [Crossref]
  230. Hardick J, Yang S, Lin S, Duncan D, Gaydos C (2003) Use of the Roche LightCycler Instrument in a Real-Time PCR for Trichomonas vaginalis in Urine Samples from Females and Males Use of the Roche LightCycler Instrument in a Real-Time PCR for Trichomonas vaginalis in Urine Samples from Females and Males. 41: 5619–5622. [Crossref]
  231. Filisetti D, Gorcii M, Pernot-marino E, Villard O, Candolfi E (2003) Diagnosis of Congenital Toxoplasmosis : Comparison of Targets for Detection of Toxoplasma gondii by PCR Diagnosis of Congenital Toxoplasmosis : Comparison of Targets for Detection of Toxoplasma gondii by PCR. 41: 4826–4828. [Crossref]
  232. Homan WL, Vercammen M, De Braekeleer J, Verschueren H (2000) Identification of a 200- to 300-fold repetitive 529 bp DNA fragment in Toxoplasma gondii, and its use for diagnostic and quantitative PCR. Int J Parasitol 30: 69–75. [Crossref]
  233. James GS, Sintchenko VG, Dickeson DJ, Gilbert GL (1996) Comparison of cell culture , mouse inoculation , and PCR for detection of Toxoplasma gondii : effects of storage conditions on sensitivity . Comparison of Cell Culture , Mouse Inoculation, and PCR for Detection of Toxoplasma gondii : Effects of Storage C. 34: 1572–1575. [Crossref]

Editorial Information

Editor-in-Chief

Nigel Silman
University of the West of England

Article Type

Perspective

Publication history

Received: May 30, 2016
Accepted: July 06, 2016
Published: July 09, 2016

Copyright

©2016 Vira H. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Citation

Hemant Vira, Vivek Bhat, Preeti Chavan (2016) Diagnostic molecular microbiology and its applications: Current and future perspectives. Clin Microbiol Infect Dis, 1: DOI: 10.15761/CMID.1000105

Corresponding author

Vivek Bhat

Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, India

E-mail : vivekbhat2005@yahoo.com

Table 1. Standardised molecular protocols in diagnostic bacteriology

Bacteria

Acceptable Specimen

Unacceptable Specimen

Limitations/Comments      

Primer Sequence/Probe and Method References

Bordetella pertussis

Nasopharyngeal aspirates, nasal swabs or throat swabs.Sputum, endotracheal tube aspirates,and BAL fluids are also occasionally processed

The potential for false B. pertussis detection due to B. Holmseii infection must also be kept in mind

-

[72-75]

Bordetella parapertussis

Nasopharyngeal aspirates, nasal swabs or throat swabs.Sputum, endotracheal tube aspirates, and BAL fluids are also occasionally processed

-

-

[72-75]

Brucellosis

Bacterial isolates, Blood (EDTA), Serum, Buffy coat, fresh tissue (liver, spleen,thyroid, bone, synovial tissue and fluid)

-

Assays were validated by testing historical culture confirmed B. suis and B. melitensis patient isolates as well as cattle derived B. abortus isolates

[76,77]

Burkholderia cepacia

Designed for the identification of suspected B. cepaciacomplex and B. gladioli cultures and has not yet been validated for the detection of these organisms in clinical samples.

-

Species specific assays are not currently available forall of the B. cepacia complex species (e.g. Burkholderia dolosa,

Burkholderia pyrrocinia)

[78-81]

Chlamydia

Acceptable specimens are urine, urethral swab, endocervical swab, dry swab (including throat, rectal and vaginal), tampon  cells collected in liquid based, cytology medium PreservCytTM (Hologic, Bedford, MA, USA) and SurePathTM (BD, Franklin Lakes, NJ, USA)

Not validated for other specimens

Most commercial assays, however, do not differentiate between LGV and other serovars of C. trachomatis

[82,83]

Chlamydophila

Respiratory material, including throat swabs, nose swabs, nasopharyngeal aspirates, bronchoalveolar lavage, whole blood (EDTA), urine and CSF, occasionally environmental/bird specimens

Wound swabs, drainage fluid, faeces

No international controls are available for C. psittaci due to restrictions on transport

of potential bioterrorism agents

[84-86]

Coxiellaburnetii

Blood, serum, bone marrow, biopsies

(Including those imbedded in paraffin) and cerebrospinal fluid.

-

-

[63]

Diarrheagenic Escherichia coli Pathotypes (DEP)

Including Enterohaemorrhagic (EHEC)/Shiga-toxin

E. coli (STEC)

Infectious colonies isolated after processing.

-

-

[87-90]

Haemophilus ducreyi

Impression smears or press slides, swabs, and biopsies only.

-

-

[91-93]

Klebsiella granulomatis.

------do-----------

-

Only K. granulomatis carries the restriction site specific to this organism

[92]

Haemophilus influenzae

– Capsulated and Non-typeable

H. influenza cultures only

Not  been validated for the detection of H. influenza in clinical samples

-

[94-96]

Mycobacterium tuberculosis Complex

Respiratory tract

Non-respiratory origin, the sensitivity and specificity of the assay has been estimated to be 69.8% and 95.6%, respectively.

Potential for environmental mycobacteria to contaminate samples

[94-96]

Mycobacteria Other Than Mycobacterium tuberculosis

Fresh tissue, cerebrospinal fluid, respiratory specimens, and body fluids, including uncoagulated blood in EDTA tubes

Formalin fixed and paraffin embedded tissue samples, clotted blood, serum, swabs,

urine and faeces

-

[97,98]

Mycoplasma genitalium

Urine and genital swabs

-

M. genitalium DNA may be detected in asymptomatic individuals

[99,100]

Mycoplasma pneumoniae

Sputum and Bronchoalveolar lavage (BAL) specimens, nasopharyngeal and throat swabs, nasopharyngeal aspirates, tracheal aspirates and pleural fluid specimens

Not been validated for any other specimen than those listed.

M. pneumonia may be detected for some time after acute infection following the resolution of clinical symptoms

[101-103]

Neisseria gonorrhoeae

Urine, urethral swab, endocervical swab, dry swab (including throat, rectal and vaginal)

Not been validated for any other specimen than those listed

PHLN guidelines for the use and interpretation of nucleic acid detection tests for Neisseria gonorrhoeae

[104-107]

Neisseria meningitidis

EDTA whole blood, CSF and aspirates from joints. Heparinised or citrated samples can be tested

Plasma and serum samples show a decreased recovery of N. meningitides DNA

The ctrAPCR assay is used as a diagnostic test for meningococcal disease

[108,109]

Pseudomonas  aeruginosa

P. aeruginosa isolates only

Used in sputum samples, but has not been validated

-

[110-113]

Rickettsia

Blood, biopsies, and cerebrospinal fluid

Clotted blood, serum, swabs and urine

-

[114]

Streptococcus pneumoniae

CSF and other sterile fluids.

Not appropriate to test respiratory specimens

ply gene is not specific for pneumococci and been detected in Streptococcus mitis and Streptococcus oralis isolates

[115]

Treponema pallidum

Swabs from skin lesions only

Not validated for other specimens

The gold standard for the detection of T. pallidum is probably rabbit inoculation

[116-118]

Table 2.  Standardised molecular protocols in diagnostic virology

Virus

Acceptable Specimen

Unacceptable Specimen

Limitations/Comments      

Primer Sequence/Probe and Protocol References

Adenovirus

Clinical specimens (throat or nasal swabs or faecal samples)

Not been validated for any other specimen than those listed

Most common technique is to passage Adenovirus in A549 cells before genetic analysis

[119-121]

Cytomegalovirus (CMV)

New born Screening card, fresh placenta, fresh umbilical cord, amniotic fluid, urine, serum, plasma, buffy coats, formalin-fixed, paraffin-embedded tissues

Not been validated for any other specimen than those listed

Generally two different CMV PCR are performed on each sample to minimise false positivity

[122-126]

Dengue Virus

Serum

Not been validated for any other specimen than those listed

‘Not detected’ should not be considered as absence of virus.

[127-131]

Enterovirus

Faeces, CSF, nasopharyngeal aspirates, cell culture supernatant, throat, vesicle and

rectal swabs

Blood and tissue samples

The specimen should be kept at 4 degree C

[132-135]

Epstein Barr Virus

CSF, Blood, Biopsies

Not been validated for any other specimen than those listed

Stored at -20 degree C

[136,137]

Flavivirus

CSF, CNS tissues and whole blood

Not been validated for any other specimen than those listed

West Nile virus assay also detects Kunjin virus.

[137]

Hepatitis A Virus

Whole blood, serum, and EDTA tubes

Heparin tubes as it is a potent inhibitor of PCR

Haemolysed samples are causes PCR inhibition

[138]

Hepatitis B Virus

-----------do-----------

-------do--------

---------------do------------------

[138]

Hepatitis C Virus

-----------do-----------

-------do---------

---------------do-------------------

[139]

Hepatitis D Virus

-----------do-----------

-------do---------

---------------do-------------------

[140]

Hepatitis E Virus

-----------do-----------

-------do---------

---------------do-------------------

[141]

Herpes Simplex Virus Type 1 and 2

 CSF, brain biopsy, FFPE

Not been validated for any other specimen than those listed

PCR detection has become a confirmatory test for HSV encephalitis

[142-147]

Human Bocavirus

Respiratory secretions, nose/throat swabs, stool samples, whole blood

Not been validated for any other specimen than those listed

-

[148]

Human Coronaviruses

Respiratory specimens (Nasopharyngeal aspirates, nose and throat swabs and bronchial samples)

Not been validated for any other specimen than those listed

-

[149-151]

Human Herpes Viruses 6, 7 and 8

EDTA blood, bone marrow and body fluids (amniotic fluid, CSF), biopsies, newborn screening cards

Not been validated for any other specimen than those listed

-

[122,124,125]

Human Papillomavirus

Vaginal swab, tampon, FFPE, cells collected in liquid based cytology medium PreservCytTM (Hologic, Massachusetts, USA) and SurePathTM (BD, New Jersey, USA)

Not been validated for any other specimen than those listed.

For keratinised tissues, specific methods should be followed

[152,153]

Human Polyomaviruses – JCV and BKV

EDTA blood, CSF, body fluids, urine, biopsies

Not been validated for any other specimen than those listed

JCV and BKV may occur at the same time in same patients

[93,154-158]

Human Polyomaviruses – KIV and WUV

Nasopharyngeal aspirates, bronchial washes and other respiratory secretions, nose/throat swabs, cerebrospinal fluid, urine, whole blood, and stool samples

Not been validated for any other specimen than those listed

-

[159-161]

Human Rhinoviruses

Nasopharyngeal aspirates, nose and

throat swabs, nasal or bronchial washings, broncho alveolar lavage or middle ear fluids

Not been validated for any other specimen than those listed

The assay may miss some viral strains

[162]

Influenza Virus A H5N1 (“Avian Influenza”)

Throat swab, nasal swab or nasal wash

Not been validated for any other specimen than those listed

Prior testing with current strains is mandatory

[130]

Influenza Virus A H1N1 (2009) (“Human Swine Influenza”)

Nasopharyngeal aspirates, bronchial washings, throat swab, nose swab

Not been validated for any other specimen than those listed

-

[113,163,164]

Influenza Type C

Respiratory secretions, bronchial lavage and washings, nose/throat swabs

Not been validated for any other specimen than those listed.

-

[165]

Measles Virus

EDTA blood, Urine, nasopharyngeal aspirates, throat and nasopharyngeal

swab

Not been validated for any other specimen than those listed

No cross-reactivity identified between human and viral DNA in the cited protocols

[166,167]

Norovirus

Faeces

Not been validated for any other specimen than those listed

[168-171]

Respiratory Syncytial Virus Types A and B

Respiratory swabs, sputa or respiratory tissues, and paranasal aspirates

Not been validated for any other specimen than those listed

The coat protein gene of MS2 RNA coliphage and A and B (RSVA and

RSVB)

[172]

Rotavirus

Faeces

Not been validated for any other specimen than those listed

RNA should be completely free from residual stool inhibitors

[173-177]

Varicella Zoster virus

CSF and CNS tissue

Not been validated for any other specimen than those listed

-

[178-180]

Table 3.  Standardised molecular protocols in diagnostic mycology and parasitology

Fungi/Parasites

Acceptable Specimen

Unacceptable Specimen

Limitations/Comments      

Primer Sequence/Probe and Method References

Aspergillus Species

BAL, EDTA whole blood, serum, fresh and paraffin embedded tissue, vitreous fluid, ascitic fluid and CSF

Sputum and plasma

Penicillium spp. can also be amplified by the assay

[181-184]

Cryptosporidium

Fresh or frozen stool samples

-

PCR has a high sensitivity

[185-196]

Cyclospora

Fresh or frozen stool samples

-

The cited assay has a detection limit of 0.5 oocyst

[189]

Dientamoeba fragilis

Fresh or frozen stool samples

-

The cited assay amplifies 18s rDNA

[197-199]

Entamoeba histolytica

Fresh or frozen stool samples

The cited assay amplifies small subunit (18S) rRNA gene

[200-211]

Giardia

Fresh or frozen stool samples

-

-

[212-216]

Malaria (P. falciparum, P. vivax, P. malariae, P. ovale)

Whole blood samples

-

-

[155,217-219]

Microsporidia

Fresh or frozen stool samples

-

-

[220-227]

Pneumocystis jirovecii

Sputum, induced sputum, bronchial washings, bronchial lavages and lung biopsies

-

The cited assay can be used only as qualitative assay

[228,229]

Trichomonas vaginalis

Genital swabs and urine

-

The cited assay targets  the β-tubulin and 18S rRNA genes

[230]

Toxoplasma gondii

CSF, CNS tissue, and FFPE

CSF supernatant, plasma, and serum

Negative results should be confirmed with serology

[231-233]