key: cord-0035140-m9fuinmu authors: L., G. title: Respiratory Syncytial Virus and Other Pediatric Respiratory Virus Infections date: 1998 journal: Diagnostic Virology Protocols DOI: 10.1385/0-89603-479-8:213 sha: c92ab6120f54c35092879ab5ea87b8e785a48a10 doc_id: 35140 cord_uid: m9fuinmu Infants and young children undergoing their primary infection with common human respiratory viruses are at risk of serious, even life-threatening, lower respiratory tract infection, A multiplicity of viruses infect the human respiratory tract but a relatively small number are responsible for the majority of significant illness. Of these the most commonly diagnosed in the pediatric population is respiratory syncytial virus (RSV), which infects essentially all children in their first or second year of life, bringing approx 1% into the hospital with bronchiolitis or pneumonia (1). Children with underlying cardiac or pulmonary disease, or born prematurely are particularly at risk. The virus also causes problems in the immunosuppressed and mortality rates are alarmingly high for RSV pneumonia after bone marrow transplantation (2). The availability of therapy with ribavirin (3) or high titer anti-RSV γ-globulin (4), which may be of benefit in these at risk groups, places a premium on rapid and accurate, but cost effective, diagnosis. 1. Introduction Infants and young children undergoing their primary infection with common human respiratory viruses are at risk of serious, even life-threatening, lower respiratory tract infection, A multiplicity of viruses infect the human respn-atory tract but a relatively small number are responsible for the majority of stgnificant illness. Of these the most commonly diagnosed in the pediatric population is respiratory syncytial virus (RSV), which infects essentially all children in their first or second year of life, bringing approx 1% into the hospital with bronchiolitis or pneumonia (I). Children with underlying cardiac or pulmonary disease, or born prematurely are particularly at risk. The virus also causes problems m the immunosuppressed and mortality rates are alarmingly high for RSV pneumonia after bone marrow transplantation (2) . The availability of therapy with ribavirin (3) or high titer anti-RSV y-globulm (4) , which may be of benefit in these at risk groups, places a premium on raptd and accurate, but cost effective, diagnosis. Significant lower respn-atory tract disease may also result from infection with the parainfluenza viruses, the influenza viruses, and adenoviruses. Measles virus, currently relatively rare in countries operating a successful vaccination regime, may also be regarded as an important respiratory pathogen. Influenza C virus, the coronaviruses, the rhmovnuses, the reovnuses, and the enterovn-uses, also commonly demonstrable in respiratory secretions, are generally considered less significant respiratory pathogens, mainly restricted to the upper respiratory tract. Nonetheless all may be associated, at least occasionally, wtth serious lower respiratory tract dtsease. There are a number of routes to the diagnoses of a respiratory virus infection, not all of which are suitable for routme pedtatrtc use. Serological diagnoses has proved unreliable m infants, the younger of whom still possess transplacentally acquired maternal antibodies that can interfere with or mask serologtcal responses (I). The detection of viral nucleic acid in nasopharyngeal secretions by polymerase chain reaction (PCR) offers great sensmvtty, but the precautions required to prevent false-postttve results, and the greater expense and time taken to achieve a dtagnosts make it currently unattractive for routme use (5) . Demonstratton of infectious virus by inoculation of secrettons mto cell cultures of high virus suscepttbthty, although necessarily labortous and slow, was for a long time the gold standard against which other techniques were measured. Although this approach can be speeded up by the tmmunofluorescence staining of early cultures with virus-specific monoclonal anttbodtes (MAbs), culture can never match the speed of direct anttgen detection m specimens using either immunofluorescence or enzyme tmmunoassay. Although m most studies vnus culture shows marginally greater sensitivity than the rapid techniques, occasionally antigen detection may succeed where vn-us culture fails, Whether antigen detection methods alone, without the backup of vn-us culture, are adequate to provide a reliable diagnosttc service remains a controverstal issue. Enzyme-linked unmunosorbent assay (ELBA) techniques are amenable to automation and attractive to laboratortes with a large throughput. Although ELISA techniques are often relatively slow and of poor sensitivity, some rapid and sensmve commercial kits are now available that should be considered where the cost can be justified (6) . Immunofluorescence staining of virus antigens m exfohated cells collected from nasopharyngeal secretions is the most widely used technique for the demonstration of respiratory viruses in pediatric populations. Where suitable antibodies, evaluated directly on clinical material, are used, sensitivity and specificity are comparable with any other technique, with the possible exception of PCR (574. In addition, the direct observation of the specimen provides useful visual feedback on specimen quality allowmg negative results to be reported with greater confidence Usmg direct unmunofluorescence wrth conjugated MAbs, results can be available within an hour with only minor sacrifice of sensmvity. Furthermore, suitable antibodies are available for all of the major and many of the mmor viral respiratory pathogens (Table 1 ) which can thus be tested for on a single multtwell slide offering great flexibility at reasonable cost. It 1s this technique, therefore, that will be detailed here. hT~o antigemcally dtstmct subgroups of RSV isolates, A and B can be differentiated with monoclonal anttbodtes. The clmical relevance of sub-grouping has yet to be established but a subgroup A-spectfic MAb 1s avatlable from Blosoft 'MAbs to paramfluenza vnus types 1,2, and 3 are generally avatlable but not to type 4 dA group-specific rhmovnus antigen has been described (9) but no monoclonal antibodies to this antigen are currently available+ Diagnosis 1s made by demonstrating the presence of virus-infected respiratory epithelial cells desquamated into the nasopharyngeal mucus of the infected child. Infected cells are sedimented from samples of mucus, fixed onto a glass slide, and stained either with virus specific antibodies conjugated to fluorescein (the direct technique) or unconjugated antibodies subsequently labeled with a fluorescein-conjugated secondary anti-immunoglobulin (the indnect technique). Fluorescing infected cells are then vtsuahzed under a UV mtcroscope. Immunofluorescence staining is also valuable m identifying viruses isolated in cell culture in tubes or shell vials, and may be carried out l-2 d postinoculation, which 1s often some time before cytopathic effect is evident (10) . The detailed descriptions of these techniques below are essentially those of Gardner and McQuillin (II), with minor modifications to take account of more recent, commercially available reagents. with carefully prepared polyclonal antisera, appropriately absorbed to remove nonspecific staining and extensively evaluated on positive and negative clinical material gives optimal sensitivity and specificity of viral antigen detection Such high quality reagents, however, are not generally available, and where they are interpretation of fluorescence staining requires an experienced eye. For the preparatton of such antisera the reader is referred to Gardner and McQmllm (II). MAbs, carefully selected for sensitivity and specificity on clinical material, are commercially available (Table 1 ) and offer an acceptable alternative m many situatrons (see Note 1) Dtrect tmmunofluorescence with fluorescein-labeled MAbs provides a simpler, more rapid, and easily interpretable test, but MAbs rarely offer the sensitivity achievable with polyclonal antrsera. The lack of sensitivity may be improved by using a pool of MAbs to different virus epitopes, preferably on different proteins, which may both increase the number of cells stained and improve the level and pattern of stammg within individual cells. 12. Fluorescence microscope. The fluorescence microscope must be equtpped with a lamp and an excitation and barrier filter system capable of lllummatmg the speci-men with light at close to 490 nm, the peak of absorptton by fluorescein, and of transmitting only apple green fluorescent light of wavelengths around 5 17 nm to the eyepiece (see Note 2) . The most suitable modern fluorescence mrcroscopes are equipped for incidence light fluorescence with high numerical aperture lowpower and high-power (x50 or x63) oil immersion objectives (see Note 3). Secretions collected from the nasopharynx of children with respiratory infections are the material of choice for the identification of virus as they contain a larger number of infected cells than nose and throat swabs and are more easily available than secretions from the lower respiratory tract. The latter, however, are preferable if available. Here, the simple aspiration of nasopharyogeal secrettons IS described although some laboratories instill a small volume of buffered saline mto the nose prior to aspiration (12) . Cough and nasal swabs are collected simultaneously as suctron for nasopharyngeal secretions is not always productive. Nasopharyngeal secrettons, cough, and nasal swabs ~111 generally be collected by ward staff who may require some training to produce material optimal for virus diagnosis (see Note 4). 1. Swab one of the child's nostrils and break the swab into 4 mL of transport medium held on melting ice 2. Swab the back of the throat until the patient gags and coughs onto the swab Break this swab into the same bottle of transport medium which 1s held in melting ice 3. Attach a sterile polythene feeding tube to the inlet of a sterile plastic mucus extractor. Attach the mucus extractor to a portable vacuum pump which has a maximum suction pressure of -26psi (239 kPa) 4. Switch on the pump and msert the feeding tube into the child's nasopharynx via each nostril in turn. Some children may show signs of distress, partrcularly if the nose is dry and secretions sparse and viscous. Secretions collect m the mucus extractor, although if few are present in the child's nose, they may lodge in the feeding tube and will require washing through with a little sterrle PBS 5. Place the mucus extractor and the cough and nasal swabs in a vacuum flask on melting ice and transport to the laboratory with mmrmum delay. 1 Repeatedly ptpet the medium over the swabs to remove adherent secretions. Remove the fluid containing the secretions to a centrifuge tube and centrifuge at 380g for 10 min at 4°C to pellet the cells. Remove the supernatant, which may be used for vmts tsolation. 2 Resuspend the cells in 3 mL of PBS by gentle pipeting and recentrifuge as above to obtain a cell pellet. 3 Centrifuge the mucus extractor containing nasopharyngeal secretions at 3808 for 10 min at 4'C to collect all the secretton at the base of the vessel A small aliquot may be removed for vnus isolation Add 2-3 mL of PBS to the remainder and disperse the mucus by gentle pipetmg with a wide-bore pipet. Add further aliquots of PBS to about 10 mL, pipeting after each aliquot until the majority of the mucus is broken up Transfer the suspension, leaving any remaining mucus lumps, to a centrifuge tube and pellet the cells at 380g for 10 min at room temperature 1. Rinse Teflon-coated glass multiwell slides in absolute alcohol and wipe dry with a Imt-free cloth. Engrave identificatron details on the free end and wipe again wtth a watermoistened cloth. 2. Place sufficient cell suspension on each well to cover the glass surface (15-35 pL depending on the size of the well). Larger wells, allowmg more cells to be examined, will give an increase in sensitivity. 3 Allow to an dry (a hairdryer on a cold setting or a fan may be used to speed drymg). 4 Immerse slides m cold acetone (4°C) for 10 mm (see Note 7) 5 Dry at room temperature and stam mrmedtately or store at or below 40°C indefimtely. (Samples stored next to the door can be repeatedly freeze/thawed on openmg and closing ) 3. Incubate for 30 mm at 37°C m a moist box. 4 Gently rmse anttbody from each slide with PBS applied via a Pasteur pipet 5. Immerse the shde m PBS m a Coplm jar or stammg trough and soak for 10 mm (see Note 10). 6 Discard and replemsh the PBS and soak for a further 10 mm Repeat, for three 1 0-min soaks total 7. Dram the slides and au dry. 8. Staining ofthe tmmunoglobulin-coated cells with the appropriate FITC-conjugated antiglobulin is carried out immediately as described below (Subheading 3.5.) for direct stammg of cells. Thrs procedure may be applied to fixed ceils already reacted with unconjugated antibodies by the indirect techntque descnbed in Subheading 3.4. using the approprtate species-specrfic FITC-conjugated anti-y-globulin. Alternatively it may be used wtth FITC-conjugated antiviral monoclonal or polyclonal anttbodtes for single-step visualization of virus-infected cells. Suitable control reagents must be included u-r each test to ensure the spectficrty of the fluorescent antrbody staining observed (see Note 11). 1. Dilute the relevant FITC-conlugated antibody to the recommended working dtlunon in Evan's blue counterstain (see Note 9) The dtluted conjugate may be kept at 4°C but only so long as tt remains sterile. Long-term storage of diluted reagent is not recommended 2. Carefully spread one drop of FITC-conjugated antibody over each cell preparation. 3 Incubate for 30 mm (see Note 12) at 37'C m a moist box 4 Rinse as in steps 4-7 of Subheading 3.4. 5 Immerse slides in dtsttlled water and soak for l-2 mm to remove PBS, which will crystallize rf allowed to dry on the slide 6 Allow to dry in air and either immediately examine mmroscopmally rmmedrately or store at 4°C in the dark m a closed container for examination the next day. Quality of stammg deteriorates on storing of unmounted preparations, particularly if rmmersron 011 has been applied Mounting in Fluokeep (Biosoft; TCS, Buckmgham, UK) under a coverslrp not only increases the intensity of fluorescence but protects the cells from the deleterious effects of immersion oil 4. Notes 1, A vast range of MAbs to viral antigens are available commerctally and through other channels, but very few of these are likely to be suitable for diagnostic purposes. Some currently available MAbs or MAb pools designed for antigen detection in clinical material are listed m Table 1 . Also noteworthy are the polyvalent pools of antibodies to RSV, the mfluenzavtruses, the paramfluenza vnuses, and adenoviruses produced by some manufacturers. These allow one-test screenmg Toms for significant respiratory virus pathogens, reducing the costly screening of negative samples against multiple type specific reagents. 2. For more information prior to purchasing consult E 0. Caul (13) 3. Not all objectives are suitable for fluorescence microscopy owing to autofluorescence of some components or excesstve internal light scattering. Manufacturers should be able to give advice on those most suitable. 4 Adequate specimens must contam a reasonable number of respiratory epithelial cells derived from the nasopharyngeal eptthelium Squamous epithelial cells, derived from the anterior an passages, are not generally mfected 5. It is advisable to carry out the handling of material likely to generate airborne pathogens in a class 2 containment cabinet. 6 Cell cultures exhibttmg extensive cytopathtc effect may detach on washing In this case, the cells should be scraped and pipeted directly into the culture medium. Cells are pelleted at 380g for 5 mm at room temperature, resuspended to original volume in PBS to wash, and recentrtfuged to obtamed a washed cell pellet. A similar procedure is adopted for vu-us-Infected cultures of nonadherent cells 7 Acetone should not be stored m conventtonal refrigerators as it poses an explosion hazard A Coplin jar of acetone in an ice bath is convenient for fixing a small number of slides. For cell culture preparations, which are free from mucus, tixanon may be reduced to 5 mm. 8. Specificity of reagents cannot be taken for granted, particularly when climcal material is being investigated and adequate controls must be included to reveal non-specific reacttons. The followmg should be Included for the indirect test a A cell preparation stmtlar to the test specimen but known to be negative for the virus m question, stained in parallel with the test specimen Once sure of all the reagents and methods and familiar with the patterns of staining they produce, experienced workers may choose to omit this control. b. A duplicate preparation of the test specimen stained with a negative antibody as similar as possible to the virus-specific antibody m use, followed by the FlTC-conjugated anttglobulin. Where a polyclonal antiserum from an animal 1s employed, the ideal control would be the preimmune serum from the same animal. This IS rarely available for commercial products and here a serum from an animal immumzed by a similar protocol with an antigen likely to be absent from the specimen (e g., another virus) must suffice-where MAbs are employed a MAb of the same rmmunoglobulin class but specific for an irrelevant antigen known to be absent from the specimen should be used c. A duplicate preparation of the test specimen stained with PBS, Instead of the vu-us specific antibody, followed by the FITC-conjugated antiglobulin. 9 Although the maker's mstructions or advice from other laboratories may be taken as a guide, optimal working dilutions vary widely from laboratory to laboratory (14) . It is always advisable to titrate new antibody preparations in your laboratory. Normally, testing preparattons fivefold either side of the recommended dtlutton on known positive control material will suffice. 10 . For cell culture preparations, in which nonspecific stainmg is less troublesome, 5-min soaks will suffice. 11. In the direct test the controls should Include: a. A cell preparation similar to the test specimen but known to be negative for the virus m question and stamed m parallel with the test specimen. b. A duplicate preparation of the test specrmen stained with a negative control comugated antibody as similar as possible to the virus-spectfic comugated antibody to be employed. Where a polyclonal antibody from an animal has been purified and comugated, the most suitable control would be the preimmune serum from the same animal similarly purltied and conjugated. Where such is not available, a similar conjugated anttserum from the same species but specific for an antigen known to be absent from the test specimen will suffice. Where conjugated MAbs are employed, a conjugated MAb of the same immunoglobulin class but specific for an antigen known to be absent from the test specimen should be used. The specificity of fluorescent staining can be confirmed m a subsequent blocking test. Here, preparations of the test specrmen are preincubated with either unconJugated virus-specrfic antrbody or UnconJugated negative-control antibody. On subsequent staining with ConJugated antibody, specific fluorescent staining will be blocked by the latter but not the former 12. In the direct immunofluorescence test this incubation period may be reduced to 15 min for many conjugated MAbs in line with manufacturer's instructions. Respiratory syncytial virus Respiratory syncytial virus pneumonia in hospitalized adult patients with leukemia Ribavirin: a clinical overview The role of RSV neutralizing antibodies m the treatment and prevention of respiratory syncytial virus infection m high-risk chtldren Detection of respiratory syncytial vu-us by reverse transcription PCR and hybridization with a DNA enzyme-immunoassay Evaluation of Abbott Testpack RSV for the diagnosis of resptratory syncytial wrus infections Evaluation of five methods for respiratory syncyttal wrus detection A comparison of commercially available monoclonal-antibodies for dtrect and indirect immunofluorescence culture confirmation and direct detection of paramfluenza vnuses Diag Rapid culture amphfied immunofluorescent test for the detection of human rhmovtruses in clmical samples: evidence of a common epitope m culture Isolation of 7 respiratory vtruses m shell veals-a practical and highly sensttive method Diagnosing respiratory syncttal virus by nasal lavage Flourescence mtcroscopy Use of monoclonal antibodies for rapid dtagnosts of respiratory viruses-memorandum from a WHO meeting Acknowledgments I wish to thank Fiona Fenwick, Rosemary McGuckin, and Professor Dick Madeley for assistance in preparing this manuscript and for constructive criticism.