U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

Institute of Medicine (US) Committee on the Assessment of Asthma and Indoor Air. Clearing the Air: Asthma and Indoor Air Exposures. Washington (DC): National Academies Press (US); 2000.

Cover of Clearing the Air

Clearing the Air: Asthma and Indoor Air Exposures.

Show details

5INDOOR BIOLOGIC EXPOSURES

The allergic constituents of indoor air are predominantly biologic in origin (Becher, 1996). As early as the sixteenth century, associations between a number of these exposures and asthma were suspected; however, the scientific data available were unable to confirm such an association. Concern in recent years regarding the potential health effects of indoor air, as well as the marked increase in the prevalence of asthma in industrialized countries, has prompted an influx of scientific data on exposure to airborne biologic agents and asthma.

The committee was charged with the task of evaluating the strength of the scientific evidence concerning the possible association between these agents and asthma prevalence and severity. The committee was also tasked with examining possible means of mitigating or preventing exposure to these agents. In this chapter the committee evaluates indoor exposure to biologic agents, addressing the following to the extent permitted by available research:

1.

which factors influence exposures to the agent;

2.

whether a relationship exists between the agent and asthma prevalence or severity, taking into account the strength of the scientific evidence and the appropriateness of the methods used to detect the relationship;

3.

what type of relationship exists between the agent and asthma;

4.

whether there are special considerations regarding the agent (e.g., subpopulations at risk and interactions with other exposures);

5.

which strategies effectively mitigate or prevent exposure to the agent;

6.

whether these strategies only reduce exposures or decrease the occurrence or exacerbation of asthma; and

7.

whether these strategies are reasonable for use by the target populations.

Each section begins by providing a definition of the agent and a summary of the factors that influence exposure. The evidence concerning the possible association between the agent and asthma is discussed, followed by the committee's conclusions regarding the health impacts. Where information is available, evidence regarding possible means of mitigating or preventing exposure to the agent is addressed. Each section concludes with any committee recommendations for areas for which additional research is needed with respect to the agent. Because there are great differences in the amount and type of information available on specific agents, the sections vary in their depth and focus.

ANIMALS

Cats

Definition of the Agent and Means of Exposure

Cats are kept as pets in 27% of U.S. households. The major cat allergen, Fel d I, is a glycoprotein structured as a heterodimer with two chains of amino acids, which have been defined by polymerase chain reaction (PCR) and subsequent DNA sequencing (Griffith et al., 1992; Morgenstern et al., 1991; Schou, 1993). It is found on cat hair and is produced in cat sebaceous, salivary, and anal glands (De Andrade et al., 1996). In male cats, Fel d I glandular production is under hormonal control and decreases after castration (Zielonka et al., 1994). As discussed in the Third International Workshop report (Platts-Mills et al., 1997), the clinical significance of this decrease in allergen production is not certain, since many symptomatic cat-allergic asthmatics in the United States have neutered cats. Further investigation of hormonal and genetic control of Fel d I production could be relevant to the control of allergen levels in homes with cats. Although 90% of patients allergic to cats make immunoglobulin E (IgE) to Fel d I (de Groot et al., 1988; Schou, 1993), making Fel d I a marker for the immune response to cat allergens, at least eight other cat allergens have been identified (Duffort et al., 1987), suggesting that protection from Fel d I exposure may not be the equivalent of protection from cat allergen exposure. This conclusion is supported by the findings that 66% of the histamine-releasing activity of cat hair and dander extract, and about 60% of the cat dander radioallergosorbent test (RAST) activity, was carried by Fel d I (Schou, 1993).

Touching the cat is only one mode of contact that may result in airborne suspension of allergen and potential direct hand-to-nose deposition of allergen-associated particles. In contrast with cockroach allergen, which is airborne only transiently during the disturbance of household dust, cat allergen can remain airborne for long periods of time, in part because Fel d I is associated to a significant extent with smaller particles of less than 5 µm (Custovic et al., 1998b). Particles on which cat allergen is carried, coming primarily from cat dander, are also very adherent. Consequently cat allergen is spread easily throughout a house, even when cats are kept out of certain rooms. Moreover, cat allergen is easily carried from home to home, office, school, or day care center by those who touch cats or visit households with cats (Custovic et al., 1998a; Dybendal and Elsayed, 1994; Warner, 1992). At trace or small amounts that may be significant for sensitization or exacerbation of disease in sensitized individuals, Fel d I in settled dust is found in most homes without cats (Bollinger et al., 1996; Chew et al., 1998), although allergen levels are generally higher in homes with cats. A Baltimore, Maryland, study found measurable levels of airborne Fel d I in 37 homes with cats (range, 1.8–578 ng/m3; median, 45.9 ng/m3) and in 10 of 40 homes without cats (range for detectable samples, 2.8–88.5 ng/m3; median, 17 ng/m3) (Bollinger et al., 1996). In 38 of 40 homes without cats, Fel d I was present in the settled dust (range, 39–3,750 ng/g; median, 258 ng/g); dust levels were weakly correlated with airborne levels. Carpeting, bedding, and upholstered furniture can be reservoirs for deposited cat allergen (Wood et al., 1989); shaking the bedding in rooms with cats resuspends cat allergen in the air.

As well as being detected in homes without cats, cat allergen has also been detected in public places such as hospitals and schools. In one British study that measured both settled and airborne cat allergen in hospitals, the amount of cat allergen in settled dust in upholstered chairs was as high as in homes with cats (geometric mean, 23 µg/g dust), but airborne levels were low (0.22 ng/m3) (Custovic et al., 1998a).

Evidence Regarding Asthma Exacerbation

In cat-sensitized asthmatics, cat allergen can induce allergic symptoms, asthmatic symptoms, and decrements in lung function. Exposure to inhaled cat allergen in an experimental cat room led to significant decreases in forced expiratory volume in one second (FEV1, range, 6–57%; mean, 25%) in a study of 13 adults with cat allergy. The percentage decrease in FEV1 did not correlate significantly with either the intradermal titration end point with cat allergen or the magnitude of the RAST response with cat allergen. Those cat-allergic subjects classified as asthmatic by methacholine challenge testing experienced almost identical responses to environmental allergen challenge in an experimental cat room and inhalation challenge with cat allergen (Sicherer et al., 1997). Norman and colleagues documented progressive increases in both nasal and lung symptom scores during a 60-minute period in a cat room (Norman et al., 1996).

While initial cat room studies involved very high airborne cat allergen levels, a later experimental cat room exposure study by Bollinger and colleagues (1996) evaluated symptom and lung function responses of cat-sensitive subjects to low-level airborne cat allergens. They demonstrated that cat-sensitive individuals can have increases in upper- (congestion, rhinorrhea, pruritus) and lower- (chest tightness, wheezing) respiratory symptoms and decrements in lung function at levels of cat allergen occurring in homes without cats. The median FEV1 change was 15% in the seven challenges with a Fel d I level less than 100 ng/m3.

In a Delaware case-control study, Gelber and colleagues (1993) studied allergen predictors of emergency room visits for asthma. This study compared 93 patients, 15 to 55 years of age, who presented with breathlessness and airway obstruction, to 93 patients presenting without breathlessness. For cat and cockroach, the combination of sensitization and the presence of allergen in the house was associated with asthma presenting to hospitals (14/93 asthmatics versus 1/93 controls). Whether other exposures potentiate the response of cat-allergic asthmatics to cat exposure is unknown. In a cross-sectional study of children in New Mexico with asthma or bronchial hyperresponsiveness, cat sensitization and exposure to cat allergen were common (Sporik et al., 1995). Among children with asthma (defined as symptomatic bronchial reactivity), 13/19 were sensitized to cat. Numbers were too small to compare symptoms in cat-sensitized asthmatics with and without significant home exposure to cat allergen (Ingram et al., 1995).

Evidence Regarding Asthma Development

Insufficient data are available to assess whether exposure to cats influences the development of asthma. Cross-sectional studies of children suggest an association between sensitization to cats and home exposure in the first six months of life (Suoniemi et al., 1981; Warner et al., 1991). These studies may be subject to recall bias. A longitudinal birth cohort study from the Isle of Wight found that the presence of a cat in the home predicted a greater risk of skin test reactivity to cat and a greater risk of any skin test reactivity by 2 years of age (Hide et al., 1994). Sensitization to cat predicts asthma development, but this may simply be a confirmation of the well-documented fact that atopic individuals are more likely to develop asthma than nonatopic individuals. In a New Zealand birth cohort study, the development of asthma by 13 years of age was associated with sensitization to cats and dogs at age 13 (Sears et al., 1989). Sensitization to cats predicted the development of bronchial hyperresponsiveness in a longitudinal study of adults in Boston, Massachusetts (Litonjua et al., 1997). However, neither of these studies provides evidence of whether exposure to cats predicts either asthma or bronchial hyperresponsiveness. Since cat allergen exposure can potentially take place either at home or in schools and public places, the relative importance of home versus community-wide exposure to cat allergen in the risk of specific sensitization to cats is unknown.

Although the field of the genetics of asthma is in its infancy, preliminary studies suggest that certain genetic phenotypes are associated with allergy to specific insects or animals (Fukuda et al., 1995; Hizawa et al., 1998; Young et al., 1992, 1993). Understanding the genetics of allergy and asthma, including understanding the phenotypes associated with allergy specificity, may eventually prove useful in understanding gene-by-environment interaction in the development of asthma.

Conclusions: Asthma Exacerbation and Development

In cat-sensitive asthmatics, cat allergen exposure leads to worsening of respiratory symptoms and to a decline in lung function. Although sensitization to cats is a prerequisite to reactivity to cat exposure, the level of airborne cat allergen that exacerbates asthma varies by individual and is not necessarily predictable by the size of the skin test reaction to cat or the titer of IgE antibody. However, specific sensitive subgroups have not been defined. The relationship between cat allergen in the home and asthma development is uncertain. Because cat allergen is frequently found outside the home and in households without cats, the assessment of individual exposures to cats is difficult, making evaluation of the association between cat allergen exposure and asthma development difficult as well. In summary:

  • There is sufficient evidence of a causal relationship between cat allergen exposure and exacerbation of asthma in individuals specifically sensitized to cats.
  • There is inadequate or insufficient evidence to determine whether or not an association exists between cat allergen exposure and the development of asthma.

Evidence Regarding Exposure Mitigation and Prevention: Homes

Removal of the cat from the home will decrease exposure to cat allergen and is widely recommended for symptomatic cat-sensitive asthmatics. However even when the owner removes the cat, cat allergen levels may remain elevated for 20 weeks or more (Wood et al., 1989). Removal of carpets and upholstery, with encasement of mattresses and pillows, may be required for diminishing cat allergen to levels commonly measured in homes without cats (Wood et al., 1989).

No studies are available that evaluate symptoms or lung function in cat-sensitive asthmatics before and after removal of the cat from the home. Nor are there studies of change in symptoms or lung function after moving from a home with a cat to a home without a cat. However, exposure studies suggest that in cat-sensitive subjects the decline in lung function associated with low-level airborne exposure to cat allergen, which can be present in a home with no cats, tends to be less extreme than the decline in lung function associated with higher-level airborne exposure (Bollinger et al., 1996). The experiments demonstrating entry into a room with a cat as a source of exacerbation of asthma in catsensitive individuals also suggest that removal of the cat from the household may decrease symptoms in cat-allergic asthmatic.

Because of the reluctance of cat-allergic symptomatic asthmatics to get rid of their cats, a number of studies have focused on the potential for lowering cat allergen levels by washing the cat. In eight households with cats, de Blay and colleagues (1991a) found that the combination of washing the cat weekly, reducing furnishings, vacuum cleaning, and air filtration reduced airborne cat allergen levels. In a second study, however, Avner and colleagues (1997) from the Platts-Mills group found that while washing cats by immersion will transiently remove significant allergen from the cat and reduce the quantity of airborne Fel d I, this reduction in allergen is not maintained by one week. Klucka and colleagues (1995) found no significant reduction of Fel d I by washing, use of Allerpet-C (a widely advertised topical spray), or acepromazine, a tranquilizer advocated as efficacious in subsedating doses. While removal of the cat from the living room and bedroom areas of the home and use of a High-Efficiency Particulate Air (HEPA) filter reduced airborne levels of cat allergen in homes with cats, the reduction was not evenly spread across the particle size range (Custovic et al., 1998b). It is unclear that the level of reduction of allergen obtained in this study is sufficient to influence symptoms in cat-sensitized asthmatics. No studies are available to assess the efficacy of recommendations to wash cats in reducing symptoms in cat allergic-asthmatics. Although the combination of HEPA filter use, mattress and pillow covers, and exclusion of cats from the bedroom reduced airborne cat allergen levels, a Maryland study detected no improvement in daily symptom scores, peak flow rates, medication use, monthly spirometry, pre-and post-study cat-specific IgE levels, and methacholine challenge studies in cat-allergic subjects (Wood et al., 1998). On the other hand, in a double-blind, placebo-controlled, cross-over study of twenty asthmatic children sensitized to cat or dog allergens, and living in homes with these animals, airway hyperresponsiveness was improved and peak flow variation was decreased during the use of air cleaners in the living room and bedroom of the child (van der Heide et al., 1999). The authors report that substantial amounts of cat and dog allergen were captured by the air cleaners; floor cat and dog allergen levels were unchanged by air cleaner use.

Evidence Regarding Exposure Mitigation and Prevention: Schools and Hospitals

Even if the cat is removed from the home, continued low-grade exposures may occur in public places or via clothes from cat owners. Since cat allergen is everywhere, there is little potential for absolute avoidance (Dybendal and Elsayed, 1994; Warner, 1992). Norwegian investigators have demonstrated the presence of Fel d 1 in schools on both smooth and carpeted floors, with approximately 11 times more allergen on the carpeted floors (Dybendal et al., 1991, 1989a, 1989b). The frequency of cleaning floors and furniture was believed to influence the level of cat and dog allergen, which were higher on chairs than in floor dust (Warner, 1992). Upholstered chairs and mattresses in hospitals are also demonstrated reservoirs for cat and dog allergen (Custovic et al., 1998a).

To lessen the risk of exacerbation of asthma in cat- or dog-sensitized asthmatics in public buildings, Warner and others (1992) have recommended the use of smooth floors and frequently cleaned wooden or plastic chairs. In their study demonstrating the presence of significant cat and dog allergen levels in a hospital in Manchester, England, Custovic and colleagues (1998a) questioned the introduction of soft furnishings and carpets into hospitals where highly cat- or dog-allergic asthmatics may come for care. Where upholstered chairs were present, they demonstrated that vacuuming three times a week significantly reduced allergen levels.

Conclusions: Exposure Mitigation and Prevention

Cat allergen levels can be reduced to levels found in homes without cats by removal of the cat from the home, but the reduction in allergen levels may require a prolonged period of time. The combination of HEPA filter use, mattress and pillow covers, and exclusion of cats from the bedroom may not reduce airborne cat allergen levels sufficiently to improve symptoms in cat-sensitive asthmatics. The absence of carpet, the use of plastic or wooden rather than upholstered chairs, and of frequent vacuuming in schools and hospitals may decrease the levels of cat allergen in public places. No studies are available to evaluate whether these measures improve symptoms or lung function in cat-sensitized asthmatics or whether they decrease the potential for sensitization in nonsensitized individuals. In summary:

  • There is sufficient evidence of an association between removal of a cat from the home and a decrease in levels of cat allergen in the home; this decrease in levels of allergen may be slow if reservoirs of cat allergen are not simultaneously removed from the home.
  • There is limited or suggestive evidence of an association between removal of a cat from the home and improvement of symptoms or lung function in cat-allergic asthmatics.
  • There is limited or suggestive evidence of an association between measures short of removal of a cat from the home (e.g., washing the cat, HEPA filter use) and some transient reduction in cat allergen levels in the home.
  • There is inadequate or insufficient evidence to determine whether or not an association exists between measures short of removal of a cat from the home (e.g., washing the cat, HEPA filter use) and improvement in symptoms in cat-allergic asthmatics.

Dogs

Definition of the Agent and Means of Exposure

Dogs are present in 31% of U.S. households and are also sources of allergens (Schou, 1993). Allergy to cats is reported to be about twice as common as allergy to dogs, despite the fact that dogs are as common in U.S. households as cats (Bollinger et al., 1996). Can f I and Can f II are purified dog allergens that have been identified (Schou, 1993). Can f I is a polypeptide whose molecular weight and structure have been partially but not fully defined (Schou, 1993). It is present in dander, pelt, hair, and saliva, but not in the urine or feces of dogs (Schou, 1993). Though there are likely to be other dog allergens, no others have been found to have clinical importance. Can f I is considered a major allergen, because it accounts for at least half of the allergenic activity in dog hair and dander. In addition, 92% of dog-allergic patients had a positive skin prick test to Can f I (Schou, 1993; Yman et al., 1973). It is still a controversial matter whether true breed-specific dog allergens exist or whether the differences observed between breeds are quantitative rather than qualitative (Schou, 1993). Hair is not the only source of dog allergen, and it is not known whether short-haired dogs are less allergenic. Cross-reactivity can be found between dog and cat allergen (Vanto and Koivikko, 1983).

Dog allergen, like cat allergen and unlike cockroach, is easily aerosolized and widely disseminated throughout the community (Custovic et al., 1997). In a Baltimore study of 42 homes, dog antigen was demonstrated in more than half of households (Lind et al., 1987; Schou, 1993). In Sweden, dog allergen has been measured in homes that have never had dogs (Munir et al., 1992). Like cat allergen, dog allergen has been found in significant amounts in public buildings such as schools (Berge et al., 1998; Dybendal et al., 1989a; Schou, 1993; Warner, 1992) and hospitals (Custovic et al., 1998a). In dust from upholstered English hospital chairs, Can f I levels (geometric mean = 22 µg/g, range, 4–63) were as high as levels in settled dust from households with dogs (Custovic et al., 1998a; Munir et al., 1994). Hospital airborne Can f I levels were detectable in 7 of 10 testing days but were lower (range 0.09–0.22 ng/m3) than those often found in homes with dogs (range 0–100 ng/m3 Can f I) (Custovic et al., 1998a; Hodson et al., 1999).

Evidence Regarding Asthma Exacerbation

The asthmatic response to bronchial provocation test (PT) with dog allergen was evaluated in a cross-sectional Finnish study of 203 asthmatic children selected from the Children's Asthma Registry (Vanto and Koivikko, 1983). Of those with a positive PT, 64% had kept dogs, whereas only 36% with a negative PT had kept dogs. A positive PT was correlated with a positive skin prick test to dog (correlation coefficient = 0.8), but not with the frequency of reported asthma symptoms. In immunotherapy trials, positive response to bronchial provocation with dog allergen has also been associated with elevated levels of IgE to dog allergen in asthmatic subjects (Hedlin et al., 1995; Valovirta et al., 1984; Vanto et al., 1980). Some investigators consider symptomatic and bronchial response to animal allergen in an experimental animal room to be more definitive proof that animal allergen triggers asthma than allergen bronchial PT. They question whether the airway response to bronchial provocation with an allergen is always an allergic rather than an irritant response. The committee could find no published studies of the response of dog-sensitized asthmatics to exposure to dogs in an experimental dog room analogous to the cat room set up by the Hopkins group (Sicherer et al., 1997).

Evidence Regarding Asthma Development

There is insufficient evidence regarding the role of dog allergen in the development of asthma. In keeping with the ecology of Los Alamos, New Mexico, which is high and dry, with less dust mite, the allergens to which asthmatics are sensitized tend to be the predominant allergens in the indoor and outdoor environment. These allergens include dog and Alternaria, as well as cat (Ingram et al., 1995; Sporik et al., 1995). A cross-sectional retrospective Finnish study suggested that dog allergy was more prevalent in children from homes where dogs were present in the first few months of life, compared to homes where the dog was introduced after the child had reached the age of 1 (Vanto and Koivikko, 1983). As mentioned above, in a New Zealand birth cohort study the development of asthma by 13 years of age was associated with sensitization to dogs at age 13 (Sears et al., 1989). In contrast, the presence of a dog in the home in childhood was negatively associated with asthma (OR = 0.85, 95% CI = 0.78–0.92) among adults reporting no parental allergy, in the cross-sectional European Community Respiratory Health Survey of 13,932 20- to 44-year-old subjects from 36 areas in Europe, New Zealand, Australia, and the United States (Svanes et al., 1999). Among 1,649 Swedish school children aged 7–13 years, a report of keeping a cat or a dog in the first year of life was also negatively associated with asthma and allergic rhinitis (Hesselmar et al., 1999).

Conclusions: Asthma Exacerbation and Development

  • There is sufficient evidence of an association between dog allergen exposure and exacerbation of asthma in individuals specifically sensitized to dogs.
  • There is inadequate or insufficient evidence to determine whether or not an association exists between dog allergen exposure and development of asthma.

Evidence Regarding Exposure Mitigation and Prevention

Because the aerodynamic properties, carrier material, and chemical composition of dog and cat allergens are similar, issues related to mitigation of dog allergen are likely to be similar. However fewer studies are available related to the mitigation of dog allergen exposure and its health consequences. In a cross-sectional study of 203 asthmatic children listed in the Finnish Asthma Register, 68 of 203 had kept a dog, but 59 of 68 (87%) had removed the dogs from their homes. Parents tended to report that removal of the dog had improved asthma symptoms and had not been detrimental because of emotional deprivation (Vanto and Koivikko, 1983). The Finnish study also demonstrated that homes with dogs had higher dog antigen levels in dust than homes without dogs, some of which had a past history of a dog in the home. Homes in which occupants had indirect contact with dogs had more dog allergen than homes in which no one reported contact with dogs. Despite reported avoidance of dogs, a rising or steadily high level of dog-specific IgE was observed in follow-up of 24 dog-allergic subjects. The authors hypothesized that dog allergen encountered outside the home might be sufficient to boost IgE synthesis in most sensitive subjects (Vanto and Koivikko, 1983).

In a study of 25 homes with dogs, Custovic found that dogs had to be washed at least twice a week to maintain a reduction in recoverable Can f I from the hair (Hodson et al., 1999). Airborne dog allergen levels were not significantly affected by washing the dog. No studies are available regarding the effect of this mitigation measure on symptoms or lung function in dog-sensitive asthmatics.

Conclusions: Exposure Mitigation and Prevention

  • There is limited or suggestive evidence of an association between removal of the dog from the home and reduction of dog allergen levels. This evidence comes from an association between the absence of a dog in the home and the measurement of low dog allergen levels in a study including homes that had a history of keeping dogs (Vanto and Koivikko, 1983).
  • There is inadequate or insufficient evidence to determine whether or not an association exists between removal of a dog from the home and improvement in symptoms or lung function in dog-sensitized asthmatics. The one epidemiologic study reporting an association between dog removal and symptom improvement in asthmatic children relies on retrospective parental reporting without measures of sensitization at the time of dog removal or measures of symptoms or lung function before and after dog removal (Vanto and Koivikko, 1983).

Rodents

Definition of the Agent and Means of Exposure

Exposure to rodents can come either from keeping pets or from their presence as pests in the home. Rodents (mouse, rat, and guinea pig) can also be found in school settings. They have been studied as sources of allergens, particularly because of their extensive use as laboratory animals (Schou, 1993). Hair and epithelial fragments carry allergenic molecules, the allergens measured are believed to be from urine, saliva, or skin. The relative importance of these allergen sources has been debated (Karn, 1994; Longbottom and Austwick, 1987; Walls and Longbottom, 1985). As described by Schou (1993), rodents have permanent proteinuria; allergenic protein from sprayed urine dries up and becomes airborne on dust particles. Airborne rodent allergen has been measured in laboratory facilities (Sakaguchi et al., 1990a; Swanson et al., 1985; Twiggs et al., 1982) and, in one study, in inner city apartments (Swanson et al., 1985). Clinically important allergens that have been identified include Mus m I and Mus m II for mouse, Rat n I for rat, and Cav p I and II for guinea pig (Schou, 1993). Assessment of rodent exposure in the home has been limited, to some extent, by limitations in the ability to measure allergens from all species of wild mice potentially present in the home.

Evidence Regarding Asthma Exacerbation

A number of cross-sectional studies document the association between handling animals in a laboratory setting and allergy (Beeson et al., 1983; Cockcroft et al., 1981; Cullinan et al., 1994; Davies and McArdle, 1981; Gross, 1980; Newman-Taylor, 1982; Schumacher et al., 1981; Venables et al., 1988). Hollander and colleagues (1996) conducted a prospective panel study of self-reported symptoms and peak flow in Dutch laboratory animal workers. Workers who reported asthmatic symptoms (chest tightness) due to working with rats had significant decreases in peak expiratory flow on days they worked with the animals; 86% of them were sensitized to rat allergens (Hollander et al., 1996). A cross-sectional study of British laboratory workers demonstrated an association between positive skin tests to animal extracts and asthmatic symptoms (Cockcroft et al., 1981). Whereas IgE antibody to rat allergen was present in only 2 of 135 laboratory workers without asthmatic symptoms, it was present in 12 of 18 laboratory workers with symptoms (Platts-Mills et al., 1987).

In the U.S. National Cooperative Inner City Asthma Study, 19% of asthmatic children were allergic to rats and 15% were allergic to mice, suggesting exposure to rat or mouse allergens in the home (Kattan et al., 1997). No data are available on rat or mouse allergen levels in the home and the exacerbation of asthma among rodent-sensitized asthmatics.

Evidence Regarding Asthma Development

Although there are retrospective reports of the incidence of asthma symptoms after beginning laboratory work with rodents (Platts-Mills et al., 1987), the committee could find no relevant studies on rodent allergen exposure and the development of asthma.

Conclusions: Asthma Exacerbation and Development

  • There is sufficient evidence of an association between exposure to rodents in a laboratory setting and exacerbation of symptoms or lung function in rodent-sensitized asthmatics.
  • There is inadequate or insufficient evidence to determine whether or not an association exists between exposure to rodents (wild or as pets) in the home and exacerbation of symptoms or lung function in rodent-sensitized asthmatics.
  • There is inadequate or insufficient evidence to determine whether or not an association exists between exposure to rodents and the development of asthma.

Evidence and Conclusions: Exposure Mitigation and Prevention

No studies are available to document the influence of rodent eradication on the level of rodent allergen or on the severity of asthma in sensitized asthmatics.

  • There is inadequate or insufficient evidence to determine whether or not an association exists between removal of rodents from the home and the reduction of rodent allergen levels.
  • There is inadequate or insufficient evidence to determine whether or not an association exists between removal of rodents from the home and improvement in symptoms or lung function in rodent-sensitized asthmatics.

Cow and Horse

Definition of the Agent and Means of Exposure

Relatively few people in the United States live on farms where exposure to cows, horses, or pigs can be significant; only 1.5% of U.S. households keep horses as pets. Cow hair and dander contain at least 17 antigens, 4 of which have been identified as allergens and 3 of which have been purified (Bos d I, II, and III). Horse hair and dander have been demonstrated to contain three important allergens (Equ c I, II, and III) (Schou, 1993).

Evidence and Conclusions: Asthma Exacerbation and Development

Allergies to cows and horses are considered occupational diseases of farm workers and veterinarians (Prahl and Roedpetersen, 1979; Schou, 1993). Data on the effect of non-occupational exposures is lacking. In summary:

  • There is inadequate or insufficient evidence to determine whether or not an association exists between cow or horse allergen in the home and the exacerbation of asthma in sensitive children or the development of asthma.

Living on a Farm and Development of Asthma

The epidemiologic literature on allergy in farmers and children from farming families was reviewed in a 1999 article demonstrating a lower prevalence of hay fever and allergic sensitization in farmer's children compared to peers from nonfarming families living in the same rural Swiss community (BraunFahrlander et al., 1999). While farm animals can be allergenic, lower rates of sensitization to pollen and animal dander have been reported in adult farmers compared to other occupational groups (Iversen and Pedersen, 1990; Kohler et al., 1983; Rautalahi et al., 1987; Sigsgaard et al., 1996). Swedish conscripts and Finnish university students raised on farms have reported fewer allergic symptoms than students from nonagricultural backgrounds (Åberg, 1989; Kilpeläinen et al., 1997). This may be the result of self-selection out of the farming community by individuals and families with a genetic predisposition toward allergy (von Mutius et al., 1994).

These findings have led investigators to question whether the farm environment itself might play a protective role in the development of allergy and allergic asthma. One hypothesis is that contact with farm animals and their bacterial products (including endotoxin, which is discussed later in this chapter) may be protective against allergy or asthma through early-life stimulation of TH1 immunity, particularly for individuals with specific genetic characteristics (Baldini et al., 1999). However, no additional evidence is available to confirm or reject the hypothesis that for some children, exposure to farm animals early in life might be protective against either allergy or asthma. The farm is a complex environment that varies by country and culture, and many other aspects of farm living may contribute to the observed epidemiologic differences between asthma or atopy in children from farming versus nonfarming families.

Birds

Definition of the Agent and Means of Exposure

Birds are kept in 5% of U.S. households. While it is clear that hypersensitivity pneumonitis can be associated with antigens from bird excreta, serum protein, and proteinaceous material in dispersed dust from birds (Christensen et al., 1975; Hendrick et al., 1978), specific bird antigens associated with allergy and asthma have not been defined with certainty. Tauer-Reich and colleagues (1994) studied five bird fanciers who complained of asthmatic symptoms during contact with their birds and also had documented bronchial hyperreactivity to acetylcholine. These individuals had positive IgE antibody reactions to bird sera as well as to extracts of feathers. Skin prick tests to mites, molds, pollen, and domestic animals other than birds were negative in all five patients.

Evidence Regarding Asthma Exacerbation and Development

A few additional case reports describe bird handlers with the combination of asthmatic symptoms in the presence of birds, symptoms of egg hypersensitivity, and specific IgE antibodies against blood serum proteins of chicken, parrot, budgerigar, or pigeon serum (de Blay et al., 1991b).

A portion of what is called bird allergy may be an expression of allergy to dust mites. Bird feathers can harbor mites (Kemp et al., 1996). The material used in skin prick testing for bird allergy may also be contaminated with mite allergen. Although clinicians have traditionally advised asthmatic patients not to use feather pillows, an epidemiologic study found an increased risk of wheeze in children using foam pillows compared to children using feather pillows (Strachan and Carey, 1995). This association could occur if parents of symptomatic children tend to provide their children with synthetic rather than feather pillows because of advice from health professionals. On the other hand, this association may relate to increase mite exposure from synthetic pillows. New Zealand researchers found that after four months of use, dust mite (Der p I) levels were significantly higher in synthetic than in feather pillows (Crane et al., 1997; Rains et al., 1999). To prevent the feathers from coming out, feather pillows may have more impermeable covers than synthetic pillows; which could result in less dust mite infestation.

Conclusions: Asthma Exacerbation and Development

  • There is limited or suggestive evidence of an association between bird exposure and exacerbation of symptoms in birdsensitized asthmatics. This association may be confounded by the allergic asthmatic response to mites harbored by birds.
  • There is inadequate or insufficient evidence to determine whether or not an association exists between bird allergen exposure and the development of asthma.
  • There is inadequate or insufficient evidence to determine whether or not an association exists between down pillows and exacerbation of symptoms or lung function in asthmatics. Pillows are believed to be a risk factor for asthma because of their documented mite content, rather than because of the presence of bird allergen.

Evidence and Conclusions: Exposure Mitigation and Prevention

There is inadequate or insufficient evidence to determine whether or not an association exists between removal of a bird from the home and reduction in bird allergen levels or improvement in symptoms or lung function in bird-sensitized asthmatics.

Research Needs

The associations between dust mite allergen, asthma exacerbation, and asthma development are much more well defined than the associations between larger animals and asthma. This is only partly a function of the number of years and intensity of efforts to investigate the health effects of dust mites. Compared with dust mite allergen, once the allergen source is present, cat and dog allergens are more easily dispersed throughout the household. Cat and dog allergens remain airborne for much longer than dust mite. The potential for exposure to allergens outside the home is markedly greater for cats and dogs than for dust mites. The potential for home exposure to cat or dog allergen in homes without cats or dogs has also been underestimated. The absence of adequate information regarding allergen exposure may, in part, account for contradictory data regarding the effects of cats or dogs in the home on the development of asthma.

Research is needed to assess whether removal of the cat or dog from the home results in sufficient reduction in overall allergen exposure to reduce symptoms and improve bronchial reactivity in specifically sensitized asthmatics. Further research is needed to assess the level of animal allergen exposure (cat and dog) in day care centers and schools. When significant levels are noted, the potential for lowering exposure should be investigated. Since so many cat- or dog-allergic asthmatics are emotionally attached to their pets, investigators should explore the success of efforts that recommend the removal of the pet for sensitized symptomatic child and adult asthmatics. Further research is also needed to evaluate the effect of mitigation measures short of animal removal on asthma symptoms, lung function, or bronchial responsiveness in specifically sensitized asthmatics. Although frequent animal washing and HEPA filter use are widely recommended, their efficacy in reducing asthma severity has not been proven.

Two retrospective cross-sectional studies suggest that exposure to cat or dog in early life may actually be protective against asthma development in some subsets of children (Hesselmar et al., 1999; Svanes et al., 1999). The relationship between cat or dog allergen exposure in early childhood, the development of sensitization, and the development of asthma merits further investigation. This investigation will require better assessment of exposure. It is likely that the genetic phenotype will modify the response to cat or dog allergen at different levels of exposure, but gene-by-environment interactions cannot be effectively explored until the genetics of asthma is better understood.

Further research is needed to evaluate rodent allergen exposure in the home as a potential factor in the exacerbation of asthma in rodent-sensitized asthmatics. Particularly in socially disadvantaged populations, research should focus on effective reduction of rodent allergen and its effect on symptoms or lung function in specifically sensitized asthmatics.

Researchers should also consider the possibility that animal (or animal allergen) exposure may be either protective or allergenic. The effects may depend on the mode of exposure, the genetic characteristics of the populations, the timing in the life cycle when exposure occurs, and many other cofactors (e.g., early-life viral, bacterial, and parasitic infection experience).

COCKROACH

Many insects have been identified as sources of inhalant allergens in case reports or small outbreaks; these include moths, crickets, locusts, beetles, “green nimitti” midges, lake flies, and houseflies (IOM, 1993). Cockroaches, however, are the only insect that has been repeatedly recognized as a common source of indoor allergens.

Definition of the Agent and Means of Exposure

Agent Definition and Biology

Cockroach is an important source of indoor allergen worldwide. Although more than 60 species have been identified, the most common indoor species in North America are the German (Blattella germanica), American (Periplaneta americana), and Oriental (Blatta orientalis). There are multiple allergens from cockroach that have been identified (Bla g I or Per a I, Bla g II, IV, and V) and cloned (Bla g II, IV, and V). Schou and colleagues (1990) purified an allergen from B. germanica and P. americana extract, which resulted in positive reaction to skin tests in 50% of patients who were allergic to cockroaches and was designated Bla g I (or Per a I). Monoclonal antibodies have been produced against extracts of both cockroach species and used for cockroach allergen identification and purification. Enzyme-linked immunosorbent assays have been developed that can be used to estimate exposure to some of these cockroach allergens (Bla g I, Per a I, and Bla g II) in the environment. Bla g I is a 25-kD (kilodaltons, a unit of molecular weight; also abbreviated kDa) cross-reacting antigen from both the German and the American cockroach. Bla g II is a 36-kD species-specific antigen derived from the German cockroach. Specific allergens have not been purified from Oriental cockroaches.

Neither the antigenic relationships between cockroach species nor the precise source of cockroach allergens are well understood. The source of the allergens derived from cockroaches is not known, but there are speculations to suggest that they may come from feces, parts of the body, or other sources in the body. Sequence homology searches are useful tools for investigation of the biologic function of cockroach allergens, and as more sequences become available it will be possible to make comparisons of biologic function and allergenicity, to compare allergen expression in different species, and to localize the source of the allergens in cockroach tissues.

At present, cockroach extracts are not standardized; however, the Food and Drug Administration's (FDA's) Center for Biologics Evaluation and Research has embarked on a program to standardize B. germanica extracts on the basis of skin testing, protein content, RAST inhibition, and specific allergen assays. Most patients in the United States appear to be sensitized to B. germanica, although there is a cross-reactivity between B. germanica and P. americana on skin tests and serum IgE antibody assay. The prevalences of IgE antibodies to Bla g I and Bla g II in patients allergic to cockroaches are 40% and 60–80%, respectively (Pollart et al., 1991a). Approximately 20% of patients lack detectable IgE antibody to either allergen, which suggests that cockroaches produce other important allergens (Chapman, 1993). However, the levels of the two allergens have been found to be highly correlated (r = 0.92; p < .01) in dust samples (Pollart et al., 1991a).

Factors Influencing Exposure

Studies have suggested that cockroach sensitization is an affliction of the inner city poor, but the complex interrelationship of race, poverty, and residence has been difficult to unravel (Bernton and Brown, 1964; Call et al., 1992; Garcia et al., 1994; Gelber et al., 1993; Koehler et al., 1987). Sarpong and colleagues (1996a) examined race and socioeconomic status (SES) as risk factors for cockroach allergen exposure and sensitization. In their cohort of 48 white and 39 African-American children, they found that both factors were independent predictors of cockroach sensitization. Among low-SES subjects, sensitization was common with 50% (4 of 8) of white participants and 75% (15 of 20) of African-American participants exhibiting a positive skin test. Cockroach antigens are widely distributed in homes and schools, and the kitchen is the most common source of cockroach allergen (Rosenstreich et al., 1997; Sarpong et al., 1996b, 1997). The cockroach allergen in school dust was similar to that reported in homes. The level of antigen reported in school dust is of concern because it may constitute a very important occupational risk to students, teachers, and other school workers. The source of the allergen is not known, but it is likely that the schools may have been infested with cockroaches since there was evidence of dead cockroaches in the schools examined (Sarpong et al., 1997).

In kitchens, food and water sources may be important factors for the proliferation of cockroaches. The humidity in the home may be an important factor for increased cockroach allergen in infested homes. However, in a study evaluating allergen levels in schools, the presence of air conditioners, which may lower the humidity, did not affect the distribution of allergen levels (Sarpong et al., 1997). Despite the evidence that cockroach allergen levels are higher in the kitchens of both asthmatic and nonasthmatic individuals, the concentration of cockroach allergen in the bedroom has been used as the surrogate marker of exposure (Eggleston et al., 1998; Rosenstreich et al., 1997; Sarpong and Han, 1999; Sarpong et al., 1996a, 1996b).

Exposure to cockroach allergens are dependent on their aerodynamic properties and the characteristics of the surface on which they are deposited. In an Ohio study, there was no difference between dust mite allergen concentrations in low-pile carpet and smooth floors, but allergen levels were significantly higher in high-pile carpets (Arlian et al., 1982). In a school study in the Baltimore metropolitan area, there was no difference in cockroach allergen, Bla g I between low-pile carpet and uncarpeted floors (Sarpong et al., 1997). However, kitchen areas are generally noncarpeted, and this may have distorted the level of allergen in uncarpeted compared to carpeted areas.

Cockroach allergens may behave like the dust mite antigen; that is, they are carried on large particles that become airborne for short periods of time during active disturbance. High concentrations of cockroach allergen are found in the kitchen, compared to the living room or bedroom. However, some studies have reported similar levels of cockroach allergen in all these sites. Taken together, the cockroach allergen may be more relevant in the bedroom than the kitchen or the living room because of close contact with the pillow while in bed. Interestingly, about 20% of homes with no evidence of cockroach infestation have significant levels of cockroach allergen in settled dust (Pollart et al., 1991b). Although the possibility of reporting bias cannot be excluded, it is likely that cockroach allergen may be present in a home long after the infestation has been controlled.

Cockroach hypersensitivity is a unique risk factor for asthma among the urban poor. Many case-control studies have documented that cockroach allergen exposure and sensitization are significantly more common in patients with asthma living in urban homes compared to those living in suburban homes (Bernton et al., 1972; Kang et al., 1993; Sarpong et al., 1996a). However, despite higher rates of exposure and sensitization among urban subjects, place of residence was not independently associated with cockroach exposure after correction for socioeconomic status; families of lower SES were likely to be exposed wherever they lived (Sarpong et al., 1996a). This agrees with the observations of Gelber and colleagues (1993), who used lack of health insurance as a marker of poverty and showed that this was a more important correlate of sensitization than urban residence per se as well as with others who have categorized patients on the basis of access to private medical care (Garcia et al., 1994). A 1999 report from Morgantown, West Virginia, documented the role of sensitization in infantile asthma (Wilson et al., 1999). These children who were at least 3 years of age with documented wheezing episodes demonstrated cockroach hypersensitivity at a rate of 25%. The rate of sensitivity in this predominantly white population was not related to low socioeconomic status.

Evidence Regarding Asthma Exacerbation and Development

Biologic Plausibility

Bernton and colleagues (1972) reported that inhalation of cockroach extract could induce an immediate asthmatic reaction in cockroach-sensitive asthmatic subjects. Subsequently, controlled inhalation challenge confirmed the production of antigenspecific, acute and late bronchospasm in cockroach-sensitive asthmatics (Kang, 1976). These antigen-induced asthmatic reactions were blocked by premedication with disodium cromoglycate. Challenge of these subjects with inhaled cockroach extract produced a significant antigen-specific peripheral eosinophilia, which progressed to its peak 24–36 hours after exposure and often more than doubled the baseline value. In animal studies there is evidence to suggest that cockroach antigen can induce lung eosinophilia (Campbell et al., 1998). Previous data in guinea pigs have shown that aerosolized cockroach antigen can be utilized to induce airway inflammation and alter airway physiology (Kang et al., 1996). The development of these models will allow the evaluation of mediators involved in both stages of cockroach allergen challenge, as well as the testing of specific therapeutic modalities.

Consistency

There is now good evidence from epidemiologic studies in several parts of the world which demonstrates that the development of immediate sensitivity to cockroach allergens is associated with asthma morbidity and that sensitization is related to the degree of allergen exposure (Eggleston et al., 1998; Rosenstreich et al., 1997; Sarpong and Han, 1999; Sarpong et al., 1996a; Sastre et al., 1996). In the original description of cockroach sensitivity in 1967, Bernton and Brown (1967) found higher rates of sensitization among Puerto Ricans and African Americans than among Jews and Italians in New York City and believed that the difference was related to the improved economic conditions of the latter groups. Shulaner (1970) also found a relationship to poverty and overcrowding, and a report by Gelber and colleagues (1993) relates exposure and sensitization to urban housing, race, and poverty as indicated by lack of medical insurance.

Strength of Association

Sensitization, with production of specific IgE antibodies to cockroach, is a strong risk factor for acute severe asthma, especially when sensitized persons are exposed to high concentration of allergen in their homes. R.P. Nelson and colleagues (1996) compared 29 children with acute asthma, ages 3–16 years, first seen in a Florida emergency department, to 25 control subjects. They found that sensitization to cockroach allergens was associated with acute asthma that required emergency treatment. Gelber and colleagues (1993) and Call and colleagues (1992) showed that cockroach sensitization was a risk factor for acute asthma in patients visiting emergency departments in Wilmington, Delaware, and Atlanta, Georgia, respectively. The multicenter National Cooperative Inner City Asthma Study (NCICAS) of the home environment of asthmatic children aged 4–9 years living in seven major urban areas and eight centers reported that cockroach allergen was the predominant indoor allergen in these homes. In the NCICAS, it was suggested that sensitization to cockroach allergen and exposure to high levels of cockroach allergen may explain the asthma-related health problems of inner city children (Rosenstreich et al., 1997).

Dose–Response

A dose–response relationship between cockroach allergen exposure and sensitization has been established in asthmatic children. Sarpong and colleagues (1996a) report that children who were exposed to Bla g I or Bla g II of 1 unit per gram (U/g) or higher demonstrated skin sensitivity to cockroach allergen. For Bla g I, all the children who were exposed to more than 10 U/g were sensitized to cockroach allergen. Similarly, 100% of the children who were exposed to more than 5 U/g of Bla g II were sensitized to cockroach allergen. Following this report, the NCICAS has suggested that cockroach allergen (Bla g I) exposure is related to sensitization in a dose–response manner (Eggleston et al., 1998). Preliminary data from a population of pregnant women have suggested that total cockroach allergen (Bla g I and Bla g II) exposure and sensitization were related in a dose–response fashion (Sarpong and Han, 1999). This relationship was demonstrated in both asthmatic and nonasthmatic controls. However, the dose of cockroach allergen concentration required to induce sensitization rates in the asthmatic population was at least a factor of ten lower than in the nonasthmatic population. A dose–response relationship was also demonstrated in the inner city asthma study between cockroach allergen (Bla g I) exposure and morbidity due to asthma. Children who were exposed to more than 8 U/g of Bla g I were more likely to be hospitalized for asthma symptoms than those who were exposed to less. In a prospective study by Gold and colleagues (1999), infants who were born in homes with Bla g I levels greater than 2 U/g reported increased risk of wheezing by the age of 1 year. A study by Litonjua and colleagues available as a conference abstract (1998) evaluated the relationship between home allergen levels and the prevalence and incidence of asthma 16 months later among 215 children younger than 5 years old. The researchers found that, among the children with new onset of asthma, 7 (87.5%) lived in homes with high Bla g I or II levels while 1 (12.5%) lived in a home with low levels (p = .028).

Although it seems clear that exposure to cockroach allergen precedes disease, it has not been shown that sensitization consistently precedes disease. However, it is the combination of strong association, biological plausibility, dose–response, and provocation experiments that creates the strength of the argument.

Conclusions: Asthma Exacerbation and Development

Cockroach antigen exposure can elicit a strong IgE immune response to induce sensitization. Sensitization to cockroach antigen has been linked to the season of birth (Sarpong and Karrison, 1998a), although this study does not necessarily support cockroach antigen as a disease promoter, it at least gives some insight that exposure during the perinatal period may be critical. Because of the likely aerodynamic nature of the cockroach allergen, it is difficult to predict whether cockroach allergen per se is an initiator of asthmatic symptoms. However, data suggest that infants who are exposed to cockroach allergen are more likely to wheeze during infancy. Moreover, researchers have shown that cockroach sensitization is a risk factor for acute asthma in patients visiting emergency departments (Call et al., 1992; Gelber et al., 1993; R.P. Nelson et al., 1996). In a retrospective study of asthmatic children in Chicago, it was suggested that children with combined sensitization to cat, dog, dust mite, and cockroach allergens were at increased risk of having more severe asthma (Sarpong and Karrison, 1998b). Researchers have also found that there is an increased risk of sensitization to cockroach allergen in asthmatic children born in the winter months (Sarpong and Karrison, 1998a). Because the risk of respiratory syncytial virus (RSV) infection is high in winter months and RSV is also known to give rise to asthma-like symptoms, others have even suggested that it may promote IgE sensitization. The level of cockroach allergen in settled dust may not demonstrate seasonality. It is therefore attractive to postulate an interaction between RSV bronchiolitis and cockroach allergeninduced sensitization. Thus, there may be virus–antigen interaction in initiating sensitization and possibly inducing asthma.

Every individual is at risk for cockroach allergen exposure and sensitization. However sensitization risk is higher among the asthmatic population. Understanding the genetics of allergy and asthma, including understanding the phenotypes associated with cockroach allergy, may eventually prove useful in the understanding of gene–environment interaction in the development of asthma.

There are no prospective data demonstrating the association of exposure to cockroach allergen with subsequent development of sensitization to cockroach allergen and asthma. However, a number of case-control studies demonstrate a link between cockroach allergen exposure in sensitized individuals and asthma morbidity, as well as a dose–response relationship between cockroach exposure and sensitization (Gelber et al., 1993; Rosenstreich et al., 1997; Sarpong et al., 1996a).

In summary:

  • There is sufficient evidence of a causal relationship between cockroach allergen exposure and exacerbation of asthma in individuals specifically sensitized to cockroaches.
  • There is limited or suggestive evidence of an association between cockroach allergen exposure and the development of asthma in preschool-aged children.
  • Inadequate or insufficient evidence exists to determine whether or not an association exists between cockroach allergen exposure and the development of asthma in older children and adults.

Evidence Regarding Exposure Mitigation and Prevention

In principle, control of exposure to cockroach allergens combines three approaches directed through an indoor environment:

1.

control of reservoirs of allergen in beds, carpets, furnishings, and clothing, which are the main sources of exposure;

2.

control of the sources of new allergen (e.g., cockroach reinfestation); and

3.

direct control of cockroach airborne allergens.

There are several potential strategies for achieving these forms of control. These are noted below, along with some of their practical limitations.

The bed is probably the most important site of cockroach allergen exposure because of the high level of exposure during sleep, the proximity of the subject to the source, the proportion of time spent indoors at this one site, and the large amounts of dust present in the bed. The relative success and simplicity of interventions directed at this site make it a key target for allergen control. Strategies known to reduce allergen exposure include encasing mattresses and pillows and laundering bedding in hot (>130°F, or 55°C) water. (This strategy is also effective for dust mite allergen.) However, there may be problems of subject compliance with regular washing of some items, such as blankets, even in the setting of a clinical trial.

Carpets serve as a major reservoir of cockroach and many other indoor allergens and may serve as an additional primary source of allergen. Replacement of fitted carpets with smooth flooring has been shown to reduce dust mite allergen levels (Hayden et al., 1997) and is often recommended as a cockroach allergen management strategy. However, this intervention is sometimes unpopular and may be impractical in rental units, public housing, and other environments where occupants do not have control over floor coverings. Dry vacuum cleaners are useful for picking up excess dust and reducing reservoirs and allergen concentration. Whether this achieves significant reductions in exposure remains to be demonstrated. Further, while wet vacuuming and steam cleaning may reduce allergen levels, they also may enhance the environment for dust mites and other potential allergens if the surface does not dry properly or promptly.

Extermination is a primary approach to control new sources. The use of avermectin or hydramethylnon has, for example, been shown to reduce cockroach populations (Cochran, 1995, 1996).

Finally, housekeeping measures aimed at limiting open food-stuffs (using sealed containers, cleaning under refrigerators and stoves, washing dishes promptly after meals, and the like) and water sources (fixing leaking faucets and pipes; eliminating pet water dishes) are often suggested as part of an overall cockroach management strategy.

As noted in Chapter 10, ventilation rates are not likely to have an appreciable direct impact on indoor concentrations of the larger particles associated with cockroach allergens.

Attempts to eliminate cockroach allergen from the environment have had limited success. A short-term effect of extermination and cleaning on cockroach antigen levels was noted in a study conducted in an urban college dormitory (Sarpong et al., 1996b). Other studies have found that the number of cockroaches seen in a dwelling can be drastically reduced with roach insecticides such as hydramethylnon. However, the elimination of cockroach sightings did not decrease the levels of Bla g I or Bla g II in vacuum dust over the next six months in single-family dwellings in North Carolina. No extra cleaning was performed in these homes (Williams et al., 1999). Eggleston and colleagues (1999) evaluated the effect of professional pest control and home cleaning on infestation and allergen concentrations in 13 inner-city homes with active infestations. Occupants were instructed on how to conduct follow-up cleaning and pest control measures, and study personnel monitored compliance over the eight months of the study. The researchers reported decreases in infestation and in allergen concentrations in settled dust. While mean allergen concentrations were reduced between 74% and 93% in various rooms of the homes, they still exceeded 20 U/g, a level associated with increased morbidity in the NCICAS study. In the NCICAS, integrated intervention to reduce cockroach allergen in homes was conducted. Despite a significant but short-lived reduction, cockroach allergen levels remained well above those previously found to be clinically significant (Gergen et al., 1999). Insufficient evidence is available to determine whether or not reduction of cockroach allergen levels in the home reduces asthma severity in cockroach-sensitized asthmatics. Additional studies are needed to evaluate, within both multi-unit and single family housing, the effectiveness of various proposed methods of reducing cockroach allergen to levels below those associated with symptoms in asthmatic subjects. Until more effective methods of reduction of cockroach allergen levels are developed, it may prove difficult to assess the effectiveness of cockroach allergen reduction programs in reducing asthma morbidity. Ongoing intervention studies may provide further data as to whether, for some subsets of sensitized asthmatics, moderate reductions in allergen levels or in allergen load influence asthma morbidity. Given the evidence that decreasing exposure to dust mites can help control the symptoms of dust mite-allergic asthmatics, it is prudent to identify patients who are allergic to cockroaches and educate them to reduce allergen exposure indoors.

Conclusions: Exposure Mitigation and Prevention

There is preliminary evidence to suggest that reduction in cockroach allergen in settled dust can be achieved with extermination on a short-term basis; there are ongoing studies to evaluate whether long-term reduction of cockroach allergen in settled dust can be achieved with extermination. It appears that the use of abatement strategies combining extermination and cleaning can reduce cockroach allergen exposure. However, research has not to date demonstrated that these strategies result in an improvement in symptoms or lung function in cockroach-sensitized asthmatics. It is important to remember that the absence of evidence does not mean an absence of effect. The science regarding indoor environmental interventions, exposure limitation, and effects on asthma outcomes is not nearly as well developed as that regarding the health effects of exposures. Given the evidence that decreasing exposure to dust mites can help control the symptoms of dust mite-allergic asthmatics, it is prudent to identify patients who are allergic to cockroaches and educate them to reduce allergen exposure indoors.

In summary:

  • There is sufficient evidence of an association between the implementation of intensive cockroach allergen exposure mitigation strategies and short-term reduction of cockroach allergen levels. Such strategies must include both removal of the allergen from reservoirs and control of sources (i.e., abatement and prevention of reinfestation) to be effective.
  • There is inadequate or insufficient information to determine whether or not an association exists between cockroach reduction interventions and improvement in symptoms or lung function in cockroach-sensitized asthmatics.

Research Needs

The preceding discussion suggests that there is still a need for fundamental research on cockroach allergens and asthma outcomes. Future research should focus on the efficacy of cockroach allergen reduction in the homes of asthmatic patients, the aerodynamic properties of cockroach allergen, the efficacy of cockroach immunotherapy, and B and T cell reactive epitopes. Further studies are also needed to better elucidate any relationship between cockroach allergen exposure and asthma development; explore the interaction of cockroach allergen with infectious agents, irritants, and other allergens in causing asthma; and examine the influences of genetics, socioeconomic status, and location on exposure and sensitization.

HOUSE DUST MITES

In 1967, Voorhorst and colleagues identified dust mites of the genus Dermatophagoides as the most important source of allergens in house dust (Voorhorst et al., 1969). Using dust mite extracts, they demonstrated that sensitization was common among children with asthma and later developed techniques for growing mites. By 1974, extracts were widely available for skin testing, and mite sensitization became recognized in many countries. Indeed, mite sensitization was found to be strongly associated with asthma in all countries that had reported increases in asthma up to 1980 (i.e., the United Kingdom, Australia, New Zealand, Japan) (Clarke and Aldons, 1979; Miyamoto et al., 1968; Smith et al., 1969).

Since the original research by Voorhurst and colleagues (1969), most of the studies on allergen avoidance have focused on miteallergic patients. Studies have included moving children from Holland to Davos (Kerrebijn, 1970), keeping mite-allergic adults in special hospital rooms (Platts-Mills et al., 1982), and controlled trials of dust mite reduction in patients' rooms (Murray and Ferguson, 1983). In each of these models, change in environment with reduction in exposure to mite allergen was associated with decreased symptoms of asthma and decreased bronchial hyperreactivity (BHR) (Platts-Mills and de Weck, 1988).

Definition of the Agent and Means of Exposure

Agent Definition and Biology

Purification of indoor allergens is dependent on the quality of source materials. Before the discovery of dust mites, several unsuccessful attempts were made to purify allergens from dust obtained from carpets and bedding. Even when dust mite cultures became available, the early attempts at purification used only a small amount of material, ~40 g. The first purification of the major allergen Der p I was by classical immunochemistry from 400 g of “spent” culture (Chapman and Platts-Mills, 1980). The spent culture was very rich in fecal material. Purification made it possible to measure the quantity of allergen in extracts, in dust samples, and airborne (Tovey et al., 1981a). In addition, the purified allergens have been used to study the immune response to allergens, as well as the properties of these proteins. Each of these approaches has relevance to the causes of lung symptoms.

Epidemiologic evidence about the relationship between sensitization to indoor allergens and asthma is based on skin tests or on serum assays of IgE antibodies. However, the immune response to inhalant allergens also includes IgG antibodies, IgA antibodies, and T cells. These T cells in allergic individuals are predominantly CD4+ cells of the TH2 type (Romagnani, 1992; Wierenga et al., 1990). There are two important questions about this response: (1) Can differences in the immune response predict which skin test-positive individuals will develop asthma? Most of the evidence suggests that skin test-positive asymptomatic individuals have very similar IgE antibodies and T cells (i.e., TH2) compared to symptomatic patients. (2) What, if anything, is the immune response to mite allergens in skin test-negative individuals? Here, the evidence is conflicting. Several groups are proposing that nonallergic individuals have made a TH1 response. This is supported by evidence that T cells in the cord blood of individuals who are going to become allergic produce less interferon (IFN-γ) in response to nonspecific stimuli, and also by evidence that it is possible to clone mite-specific TH1 T cells from the peripheral blood of nonallergic individuals (Miles et al., 1996; Prescott et al., 1998; Warner et al., 1997; Wierenga et al., 1990). On the other hand, most investigators have not found evidence of an immune response to mite proteins in nonallergic individuals. For example, most skin test-negative individuals have no detectable IgG antibodies to purified antigens, have poor or no in vitro T cell responses, and have no immediate or delayed skin responses. These results suggest (1) that the primary issue is who makes an IgE antibody response and (2) that most individuals who are nonallergic have not made any immune response to dust mites.

Almost all of the well-defined allergens are proteins or glyco-proteins, and it is not surprising that many of them have amino acid sequence homology with known enzymes (Arruda et al., 1997; Stewart and Thompson, 1996). In some cases these proteins have enzymatic activity that could play a role in their immunogenicity. For example, Der p I is a potent protease that has been shown to cleave CD23 and CD25 on the surface of lymphocytes in vitro (Hewitt et al., 1995; Schulz et al., 1998). Furthermore, in animal experiments, it appears that the enzymatic activity of Der p I can influence immunogenicity (Comoy et al., 1998). However, enzymatic activity is not a prerequisite for a protein to be a significant allergen, and there are considerable doubts about whether these enzymes are active in vivo. Some of the allergens that have homology with enzymes have not been shown to have enzymatic activity. More significantly, it has not been established that any of these proteins have enzymatic activity under the conditions that occur in the human respiratory tract. In addition, many important allergens do not have homology with enzymes, including Der p II. It is certainly possible that enzymatic activity is relevant to the effects of some allergens; however, it is unlikely that this property plays an important role either in inducing IgE antibody responses or in causing symptoms.

Factors Influencing Exposure

The quantities of dust mite allergen that have been found in the air of houses range from <0.2 to ≥100 ng/m3. Thus, accurate determination of the quantity and particle size of airborne allergen is dependent on immunoassays capable of accurately measuring quantities as small as 1 ng (Chapman et al., 1987; Luczynska et al., 1989; Sakaguchi et al., 1990b). After it became clear that Der p I was concentrated in mite fecal particles, experiments were designed to answer whether the allergen became airborne in this form. Using various approaches to air sampling, several conclusions became clear: (1) there is very little or no airborne mite allergen in an undisturbed room; (2) the allergen that becomes airborne during disturbance is predominantly on particles ≥10 µm; and (3) allergen falls rapidly after disturbance, in keeping with an aerodynamic size of ~10 µm (Tovey et al., 1981b). In addition, allergen-containing particles identified using a microimmunodiffusion technique have been shown to be mite fecal particles (Tovey et al., 1981a). These properties are strikingly different from those of other indoor allergens (e.g., of cat or dog). However, the size of the particle is very similar to pollen grains. A small proportion (i.e., 5–15%) of the fecal particles that are breathed through the mouth would be expected to enter the lungs, and we assume that these particles are assumed to produce the inflammatory response (Bates et al., 1966; Svartengren et al., 1987). At present the question of how allergen particles enter the lungs is not resolved, and this issue is of considerable importance since it may well define the distribution of “inflammation” and airway obstruction.

Evidence Regarding Asthma Exacerbation and Development

Association with Sensitization

Many studies have shown an association between dust mite sensitization and asthma (Platts-Mills et al., 1997). These studies include case-control studies in clinics, emergency rooms, and hospitals (Gelber et al., 1993); population-based studies in schools (Peat et al., 1996; Squillace et al., 1997); and prospective studies (Sears et al., 1989; Sporik et al., 1990). The results consistently show odds ratios for asthma of 6 to >12 in individuals with dust mite sensitization. However, in all studies there are a significant number of individuals who are skin test positive but not symptomatic. Using multiple regression analysis, a prospective study in New Zealand demonstrated that mite sensitization was an independent risk factor for asthma (Sears et al., 1989). In the same analysis, pollen sensitization was not significantly associated with bronchial reactivity. This pattern has now been seen in many studies and in several different countries (Table 5-1). Furthermore, similar results have been found in areas where other indoor allergens are most important (Sporik et al., 1995). That is, the relationship between sensitization and asthma is not for allergens in general but specifically for those allergens to which patients are exposed perennially in the environment. Although most of the perennial allergens are found indoors, the airborne fungus Alternaria is also an important factor in areas of the world where exposure persists for many months of the year (Halonen et al., 1997).

TABLE 5-1. Sensitization as a Risk Factor for Asthma (symptomatic bronchial hyperreactivity).

TABLE 5-1

Sensitization as a Risk Factor for Asthma (symptomatic bronchial hyperreactivity).

In many of these studies the odds ratios for asthma for individuals who are allergic to dust mites are very high (i.e., ≥6) (Table 5-1 ). However there are several features of the prospective and case control studies that need to be emphasized. In all reported studies, there were a significant number of individuals who were skin test positive to dust mites (or other indoor allergens) but did not have symptoms. In many countries or areas, dust mites are the most important source of allergens. In the south, the south-east, and the west coast of the United States, as well as in New Zealand, Australia, Japan, and the UK, mites of the genus Dermatophagoides are dominant. By contrast in areas where dust mites do not thrive, other allergens are important (e.g., animal dander in the mountain states of the United States and in Scandinavia; or cockroach in the apartments of Chicago, Boston, New York, and Philadelphia). In addition the studies shown in Table 5-1 illustrate the fact that in most areas sensitization to outdoor pollens is not significantly associated with asthma.

Sensitization to storage mites (Lepidoglyphus destructor, Tyrophagus putrescentiae, Acarus siro, and others) is an important risk factor in occupational asthma in agricultural settings (ATS, 1998). These mites may also play a role in nonoccupational asthma from exposures in indoor environments in rural and perhaps other areas, although research characterizing exposure levels and responses is lacking.

Association with Exposure

Sampling. The measurement of allergen entering the lungs or even the measurement of dust mite allergen inhaled has proved very difficult. The essential problem is that the allergen is carried on particles that behave aerodynamically as if they are 10–25 µm in diameter. This means that in still air, they remain airborne only for a few minutes. Thus, all measurements of airborne dust mite have required artificial disturbance, and it has not proved possible to standardize disturbance (Sakaguchi et al., 1990b; Tovey et al., 1981b). Because of this, other measurements of mite allergen have been used as an index of exposure. The accepted measurement is the concentration of Group 1 mite allergen, which is Der p I + Der f I in house dust expressed as micrograms per gram of dust. The immunoassays available use monoclonal antibodies in a two-site enzyme-linked immunosorbant assay (ELISA). The significance of this measurement has been endorsed by three international workshops (Platts-Mills et al., 1997). While there are many different ways of obtaining samples, the most widely used technique is to sample reservoir dust from bedding, bedroom floors, and the living room with a hand-held vacuum cleaner.

In areas of the world that are humid persistently or for at least eight months of the year, mites flourish inside houses, and it is not unusual to find 100 or even 500 mites per gram of dust. This translates to 2 or 10 µg of Group 1 allergen per gram of dust, respectively. In areas where most houses (i.e., at least 80%) have greater than 2 µg of mite allergen per gram of dust, sensitization to these allergens has consistently been found in a large proportion (45–85%) of children with asthma. In some studies the concentration in individual houses has been shown to be an important predictor of sensitization (Peat et al., 1996; Sporik et al., 1990). In Germany, Kuehr and his colleagues demonstrated that the concentration necessary to convert “non-atopic” children to positive skin tests to dust mite was ~60 µg/g. In the same study, children who already had a positive skin test to another allergen were at risk of becoming sensitized with exposures greater than 2 µg/g (Kuehr et al., 1994). Taken together, the results strongly support a dose–response relationship between exposure and development of sensitization, with an approximate threshold of 2 µg/g. This threshold is a statistical concept representing the concentration at which exposure to mite allergen becomes a risk for sensitization. However, there is no sense in which the threshold for mite exposure is comparable to thresholds for airborne toxic gases: (1) high concentrations of mite allergen are not toxic to nonallergic individuals; (2) concentrations below this level may cause symptoms for highly allergic or highly reactive asthmatics; and (3) the measurement is an index of exposure, which may not reveal high levels of exposure elsewhere in the house and has only an indirect relationship to exposure of the lungs.

Experimental Evidence

The earliest experiments on bronchial provocation confirmed that allergens inhaled into the lungs could produce an asthmatic response (Blackley, 1873). With the development of the nebulizer it became much easier to challenge the lungs. The exposure represents ~108 droplets with 0.1–10 µg Der p I per milliliter in a 2-minute challenge giving approximately 0.5 mL inhaled. In allergic individuals this challenge consistently gives rise to an immediate fall in FEV1 and often produces a late response as well (Cockcroft et al., 1979). Following a bronchial challenge, eosinophils are recruited into the lungs in keeping with the events that are thought to occur in asthma. These changes can also be produced by a segmental challenge, and in these experiments the eosinophils may persist in the lung for up to two weeks after challenge (Shaver et al., 1997).

The lungs of patients with asthma are characterized by inflammation and nonspecific BHR. In several different types of studies it has been shown that these changes can be reversed under conditions that include reduction of exposure to mite allergens. Mite allergic children moved from the Netherlands; Marseille, France; or Verona, Italy to sanatoriums in the Alps have consistently shown clinical improvement and decreased nonspecific BHR. These sanatoriums have many features that are different from the children's homes, including very low mite allergen levels (Platts-Mills and de Weck, 1988). Piacentini and his colleagues (1996) in Italy have also shown that the children who spend three months in the Dolomite Mountains have a decrease in eosinophils and eosinophil products in induced sputum in parallel with decreased BHR. With adults, similar data have been obtained by moving patients from their homes in London to mitefree hospital rooms. Again, there were many other changes associated with the move to mite-free rooms (e.g., no animals, very low spore counts, increased physical activity as patients felt better). However, it is clear that BHR is reversible in many mite-allergic patients.

Conclusions: Asthma Exacerbation and Development

In mite-sensitive asthmatics, continual exposure to mite allergens is a contributing cause of exacerbations and chronic bronchial hyperreactivity. The risk of disease, however, is not a function of either the magnitude of the positive response to skin tests or the level of IgE antibody measured in serum. Therefore, in summary:

  • There is sufficient evidence of a causal relationship between dust mite allergen exposure and exacerbations of asthma individuals specifically sensitized to dust mites. Continual exposure to dust mite allergens is also a contributing cause of chronic bronchial hyperreactivity.
  • There is sufficient evidence of a causal relationship between dust mite allergen exposure and the development of asthma in susceptible children.

It is very difficult to prove a causal relationship between an agent that is present in the environment every day and a chronic disease. Certainly, none of the studies reported here taken on its own can be considered to prove that dust mite antigens cause asthma. However, when all of these studies are taken together the case becomes compelling. In 1965, Bradford Hill presented the argument that many different types of study should be considered together to make a case for causality. He was addressing the specific case of a dust causing chronic lung disease (Hill, 1965). When these criteria are applied to the role of dust mites in asthma, the case becomes very strong (Box 5-1). It is the combination of association, biological plausibility, provocation experiments, and the results of avoidance that create the strength of the argument.

Box Icon

BOX 5-1

Evidence That the Relationship Between Dust Mite Exposure and Asthma Is Causal. The association between sensitization and asthma as judged by odds ratios (i.e., ≥6) is very strong in: population-based studies;

Evidence Regarding Exposure Mitigation and Prevention

Distribution of Mites in Houses

Most houses contain at least three of the four requirements for mite growth: (1) there are multiple sites that can provide a nest for mites (i.e., carpets, sofas, mattresses, pillows, bedding); (2) the presence of humans guarantees an abundant food source in the form of skin scales; (3) in the latter half of the twentieth century, most houses are kept close to the optimal temperature for mite growth. For these reasons, the fourth requirement—humidity—is the major and often the only factor that determines whether a house has high concentrations of mites and mite allergen. In areas where humidity is high for most of the year, mites will grow well and may be found in almost any fabric including drapes and clothing, as well as traditional nests. By contrast, in truly dry areas (e.g., the mountain states [≥5,000 feet elevation]), and in Chicago in winter, mites cannot grow and houses are generally devoid of mites (Rosenstreich et al., 1997; Sporik et al., 1995). It is in areas where humidity is marginal or raised for a prolonged season that the structural features of a house may make a large difference in the concentration of mites (see Box 5-2). The three biggest factors are the position of the living area within a building; the presence of carpeting on unventilated floors, which will both trap water leaks and lead to local condensation; and buildings whose ventilation rates are so low that humidity produced by the occupants will accumulate even though the outdoor humidity is not high. The scale of these effects is well illustrated by the fact that a sample of houses and duplexes in Boston was between two and three times more likely to have “high” (in excess of 10 µg/g) dust mite allergen concentrations than apartments in the same city (Chew et al., 1998).

Box Icon

BOX 5-2

Features of Houses That Can Produce Major Changes in Mite Growth in “Marginal” Climates. Structural Features Ground floor level

Strategies to Control Mite Growth According to Climate

Humid Climates (i.e., eight months of the year or more with outdoor air water content of ≥5 g/g). In these climates, controlling mite growth can be achieved only by air conditioning or reducing the nests for mite growth. Air conditioning to maintain indoor relative humidity below 50% requires tight housing and is expensive in terms of energy costs. Controlling or minimizing nests can be achieved by removing carpets; reducing furniture or using surfaces such as leather; and avoiding unnecessary fabrics such as drapes, soft toys, and excess unenclosed clothing. In these areas of the country, the normal practices for bedding are effective (e.g., covering mattresses and pillows; regular hot [>130°F or 55°C] washing of all bedding).

Areas of Moderate or Seasonal Humidity. Many strategies can be helpful in controlling mite growth in these areas (see Box 5-3). During dry seasons, simply opening windows for one hour per day will ensure removal of humidity from the house (Harving et al., 1994). In addition, moving to an upper-level apartment can dramatically decrease mite exposure. The simple physical strategies normally advised are effective. In particular, the recommendations for bedding, carpets, and reducing furnishings can all help. Chemical treatment of a carpet using benzyl benzoate can be effective in controlling mite growth for a period of months (Hayden et al., 1992).

Critical issues include the nature of the fabrics used to cover mattresses and pillows and whether fabrics that breathe but block passage of mites can prevent colonization of mattresses, pillows, or furniture (Vaughan et al., 1999).

Dry Areas. Here, control of mites can be achieved simply by ensuring ventilation on a daily basis. In these areas, only very tight housing will allow accumulation of humidity within the building. Thus, this is more likely to be a problem in cold areas where the temptation to control heat loss may prevent loss of water produced by the inhabitants of a house. In the Mountain States and the Southwest, mite growth in houses is unusual.

Conclusions: Exposure Mitigation and Prevention

  • There is sufficient evidence of an association between the use of a combination of the physical measures described above and a reduction in dust mite allergen levels. As noted, the most appropriate measures vary according to the type and characteristics of the indoor environment and the prevailing climate.
  • There is sufficient evidence of an association between the use of a combination of the physical measures described above and an improvement in symptoms or lung function in mite-sensitized asthmatics. These have been shown to be effective at reducing symptoms in controlled trials and should be part of normal management of asthma in mite-allergic individuals.
  • There is inadequate or insufficient evidence to determine whether dust mite allergen mitigation strategies have an effect on asthma development. Because dust mites cannot survive in dry environments, living in such conditions does limit the opportunity for sensitization to mite allergens.

Summary

There are many reasons why dust mites of the genus Dermatophagoides have played such an important role in the evidence about the role of indoor allergens in asthma. These eight-legged arthropods are invisible to the eye and do not produce any odor that we can detect. Nonetheless, they grow extremely well in houses, requiring only humidity and a nest such as bedding, mattresses, or a carpet. As a result, a large proportion of the population is unknowingly exposed to high concentrations of these highly immunogenic proteins. In keeping with experimental responses to repeated low-dose antigen, the immune response is characterized by IgE antibodies. The factors that influence the development of asthma in allergic individuals are not fully understood. However, most of the allergens associated with asthma are perennial and are predominantly indoors. Indoor or perennial exposure characteristically produces symptoms that persist for much of the day and cannot be related to exposure. Indeed there is no characteristic history of symptoms that can be used to identify patients who are allergic to dust mites. It has been suggested that year-round exposure to dust mite allergens, which are inhaled as a “few” particles per day (i.e., ~100), may be the ideal way to establish chronic inflammation of the lungs and the associated bronchial hyperreactivity, without individuals' being aware that they are exposed. Given the evidence that decreasing exposure can help control the symptoms of an allergic patient, patients who are allergic should be identified and educated. However, it is equally important to try to identify the factors that have led to an increase in the number of individuals who have asthma associated with immediate hypersensitivity to dust mites. Although this may in part reflect an increased number of allergic individuals, it appears that much of the rise is due to an increase in asthma among allergic individuals.

Research Needs

Although more is known about dust mite allergen and its impact on asthma than most indoor exposures, research remains to be done. Particularly important is additional work on the effectiveness of specific environmental interventions in limiting asthma exacerbations and development (rather than simple measurement of allergen levels). Several studies now under way are evaluating whether aggressive allergen avoidance regimes have an effect on the subsequent development of asthma (Tovey and Marks, 1999). The results of such studies will inform the question of whether primary prevention of dust mite-induced asthma is possible, although the burdensome nature of such interventions suggests they may be difficult to implement in many circumstances. The development of methods to identify individuals, especially infants, at high risk would provide the information needed to focus primary prevention activities. A major issue in this regard is whether sensitization can occur before birth.

ENDOTOXINS

Endotoxins are components of some bacterial cell walls. They are released when the bacteria die or when the cell wall is damaged. Endotoxins originally came to the attention of physicians because of their potential to cause fevers; more recently it has been established that they can cause airway inflammation and airflow obstruction at higher exposure levels.

Research on endotoxin exposure and asthma is not as mature as that for many of the other exposures addressed in this chapter. Accordingly, this section focuses primarily on the background biologic information that underlies current research into endotoxin's role in the pathogenesis of asthma.

Definition of Agent and Means of Exposure

Agent Definition and Biology

Endotoxin is the substance responsible for certain characteristic toxic effects of gram-negative bacteria. The toxic compound, lipopolysaccharide (LPS), is a structural component of the outer membrane of these bacteria, the polysaccharide portion of which represents the antigenic surface (Sonesson et al., 1994). The lipid portion of the molecule (lipid A) is essential for its characteristic toxicity. The outer portion of the polysaccharide (O-specific antigen) varies among serotypes of a single bacterial species. The core polysaccharide and lipid A are conserved within species but vary in structure and composition between species and to a greater extent between genera. Variations in lipid A structure are associated with variations in toxic potency over a wide range, and there is some evidence for qualitative variations in toxicity. Gram-negative bacterial endotoxin should not be confused with Bacillus thuringensis delta endotoxin (Du et al., 1999; Potekhin et al., 1999), a protein from a gram-positive bacterium that has recently been genetically engineered into certain crops.

Factors Influencing Exposure

Because gram-negative bacteria are the natural surface flora for plants and are abundant in soil (Edmonds, 1979), endotoxin is ubiquitous in the outdoor environment, particularly during the growing season. High-level exposures to endotoxin occur when organic dust is generated in agriculture and related industries such as animal feed production and in cotton mills (Rylander and Morey, 1982). Recirculating water systems can also be sources of endotoxin, and high level exposures have been recorded in industries where machining fluids and recirculated wash water are used (Milton and Johnson, 1995; Walters et al., 1994). Humidification systems are also potentially abundant reservoirs for gram-negative bacteria and thus for endotoxin (Flaherty et al., 1984; Rylander et al., 1978). Therefore, even home humidifiers can generate high levels of endotoxin exposure (Tyndall et al., 1995).

Measurement of endotoxin exposure in homes is usually performed with a Limulus amebocyte lysate assay because this bioassay correlates well with endotoxin measured in the rabbit pyrogen assay. However, Limulus-based assays are prone to interference (Milton et al., 1997) and likely underestimate exposures in organic dusts, including house dust, compared to chemical assay (Saraf et al., 1999). Furthermore, endotoxin in organic dust contains a wide variety of lipid A structures, not all of which are equally reactive in the Limulus-based assay (Saraf et al., 1997). These compounds may not stimulate significant production of the inflammatory cytokine interleukin-1 (IL-1), but may be able to stimulate other effects such as reversing tolerance to certain antigens (Baker et al., 1990, 1992) that may be important in directing immune responses.

Endotoxin exposures in homes have not been extensively studied. A doctoral thesis described the association of airborne endotoxin levels with home characteristics recorded on questionnaires (Park, 1999). The strongest predictors of increased endotoxin in living room air were the presence of a dog, signs of mice, a concrete floor in the living room, and mold or mildew in the bedroom during the past year. Dehumidifiers were associated with reduced airborne endotoxin. However, airborne endotoxin levels in homes were similar to those in outdoor air. The particle size distribution of endotoxin in ambient and indoor air is not known. Inhaling typical home air, containing one endotoxin unit (EU) per cubic meter, for 24 hours (a total daily dose of approximately 1 EU in a small child and 10 EU in an adult) may be the source of the low levels of endotoxin present in bronchoalveolar lavage fluid (0.1 ng/ml) (Dubin et al., 1996).

In addition to endotoxin exposure from environmental sources, endogenous endotoxin exposure may arise from infection. It has been suggested that a strong association between periodontal disease and premature birth may be caused by endotoxin exposure from gram-negative bacteria present in subclinical infections (Damare et al., 1997). While periodontal disease is not likely a source of endotoxin exposure in children, other infectious agents common in children, such as Hemophilus sp., may be important sources of exposure, as may commensal coliforms.

Evidence Regarding Asthma Exacerbation and Development

Biologic Evidence

It has long been recognized that endotoxin is a potent stimulus for macrophage production of (TNFα), IL-1, and a variety of other cytokines; arachidonic and linoleic acid metabolites, and reactive oxygen species (Rietschel and Brade, 1992). The receptors responsible for binding endotoxin and triggering responses are still in the process of being described. The pathway for endotoxin binding and cell activation includes an opsonin, LPS binding protein (LBP), which presents LPS to CD14 (Wright et al., 1990). After binding LPS–LBP complexes, membrane-bound CD14, attached to myeloid cells via a glycosylphosphatidyl–inositol (GPI) anchor, activates myeloid cells and soluble CD14 activates nonmyeloid (endothelial or epithelial) cells (Pugin et al., 1993). The signal is apparently transduced via the human toll-like receptors TLR4 and TLR2 in myeloid and nonmyeloid cells, respectively (Chow et al., 1999; Kirschning et al., 1998; Ulevitch, 1999; Wright, 1999; Yang et al., 1998). There may also be a role in signaling for heterotrimeric G proteins associated with the GPI anchors for CD14 (Solomon et al., 1998).

CD14 and the toll-like receptors are pattern receptors and are also involved in the recognition of peptidoglycan and lipoteichoic acid, and other microbial products (Cleveland et al., 1996; Dziarski et al., 1998; Schwandner et al., 1999). However, opsonization by LBP appears to confer approximately a hundredfold greater sensitivity to LPS compared with peptidoglycan or lipoteichoic acid (Kirschning et al., 1998; Schwandner et al., 1999; Sugawara et al., 1999).

The monocyte lineage, including macrophages and dendritic cells, has abundant membrane-bound CD14 and exhibits sensitive and prolific responses to endotoxin. Cytokines produced include IL-1, 6, 8, 10, 12; TNFα, INF-β, TGF-β; MIP-1α; CSF-1; and GM-CSF. Expression of MHC-II and B7.1 are also upregulated (Medzhitov et al., 1997; Santiago-Schwarz et al., 1989; Weatherstone and Rich, 1989). LPS also is a B cell mitogen and promotes isotype switching from IgM to IgE in the presence of IL4 (Snapper et al., 1991). Thus, endotoxin not only serves as a potent stimulus to innate immune responses but also serves as a stimulus and bridge to cognitive immunity.

The gene for CD14 maps to chromosome 5q31.1, a candidate region for loci regulating IgE expression. Baldini and colleagues (1999) observed that a C to T transposition in the promoter region of CD14 (bp-159) was associated, in TT homozygotes, with significantly higher sCD14 levels. Among white children with positive skin tests to local antigens, TT homozygotes had lower total IgE levels. Among those with any positive skin test, the TT homozygotes had significantly fewer positive tests.

Much experimentation suggests that the net effect of endotoxin exposure is to promote TH1-type immune responses such as those typically seen in bacterial infections (Baldini et al., 1999; Fearon and Locksley, 1996) (see also Chapter 4). However, endotoxin is noted for the peculiar patterns of response to repeated exposures known as the Schwartzman phenomenon and endotoxin tolerance, for which cellular mechanisms have been described (Berg et al., 1995; Bohuslav et al., 1998). It is clear from mechanistic studies of repeated exposure that endotoxin stimulation of production of IL-10, which suppressed the TH1 response, is at least as important a phenomenon as stimulation of IL-12, which enhances this response. Furthermore, much of the data indicating that endotoxin promotes TH1 responses, except Baldini et al. (1999), come from single-exposure in vitro studies. Human data on TH1/TH2 responses to endotoxin and studies of repeated experimental exposure in vivo are sparse. Two studies suggest that under at least certain circumstances, endotoxin promotes TH2 immune responses. A study of human volunteers injected with 4 ng/kg of endotoxin (Zimmer et al., 1996) demonstrated that IL-12 levels were unchanged following injection but IL-10 levels increased, suggesting that systemic endotoxin exposure promoted a TH2 immune response. A mouse model for periodontal disease, produced by repeatedly injecting LPS at 48-hour intervals, found that after the first several injections, gingival T cells were primarily of the TH1 subgroup, based on in situ hybridization. However, after 10 injections, T cell subgroups changed from a TH1 to a TH2 predominance (Iwasaki et al., 1998). Thus, chronic exposure may produce qualitatively different responses from a single acute exposure. The route of exposure as well as dose rate may be important factors, so that the net effect on development of immune responses following chronic low-level, airborne endotoxin exposure may be difficult to predict.

Epidemiologic Evidence

Several laboratory and cross-sectional epidemiologic studies suggest that asthmatics may be sensitive to the proinflammatory effects of endotoxin at lower levels of exposure than nonasthmatics. After endotoxin exposure, increased bronchial responsiveness to histamine challenge was observed among asthmatics, but not among nonasthmatic adults (Michel et al., 1989). In a cross-sectional study of asthmatic adults, the endotoxin content of house dust was associated with increased asthma severity (Michel et al., 1996).

High levels of endotoxin in agriculture and industry are associated with both acute and chronic effects on respiratory symptoms and lung function, regardless of preexisting lung disease. The increasing body of epidemiologic evidence for respiratory effects of occupational exposure to endotoxin at 10 or more times the ambient outdoor levels has been reviewed by Douwes and Heederik (1997) and Milton (1999). One occupational study suggested that the chronic airways disease associated with daily exposure to high levels of endotoxin is characterized by an increased peak flow amplitude and thus may represent a form of asthma (Milton et al., 1996). However, except for the use of contaminated humidifiers, most homes have airborne endotoxin levels similar to those found in ambient urban and suburban air. Thus, it is not clear what effect if any exposure at these levels can have on persons who do not already possess inflamed airways with increased LBP and sCD14.

If endotoxin influences the development of the asthmatic inflammatory response, it is unclear whether the influence will be as a risk factor or a protective factor; basic laboratory research (see above) suggests that either is possible. Both German and Swiss studies suggest associations between living on a farm and decreased risk of asthma (Braun-Fahrlander, 1999; von Mutius, 1994). The authors suggest that exposure to endotoxin early in life may be protective against asthma development and that there may be a gene-by-environment interaction in creating tolerance. At this time there are no data on home endotoxin exposure and the risk of asthma development in children.

Conclusions: Asthma Exacerbation and Development

Endotoxin is associated with occupational lung disease among workers exposed to high levels. Although endotoxin is suspected at lower levels to be a trigger for asthma, and may be either a disease promoter or a beneficial exposure depending on time course, dose, and route of exposure, there are too few data on low-level exposures to draw any firm conclusions at this time. Therefore, the committee concludes:

  • There is inadequate or insufficient information to determine whether or not an association exists between low-level indoor endotoxin exposure and asthma exacerbation or development.

Evidence Regarding Exposure Mitigation and Prevention

There are few data on sources in the home environment and none on the effects of interventions aimed at altering domestic endotoxin exposure. However, experimental data do exist to demonstrate that cool mist (spinning disk and ultrasonic) humidifiers can emit very high levels of endotoxin aerosol, that filtration of the mist is not effective, and that warm mist or steam humidifiers do not emit aerosols of endotoxin or other pathogenic organisms (Tyndall et al., 1995). Attempts to prevent microbial contamination of cool mist humidifiers with antifouling agents have not been successful to date (Burge et al., 1980). Thus, prevention of high-level endotoxin exposure from humidifiers would appear to be best accomplished by elimination of cool mist units. However, the impact of these units on the severity and occurrence of asthma is not known. In summary:

  • There is inadequate or insufficient evidence to determine whether or not an association exists between endotoxin interventions and reduction of endotoxin levels.

Conclusions: Exposure Mitigation and Prevention

No general conclusions about the means of altering exposure to low levels of endotoxin can be drawn at the present time. However, avoiding the use of cool mist humidifiers would appear to be a simple and effective means of eliminating the risk of high-level exposure to endotoxin at home as well as to organisms associated with hypersensitivity pneumonitis (Burke et al., 1977; Ganier et al., 1980; Seabury et al., 1976; Suda et al., 1995).

Research Needs

Given the significant body of data on the exquisite sensitivity of the innate immune system to small quantities of endotoxin, the hypotheses that domestic endotoxin exposure may influence the development of the immature immune system or affect the severity of asthma warrant further investigation.

This review suggests several avenues of research directed at understanding the role of endotoxin exposure and endotoxin susceptibility in the pathogenesis of asthma. These include studies of gene–environment interactions and the risk of developing atopy or asthma, preferably with prospective assessment of endotoxin exposure from birth, improved endotoxin exposure assessment across populations likely to have significant differences in exposure, and studies of endotoxin exposure and asthma severity.

Gene–environment interactions between the CD14 polymorphism and endotoxin exposure should take into account that CD14 is a pattern receptor and thus not specific for LPS–LBP complexes. Thus, future studies should include an assessment of exposure to other bacterial products that stimulate innate immunity via CD14 such as peptidoglycan. Prospective studies will be required to determine whether endotoxin exposure early in life plays a role in determining the direction of immune system development. Studies that can compare populations with possibly larger variations in exposure to endotoxin and other components of organic dusts than can be found within an urban or suburban area would likely have increased power to detect the effects of endotoxin exposure. Because the CD14 polymorphism is associated with atopy, a focus on specific and nonspecific IgE and TH phenotypes will likely be the most important variables for these studies.

Given that the Limulus bioassay has limitations and that “unusual” lipid A structures dominate the composition of house and other organic dusts, additional exposure assessment methods that can detect the range of environmental LPS should be employed along with Limulus assays in future studies. The possibility that endogenous sources of endotoxin exposure may be important in modulating the level of tolerance to environmental exposure (or vice versa) should also be examined.

FUNGI

Definition of Agent and Means of Exposure

Introduction

There are more than 1,000,000 species of fungi, 200 different types to which people are routinely exposed. Exposure occurs universally, both outdoors and indoors, and is impossible to avoid completely. Fungal exposure (even to one type of fungus) is complex with respect to disease agents and usually includes allergens, irritants, toxins, and sometimes potentially infectious units.

These factors have led to confusion, poorly constructed studies on the role of fungi especially in the area of allergic disease, inconclusive results, and avoidance of the field by the best investigators. However, clearly and unequivocally, fungal exposure does cause allergic, toxic, and infectious disease, and it remains only to document the extent of the problem, factors leading to disease, and approaches for control.

Fungi may play a role in asthma in several ways. The most obvious of these is via fungal allergen exposure that leads to sensitization, perhaps leads to the development of asthma, and exacerbates symptoms in sensitized people. Fungi also contain and release irritants that may enhance the potential for sensitization, potentiate allergen-induced symptoms, and (possibly) exacerbate asthma in nonsensitized people. Finally, fungal toxins could play a role in modulating the immune response and may cause direct lung damage leading to pulmonary diseases other than asthma.

Definition of Agents

Nature of Fungi Fungi are eukaryotic organisms characterized primarily by their filamentous morphology and saprobic life-style. Fungal cells are bounded by rigid cell walls that are usually composed of chitin fibrils embedded in a matrix of (1→3) β-D-glucans and/or mannans. Cell walls may be coated externally with waxes or hydrophilic extracellular polysaccharides that carry various levels of antigenic specificity. To digest food, fungi excrete enzymes into the environment initially as probes to evaluate food availability, then to digest complex carbon compounds. These enzymes are some of the major fungal allergens. While processing organic material, the fungi produce many ancillary metabolites, some of which are highly toxic (antibiotics, mycotoxins). These compounds may accumulate in the fungal body, in spores, and in the environment. The primary mode of fungal reproduction is by airborne spores, which form a major fraction of both the outdoor and the indoor large-particle aerosol. Fungi also colonize manmade environments, releasing both spores and metabolic materials.

Fungal Allergens Fungi produce an enormous array of compounds that are potentially allergenic. Each fungus produces many different allergens of a range of potency. Table 5-2 lists the major defined allergens isolated from fungi. Others have been identified, but they are generally “minor” (i.e., few patients react to them). Many others remain to be identified.

TABLE 5-2. Major Defined Allergens Isolated from Fungi.

TABLE 5-2

Major Defined Allergens Isolated from Fungi.

Sources and Variability Fungal allergen production varies by isolate (strain), species, and genera (Burge et al., 1989). Different allergen amounts and profiles are contained within spores, mycelium, and culture medium (Cruz et al., 1997; Fadel et al., 1992). In addition, substrate (growth) medium strongly influences the amount and patterns of allergen production. For example, the allergen content of Alternaria spores produced on ceiling tiles probably differs from that of spores produced on dead grass. Fungi release proteases during germination and growth, and fungal extracts contain sufficient protease to denature other allergens in mixtures. This has been demonstrated clearly for Alternaria extracts (Nelson HS et al., 1996).

Cross-Reactivity Patterns Patterns of cross-reactivity among fungal allergens have been examined using in vitro methods in which inhibition of heterologous assays is assumed to indicate cross-reactivity (although nonspecific assay inhibition must be ruled out). RAST is the most often used assay (O'Neil et al., 1990), although allergens derived from many genera of fungi have also been shown to cross-react using IgE immunoblot techniques (Verma et al., 1995). In addition, cross-reactivity has also been inferred from patterns of skin reactivity to fungal extracts. Although this latter case could result from cross-reactivity, it could also happen with multiple exposures leading to (separate) multiple sensitizations (O'Neil et al., 1990). Unfortunately, data on fungal allergen cross-reactivity are inconsistent and appear to depend on the specific strains used and on methods of allergen extraction.

Other Fungal Agents Exposure to some kinds of fungal spores induces inflammatory changes in the lung independent of allergy or sensitization (Rao, 1999; Shahan et al., 1998). MIP-2, lactase dehydrogenase, and myeloperoxidase are released in response to fungal exposure, and blood cell patterns change. These effects are dependent on the type of fungus and on the concentration of spores administered. The role these processes might play in the development or exacerbation of asthma remains unknown.

Glucans Fungal cell walls are composed of acetylglucosamine polymer (chitin) fibrils embedded in a matrix of glucose polymers ((1→3) β-D-glucans). The glucans may be chemically bound to the chitin or may form a soluble matrix (Sietsma and Wessels, 1981). Potent T cell adjuvants, the (1→3) β-D-glucans have been investigated as antitumor agents (Kiho et al., 1991; Kitamura et al., 1994; Kraus and Franz, 1991). They increase resistance to gram-negative bacterial infection by stimulating macrophages and affecting the release of TNFα mediated by endotoxin (Adachi et al., 1994a, 1994b; Brattgjerd et al., 1994; Saito et al., 1992; Sakurai et al., 1994; Zhang and Petty, 1994). Soluble glucans have an effect in the lung similar to that of endotoxin (Fogelmark et al., 1994). Glucans may be involved in the development of fungal-induced hypersensitivity pneumonitis by affecting the inflammation-regulating capacity of airway macrophages. They also probably play a role in organic dust toxic syndrome in workers exposed to dust that includes high concentrations of fungal spores. The possible role of glucans in the development and/or exacerbation of asthma has not been studied.

Mycotoxins Most mycotoxins are cytotoxic and interfere with protein synthesis, causing cell lysis and death. Some mycotoxins are potent carcinogens, and a few affect cell division (cytochalasins) or are estrogenic (zearalenone) or vasoactive (ergot alkaloids). Some cross the blood–brain barrier and affect the central nervous system. Some mycotoxins selectively kill macrophages (Gerberick et al., 1984; Jakab et al., 1994; Nikulin et al., 1996, 1997; Richard and Thurston, 1975; Sorenson and Simpson, 1986; Sorenson et al., 1985, 1986). A toxin produced by Aspergillus fumigatus inhibits macrophage functioning and may play a role in allergic bronchopulmonary aspergillosis (ABPA) by facilitating colonization of the airways of asthmatics (Amitani et al., 1995; Murayama et al., 1996). Gliotoxin, another A. fumigatus toxin, causes fragmentation of DNA, especially in thymocytes, and may facilitate tissue invasion leading to aspergillosis in immunosuppressed patients (Sutton et al., 1996; Waring and Beaver, 1996; Waring et al., 1997). Some fungal components directly cause the release of mediators of inflammation, including cytokines, reactive oxygen metabolites, and chemotactic factors (Shahan et al., 1998). The role of mycotoxins in the development and exacerbation of asthma has not been studied.

Means of Exposure

Measurement of Fungal Exposure Visual observation is the most frequently used approach to estimate potential for fungal exposure, with observational data often obtained from occupant questionnaires (e.g., Brunekreef et al., 1989). Observational data are limited by the fact that fungi are microscopic and are not visible until growth is extensive. Culture of air or dust samples is also often used indoors (e.g., ACGIH, 1999; Burge and Solomon, 1987; Su et al., 1992; Verhoeff et al., 1992). Since the presence of allergens does not always depend on culturability, this type of measure is likely to underestimate actual allergen exposure, as well as (possibly) confounding results with high levels of nonallergenic types. Microscopic identification and counting of spores from air samples (e.g., Delfino et al., 1997) represent the usual approach for outdoor samples and constitute the only currently available method that assesses nonculturable spores. The method is limited by the relatively few species that can be identified and the time commitment required for analysis. Although a few assays are available, no epidemiological studies have emerged that use fungal allergen measures of exposure. Other methods have been proposed that evaluate total fungal biomass (ergosterol, glucan assays) or that identify the presence of specific fungi (PCR techniques). None of these has been used in studies of asthma.

Factors Influencing Exposure

Nature of the Particles Spore walls may be hydrophobic with a waxy outer coat, or hydrophilic with an outer surface of water-soluble polysaccharides. Most fungi release single spores that range in size from 2 to 10 µm. Some spores can exceed 100 µm in length, although aerodynamic diameters are usually much smaller. Some spores are released as chains or clumps. Cladosporium spores are frequently encountered as large branching chains, as well as individual spores, so that particle sizes of Cladosporium units can range from near 1 µm to many hundreds of micrometers in diameter. However, air sampling with particle size-separating cascade impactors indicates that most fungal spores are <5 µm in aerodynamic diameter. Fungal hyphae also become airborne with disturbances such as high winds. Studies are not available that document the natural presence (or absence) of fungal allergens on particles other than intact fungal spores.

Studies of Fungal Aerosol Release Fungal aerosols may be produced through intrinsic spore discharge mechanisms or mechanical agitation (Ingold, 1971). Most indoor release relies on active disturbance. Few studies document actual types and intensities of mechanical disturbance necessary to release spores from surface growth. At least for some types, strong agitation or even direct abrasion is necessary (e.g., Stachybotrys chartarum), whereas for others, air movement such as that produced by a fan may be sufficient (Madelin and Johnson, 1992).

Outdoor Exposure to Fungi Many studies document the almost continuous presence of fungal spores in outdoor air, and the factors affecting prevalence for different types (e.g., AAAAI, 1998; Cross, 1997; Li and Kendrick, 1995; Munuera et al., 1998; Takahashi, 1997). Fungal spores are always present in outdoor air, although levels can be very low during periods of snow cover (Cross, 1997). Patterns of prevalence depend on seasonal and climatic factors, geography, and to some extent, human activity, especially that associated with agriculture. Because fungi are continuously present outdoors, often in concentrations far higher than those indoors, it is difficult to rule out outdoor exposure as the primary determinant of sensitization and symptoms. It is also difficult to document the presence of indoor growth using air sampling.

How Does the Indoor Environment Affect or Influence Exposure?

Penetration of Outdoor Aerosols Fungal particles probably penetrate indoor environments following the same physical principles as other types of particles. Published studies that compare indoor–outdoor relationships for fungi have not clearly addressed the relative contribution of the outdoor aerosol. Although outdoor fungal spores readily enter through open windows, few penetrate into closed environments (e.g., buildings and automobiles) (Muilenberg et al., 1991; Solomon et al., 1980).

Indoor Sources Fungi are always present in dust and on surfaces. Fungal growth occurs only in the presence of moisture. Food materials and temperature affect the amount of water required, as does the strain of fungus. Several reports present data on laboratory conditions that lead to fungal growth on building materials (Chang et al., 1996; Grant et al., 1989). With relatively concentrated inocula, some xerophilic fungi increase at relatively low substrate water activities. However, extensive growth was reported only at humidities near 100%, and most fungi require very wet conditions (near saturation), lasting for many days, to extensively colonize an environment. Human habits may affect the numbers and types of active fungal sources in buildings. Poorly maintained water-based appliances (e.g., humidifiers, vaporizers) harbor and release fungi. Wood stored indoors contains many fungi, although exposure from this source has not been studied. Moldy food is an obvious source, and indoor recoveries from air may be dominated by these small sources if recently disturbed.

Evidence Regarding Asthma Exacerbation and Development

Sensitization to Fungal Allergens

Exposure to fungi clearly plays a role in asthma. Good-quality studies have been reported that document the sensitizing potential of fungal allergens and relate fungal sensitization to the existence of asthma. Challenge studies have documented asthmatic responses in sensitized patients, and several studies have begun to make the connection between natural exposure and symptom development.

Prevalence of Sensitization

It has been estimated that about 6–10% of the population and 15–50% of atopics are sensitized to fungal allergens (Table 5-3). In studies where Alternaria extracts were used alone (i.e., not in a mixture), overall rates for allergy patients range from 3 to 36% and for asthmatics, 7 to 39%.

TABLE 5-3. Immediate Skin Reactivity to Fungi.

TABLE 5-3

Immediate Skin Reactivity to Fungi.

Skin test surveys most often focus on broad panels of allergens, with fungi included either as a single representative extract (usually Alternaria), two or three extracts, or mixtures of several different fungi. However, Galant et al. (1998), in a survey of California allergy patients, revealed that nine different fungal extracts were necessary to detect 90% of mold allergy patients. Interestingly, most studies that focus specifically on fungal sensitivity also use only a few extracts, with Alternaria again being the dominant type. One study (Szantho et al., 1992) suggests that the prevalence of sensitivity to fungi is increasing and attributes the increase to an increase in concentrations of outdoor fungi due to growth on pollution-weakened plants. However, no monitoring data support this hypothesis. Fungal skin sensitivity rates increase with age (Erel et al., 1998). Production of IgG antibodies as a result of allergen exposure may block the skin test response even in patients clearly experiencing symptoms with exposure (Witteman et al., 1996). Fungal allergens can produce a strong IgG response, possibly making reported incidences of skin reactivity underestimates.

Fungal Sensitization and Asthma

In a population of adults in Sweden, sensitivity to Cladosporium or Alternaria (but not dust mites) was associated with a current asthma odds ratio (OR) of 3.4 (1.4–8.5) (Norbäck et al., 1999). In an Arizona population, responses to A. alternata were the most frequent among asthmatic children, and a positive Alternaria skin test was the only independent association with asthma (Halonen et al., 1997). In a University of Virginia study, Alternaria sensitivity was also a significant independent risk factor for asthma in school children in Charlottesville, Virginia, and Los Alamos, New Mexico, but not in Albemarle County, Virginia (Perzanowski et al., 1998). Of the 1,218 children born on the Isle of Wight, 6% of the 918 4-year-old children tested had positive skin tests to Alternaria and/or Cladosporium. In these 61 children, a positive test to Alternaria was associated with a diagnosis of asthma (Tariq et al., 1996). In Costa Rica, where the prevalence of childhood asthma is 20–30%, dust mite, cat, Alternaria, and Cladosporium-specific IgEs predict asthma (Soto-Quiros et al., 1998). A case-control study in Denver (acute asthmatics 3–16 years old) versus matched controls used RAST to document specific IgE levels: 45% of asthmatics versus 4% of controls had high IgE to Alternaria (Nelson RP et al., 1996). In a large (6,394 children) questionnaire-based study, Peat et al. (1995) reveal that among asthmatic children, Alternaria sensitivity rates are higher inland in New South Wales, but in the damp coastal climate, sensitivity to dust mites is most prevalent. In 4,295 6- to 25-year-olds in the second National Health and Nutrition Examination Survey (NHANES II) cohort, asthma was associated with sensitivity to dust mite (OR = 2.9 [1.7–5]); and Alternaria (OR = 5.1 [2.9–8.9]) (Gergen and Turkeltaub, 1992). Retrospective investigation of asthma deaths in teenagers revealed that a large majority had positive skin tests to Alternaria, and the authors concluded that sensitivity to Alternaria is a risk for severe asthma and death from an attack (O'Hollaren et al., 1991). (The authors concluded that exposure to Alternaria is a risk factor, although they did not measure exposure.) Many studies focusing on asthma ignore fungi completely (e.g., Gottlieb et al., 1996).

Immunotherapy Data

Further evidence for the role of fungal sensitization in symptomatic asthma is provided in immunotherapy data. Bousquet and Michel (1994) have reviewed the overall literature on immunotherapy. The literature on mold immunotherapy has been reviewed by Dhillon (1991) and Bonifazi (1994). Immunotherapy with partially characterized and standardized A. alternata allergens in a small case-control study has been shown to decrease asthma symptoms overall and during challenge with relevant allergens (Cantani et al., 1988; Horst et al., 1990). In both of these studies, children who were sensitive only to Alternaria (by skin prick test) were treated. Dreborg et al. (1986) selected 30 children who were skin test, RAST, and provocation positive to Cladosporium. Most had other sensitivities as well. He treated 16 of these children; the remaining 14 served as controls. Although symptom scores remained similar for the two groups (probably because of multiple sensitivities), nasal and bronchial challenge responses and medication usage were significantly lower in the treated versus the control group.

Of the 16 children treated in the Dreborg (1986) study, 13 experienced general reactions during treatment. Kaad and Ostergaard (1982) report that 19% of 38 children treated with fungal extracts had to be withdrawn from immunotherapy because of apparent type III reactions. All had slightly elevated IgG (precipitating antibodies) to the relevant extracts before hyposensitization was begun and developed increased titers during therapy.

Overall, carefully controlled studies of mold immunotherapy show good effect. Negative studies are small, with too few patients and nonstandardized allergens. These data support the notion that there is a relationship between fungal allergen sensitization and symptoms of asthma. However, there are too few studies to confirm that mold immunotherapy is an effective public health intervention.

Fungal Exposure and Asthma

Challenge Experiments Challenge tests with Stemphylium in children with positive skin and RAST tests to Stemphylium extracts resulted in bronchial responses in 13 of 59 children (12 others had nasal responses) (Lelong et al., 1986). Malling (1986) used positive bronchial challenge in adult asthmatics with Cladosporium as a patient selection criterion for his studies on relationships between natural exposure and symptoms. Licorish et al. (1985) reproduced symptoms of asthma with Penicillium spore challenges.

Measured Natural Exposure

Outdoors Neas and colleagues (1996) associated pulmonary function changes in Pennsylvania school children with measured outdoor concentrations of several kinds of fungal spores. Peak flows, symptoms and time spent outdoors were recorded daily, and 24-hour average outdoor spore concentrations were measured using a Burkard spore trap. Changes in morning peak flow of –1 liter/min (CI –1.9 to 0.2) were associated with increments of 10,000 Cladosporium spores in unselected school children. Likewise, increases of 50 Epicoccum spores was associated with decrements in morning peak flows of 1.5 liter/min (CI –2.8 to –0.2). Epicoccum exposure was also associated with incidence of morning cough (OR = 1.8, CI = 1.0–3.2). Delfino et al. (1997) report associations between asthma severity (symptom scores and peak expiratory flow rates) and exposure to outdoor fungal spores measured with a spore trap. Symptoms were associated with total fungal spore increases of 4,000 spores per cubic meter, with a decrease in evening peak flow of 12 L per minute. Analysis of specific fungal genera increased the associations. The most important fungal correlates were Alternaria, basidiospores, and hyphal fragments. Malling (1986) related daily symptom scores to Cladosporium counts during the autumn mold season. He studied 24 adult asthmatics with positive provocation tests to Cladosporium extracts. Significant associations with spore counts were found for symptom scores and medication use. Although some patients were also sensitive to Alternaria, Alternaria counts did not relate to health outcomes in this population.

Indoors Su et al. (1992) describe relationships between indoor measured levels of culturable Aspergillus and asthma in Topeka, Kansas schoolchildren. They used culture plate impactors to collect short “grab” samples in homes of children recruited through the public schools. The relationship between Aspergillus and asthma symptoms was non-linear, possibly reflecting the different species of Aspergillus that are common in indoor environments. Garrett et al. (1998) report associations between measured concentrations of fungal spores (counted) and culturable fungi, observed indicators of dampness and molds, fungal sensitivity, and respiratory symptoms. Eighty households with 148 children (36% asthmatic) were sampled 6 times for airborne fungi. Penicillium exposure was associated with asthma, and Aspergillus exposure with atopy. Cladosporium and Penicillium exposure were associated with the presence of fungal allergy. No associations were seen for total fungal counts. Exposure measures to specific fungi showed a stronger association with symptoms than did dampness indicators.

Other studies of indoor fungi and asthma report comparisons between fungal levels in homes of asthmatics compared to homes of control subjects. Li and Hsu (1997) compared culturable fungi in homes of 46 asthmatic children, 20 atopic (presumably nonasthmatic) children, and 26 nonatopic controls. Although fungal concentrations were highest in the asthmatic and control homes, Cladosporium concentrations were higher in asthma homes than in control homes. In a study by Horak et al. (1996), Bacillus, Aspergillus, and total fungal concentrations were higher in homes of asthmatics than in control homes. These kinds of studies are compromised by the fact that families of asthmatic children often take measures to reduce exposure to allergens.

Visible Mold, Odors, and Dampness

The majority of studies that attempt to associate fungal exposure and asthma rely on reports of visible mold or other dampness indicators. Dampness is clearly associated with a variety of respiratory symptoms.

In addition, Williamson et al. (1997) report a correlation between mold growth indicators and asthma (196 age- and sex-matched subjects); r = 0.23, p < .035). These researchers also identified a relationship between severity of asthma and total dampness (r = 0.3, p = .006), and mold growth (r = 0.23, p = .35). A reduction in FEV1 of 10.6% (CI 1.0–20.3) was associated with living in a damp home.

Dampness and even visible mold growth could be indicators for dust mite allergen exposure as well as fungal exposure. However, Garrett et al. (1998) report that homes with musty odor, water intrusion, high humidity, limited ventilation, and visible mold growth had higher fungal spore concentrations than dry homes. Visible mold was associated with high concentrations of Cladosporium spores, but not total spores. Pasanen et al. (1992) report that airborne Cladosporium and yeast counts were significantly higher in damp than in dry residences. Cladosporium, in particular, appeared to be derived from indoor sources. Nicolai et al. (1998) report that the relationship between dampness and bronchial hyperreactivity in adolescents in Munich remains when controlled for dust mite allergen exposure, providing indirect evidence for a possible role of fungi.

TABLE 5-4Dampness and Mold Indicators and Respiratory Symptoms

ReferencePopulationEnvironmental IndicatorSymptomOR (95% CI)
Jaakkola et al., 19932,568 childrenMold odorAny respiratory symptom2.54–8.67
Any dampness or moldWheezing2.62 (1.39–4.39)
Phlegm2.20 (1.27–3.82)
Persistent cough2.17 (1.39–3.39)
Slezak et al., 19981,085 head start childrenSelf-reported dampness or moldSelf-reported diagnosed asthma1.94 (1.23–3.04)
Hu et al., 19972,041 young adultsSelf-reported mold growthAsthma prevalence2.0 (1.2–3.2)
Brunekreef et al., 19894,625 childrenReported moldsSymptoms and pulmonary function1.27–2.12
FEF 25-75–1.6%
Strachan, 1988873 childrenVisible moldWheeze in past year3.0 (1.72–5.25)
Nicolai et al., 1998155 children with asthmaDampnessBronchial hyperreactivity in adolescence16.14 (3.53–73.71)
Andriessen et al., 19981,614 childrenSelf-reported moldsPEF variability1.92 (1.18–3.12) current home
Williamson et al., 1997102 asthmatics,Self-reported dampnessAsthma2.11 (1.29–3.47), past home
196 matched controlsObserved dampness3.03 (1.65–5.57)

NOTE: CI = confidence interval; FEF = forced expiratory flow; OR = odds ratio; and PEF = peak expiratory flow.

Conclusions: Asthma Exacerbation and Development

The extent of fungal-related asthma remains unknown, and the exposure parameters leading to fungal-related asthma development or exacerbation are not clear. This is due (in part) to inadequate diagnostic and environmental testing procedures and lack of adequate study. Sensitization (i.e., positive skin test) to fungal allergens is associated with the presence of asthma in both children and adults. Although the evidence regarding asthma development is provocative, it is inclusive. In summary:

  • There is sufficient evidence of an association between fungal exposure and symptom exacerbation in sensitized asthmatics. Exposure may also be related to nonspecific chest symptoms.
  • There is inadequate or insufficient evidence to determine whether or not there is an association between fungal exposure and the development of asthma.

Evidence and Conclusions: Exposure Mitigation and Prevention

As with any relatively large-particle aerosol, fungal spores can be prevented from entering enclosed spaces. However, the health effects of such prevention have not been studied. Fungi are difficult to kill, and dead fungal material probably contains allergens, although this has not been thoroughly tested. Terleckyj and Axler (1987) discuss the effects of various disinfectants on fungi. Chlorine dioxide, glutaraldehyde, and ethyl alcohol affected most fungi after 15 minutes of contact time. However, Aspergillus fumigatus was highly resistant to most disinfectants, and a quaternary ammonium compound and an iodophor were the least effective for all fungi.

It is possible to physically remove active fungal growth from indoor environments. While such removal is undoubtedly advisable, its health impact has not been studied. Rautiala et al. (1998) studied spore levels during removal of fungal-contaminated material. They found that negatively pressurized containment is necessary to prevent the spread of spores to unaffected parts of the building and that, within the containment field, spore levels may remain high and workers should wear protection. Overall, mitigation and prevention of fungal growth in the indoor environment has not been studied, either from the viewpoint of effectiveness or with respect to health impact. In summary:

  • There is limited or suggestive evidence of an association between the fungal removal measures described above and a reduction in the levels of fungi in the indoor environment. The paucity of studies with adequate exposure characterization and the possibility that cleaning may not eliminate the problematic component of fungal exposures prevent a more confident conclusion from being drawn.
  • There is inadequate or insufficient evidence to determine whether or not an association exists between fungal control measures and improvement in symptoms or lung function in sensitized asthmatics.

Research Needs

Few fungal allergens have been identified, and patterns of cross-reactivity among fungal allergens have not been documented. Standardized methods for assessing exposure to fungal allergens are essential, preferably based on measurement of allergens rather than culturable or countable fungi. Acquisition of these data is a necessary step before adequate estimates of the role of fungal allergen in asthma can be documented.

Studies seeking to find environmental factors that either lead to the development of asthma or precipitate symptoms in existing asthmatics must include good measures of fungal exposure. No studies have attempted to control exposure to fungal allergens either indoors or out. Intervention studies that seek to control indoor exposure to fungi are especially needed.

INFECTIOUS AGENTS

Infectious agents are unlike other indoor exposures addressed in this report because their source is the occupants themselves. They are nonetheless an appropriate exposure to address because they contribute to the overall risk of asthma disease outcomes from indoor exposures and may have some role in the observed increase in asthma incidence and mortality.

There are a number of infectious agents that have been associated with asthma (Johnston et al., 1995; Pattemore et al., 1992; Shaheen, 1995). This section focuses on four that have received particular attention from researchers: two viral agents—rhinovirus and respiratory syncytial virus—and two bacterial agents— chlamydia and mycoplasma.

Rhinovirus

Definition of the Agent and Means of Exposure

Rhinovirus is the medical term used to designate a large group of viruses responsible for a variety of respiratory infections including the common cold. There are more than 120 serotypes of the rhinovirus, of which few have been associated with lower-respiratory abnormality. Rhinovirus is transmitted through the respiratory route, with virus particles shed in nasal secretions and spread by coughing and sneezing. It can also be transmitted through direct contact with tissues or hands.

Evidence Regarding Asthma Exacerbation and Development

Widespread clinical experience and epidemiologic research indicate that rhinovirus is associated with wheezing and exacerbations of asthma in established asthmatics (Johnston, 1997; Johnston et al., 1995; Lemanske et al., 1989; Micillo et al., 1998; Rakes et al., 1999). The evidence of this is especially strong for children. The mechanism by which the virus traffics from the upper airway to the lower airway to cause asthma symptoms is not known. It is speculated that individuals with asthma may make more of a specific cytokine than those without asthma, leading to increased airway inflammation and the development of symptoms. Alternatively, individuals with asthma and those without asthma may make the same amount of cytokines in response to viral infections, but these cytokines may react differently in individuals with asthma and produce different effects in the airway. Individuals with asthma have a different cytokine profile from those without asthma, which may result in a different clinical phenotype.

Studies have not identified an association between rhinovirus infection and asthma development. Martinez (1995) observes that most infants who wheeze during viral infections become symptom free later in life and suggests that viral infections in infants are likely to play a minor role in the subsequent development of asthma. There is burgeoning research interest in this issue, however, with investigators suggesting both the possibility of a mechanistic association and the prospect that certain viruses may have a protective effect (Grunberg and Sterk, 1999; Martinez, 1994).

Respiratory Syncytial Virus

Definition of the Agent and Means of Exposure

Respiratory Syncytial Virus (RSV) belongs to the Paramyxoviridae family and to the genus Pneumovirus. Human paramyxoviruses are a common cause of respiratory disease in children, with RSV particularly important in infants. RSV has been found in every geographical area examined for evidence of infection (Glezen et al., 1981; Hall et al., 1979), and RSV infection appears to have similar characteristics in areas with widely differing climates. Outbreaks of infection occur yearly, typically in the spring and winter. Indeed, RSV is the only viral respiratory agent that can be relied on to produce a sizable crop of infection each year. Socioeconomic status and race or ethnicity appear to influence the risk of RSV bronchiolitis (Glezen et al., 1981).

Evidence Regarding Asthma Exacerbation and Development

The study of RSV and asthma exacerbations is complicated by the problematic diagnosis of asthma in infants and young children. However, viruses in general have been identified as responsible for most wheezing illnesses and asthma exacerbations occurring in childhood, with RSV among the predominant organisms recognized (Hegele, 1999; Pattemore et al., 1992). An association has also been noted for adults (Nicholson et al., 1993).

Significant attention has been paid to the possibility of an association between RSV bronchiolitis in infancy and later development of asthma. RSV bronchiolitis is associated with the development of high titers of virus-specific IgE in respiratory secretions during both the acute and the convalescent stages of illness in some patients (Robinson et al., 1993; Welliver and Duffy, 1993). An exaggerated RSV IgE response at the time of RSV bronchiolitis in infancy is associated with recurrent wheezing in later childhood (Scott et al., 1984). Various mechanisms may be proposed to explain this association. These include direct induction of release of inflammatory mediators from pulmonary mast cells, participation in a cascade of mediators of airway obstruction, reflection of general IgE hyperresponsiveness to multiple allergens, and induction of persistent T helper type 2 responses (Roman et al., 1997). Although an exaggerated RSV IgE response to RSV infection during infancy may correlate with recurrent wheezing during a child's early years, this may not represent asthma per se but rather repeated similar responses to multiple infections with RSV and other respiratory pathogens (Welliver et al., 1986). A study by Sigurs and associates (1995) tends to support the hypothesis that RSV infection may cause or promote asthma by inducing atopy in infected individuals. Since the rate of sensitization among controls with and without IgG antibodies to RSV is similar, an encounter with the virus without development of bronchiolitis does not seem sufficient to increase the risk of sensitization. Other investigators have reached different conclusions about the relationship between RSV infection in infancy and the development of atopy and asthma later in childhood. In a study by Pullan and Hey (1982), the frequency of positive skin tests for common allergens was similar for individuals with previous bronchiolitis and for controls. Murray and colleagues (1992) found no difference in skin test reactivity to dust mites between bronchiolitis patients and controls.

A precise mechanism by which RSV bronchiolitis might induce allergies and asthma has not been elucidated (Dezateux et al., 1997). However, one possible pathogenic mechanism could be through increasing the synthesis of IgE since it has been demonstrated that RSV bronchiolitis can induce an IgE antivirus response and that RSV-specific IgE responses in infancy are associated with later recurrence of wheezing. Welliver and colleagues (1981) found that IgE titers to RSV in nasal secretions were highest in patients with evidence of airway obstruction. Chang and colleagues (1990) prospectively followed 38 of these infants for 48 months after an initial episode of RSV bronchiolitis. Only 20% of infants with undetectable titers of RSV IgE had subsequent episodes of documented wheezing. In contrast, 70% of children with high RSV IgE antibody titers experienced wheezing. In a four-year prospective study of 13 infants of history-positive bilaterally allergic parents, Frick and colleagues (1979) noted that viral infections including RSV occurred one to two months before the onset of allergic sensitization in 10 of the 11 children who subsequently became atopic.

Despite the strong correlation between virus-specific IgE, mediator release, and the clinical pattern of acute and recurrent disease, it is premature to conclude that virus-specific IgE is causal in this process. Moreover, whether those infants who had persistent wheezing and/or airway obstruction were concurrently both sensitized and exposed to indoor allergens is not known.

Chlamydia

Definition of the Agent and Means of Exposure

There are two forms of the bacterium Chlamydia that have been examined in relation to asthma. Chlamydia trachomatis is a sexually transmitted infectious disease that may be spread from mother to child during birth and perhaps through close contact with secretions of an infected individual. Chlamydia pneumoniae is an important cause of adult respiratory disease including pneumonia, bronchitis, sinusitis, and pharyngitis and may be associated with atherosclerosis (Kalman et al., 1999). A 1991 editorial in the Journal of the American Medical Association suggested that C. pneumoniae infection might provide an explanation for the increased incidence of asthma (Bone, 1991).

Evidence Regarding Asthma Exacerbation and Development

An anecdotal report suggests that C. trachomatis produces asthma-like symptoms in infants that may be treated successfully with antichlamydial antibiotic therapy (Bavastrelli et al., 1992). Björnsson and colleagues (1996) report that serological signs of a previous C. trachomatis infection were found significantly more often in subjects who reported having had asthma at some time, asthma during the past year, wheezing during the past year, and bronchial hyperresponsiveness. It is unclear, however, whether such reports uniquely implicate C. trachomatis or are unrecognized C. pneumoniae infections since the two species are cross-reactive.

Chlamydia pneumoniae infection has been associated with asthma exacerbations in both adults and children. Björnsson and colleagues (1996) found a statistically significant relationship between current or recent C. pneumoniae infection and wheezing. Johnston (1997) found that immune responses to C. pneumoniae were positively associated with an increased frequency of asthma exacerbations in a group of 9- to 11-year-olds. Cunningham and colleagues (1998) report increased numbers of asthma exacerbations in children with higher local antibody responses to C. pneumoniae. Miyashita and colleagues (1998) found a significantly higher frequency and geometric mean titer of C. pneumoniae antibodies in adult patients with exacerbations of asthma. However, Cook and colleagues (1998) did not find an association between asthma exacerbations (termed “acute asthma” in the study) and seropositivity in a two-year study of 123 adult asthmatics and 1,518 controls.

Several groups of researchers have reported a serological association between C. pneumoniae and chronic adult asthma in studies that controlled for one or more known confounders. Hahn and colleagues (1991) found that patients with serologically confirmed C. pneumoniae infection were more likely than seronegative patients to develop bronchial asthma subsequent to their infection. A later study of 163 adolescents and adults by Hahn and McDonald (1998) had the same findings, leading the authors to conclude that acute C. pneumoniae respiratory tract infections in previously unexposed nonasthmatic individuals can result in chronic asthma. Cook and colleagues (1998) found a statistically significant association between severe chronic asthma and seropositivity in their two-year study of 123 adult asthmatics and 1,518 controls. Johnston (1999) reports a high prevalence of C. pneumoniae infection in a group of 9- to 11-year-old asthmatics. Von Hertzen and colleagues (1999) found asthma to be significantly associated with IgG antibody levels to C. pneumoniae, with the strongest association for nonatopic, long-standing asthma.

However, not all studies find this association. Kraft and colleagues (1998) did not find C. pneumoniae in the lungs or airways of 18 asthmatic subjects. Larsen and colleagues (1998) found that a group of 22 asthmatics did not differ from 55 controls with relation to two markers of C. pneumoniae infection. The authors note two weaknesses in many of the studies reporting a relationship: (1) a diagnosis of asthma was seldom well-established in these cases, and (2) no pathobiologic mechanism was proposed to account for initiation in adults (production of specific IgE was noted as a possible initiating mechanism in children).

At present (late 1999) there is a debate in the literature over the meaning of these studies. Although their results are consistent with an association between C. pneumoniae (and perhaps C. trachomatis) and asthma development, present data are insufficient to distinguish this premise from other reasonable hypotheses, notably whether asthma predisposes individuals to other chronic respiratory infections.

Mycoplasma

Definition of the Agent and Means of Exposure

Mycoplasma pneumoniae is a bacterial infection that is responsible for a number of respiratory diseases including tracheobronchitis, rhinitis, pharyngitis, otitis, and a form of pneumonia. Over the past several years there have been multiple anecdotal reports and epidemiologic studies suggesting an association between M. pneumoniae infection and exacerbations of asthma (e.g., Seggev et al., 1986). Among the more recent studies, Freymuth and colleagues (1999) found that C. pneumoniae and M. pneumoniae infections—alone or in combination—were present in nearly 82% of asthmatic exacerbations. However, Cunningham and colleagues (1998) and Johnston (1997, 1999) found no evidence of a role for M. pneumoniae in acute asthma exacerbations.

Evidence Regarding Asthma Exacerbation and Development

The literature regarding M. pneumoniae and asthma development is sparse. Yano and colleagues (1994) suggested in a case report that an adult male may have developed asthma as a result of M. pneumoniae infection. Kraft and colleagues (1998) found M. pneumoniae present in the lower airways of chronic stable asthmatics with significantly greater frequency (10 of 18) than in controls (1 of 11).

Conclusions: Asthma Exacerbation and Development

  • There is sufficient evidence of an association between infection with rhinovirus and exacerbation of asthma.
  • There is limited or suggestive evidence of an association between infection with RSV and exacerbation of asthma.
  • There is limited or suggestive evidence of an association between infection with Chlamydia pneumoniae and the exacerbation of asthma.
  • There is inadequate or insufficient evidence to determine whether or not there is an association between infection with Chlamydia trachomatis and exacerbation of asthma.
  • There is limited or suggestive evidence of an association between Mycoplasma pneumoniae infection and exacerbation of asthma.
  • There inadequate or insufficient information to determine whether or not there is an association between infection with rhinovirus and the development of asthma in infants. There is limited or suggestive evidence of no association between infection with rhinovirus and the development of asthma in older children and adults.
  • There is limited or suggestive evidence of an association between infection with RSV and the development of asthma.
  • There is inadequate or insufficient information to determine whether or not there is an association between Chlamydia trachomatis or Chlamydia pneumoniae infections and the development of asthma.
  • There is inadequate or insufficient information to determine whether or not there is an association between Mycoplasma pneumoniae infection and the development of asthma.

Evidence Regarding Exposure Mitigation and Prevention

A primary infection prevention strategy is the avoidance of transmission and exposure through proper personal hygiene practices. Vaccination has also been mentioned as a means of prevention. Although the data are conflicting, breast feeding offers a possible mechanism of passive immunization of infants that may play some role in protection against infection. Further studies are needed to evaluate this notion. A detailed discussion of these strategies and their effectiveness is outside the scope of this report.

Some data are available on the characteristics of RSV that may inform infection mitigation and prevention strategies for this agent. RSV withstands changes in temperature and pH relatively poorly. Only 10% of RSV remained after the virus was exposed to 55°C for 5 minutes. At 37°C, the virus was stable for 1 hour, and only 10% of the infectivity remained after 24 hours. RSV also withstands an acid medium poorly, and the optimal pH is 7.5. Ether, chloroform, and a variety of detergents such as sodium dodecyl sulfate quickly inactivate the virus. At room temperature, RSV in the secretions of patients may survive on nonporous surfaces, such as countertops for 3–30 hours, depending on the humidity. On porous surfaces such as cloth and paper, survival is shorter, usually lasting less than an hour. The infectivity of RSV on the hands is variable from person to person but usually lasts less than one hour. The survival of RSV in the environment appears to depend in part on the drying time as well as on the humidity. These data suggest that proper cleaning of objects and surfaces exposed to RSV may serve to limit exposure to the virus.

Aspects of the indoor environment may also increase the risk of respiratory infection. Building characteristics would have an indirect effect on asthma symptoms if these characteristics influenced the prevalence of the relevant respiratory illnesses among building occupants. Crowded living conditions—found more often among urban residents and lower-income individuals in the United States—may facilitate the spread of infectious disease. Certain indoor spaces such as day care facilities and schools may also present more favorable conditions for their spread. One study suggested higher rates of transmission of infectious pulmonary disease in more tightly constructed military barracks (Brundage et al., 1988). As reviewed by Fisk (1999), the results of ten studies suggest that the prevalence of respiratory illness can be influenced significantly by building characteristics such as ventilation rate, space sharing, or occupant density, with relative risks typically between 1.2 and 1.5. None of these studies have confirmed that the responsible agents were rhinoviruses, and only one older study involved children; therefore, this indirect linkage of building characteristics to asthma exacerbation remains theoretical. Nevertheless, the evidence of an indirect link is sufficient to warrant further investigation.

Conclusions: Exposure Mitigation and Prevention

  • The committee declined to draw conclusions about the effectiveness of specific personal hygiene or medical practices in mitigating or preventing exposure to infectious agents because it believes this to be outside the scope of this report.
  • There is inadequate or insufficient information to determine whether or not there is an association between building characteristics that reduce close contact between individuals and decreased spread of infectious diseases. There is also inadequate or insufficient evidence of an association with ventilation rates.

Research Needs

Numerous studies suggest an association between the infections discussed in this section and asthma exacerbations, although uncertain ascertainment of asthma and questions about the identity of the specific infections responsible limit the confidence with which some conclusions can be drawn. Advances in analysis techniques that allow more sensitive and confident identification of viruses, such as PCR and ELISA, will facilitate research on this topic. These advances will also aid studies of other viruses that may be associated with asthma such as adenovirus, coronavirus, cytomegalovirus, and parainfluenza.

Research on the possible association between infectious agents and asthma development is continuing and is encouraged. There are gaps in the knowledge concerning the mechanism(s) by which agents may promote asthma and whether particular interventions aimed at limiting infections result in decreased rates of asthma. Among the interesting questions are whether the lower respiratory tract acts as a potential reservoir for common respiratory viruses and whether maternal immunization has the potential to protect both the mother and the infant. Research on the impact of building characteristics on the transmission of infectious agents, which is in its infancy, may yield important public health benefits.

HOUSEPLANTS

Definition of the Agent and Means of Exposure

Houseplants have the potential to release pollen, sap, and other plant parts; arthropod pest allergens; and fungi.

Houseplant Pollen

The data on houseplant pollen allergy is restricted to the occupational literature and to case reports. Allergy to some flower pollens has been clearly documented in occupational situations and could occur in residential environments with cut flowers (de Jong et al., 1998). Co-sensitization with some houseplant pollen allergens and some outdoor allergens is extremely common (e.g., mugwort, daffodil) and may indicate that these plants share allergens. In cases of this sort, asthma related to outdoor pollen exposure could be exacerbated by houseplant pollen, although very little pollen is produced by most houseplants and close contact would be required.

Other Plant Parts and Fluids

Exposure to plant materials during the production of plant extracts as medicines has led to occupational asthma (Giavina-Bianchi et al., 1997). The expanding use of herbal medicines may lead to an increase in these types of exposures. Several papers report cases of houseplant (in particular, Ficus) latex causing contact sensitization and even inhalation allergy. The available data are restricted entirely to the case study literature, and only two reports were retrieved (Diez-Gomez et al., 1998; Schenkelberger et al., 1998).

Arthropods

Arthropods that inhabit plants (particularly spider mites) may release allergens. However, again, all data are in the occupational case study literature, and no information is available on the extent of the problem (Orta et al., 1998). The potential for cross-reactivity with dust mite allergens has not been evaluated.

Fungi

Several reports implicate houseplants as sources of fungi in indoor environments, with a focus on human infectious agents rather than allergens. These studies, in general, rely on source (soil) sampling and do not relate exposure to any allergic disease (Staib, 1992, 1996; Staib et al., 1978a, b, 1980; Summerbell et al., 1989). Burge and colleagues (1982) found no relationship between the presence of houseplants and either concentration or type of fungal spores or pollen in the air.

Conclusions: Asthma Exacerbation and Development

There is no evidence that exposures from houseplants lead to the development of asthma. Although patients occasionally complain of worsening symptoms related to houseplants or exposure to cut flowers, no studies have been conducted to document this connection. While some epidemiologic studies suggest that occupational exposure to plant extracts may lead to allergic diseases including asthma, the relevance of this literature to casual, nonoccupational exposure is not clear. Mites or fungi that may be associated with houseplants could be involved in asthma development or exacerbation, but there is no evidence bearing on this hypothesis.

In summary:

  • There is inadequate or insufficient evidence to determine whether or not an association exists between exposures from houseplants and the exacerbation or development of asthma.

Evidence and Conclusions: Exposure Mitigation and Prevention

The committee did not identify any literature regarding the effectiveness or impact on asthma outcomes of measures intended to limit houseplant exposure.

Research Needs

Further research is needed to determine whether or not houseplants release fungal spores into the air. This research will benefit both the allergy community and the infectious disease literature. Additionally, research should be conducted to determine what risks, if any, are associated with occupational exposure to plant materials.

POLLEN

Definition of the Agent and Means of Exposure

Introduction

Pollen exposure has long been recognized as a stimulant for symptoms of allergic disease. Allergic rhinitis (hay fever) has been considered the primary outcome, and outdoor environments are the major source of exposure. The role of pollen in the development and exacerbation of asthma has received relatively little direct attention, and very few studies are available that document indoor exposure to pollen allergens.

Nature of the Agent

Structure and Function Pollen is the male reproductive structure of flowering plants and gymnosperms. Pollen grains are usually unicellular, with a rigid and highly resistant cell wall formed of a complex polysaccharide-based substance called sporopollenin. Each pollen grain contains the systems required for recognition of genetically relevant female flowers and the production of a pollen tube that grows into contact with the egg cell to effect fertilization.

Pollen Allergens

The list of pollens from which extracts have been derived that produce positive skin tests in some fraction of the population is long. In one California study, 57 skin test allergens were necessary to detect 90% of the atopic patients. This set included 2 grasses, 16 weeds, and 27 trees (Galant et al., 1998).

The list of purified allergens from pollens is also growing. These include Amb a I (short ragweed, Ambrosia artemisiifolia), Bet v I (birch pollen, Betula verrucosa), and Lol p I (ryegrass, Lolium perenne). One study has estimated that a single pollen grain of B. verrucosa contains 0.006 ng Bet v I (Schappi et al., 1997).

Other Pollen-Associated Agents

Serine proteases that could directly affect respiratory function apart from allergic reactions have been detected in ragweed and mesquite pollen (Bagarozzi and Travis, 1998; Travis et al., 1996).

Characteristics of Pollen Exposure

Particle Size Considerations Intact pollen grains range from about 10 to 100 µm, with the most common types in the range of 15–30 µm. However, pollen allergens have been documented in air on much smaller particles. During light rain, 1.2 ng Bet v I/per cubic meter (200 birch pollen equivalents) were present on the particle fraction less than 7.5 µm in outdoor air (Schappi et al., 1997). Grass pollen grains have been shown to rupture during rainfall, and allergen (Lol p V) has been recovered in particle fractions less than 5 µm both as free molecules and attached to other particles such as starch grains and combustion (diesel exhaust) particles (Suphioglu, 1998). Pollen allergens have been visualized on the surface of diesel particles under laboratory conditions (Knox et al., 1997). Ragweed allergens have also been recovered from small particles (Habenicht et al., 1984; Solomon et al., 1983).

Patterns of Pollen Prevalence

Outdoors An enormous body of literature documents prevalence patterns for pollens throughout the world. In the United States, the American Academy of Allergy, Asthma, and Immunology has been collecting pollen prevalence data for more than 30 years and currently publishes an annual report containing data from about 100 stations across the nation (e.g., AAAAI, 1998). In the United States, the pollen types most often considered important are mountain cedar, birch, and oak (trees); grasses (which cross-react broadly across genera); and ragweed (a late-summer weed).

Pollen is produced seasonally. In general, tree pollens are released early in the year, grasses during late spring and early summer, and weed pollens in the late summer and fall. Major exceptions occur. For example, some grass pollen is produced throughout the year in some areas.

Meteorological variables directly affect pollen concentrations in air (in addition to their effects on pollen production). Most pollen types are released during the morning hours, but dispersal may occur later in the day with increase in wind and other disturbances. Wind is a well-known dispersal agent for pollens, which may travel long distances (hundreds of miles) in traveling air masses (Levetin and Buck, 1986). Rain generally washes pollen from the air.

Indoors Outdoor infiltrate is the primary source of pollen in indoor environments. Birch pollen allergen (Bet v I) has been found associated with suspended particles indoors (Holmquist and Vesterberg, 1999; Ormstad et al., 1998).

Evidence Regarding Asthma Exacerbation and Development

Pollen Exposure and Asthma

Sensitization to Pollen Allergens In 3,371 Canadian allergy patients, skin tests indicated that 52% were sensitive to grass allergens and 45% to ragweed allergens (Boulet et al., 1997). In the Swiss population (random, more than 8,000 individuals studied) 32% were atopic, with sensitivity to grass allergen the most prevalent (12%), followed by dust mite (8.9%) and birch pollen (7.9%) (Wuthrich et al., 1995). In 1,159 random participants in Hamburg and Ehrfurt, respectively, grass and birch allergen sensitivities were 24 versus 19% and 19 versus 8% (Nowak et al., 1996). Of inner city asthmatics in Chicago, 45% were sensitized to ragweed pollen (76% were sensitive to indoor allergens) (Kang et al., 1993). In 7,079 retrospectively observed patients with asthma and/or rhinitis, 44% had one or more positive skin tests, with grass, cat, and birch allergens eliciting the greatest number of positive tests. Of the 35% of sensitized patients who were monosensitized, 7.4% reacted only to mite allergens and 7.0% to grass allergen (Eriksson and Holmen, 1996). On the other hand, Subiza and colleagues (1994) report that 92% of Madrid allergy patients are sensitive to grass allergens, 63% to olive allergens, and 56% to sycamore (Platanus) allergens.

Relationships to Asthma

Pollen Sensitization Related to Asthma In general, pollen sensitization has been associated primarily with hay fever rather than asthma, and this opinion is supported in several studies. In a study reported by Eriksson and Holmen (1996), patients with grass allergen sensitivity were more likely to have rhinitis than asthma. In a Tucson population, sensitivity to ryegrass and mulberry pollen independently predicted rhinitis but not asthma (Halonen et al., 1997). In a case-control study of 343 random children, 35% were atopic, and 90% of those with five or more episodes of wheezing had one or more positive skin tests (Henderson et al., 1995). Pollen sensitivity was not associated with wheezing in this population.

On the other hand, in an Australian population of 745 young adults, sensitivity to ryegrass pollen was among the four top allergen sensitivities that posed a risk for the existence of current asthma (Abramson et al., 1996). Winter respiratory symptoms (including asthma) have been related to positive prick, RAST, or endonasal challenge tests with alder and hazel pollen allergens (Laurent et al., 1994). Grass allergen elicited the most positive skin tests among black asthmatics in Johannesburg (Luyt et al., 1995).

Pollen Exposure and Asthma Ordaz and colleagues (1998) followed symptoms in 104 asthmatic patients and related exacerbation events to airborne pollen counts. When infectious disease-related events were excluded, there was a good correlation between event days and pollen counts (r = 0.7, p < .01). In a prospective study of mild to moderate asthmatics (N = 139), Epton and colleagues (1997) were unable to distinguish a relationship between measured pollen concentrations and measured peak flow.

Baraldi and colleagues (1999) documented a twofold increase in exhaled NO in grass-sensitive asthmatic children during the grass pollen season. Natural allergen exposure was shown to induce T cell, mast cell, and eosinophil inflammatory response in grass-sensitive patients undergoing seasonal exacerbation (N = 17) (Djukanovic et al., 1996). Rosas and colleagues (1998) report an association between grass pollen counts and asthma admissions in Mexico City in both dry and wet seasons. Celenza and colleagues (1996) related changes in grass pollen concentrations and decreases in temperature to epidemic asthma. Their patients, who were involved in a thunderstorm-associated epidemic in England, had high serum IgE levels specific for grass pollen allergens (Venables et al., 1997). Newson and colleagues (1997) counted asthma admissions in two age groups (0–14 and >14 years) and related these events to lightning episodes and grass pollen concentrations. Although there were excess densities of lightning and grass pollen counts in epidemics, many (in fact, most) epidemics were not preceded by thunderstorms. In a follow-up study, they report that thunderstorms following periods of high pollen counts are more likely to lead to asthma epidemics (Newson et al., 1998).

In an analysis of 59,624 asthma hospitalizations in Finland, a peak was observed in May that was attributed to the exposure of sensitized people to birch pollen allergens (Harju et al., 1997). As part of the increase in doctor-diagnosed asthma and episodes of breathlessness in children in Norway, symptoms with exposure to birch pollen increased from 3.7 to 6.1% of children between 1981 and 1993 (Skjonsberg et al., 1995).

Woodcock and Custovic (1998) suggest that exposure to ragweed allergen reduces corticosteroid binding capacity in sensitized asthmatics, contributing to poor asthma control.

In 20 subjects, airway responsiveness to histamine challenge was highest during the privet season, and symptom scores and bronchodilator use increased. However, challenge with privet pollen allergens induced no immediate asthmatic responses, and late responses occurred in only 6 of 17 tested patients (Richards et al., 1995).

Pollen–Air Pollutant Interactions

Laboratory Studies Strand and colleagues (1997) report that in a group of 18 pollen-sensitive asthmatics, short exposure in the laboratory to concentrations of NO2 representative of those that occur in outdoor air enhances pollen allergen-induced late asthmatic responses. Repeated short exposures to NO2 enhanced effects of otherwise nonsymptomatic doses of pollen allergen (Strand et al., 1998). However, in similar studies, Hanania and colleagues (1998) found no change in allergen response following ozone challenge.

Field Studies Anderson and colleagues (1998) report a synergistic effect for pollen and SO2 with respect to hospital admissions of children (but not adults) for asthma. No other pollen–pollutant interactions were observed. In a comparison between randomly selected participants in Hamburg and Ehrfurt, Germany, the prevalence of atopy and pollen sensitization was highest in Hamburg, while air pollutant exposures were highest in Ehrfurt (Nowak et al., 1996).

Conclusions: Asthma Exacerbation and Development

Evidence from studies of outdoor exposure indicates that pollen exacerbates existing asthma in sensitized individuals. Information does not permit a conclusion concerning whether or not there is an overall role for pollen allergens in the development of asthma. Although pollen allergens have been documented in both dust and indoor air, data is lacking to draw a informed conclusion. Thus:

There is inadequate or insufficient evidence to determine whether or not an association exists between pollen exposure in the indoor environment and the exacerbation or development of asthma.

The committee concludes:

  • There is inadequate or insufficient evidence to determine whether or not an association exists between interventions to lower pollen concentrations in indoor environments and improvement of symptoms or lung function in pollen-allergic asthmatics. Indoors is typically a protective environment for such individuals.

Evidence and Conclusions: Exposure Mitigation and Prevention

There is relatively little information on the impact of ventilation and air-cleaning measures on indoor pollen levels, although it is axiomatic that shutting windows and other measures that generally limit outdoor infiltrate can be effective. Chapter 10 discusses the studies identified by the committee and draws conclusions about the effectiveness of ventilation and air-cleaning measures in reducing indoor concentrations in general.

Research Needs

Studies should be conducted to evaluate the ambiguous relationship between pollen exposure, sensitivity, and asthma. Additional research is also needed to discover the extent of indoor pollen allergen exposure and the interactions between pollen sensitivity and air pollutants.

REFERENCES

  • AAAAI. 1998. Pollen and Spore Report. American Academy of Allergy, Asthma, and Immunology, Milwaukee, WI.
  • Åberg N. 1989. Asthma and allergic rhinitis in Swedish conscripts. Clinical and Experimental Allergy 19(1):59–63. [PubMed: 2784709]
  • Abramson M, Kutin JJ, Raven J, Lanigan A, Czarny D, Walters EH. 1996. Risk factors for asthma among young adults in Melbourne, Australia. Respirology 1(4):291–297. [PubMed: 9441118]
  • ACGIH. 1999. Bioaerosols: Assessment and Control. American Conference of Governmental Industrial Hygienists, Cincinnati.
  • Adachi Y, Ohno N, Yadomae T. 1994. a. Preparation and antigen specificity of an anti-(1→3)-beta-D-glucan antibody. Biological and Pharmaceutical Bulletin 17(11):1508–1512. [PubMed: 7703974]
  • Adachi Y, Okazaki M, Ohno N, Yadomae T. 1994. b. Enhancement of cytokine production by macrophages stimulated with (1→3)-beta-D-glucan, grifolan (GRN) isolated from Grifola frondosa. Biological and Pharmaceutical Bulletin 17(12):1554–1560. [PubMed: 7537572]
  • Amitani R, Taylor G, Elezis EN, Llewellyn-Jones C, Mitchell J, Kuze F, Cole PJ, Wilson R. 1995. Purification and characterization of factors produced by Aspergillus fumigatus which affect human ciliated respiratory epithelium. Infection and Immunity 63(9):3266–3271. [PMC free article: PMC173450] [PubMed: 7543879]
  • Anderson HR, Ponce de Leon A, Bland JM, Bower JS, Emberlin J, Strachan DP. 1998. Air pollution, pollens, and daily admissions for asthma in London 1987– 92. Thorax 53(10):842–848. [PMC free article: PMC1745078] [PubMed: 10193370]
  • Andriessen JW, Brunekreef B, Roemer W. 1998. Home dampness and respiratory health status in European children. Clinical and Experimental Allergy 28(10):1191–1200. [PubMed: 9824385]
  • Angrisano A, Di Berardino L, Montrasio G, Compostella G. 1987. Allergy caused by Alternaria in children (Italian). Pediatria Medica e Chirurgica 9(2):159–160. [PubMed: 3658799]
  • Arlian LG, Bernstein IL, Gallagher JS. 1982. The prevalence of house dust mites Dermatophagoides spp, and associated environmental conditions in homes in Ohio. Journal of Allergy and Clinical Immunology 69(6):527–532. [PubMed: 7076994]
  • Arruda LK, Platts-Mills TAE, Fox JW, Chapman MD. 1990. Aspergillus fumigatus. Allergen I, a major IgE-binding protein, is a member of the mitogillin family of cytotoxins. Journal of Experimental Medicine 172(5):1529–1532. [PMC free article: PMC2188668] [PubMed: 2230656]
  • Arruda LK, Vailes LD, Platts-Mills TA, Hayden ML, Chapman MD. 1997. Induction of IgE antibody responses by glutathione-S-transferase from the German cockroach (Blattella germanica). Journal of Biological Chemistry 272(33):20907–20912. [PubMed: 9252418]
  • ATS (American Thoracic Society). 1998. Respiratory health hazards in agriculture (Official conference report). American Journal of Respiratory and Critical Care Medicine 158(5):S1–S76. [PubMed: 9817727]
  • Aukrust L, Borch SM. 1979. Partial purification and characterization of two Cladosporium herbarum allergens. International Archives of Allergy and Applied Immunology 60(1):68–79. [PubMed: 572347]
  • Avner DB, Perzanowski MS, Platts-Mills TA, Woodfolk JA. 1997. Evaluation of different techniques for washing cats: quantitation of allergen removed from the cat and the effect on airborne Fel d I [see comments]. Journal of Allergy and Clinical Immunology 100(3):307–312. [PubMed: 9314341]
  • Bagarozzi DA Jr, Travis J. 1998. Ragweed pollen proteolytic enzymes: possible roles in allergies and asthma. Phytochemistry 47(4):593–598. [PubMed: 9461675]
  • Baker PJ, Taylor CE, Stashak PW, Fauntleroy MB, Haslov K, Qureshi N, Takayama K. 1990. Inactivation of suppressor T cell activity by the nontoxic lipopolysaccharide of Rhodopseudomonas sphaeroides. Infection and Immunity 58(9):2862–2868. [PMC free article: PMC313579] [PubMed: 2143752]
  • Baker PJ, Hraba T, Taylor CE, Myers KR, Takayama K, Qureshi N, Stuetz P, Kusumoto S, Hasegawa A. 1992. Structural features that influence the ability of lipid A and its analogs to abolish expression of suppressor T cell activity. Infection and Immunity 60(7):2694–2701. [PMC free article: PMC257223] [PubMed: 1535339]
  • Baldini M, Carla Lohman I, Halonen M, Erickson RP, Holt PG, Martinez FD. 1999. A Polymorphism* in the 5' flanking region of the CD14 gene is associated with circulating soluble CD14 levels and with total serum immunoglobulin E. American Journal of Respiratory Cell and Molecular Biology 20(5):976–983. [PubMed: 10226067]
  • Baraldi E, Carra S, Dario C, Azzolin N, Ongaro R, Marcer G, Zacchello F. 1999. Effect of natural grass pollen exposure on exhaled nitric oxide in asthmatic children. American Journal of Respiratory and Critical Care Medicine 159(1):262–266. [PubMed: 9872848]
  • Bates DV, Fish BR, Hatch TF, Mercer TT, Morrow PE. 1966. Deposition and retention models for internal dosimetry of the human respiratory tract. Task group on lung dynamics. Health Physics 12(2):173–207. [PubMed: 5916786]
  • Bavastrelli M, Midulla M, Rossi D, Salzano M. 1992. Chlamydia trachomatis infection in children with wheezing simulating asthma. Lancet 339(8802):1174. [PubMed: 1349398]
  • Becher R, Hongslo JK, Jantunen MJ, Dybing E. 1996. Environmental chemicals relevant for respiratory hypersensitivity: the indoor environment. Toxicology Letters 86(2–3):155–162. [PubMed: 8711767]
  • Beeson MF, Dewdney JM, Edwards RG, Lee D, Orr RG. 1983. Prevalence and diagnosis of laboratory animal allergy. Clinical Allergy 13(5):433–442. [PubMed: 6627621]
  • Berg DJ, Kuhn R, Rajewsky K, Muller W, Menon S, Davidson N, Grunig G, Rennick D. 1995. Interleukin-10 is a central regulator of the response to LPS in murine models of endotoxic shock and the Shwartzman reaction but not endotoxin tolerance. Journal of Clinical Investigation 96(5):2339–2347. [PMC free article: PMC185885] [PubMed: 7593621]
  • Berge M, Munir AK, Dreborg S. 1998. Concentrations of cat (Fel d I), dog (Can f I) and mite (Der f I and Der p I) allergens in the clothing and school environment of Swedish school children with and without pets at home. Pediatric Allergy and Immunology 9(1):25–30. [PubMed: 9560839]
  • Bernton HS, Brown H. 1964. Insect allergy: preliminary studies of the cockroach. Journal of Allergy 35:506–513. [PubMed: 14226309]
  • Bernton HS, Brown H. 1967. Cockroach allergy II: the relation of infestation to sensitization. Southern Medical Journal 60(8):852–855. [PubMed: 6036654]
  • Bernton HS, McMahon TF, Brown H. 1972. Cockroach asthma. British Journal of Diseases of the Chest 66(1):61–66. [PubMed: 5017934]
  • Björnsson E, Hjelm E, Janson C, Fridell E, Boman G. 1996. Serology of chlamydia in relation to asthma and bronchial hyperresponsiveness. Scandinavian Journal of Infectious Disease 28(1):63–69. [PubMed: 9122636]
  • Blackley CH. 1873. Experimental research in the causes and nature of catarrhus aestivus (hay fever or hay asthma). London, England: Balliere, Tindall and Cox.
  • Bohuslav J., Kravchenko VV, Parry GC, Erlich JH, Gerondakis S, Mackman N, Ulevitch RJ. 1998. Regulation of an essential innate immune response by the p50 subunit of NF-kappaB. Journal of Clinical Investigation 102(9):1645–1652. [PMC free article: PMC509112] [PubMed: 9802878]
  • Bollinger ME, Eggleston PA, Flanagan E, Wood RA. 1996. Cat antigen in homes with and without cats may induce allergic symptoms. Journal of Allergy and Clinical Immunology 97(4):907–914. [PubMed: 8655885]
  • Bone RC 1991. Chlamydial pneumonia and asthma: a potentially important relationship. Journal of the American Medical Association 266(2):265. [PubMed: 2056629]
  • Bonifazi F. 1994. Immunotherapy in pollen and mould asthma [Review]. Monaldi Archives for Chest Disease 49(2):150–153. [PubMed: 8049700]
  • Boulet LP, Turcotte H, Laprise C, Lavertu C, Bedard PM, Lavoie A, Hebert J. 1997. Comparative degree and type of sensitization to common indoor and outdoor allergens in subjects with allergic rhinitis and/or asthma. Clinical and Experimental Allergy 27(1):52–59. [PubMed: 9117881]
  • Bousquet J, Michel FB. 1994. Specific immunotherapy in asthma [Review]. Allergy Proceedings 15(6):329–33. [PubMed: 7721083]
  • Brattgjerd S, Evensen O, Lauve A. 1994. Effect of injected yeast glucan on the activity of macrophages in Atlantic salmon, Salmo salar L., as evaluated by in vitro hydrogen peroxide production and phagocytic capacity. Immunology 83(2):288–294. [PMC free article: PMC1414938] [PubMed: 7835949]
  • Braun-Fahrlander C, Gassner M, Grize L, Neu U, Sennhauser FH, Varonier HS, Vuille JC, Wuthrich B. 1999. Prevalence of hay fever and allergic sensitization in farmer's children and their peers living in the same rural community. SCARPOL team. Swiss Study on Childhood Allergy and Respiratory Symptoms with Respect to Air Pollution. Clinical and Experimental Allergy 29(1):28–34. [PubMed: 10051699]
  • Brundage JF, Scott RM, Lednar WM, Smith DW, Miller RN. 1988. Building-associated risk of febrile acute respiratory diseases in Army trainees. Journal of the American Medical Association 259(14):2108–2112. [PubMed: 3346987]
  • Brunekreef B, Dockery DW, Speizer FE, Ware JH, Spengler JD, Ferris BG. 1989. Home dampness and respiratory morbidity in children. American Review of Respiratory Disease 140(5):1363–1367. [PubMed: 2817598]
  • Burge HA, Solomon WR, Boise JR. 1980. Microbial prevalence in domestic humidifiers. Applied and Environmental Microbiology 39(4):840–844. [PMC free article: PMC291430] [PubMed: 7377779]
  • Burge HA, Solomon WR, Muilenberg ML. 1982. Evaluation of indoor plantings as allergen exposure sources. Journal of Allergy and Clinical Immunology 70(2):101–108. [PubMed: 7096819]
  • Burge HA, Solomon WR. 1987. Sampling and analysis of biological aerosols. Atmospheric Science 21(2):451.
  • Burge HA, Hoyer ME, Solomon WR, Simmons EG, Gallup J. 1989. Quality control factors for Alternaria allergens. Mycotaxon 34(1):55–63.
  • Burke GW, Carrington CB, Strauss R, Fink JN, Gaensler EA. 1977. Allergic alveolitis caused by home humidifiers. Unusual clinical features and electron microscopic findings. Journal of the American Medical Association 238(25): 2705–2708. [PubMed: 579209]
  • Call RS, Smith TF, Morris E, Chapman MD, Platts-Mills TA. 1992. Risk factors for asthma in inner city children. Journal of Pediatrics 121(6):862–866. [Comment in: J Pediatr 1993. 123(1):171.] [PubMed: 1447646]
  • Campbell EM, Kunkel SL, Strieter RM, Lukacs NW. 1998. Temporal role of chemokines in a murine model of cockroach allergen-induced airway hyperreactivity and eosinophilia. Journal of Immunology 161(12):7047–7053. [PubMed: 9862742]
  • Cantani A, Businco E, Maglio A. 1988. Alternaria allergy: a three-year controlled study in children treated with immunotherapy. Allergologia et Immunopathologia 16(1):1–4. [PubMed: 3381709]
  • Celenza A, Fothergill J, Kupek E, Shaw RJ. 1996. Thunderstorm associated asthma: a detailed analysis of environmental factors. British Medical Journal 312(7031):604–607. [Comment in BMJ 1990. 312(7031):590–591.] [PMC free article: PMC2350378] [PubMed: 8595333]
  • Chang JCS, Foarde KK, Van Osdell DW. 1996. Assessment of fungal (Penicillium chrysogenum) growth on three HVAC duct materials. Environment International 22(4):425–431.
  • Chang TL, Shea CM, Urioste S, Thompson RC, Boom WH, Abbas AK. 1990. Heterogeneity of helper/inducer T lymphocytes. III. Responses of IL-2- and IL-4- producing (TH1 and TH2) clones to antigens presented by different accessory cells. Journal of Immunology 145(9):2803–2808. [PubMed: 1976705]
  • Chapman, MD. 1993. Dissecting cockroach allergens. Clinical and Experimental Allergy 23(6):459–461. [PubMed: 8369971]
  • Chapman MD, Platts-Mills TA. 1980. Purification and characterization of the major allergen from Dermatophagoides pteronyssinus-antigen P1. Journal of Immunology 125(2):587–592. [PubMed: 6771329]
  • Chapman MD, Heymann PW, Wilkins SR, Brown MJ, Platts-Mills TA. 1987. Monoclonal immunoassays for the major dust mite (Dermatophagoides) allergens, Der p I and Der f I, and quantitative analysis of the allergen content of mite and house dust extracts. Journal of Allergy and Clinical Immunology 80(2):184–194. [PubMed: 3611539]
  • Chew GL, Burge HA, Dockery DW, Muilenberg ML, Weiss ST, Gold DR. 1998. Limitations of a home characteristics questionnaire as a predictor of indoor allergen levels. American Journal of Respiratory and Critical Care Medicine 157(5 Pt 1):1536–1541. [PubMed: 9603135]
  • Chow JC, Young DW, Golenbock DT, Christ WJ, Gusovsky F. 1999. Toll-like receptor-4 mediates lipopolysaccharide-induced signal transduction. Journal of Biological Chemistry 274(16):10689–10692. [PubMed: 10196138]
  • Christensen LT, Schmidt CD, Robbins L. 1975. Pigeon breeders' disease—-a prevalence study and review. Clinical Allergy 5(4):417–430. [PubMed: 1053441]
  • Clarke CW, Aldons PM. 1979. The nature of asthma in Brisbane. Clinical Allergy 9(2):147–152. [PubMed: 445783]
  • Cleveland MG, Gorham JD, Murphy TL, Tuomanen E, Murphy KM. 1996. Lipoteichoic acid preparations of gram-positive bacteria induce interleukin-12 through a CD14-dependent pathway. Infection and Immunity 64(6):1906– 1912. [PMC free article: PMC174015] [PubMed: 8675286]
  • Cochran DG. 1995. Toxic effects of boric acid on the German cockroach. Experimentia 51(6):561–563. [PubMed: 7607296]
  • Cochran DG. 1996. Relevance of resistance ratios to operational control in the German cockroach (Dictyoptera, Blattellidae). Journal of Economic Entomology 89(2):318–321.
  • Cockcroft DW, Ruffin RE, Frith PA, Cartier A, Juniper EF, Dolovich J, Hargreave FE. 1979. Determinants of allergen-induced asthma: dose of allergen, circulating IgE antibody concentration, and bronchial responsiveness to inhaled histamine. American Review of Respiratory Disease 120(5):1053–1058. [PubMed: 507521]
  • Cockcroft A, Edwards J, McCarthy P, Andersson N. 1981. Allergy in laboratory animal workers. Lancet 1(8224):827–830. [PubMed: 6111683]
  • Comoy EE, Pestel J, Duez C, Stewart GA, Vendeville C, Fournier C, Finkelman F, Capron A, Thyphrontis G. 1998. The house dust mite allergen, Dermatophagoides pteronyssinus, promotes type 2 responses by modulating the balance between IL-4 and IFN-gamma. Journal of Immunology 160(5):2456–2462. [PubMed: 9498790]
  • Cook PJ, Davies P, Tunnicliffe W, Ayres JG, Honeybourne D, Wise R. 1998. Chlamydia pneumoniae and asthma. Thorax 53(4):254–259. [Comment in: Thorax 1998 December 53(12):1095–1096.] [PMC free article: PMC1745183] [PubMed: 9741366]
  • Corey JP, Kaiseruddin S, Gungor A. 1997. Prevalence of mold-specific immunoglobulins in a midwestern allergy practice. Otolaryngology—Head and Neck Surgery 117(5):516–520. [PubMed: 9374177]
  • Corsico R, Cinti B, Feliziani V, Gallesio MT, Liccardi G, Loreti A, Lugo G, Marcucci F, Marcer G, Meriggi A, Minelli M, Gherson G, Nardi G, Negrini AC, Piu G, Passaleva A, Pozzan M, D'Ambrosio FP, Venuti A, Zanon P, Zerboni R. 1998. Prevalence of sensitization to Alternaria in allergic patients in Italy. Annals of Allergy, Asthma, and Immunology 80(1):71–76. [PubMed: 9475571]
  • Crameri R. 1998. Recombinant Aspergillus fumigatus allergens: from nucleotide sequences to clinical applications. International Archives of Allergy and Immunology 115(2):99–114. [PubMed: 9482698]
  • Crane J, Kemp T, Siebers R, Rains N, Fishwick D, Fitzharris P. 1997. Increased house dust mite allergen in synthetic pillows may explain increased wheezing [letter]. British Medical Journal 314(7096):1763–1764. [PMC free article: PMC2126886] [PubMed: 9202523]
  • Cross S. 1997. Mould spores: the unusual suspects in hay fever. Community Nurse 3(4):25–26. [PubMed: 9451141]
  • Cruz A, Saenz de Santamaria M, Martinez J, Martinez A, Guisantes J, Palacios R. 1997. Fungal allergens from important allergenic fungi imperfecti [Review]. Allergologia et Immunopathologia 25(3):153–158. [PubMed: 9208052]
  • Cuijpers CE, Swaen GM, Wesseling G, Sturmans F, Wouters EF. 1995. Diverse effects of the indoor environment on respiratory health in primary school children. Environmental Research 68(1):11–23. [PubMed: 7729382]
  • Cullinan P, Lowson D, Nieuwenhuijsen MJ, Gordon S, Tee RD, Venables KM, McDonald JC, Newman Taylor AJ. 1994. Work related symptoms, sensitisation, and estimated exposure in workers not previously exposed to laboratory rats. Occupational and Environmental Medicine 51(9):589–592. [PMC free article: PMC1128051] [PubMed: 7951789]
  • Cunningham AF, Johnston SL, Julious SA, Lampe FC, Ward ME. 1998. Chronic Chlamydia pneumoniae infection and asthma exacerbations in children. European Respiratory Journal 11(2):345–349. [PubMed: 9551736]
  • Custovic A, Green R, Fletcher A, Smith A, Pickering CA, Chapman MD, Woodcock A. 1997. Aerodynamic properties of the major dog allergen Can f I: distribution in homes, concentration, and particle size of allergen in the air. American Journal of Respiratory and Critical Care Medicine 155(1):94–98. [PubMed: 9001295]
  • Custovic A, Fletcher A, Pickering CA, Francis HC, Green R, Smith A, Chapman M, Woodcock A. 1998. a. Domestic allergens in public places. III: house dust mite, cat, dog and cockroach allergens in British hospitals. Clinical and Experimental Allergy 28(1):53–59. [PubMed: 9537780]
  • Custovic A, Simpson A, Pahdi H, Green RM, Chapman MD, Woodcock A. 1998. b. Distribution, aerodynamic characteristics, and removal of the major cat allergen Fel d I in British homes. Thorax 53(1):33–38. [PMC free article: PMC1758692] [PubMed: 9577519]
  • Damare SM, Wells S, Offenbacher S. 1997. Eicosanoids in periodontal diseases: potential for systemic involvement. Advances in Experimental Medicine and Biology 433:23–35. [PubMed: 9561098]
  • Davies GE, McArdle LA. 1981. Allergy to laboratory animals: a survey by questionnaire. International Archives of Allergy and Applied Immunology 64:302–307. [PubMed: 7216507]
  • De Andrade AD, Birnbaum J, Magalon C, Magnol JP, Lanteaume A, Charpin D, Vervloet D. 1996. Fel d I levels in cat anal glands. Clinical and Experimental Allergy 26(2):178–180. [PubMed: 8835125]
  • de Blay F, Chapman MD, Platts-Mills TA. 1991. a. Airborne cat allergen (Fel d I). Environmental control with the cat in situ [see comments]. American Review of Respiratory Disease 143(6):1334–1339. [PubMed: 2048821]
  • de Blay F, Pauli G, Bessot JC. 1991. b. Cross-reactions between respiratory and food allergens. Allergy Proceedings 12(5):313–317. [PubMed: 1720408]
  • de Groot H, van Swieten P, van Leeuwen J, Lind P, Aalberse RC. 1988. Monoclonal antibodies to the major feline allergen Fel d I. I. Serologic and biologic activity of affinity-purified Fel d I and of Fel d I-depleted extract. Journal of Allergy and Clinical Immunology 82(5 Pt 1):778–786. [PubMed: 2461402]
  • de Jong NW, Vermeulen AM, Gerth van Wijk R, de Groot H. 1998. Occupational allergy caused by flowers. Allergy 53(2):204 209. [PubMed: 9534922]
  • Delfino RJ, Zeiger RS, Seltzer JM, Street DH, Matteucci RM, Anderson PR, Koutrakis P. 1997. The effect of outdoor fungal spore concentrations on daily asthma severity. Environmental Health Perspectives 105(6):622–635. [PMC free article: PMC1470068] [PubMed: 9288497]
  • Deuell B, Arruda LK, Hayden ML, Chapman MD, Platts-Mills TAE. 1991. Trichophyton tonsurans allergen I. Characterization of a protein that causes immediate but not delayed hypersensitivity. Journal of Immunology 147(1): 96–101. [PubMed: 1711086]
  • Dezateux C, Fletcher ME, Dundas I, Stocks J. 1997. Infant respiratory function after RSV-proven bronchiolitis. American Journal of Respiratory and Critical Care 155(4):1349–1355. [Published erratum appears in Am J Respir Crit Care Med 1997. 156(2 Pt 1):675.] [PubMed: 9105078]
  • Dhillon M. 1991. Current status of mold immunotherapy [Review]. Annals of Allergy 66(5):385–392. [PubMed: 2035901]
  • Diez-Gomez ML, Quirce S, Aragoneses E, Cuevas M. 1998. Asthma caused by Ficus benjamina latex: evidence of cross-reactivity with fig fruit and papain. Annals of Allergy, Asthma, and Immunology 80(1):24–30. [PubMed: 9475562]
  • Djukanovic R, Feather I, Gratziou C, Walls A, Peroni D, Bradding P, Judd M, Howarth PH, Holgate ST. 1996. Effect of natural allergen exposure during the grass pollen season on airways inflammatory cells and asthma symptoms. Thorax 51(6):575–581. [PMC free article: PMC1090485] [PubMed: 8693436]
  • Douwes J, Heederik D. 1997. Epidemiologic investigations of endotoxins. International Journal of Occupational Environmental Health 3(1):S26–S31.
  • Dreborg S, Agrell B, Foucard T, Kjellman NI, Koivikko A, Nilsson S. 1986. A double-blind, multicenter immunotherapy trial in children, using a purified and standardized Cladosporium herbarum preparation. I. Clinical results. Allergy 41(2):131–140. [PubMed: 3518526]
  • Du J, Knowles BH, Li J, Ellar DJ. 1999. Biochemical characterization of Bacillus thuringiensis cytolytic toxins in association with a phospholipid bilayer. Biochemical Journal 338(Pt 1):185–193. [PMC free article: PMC1220041] [PubMed: 9931315]
  • Dubin W, Martin TR, Swoveland P, Leturcq DJ, Moriarty AM, Tobias PS, Bleecker ER, Goldblum SE, Hasday JD. 1996. Asthma and endotoxin: lipopolysaccharide-binding protein and soluble CD14 in bronchoalveolar compartment. American Journal of Physiology 270(5 Pt 1):L736–744. [PubMed: 8967507]
  • Duffort O, Carreira J, Lombarder M. 1987. Characterization of the main IgE binding components of cat dander. International Archives of Allergy and Applied Immunology 84:339–344. [PubMed: 3679560]
  • Dybendal T, Hetland T, Vik H, Apold J, Elsayed S. 1989. a. Dust from carpeted and smooth floors. I. Comparative measurements of antigenic and allergenic proteins in dust vacuumed from carpeted and non-carpeted classrooms in Norwegian schools. Clinical and Experimental Allergy 19(2):217–224. [PubMed: 2752322]
  • Dybendal T, Vik H, Elsayed S. 1989. b. Dust from carpeted and smooth floors. II. Antigenic and allergenic content of dust vacuumed from carpeted and smooth floors in schools under routine cleaning schedules. Allergy 44(6):401–411. [PubMed: 2478043]
  • Dybendal T, Wedberg WC, Elsayed S. 1991. Dust from carpeted and smooth floors. IV. Solid material, proteins and allergens collected in the different filter stages of vacuum cleaners after ten days of use in schools. Allergy 46(6):427– 435. [PubMed: 1720284]
  • Dybendal T, Elsayed S. 1994. Dust from carpeted and smooth floors. VI. Allergens in homes compared with those in schools in Norway. Allergy 49(4):210–216. [PubMed: 8037353]
  • Dziarski R, Tapping RI, Tobias PS. 1998. Binding of bacterial peptidoglycan to CD14. Journal of Biological Chemistry 273(15): 8680–8690. [PubMed: 9535844]
  • Edmonds RL, editor. , Ed. 1979. Aerobiology, The Ecological Systems Approach.. Stroudsburg: Dowden, Hutchinson & Ross.
  • Eggleston PA, Rosenstreich D, Lynn H, Gergen P, Baker D, Kattan M, Mortimer KM, Mitchell H, Ownby D, Slavin R, Malveaux F. 1998. Relationship of indoor allergen exposure to skin test sensitivity in inner-city children with asthma. Journal of Allergy and Clinical Immunology 102(4 Pt 1):563–570. [PubMed: 9802363]
  • Eggleston PA, Wood RA, Rand C, Nixon WJ, Chen PH, Lukk P. 1999. Removal of cockroach allergen from inner-city homes. Journal of Allergy and Clinical Immunology 104(4 Pt 1):842-846. [PubMed: 10518830]
  • Epton MJ, Martin IR, Graham P, Healy PE, Smith H, Balasubramaniam R, Harvey IC, Fountain DW, Hedley J, Town GI. 1997. Climate and aeroallergen levels in asthma: a 12 month prospective study. Thorax 52(6):528–534. [PMC free article: PMC1758584] [PubMed: 9227719]
  • Erel F, Karaayvaz M, Caliskaner Z, Ozanguc N. 1998. The allergen spectrum in Turkey and the relationships between allergens and age, sex, birth month, birthplace, blood groups and family history of atopy. Journal of Investigational Allergology and Clinical Immunology 8(4):226–233. [PubMed: 9777537]
  • Eriksson NE, Holmen A. 1996. Skin prick tests with standardized extracts of inhalant allergens in 7,099 adult patients with asthma or rhinitis: cross-sensitizations and relationships to age, sex, month of birth and year of testing. Journal of Investigational Allergology and Clinical Immunology 6(1):36–46. [PubMed: 8833167]
  • Fadel R, David B, Paris S, Guesdon JL. 1992. Alternaria spore and mycelium sensitivity in allergic patients: in vivo and in vitro studies. Annals of Allergy 69(4):329–335. [PubMed: 1384402]
  • Fearon DT, Locksley RM. 1996. The instructive role of innate immunity in the acquired immune response. Science 272(5258):50–53. [PubMed: 8600536]
  • Fisk WJ. 1999. Estimates of potential nationwide productivity and health benefits from better indoor environments: an update. In: Indoor Air Quality Handbook. Spengler J, editor; , Samet JM, editor; , McCarthy JF, editor. , Eds. New York: McGraw Hill.
  • Flaherty DK., Deck FH, Cooper J, Bishop K, Winzenburger PA, Smith LR, Bynum L, Witmer WB. 1984. Bacterial endotoxin isolated from a water spray air humidification system as a putative agent of occupation-related lung disease. Infection and Immunity 43(1):206–212. [PMC free article: PMC263411] [PubMed: 6690401]
  • Fogelmark B, Sjostrand M, Rylander R. 1994. Pulmonary inflammation induced by repeated inhalations of beta(1,3)-D-glucan and endotoxin. International Journal of Experimental Pathology 75(2):85–90. [PMC free article: PMC2002110] [PubMed: 8199009]
  • Freymuth F, Vabret A, Brouard J, Toutain F, Verdon R, Petitjean J, Gouarin S, Duhamel JF, Guillois B. 1999. Detection of viral, Chlamydia pneumoniae and Mycoplasma pneumoniae infections in exacerbations of asthma in children. Journal of Clinical Virology 13(3):131–139. [PMC free article: PMC7129880] [PubMed: 10443789]
  • Frick OL, German DF, Mills J. 1979. Development of allergy in children. I. Association with virus infections. Journal of Allergy and Clinical Immunology 63(4):228–241. [PMC free article: PMC7133211] [PubMed: 85648]
  • Frost A. 1988. Frequency of allergy to Alternaria and Cladosporium in a specialist clinic. Allergy 43(7):504–507. [PubMed: 3232761]
  • Fukuda T, Mochida S, Fuckushima Y, Makino S. 1995. Detection of allergeninduced genes in peripheral blood mononuclear cells of patients with allergic asthma using subtractive hybridization. Journal of Allergy and Clinical Immunology 96(6 Pt 2):1076–1082. [PubMed: 8543765]
  • Galant S, Berger W, Gillman S, Goldsobel A, Incaudo G, Kanter L, Machtinger S, McLean A, Prenner B, Sokol W, Spector S, Welch M, Ziering W. 1998. Prevalence of sensitization to aeroallergens in California patients with respiratory allergy. Allergy Skin Test Project Team. Annals of Allergy, Asthma, and Immunology 81(3):203–210. [PubMed: 9759795]
  • Ganier M, Lieberman P, Fink J, Lockwood DG. 1980. Humidifier lung. An outbreak in office workers. Chest 77(2):183–187. [PubMed: 6766370]
  • Garcia DP, Corbett ML, Sublett JL, Pollard SJ, Meiners JF, Karibo JM, Pence HL, Petrosko JM. 1994. Cockroach allergy in Kentucky: a comparison of inner city, suburban, and rural small town populations. Annals of Allergy 72(3):203–208. [PubMed: 8129212]
  • Garrett MH, Rayment PR, Hooper MA, Abramson MJ, Hooper BM. 1998. Indoor airborne fungal spores, house dampness and associations with environmental factors and respiratory health in children. Clinical and Experimental Allergy 28(4):459–467. [PubMed: 9641573]
  • Gelber LE, Seltzer LH, Bouzoukis JK, Pollart SM, Chapman MD, Platts-Mills TA. 1993. Sensitization and exposure to indoor allergens as risk factors for asthma among patients presenting to hospital. American Review of Respiratory Disease 147(3):573–578. [PubMed: 8442589]
  • Gerberick GF, Sorenson WG, Lewis DM. 1984. The effects of T-2 toxin on alveolar macrophage function in vitro. Environmental Research 33(1):246–260. [PubMed: 6363056]
  • Gergen PJ, Turkeltaub PC. 1992. The association of individual allergen reactivity with respiratory disease in a national sample: data from the second National Health and Nutrition Examination Survey, 1976–80 (NHANES II). Journal of Allergy and Clinical Immunology 90(4 Pt 1):579–588. [PubMed: 1401641]
  • Gergen PJ, Mortimer KM, Eggleston PA, Rosenstreich D, Mitchell H, Ownby D, Kattan M, Baker D, Wright EC, Slavin R, Malveaux F. 1999. Results of the National Cooperative Inner-City Asthma Study (NCICAS) environmental intervention to reduce cockroach allergen exposure in inner-city homes. Journal of Allergy and Clinical Immunology 103(3 Pt 1):501–506. [PubMed: 10069886]
  • Giavina-Bianchi PF Jr, Castro FF, Machado ML, Duarte AJ. 1997. Occupational respiratory allergic disease induced by Passiflora alata and Rhamnus purshiana. Annals of Allergy, Asthma, and Immunology 79(5):449–454. [PubMed: 9396980]
  • Glezen WP, Paredes A, Allison JE, Taber LH, Frank AL. 1981. Risk of respiratory syncytial virus infection for infants from low-income families in relationship to age, sex, ethnic group, and maternal antibody level. Journal of Pediatrics 98(5):708–715. [PubMed: 7229749]
  • Gold DR, Burge HA, Carey V, Milton DK, Platts-Mills T, Weiss ST. 1999. Predictors of repeated wheeze in the first year of life. The relative roles of cockroach, birth weight, acute lower respiratory illness, and maternal smoking. American Journal of Respiratory and Critical Care Medicine 160(1):227–236. [PubMed: 10390405]
  • Gottlieb DJ, Sparrow D, O'Connor GT, Weiss ST. 1996. Skin test reactivity to common aeroallergens and decline of lung function. The Normative Aging Study. American Journal of Respiratory and Critical Care Medicine 153(2):561– 566. [PubMed: 8564098]
  • Grant C, Hunter CA, Flannigan B, Bravery AF. 1989. The moisture requirements of moulds isolated from domestic dwellings. International Biodeterioration and Biodegradation 25:259–284.
  • Griffith IJ, Craig S, Pollock J, Yu XB, Morgenstern JP, Rogers BL. 1992. Expression and genomic structure of the genes encoding FdI, the major allergen from the domestic cat. Gene 113(2):263–268. [PubMed: 1572548]
  • Gross NJ. 1980. Allergy to laboratory animals: epidemiologic, clinical, and physiologic aspects, and a trial of cromolyn in its management. Journal of Allergy and Clinical Immunology 66(2):158–165. [PubMed: 6772703]
  • Grunberg K, Sterk PJ. 1999. Rhinovirus infections: induction and modulation of airways inflammation in asthma. Clinical and Experimental Allergy. 29(Supplement 2):65–73. [PMC free article: PMC7162073] [PubMed: 10421825]
  • Guneser S, Atici A, Koksal F, Yaman A. 1994. Mold allergy in Adana, Turkey. Allergologia et Immunopathologia 22(2):52–54. [PubMed: 8059675]
  • Habenicht HA, Burge HA, Muilenberg ML, Solomon WR. 1984. Allergen carriage by atmospheric aerosol. II. Ragweed-pollen determinants in submicronic atmospheric fractions. Journal of Allergy and Clinical Immunology 74(1):64– 67. [PubMed: 6203953]
  • Hahn DL, Dodge RW, Golubjatnikov R. 1991. Association of Chlamydia pneumoniae (strain TWAR) infection with wheezing, asthmatic bronchitis, and adult-onset asthma. Journal of the American Medical Association 266(2):225–230. [Comment in: JAMA 1991. 266(2):265.] [PubMed: 2056624]
  • Hahn DL, McDonald R. 1998. Can acute Chlamydia pneumoniae respiratory tract infection initiate chronic asthma? Annals of Allergy, Asthma, and Immunology 81(4):339–344. [PubMed: 9809498]
  • Hall CB, Kopelman AE, Douglas RG Jr, Geiman JM, Meagher MP. 1979. Neonatal respiratory syncytial virus infection. New England Journal of Medicine 300(8):393–396. [PubMed: 759915]
  • Halonen M, Stern DA, Wright AL, Taussig LM, Martinez FD. 1997. Alternaria as a major allergen for asthma in children raised in a desert environment. American Journal of Respiratory and Critical Care Medicine 155(4):1356–1361. [PubMed: 9105079]
  • Hanania NA, Tarlo SM, Silverman F, Urch B, Senathirajah N, Zamel N, Corey P. 1998. Effect of exposure to low levels of ozone on the response to inhaled allergen in allergic asthmatic patients. Chest 114(3):752–756. [PubMed: 9743162]
  • Harju T, Keistinen T, Tuuponen T, Kivela SL. 1997. Seasonal variation in childhood asthma hospitalisations in Finland, 1972–1992. European Journal of Pediatrics 156(6):436–439. [PubMed: 9208236]
  • Harving H, Korsgaard J, Dahl R. 1994. House-dust mite exposure reduction in specially designed mechanically ventilated “healthy” homes. Allergy 49(9): 713–718. [PubMed: 7695059]
  • Hayden ML, Rose G, Diduch KB, Domson P, Chapman MD, Heymann PW, Platts-Mills TA. 1992. Benzyl benzoate moist powder: investigation of acaricidal activity in cultures and reduction of dust mite allergens in carpets. Journal of Allergy and Clinical Immunology 89(2):536–545. [PubMed: 1740584]
  • Hayden ML, Perzanowski M, Matheson L, Scott P, Call RS, Platts-Mills TA. 1997. Dust mite allergen avoidance in the treatment of hospitalized children with asthma. Annals of Allergy, Asthma, and Immunology 79(5):437–442. [PubMed: 9396978]
  • Hedlin G, Heilborn H, Lilja G, Norrlind K, Pegelow KO, Schou C, Lowenstein H. 1995. Long-term follow-up of patients treated with a three-year course of cat or dog immunotherapy. Journal of Allergy and Clinical Immunology 96(6 Pt 1):879–885. [PubMed: 8543744]
  • Hegele RG. 1999. Infection by asthma-associated viruses: clinical implications. Medscape Respiratory Care 3(1). URL:http://www​.medscape.com​/Medscape/RespiratoryCare​/journal/1999/v03​.n01/mrc4638.volc/mrc5212​.hege/mrc5212.hege-01.html. Accessed August 25, 1999.
  • Helbling A, Gayer F, Pichler WJ, Brander KA. 1998. Mushroom (Basidiomycete) allergy: diagnosis established by skin test and nasal challenge. Journal of Allergy and Clinical Immunology 102(5):853–858. [PubMed: 9819305]
  • Henderson FW, Henry MM, Ivins SS, Morris R, Neebe EC, Leu SY, Stewart PW. 1995. Correlates of recurrent wheezing in school-age children. The Physicians of Raleigh Pediatric Associates. American Journal of Respiratory and Critical Care Medicine 151(6):1786–1793. [PubMed: 7767521]
  • Hendrick DJ, Faux JA, Marshall R. 1978. Budgerigar-fancier's lung: the commonest variety of allergic alveolitis in Britain. British Medical Journal 2(6130):81–84. [PMC free article: PMC1605890] [PubMed: 566603]
  • Hesselmar B, Åberg N, Åberg B, Eriksson B, Björkstén B. 1999. Does early exposure to cat or dog protect against later allergy development? Clinical and Experimental Allergy 29(5):611–617. [PubMed: 10231320]
  • Hewitt CR, Brown AP, Hart BJ, Pritchard DI. 1995. A major house dust mite allergen disrupts the immunoglobulin E network by selectively cleaving CD23: innate protection by antiproteases. Journal of Experimental Medicine 182(5):1537–1544. [PMC free article: PMC2192194] [PubMed: 7595223]
  • Hide DW, Matthews S, Matthews L, Stevens M, Ridout S, Twiselton R, Gant C, Arshad SH. 1994. Effect of allergen avoidance in infancy on allergic manifestations at age two years. Journal of Allergy and Clinical Immunology 93(5):842–846. [PubMed: 8182225]
  • Hill AB. 1965. The environment and disease: association or causation. Proceedings of the Royal Society of Medicine 58:295–300. [PMC free article: PMC1898525] [PubMed: 14283879]
  • Hizawa N, Freidhoff LR, Ehrlich E, Chiu YF, Duffy DL, Schou CP, Dunston GM, Beaty TH, Marsh DG, Barnes KC, Huang SK. 1998. Genetic influences of chromosomes 5q31-q33 and 11q13 on specific IgE responsiveness to common inhaled allergens among African American families. Collaborative Study on the Genetics of Asthma (CSGA). Journal of Allergy and Clinical Immunology 102(3):449–453. [PubMed: 9768587]
  • Hodson T, Custovic A, Simpson A, Chapman M, Woodcock A, Green R. 1999. Washing the dog reduces dog allergen levels, but the dog needs to be washed twice a week. Journal of Allergy and Clinical Immunology 103(4):581–585. [PubMed: 10200004]
  • Hollander A, Doekes G, Heederik D. 1996. Cat and dog allergy and total IgE as risk factors of laboratory animal allergy. Journal of Allergy and Clinical Immunology 98(3):545–554. [PubMed: 8828532]
  • Holmquist L, Vesterberg O. 1999. Quantification of birch and grass pollen allergens in indoor air. Indoor Air 9(2):85–91. [PubMed: 10390933]
  • Horak B, Dutkiewicz J, Solarz K. 1996. Microflora and acarofauna of bed dust from homes in Upper Silesia, Poland. Annals of Allergy, Asthma, and Immunology 76(1):41–50. [PubMed: 8564627]
  • Horner WE, Reese G, Lehrer SB. 1995. Identification of the allergen Psi c 2 from the basidiomycete Psilocybe cubensis as a fungal cyclophilin. International Archives of Allergy and Immunology 107(1–3):298–300. [PubMed: 7542081]
  • Horst M, Hejjaoui A, Horst V, Michel FB, Bousquet J. 1990. Double-blind, placebocontrolled rush immunotherapy with a standardized Alternaria extract. Journal of Allergy and Clinical Immunology 85(2):460–472. [PubMed: 2406323]
  • Hu FB, Persky V, Flay BR, Richardson J. 1997. An epidemiological study of asthma prevalence and related factors among young adults. Journal of Asthma 34(1):67–76. [PubMed: 9033442]
  • Ingold CT. 1971. Fungal spores: their liberation and dispersal. Oxford: Clarenden Press.
  • Ingram JM, Sporik R, Rose G, Honsinger R, Chapman MD, Platts-Mills TA. 1995. Quantitative assessment of exposure to dog (Can f I) and cat (Fel d I) allergens: relation to sensitization and asthma among children living in Los Alamos, New Mexico. Journal of Allergy and Clinical Immunology 96(4):449–456. [PubMed: 7560654]
  • IOM (Institute of Medicine). 1993. Indoor Allergens. Pope AM, editor; , Patterson R, editor; , Burge H, editor. , eds. Washington, DC. National Academy Press.
  • Iversen M, Pedersen B. 1990. The prevalence of allergy in Danish farmers. Allergy 45(5):347–353. [PubMed: 2378438]
  • Iwasaki Y, Hara Y, Koji T, Shibata Y, Nakane PK, Kato I. 1998. Differential expression of IFN-gamma, IL-4, IL-10, and IL-1beta mRNAs in decalcified tissue sections of mouse lipopolysaccharide-induced periodontitis mandibles assessed by in situ hybridization. Histochemistry and Cell Biology 109(4):339– 347. [PubMed: 9562383]
  • Jaakkola JJ, Jaakkola N, Ruotsalainen R. 1993. Home dampness and molds as determinants of respiratory symptoms and asthma in pre-school children. Journal of Exposure Analysis and Environmental Epidemiology 3(Suppl 1):129–142. [PubMed: 9857299]
  • Jakab GJ, Hmieleski RR, Zarba A, Hemenway DR, Groopman JD. 1994. Respiratory aflatoxicosis—suppression of pulmonary and systemic host defenses in rats and mice. Toxicology and Applied Pharmacology 125(2): 198–205. [PubMed: 8171428]
  • Johnston SL. 1997. Influence of viral and bacterial respiratory infections on exacerbations and symptom severity in childhood asthma. Pediatric Pulmonology Supplement 16:88–89. [PubMed: 9443219]
  • Johnston SL. 1999. The role of viral and atypical bacterial pathogens in asthma pathogenesis. Pediatric Pulmonology Supplement 18:141–143. [PubMed: 10093125]
  • Johnston SL, Pattemore PK, Sanderson G, Smith S, Lampe F, Josephs L, Symington P, O'Toole S, Myint SH, Tyrrell DA, Holgate ST. 1995. Community study of role of viral infections in exacerbations of asthma in 9–11 year old children. British Medical Journal 310(6989):1225–1229. [Comment in BMJ 1995. 311(7005):629–630.] [PMC free article: PMC2549614] [PubMed: 7767192]
  • Kaad PH, Ostergaard PA. 1982. The hazard of mould hyposensitization in children with asthma. Clinical Allergy 12(3):317–320. [PubMed: 7105396]
  • Kalman S, Mitchell W, Marathe R, Lammel C, Fan J, Hyman RW, Olinger L, Grimwood J, Davis RW, Stephens RS. 1999. Comparative genomes of Chlamydia pneumoniae and C. trachomatis. Nature Genetics 21(4):385–389. [PubMed: 10192388]
  • Kang B. 1976. Study on cockroach antigen as a probable causative agent in bronchial asthma. Journal of Allergy and Clinical Immunology 58(3):357–365. [PubMed: 965615]
  • Kang BC, Johnson J, Veres-Thorner C. 1993. Atopic profile of inner–city asthma with a comparative analysis on the cockroach-sensitive and ragweed-sensitive subgroups. Journal of Allergy and Clinical Immunology 92(6):802–811. [PubMed: 8258614]
  • Kang BC, Zhou K, Lai YL, Hong CB. 1996. Experimental asthma developed by room air contamination with cockroach allergen. International Archives of Allergy and Immunology 111(3):299–306. [PubMed: 8917126]
  • Karn RC. 1994. The mouse salivary androgen-binding protein (ABP) alpha subunit closely resembles chain 1 of the cat allergen Fel d I. Biochemical Genetics 32(7–8):271–277. [PubMed: 7826314]
  • Kattan M, Mitchell H, Eggleston P, Gergen P, Crain E, Redline S, Weiss K, Evans R III, Kaslow R, Kercsmar C, Leickly F, Malveaux F, Wedner HJ. 1997. Characteristics of inner-city children with asthma: the National Cooperative Inner-City Asthma Study. Pediatric Pulmonology 24(4):253–262. [PubMed: 9368259]
  • Katz Y, Verleger H, Barr J, Rachmiel M, Kiviti S, Kuttin ES. 1999. Indoor survey of moulds and prevalence of mould atopy in Israel. Clinical and Experimental Allergy 29(2):186–192. [PubMed: 10051722]
  • Kemp TJ, Siebers RW, Fishwick D, O'Grady GB, Fitzharris P, Crane J. 1996. House dust mite allergen in pillows [see comments]. British Medical Journal 313(7062):916. [PMC free article: PMC2352227] [PubMed: 8876094]
  • Kerrebijn KF. 1970. Endogenous factors in childhood CNSLD: methodological aspects in population studies. In: Bronchitis III, editor; . Orie NGM, editor; , van der Lende R, editor. , Eds. The Netherlands: Royal Vangorcum Assen, pp.38–48.
  • Kiho T, Sakushima M, Wang SR, Nagai K, Ukai S. 1991. Polysaccharides in fungi. XXVI. Two branched (1→3)-beta-D-glucans from hot water extract of Yu er. Chemical and Pharmaceutical Bulletin 39(3):798–800. [PubMed: 2070468]
  • Kilpeläinen M, Terho EO, Koskenvuo M. 1997. Asthma and atopic diseases among Finish university students. European Respiratory Journal 10(Supplement):143s.
  • Kirschning CJ, Wesche H, Merrill Ayres T, Rothe M. 1998. Human toll-like receptor 2 confers responsiveness to bacterial lipopolysaccharide. Journal of Experimental Medicine 188(11):2091–2097. [PMC free article: PMC2212382] [PubMed: 9841923]
  • Kitamura S, Hori T, Kurita K, Takeo K, Hara C, Itoh W, Tabata K, Elgasaeter A, Stokke BT. 1994. An antitumor, branched (1→3)-beta-D-glucan from a water extract of fruiting bodies of Cryptoporus volvatus. Carbohydrate Research 263(1):111–121. [PubMed: 7982226]
  • Klucka CV, Ownby DR, Green J, Zoratti E. 1995. Cat shedding of Fel d I is not reduced by washings, Allerpet-C spray, or acepromazine. Journal of Allergy and Clinical Immunology 95(6):1164–1171. [PubMed: 7797784]
  • Knox RB, Suphioglu C, Taylor P, Desai R, Watson HC, Peng JL, Bursill LA. 1997. Major grass pollen allergen Lol p I binds to diesel exhaust particles: implications for asthma and air pollution. Clinical and Experimental Allergy 27(3):246–251. [PubMed: 9088650]
  • Koehler PG, Patterson RS, Brenner RJ. 1987. German cockroach (Orthoptera, Blattellidae) infestations in low-income apartments. Journal of Economic Entomology 80(2):446–450. [PubMed: 3584641]
  • Kohler F, Kohler C, Patris A, Grillat JP. 1983. Fréquence de l'allergie pollinique chez les agriculteurs par rapport aux autres catégories socio-professionnelles. Revue Francaise D Allergologie 23(119–124).
  • Kongpanichkul A, Vichyanond P, Tuchinda M. 1997. Allergen skin test reactivities among asthmatic Thai children. Journal of the Medical Association of Thailand 80(2):69–75. [PubMed: 9078689]
  • Kraft M, Cassell GH, Henson JE, Watson H, Williamson J, Marmion BP, Gaydos CA, Martin RJ. 1998. Detection of Mycoplasma pneumoniae in the airways of adults with chronic asthma. American Journal of Respiratory and Critical Care Medicine 158(3):998–1001. [Published erratum appears in Am J Respir Crit Care Med 1998. 158(5 Pt 1):1692.] [PubMed: 9731038]
  • Kraus J, Franz G. 1991. β(1-3)Glucans: anti-tumor activity and immunostimulation. In: Fungal cell wall and immune response. Latge JP, editor; , Boucias D, editor. , Eds. Berlin, Heidelberg: SpringerVerlag. NATO ASI series, H53:431–444.
  • Kuehr J, Frischer T, Meinert R, Barth R, Forster J, Schraub S, Urbanek R, Karmaus W. 1994. Mite allergen exposure is a risk factor for the incidence of specific sensitization. Journal of Allergy and Clinical Immunology 94(1):44–52. [PubMed: 8027498]
  • Larsen FO, Norn S, Mordhorst CH, Skov PS, Milman N, Clementsen P. 1998. Chlamydia pneumoniae and possible relationship to asthma. Serum immunoglobulins and histamine release in patients and controls. APMIS 106(10):928– 934. [PubMed: 9833693]
  • Laurent J, Decoux L, Ickovic MR, Le Gall C, Gacouin JC, Sauvaget J, Lafay M. 1994. Winter pollinosis in Paris. Allergy 49(9):696–701. [PubMed: 7695057]
  • Lehrer SB, Lopez M, Butcher BT, Olson J, Reed M, Salvaggio JE. 1986. Basidiomycete mycelia and spore-allergen extracts: skin test reactivity in adults with symptoms of respiratory allergy. Journal of Allergy and Clinical Immunology 78(3 Pt 1):478–485. [PubMed: 3760405]
  • Lelong M, Henard J, Wattre P, Duprey J, Thelliez P, Miersman R. 1986. Does immediate-type respiratory allergy occur regarding Stemphylium? Evaluation of 39 challenge tests [French]. Allergie et Immunologie 18(8):21, 23,25–6. [PubMed: 3453724]
  • Lemanske RF Jr, Dick EC, Swenson CA, Vrtis RF, Busse WW. 1989. Rhinovirus upper respiratory infection increases airway hyperreactivity and late asthmatic reactions. Journal of Clinical Investigation. 83(1):1–10. [PMC free article: PMC303635] [PubMed: 2536042]
  • Levetin E, Buck P. 1986. Evidence of mountain cedar pollen in Tulsa. Annals of Allergy 56(4):295–299. [PubMed: 3963520]
  • Li CS, Hsu LY. 1997. Airborne fungus allergen in association with residential characteristics in atopic and control children in a subtropical region. Archives of Environmental Health 52(1):72–79. [PubMed: 9039862]
  • Li DW, Kendrick B. 1995. A year-round study on functional relationships of airborne fungi with meteorological factors. International Journal of Biometeorology 39(2):74–80. [PubMed: 8530208]
  • Licorish K, Novey HS, Kozak P, Fairshter RD, Wilson AF. 1985. Role of Alternaria and Penicillium spores in the pathogenesis of asthma. Journal of Allergy and Clinical Immunology 76(6):819–825. [PubMed: 4067131]
  • Lind P, Norman PS, Newton M, Lowenstein H, Schwartz B. 1987. The prevalence of indoor allergens in the Baltimore area: house dust-mite and animal-dander antigens measured by immunochemical techniques. Journal of Allergy and Clinical Immunology 80(4):541–547. [PubMed: 3668118]
  • Litonjua AA, Sparrow D, Weiss ST, O'Connor GT, Long AA, Ohman JL Jr. 1997. Sensitization to cat allergen is associated with asthma in older men and predicts new-onset airway hyperresponsiveness. The Normative Aging Study. American Journal of Respiratory and Critical Care Medicine 156(1):23–27. [PubMed: 9230721]
  • Longbottom JL, Austwick PK. 1987. Allergy to rats: quantitative immunoelectrophoretic studies of rat dust as a source of inhalant allergen. Journal of Allergy and Clinical Immunology 80(3 Pt 1):243–251. [PubMed: 3624679]
  • Luczynska CM, Arruda LK, Platts-Mills TA, Miller JD, Lopez M, Chapman MD. 1989. A two-site monoclonal antibody ELISA for the quantitation of the major Dermatophagoides spp. allergens, Der p I and Der f I. Journal of Immunological Methods 118(2):227–235. [PubMed: 2926155]
  • Luyt DK, Davis G, Dance M, Simmank K, Patel D. 1995. Clinical characteristics of black asthmatic children. South African Medical Journal 85(10):999–1001. [PubMed: 8596993]
  • Madelin TM, Johnson HE. 1992. Fungal and actinomycete spore aerosols measured at different humidities with an aerodynamic particle sizer. Journal of Applied Bacteriology 72:400–409. [PubMed: 1618718]
  • Malling HJ. 1986. Diagnosis and immunotherapy of mould allergy. IV. Relation between asthma symptoms, spore counts and diagnostic tests. Allergy 41(5):342–350. [PubMed: 2429577]
  • Martinez, FD. 1994. Role of viral infections in the inception of asthma and allergies during childhood: Could they be protective? Thorax 49(12):1189– 1191. [PMC free article: PMC475320] [PubMed: 7878550]
  • Martinez FD. 1995. Viral infections and the development of asthma. American Journal of Respiratory and Critical Care Medicine 151(5):1644–1648. [PubMed: 7735627]
  • Medzhitov R., Preston-Hurlburt P, Janeway CA Jr. 1997. A human homologue of the Drosophila Toll protein signals activation of adaptive immunity. Nature 388(6640):394–397. [PubMed: 9237759]
  • Michel O, Duchateau J, Sergysels R. 1989. Effect of inhaled endotoxin on bronchial reactivity in asthmatic and normal subjects. Journal of Applied Physiology 66(3):1059–1064. [PubMed: 2708231]
  • Michel O, Kips J, Duchateau J, Vertongen F, Robert L, Collet H, Pauwels R, Sergysels R. 1996. Severity of asthma is related to endotoxin in house dust. American Journal of Respiratory and Critical Care Medicine 154:1641–1646. [PubMed: 8970348]
  • Micillo E, Marcatili P, Palmieri S, Mazzarella G. 1998. Viruses and asthmatic syndromes. Monaldi Archives for Chest Disease 53(1):88–91. [PubMed: 9632915]
  • Miles EA, Warner JA, Jones AC, Colwell BM, Bryant TM, Warner JO. 1996. Peripheral blood mononuclear cell proliferative responses in the first year of life in babies born to allergic parents. Clinical and Experimental Allergy 26(7):780–788. [PubMed: 8842551]
  • Milton DK. 1999. Endotoxin and other bacterial cell-wall components. In: Bioaerosols: Assessment and Control. Macher J, editor; , Milton DK, editor; , Burge HA, editor; , Morey P, editor. , Eds. Cincinnati, OH: American Conference of Governmental Industrial Hygienists.
  • Milton DK, Johnson DK. 1995. Endotoxin exposure assessment in machining operations. In: The Industrial Metalworking Environment: Assessment and Control. Dearborn, MI: American Automobile Manufacturers Association.
  • Milton DK, Wypij D, Kriebel D, Walters M, Hammond SK, Evans JS. 1996. Endotoxin exposure–response in a fiberglass manufacturing plant. American Journal of Industrial Medicine 29(1):3–13. [PubMed: 8808037]
  • Milton DK, Johnson DK, Park JH. 1997. Environmental endotoxin measurement: interference and sources of variation in the Limulus assay of house dust. American Industrial Hygiene Association Journal 58:861–867. [PubMed: 9425646]
  • Miyamoto T, Oshima S, Ishizaka T, Sato SH. 1968. Allergenic identity between the common floor mite (Dermatophagoides farinae, Hughes 1961) and house dust as a causative antigen in bronchial asthma. Journal of Allergy and Clinical Immunology 42(1):14–28. [PubMed: 5243795]
  • Miyashita N, Kubota Y, Nakajima M, Niki Y, Kawane H, Matsushima T. 1998. Chlamydia pneumoniae and exacerbations of asthma in adults. Annals of Allergy, Asthma, and Immunology 80(5):405–409. [PMC free article: PMC7128287] [PubMed: 9609611]
  • Morgenstern JP, Griffith IJ, Brauer AW, Rogers BL, Bond JF, Chapman MD, Kuo MC. 1991. Amino acid sequence of Fel d I, the major allergen of the domestic cat: protein sequence analysis and cDNA cloning. Proceedings of the National Academy of Sciences of the United States of America 88(21):9690–9694. [PMC free article: PMC52784] [PubMed: 1946388]
  • Muilenberg ML, Skellenger WS, Burge HA, Solomon WR. 1991. Particle penetration into the automotive interior. I. Influence of vehicle speed and ventilatory mode. Journal of Allergy and Clinical Immunology 87(2):581–585. [PubMed: 1993816]
  • Munir AK, Andersson R, Einersson R, Schou C, Dreborg S. 1992. The amount of dog allergen in dust from Swedish schools is much higher than that of cat. Allergy 47:223.
  • Munir AK, Bjorksten B, Einarsson R, Schou C, Ekstrand-Tobin A, Warner A, Kjellman NI. 1994. Cat (Fel d I), dog (Can f I), and cockroach allergens in homes of asthmatic children from three climatic zones in Sweden. Allergy 49(7):508– 516. [PubMed: 7825716]
  • Munuera Giner M, Carrion Garcia JS, Garcia Selles J. 1998. Incidence of Alternaria spores in the atmosphere of Murcia (SE Spain). Seasonal, monthly and intradiurnal variations. Journal of Investigational Allergology and Clinical Immunology 8(5):304–308. [PubMed: 9827428]
  • Murayama T, Amitani R, Ikegami Y, Nawada R, Lee WJ, Kuze F. 1996. Suppressive effects of Aspergillus fumigatus culture filtrates on human alveolar macrophages and polymorphonuclear leucocytes. European Respiratory Journal 9(2):293–300. [PubMed: 8777967]
  • Murray AB, Ferguson AC. 1983. Dust-free bedrooms in the treatment of asthmatic children with house dust or house dust mite allergy: a controlled trial. Pediatrics 71(3):418–422. [PubMed: 6338475]
  • Murray M, Webb MS, O'Callaghan C, Swarbrick AS, Milner AD. 1992. Respiratory status and allergy after bronchiolitis. Archives of Disease in Childhood 67(4):482–487. [PMC free article: PMC1793348] [PubMed: 1580676]
  • Neas LM, Dockery DW, Burge H, Koutrakis P, Speizer FE. 1996. Fungus spores, air pollutants, and other determinants of peak expiratory flow rate in children. American Journal of Epidemiology 143(8):797–807. [PubMed: 8610690]
  • Nelson HS, Ikle D, Buchmeier A. 1996. Studies of allergen extract stability: the effects of dilution and mixing. Journal of Allergy and Clinical Immunology 98(2):382–388. [PubMed: 8757215]
  • Nelson RP Jr, DiNicolo R, Fernandez-Caldas E, Seleznick MJ, Lockey RF, Good RA. 1996. Allergen-specific IgE levels and mite allergen exposure in children with acute asthma first seen in an emergency department and in nonasthmatic control subjects. Journal of Allergy and Clinical Immunology 98(2):258–263. [PubMed: 8757201]
  • Newman-Taylor AJ. 1982. Laboratory animal allergy. European Journal of Respiratory Diseases 63:60–64. [PubMed: 6962083]
  • Newson R, Strachan D, Archibald E, Emberlin J, Hardaker P, Collier C. 1997. Effect of thunderstorms and airborne grass pollen on the incidence of acute asthma in England, 1990–94. Thorax 52(8):680–685. [Comment in Thorax 1997. 52(8):669–670.] [PMC free article: PMC1758625] [PubMed: 9337825]
  • Newson R, Strachan D, Archibald E, Emberlin J, Hardaker P, Collier C. 1998. Acute asthma epidemics, weather and pollen in England, 1987–1994. European Respiratory Journal 11(3):694–701. [PubMed: 9596123]
  • Nicholson KG, Kent J, Ireland DC. 1993. Respiratory viruses and exacerbations of asthma in adults. British Medical Journal 307(6910):982–986. [PMC free article: PMC1679193] [PubMed: 8241910]
  • Nicolai T, Illi S, von Mutius E. 1998. Effect of dampness at home in childhood on bronchial hyperreactivity in adolescence. Thorax 53(12):1035–1040. [PMC free article: PMC1745148] [PubMed: 10195075]
  • Nikulin M, Reijula K, Jarvis BB, Hintikka EL. 1996. Experimental lung mycotoxicosis in mice induced by Stachybotrys atra. International Journal of Experimental Pathology 77(5):213–218. [PMC free article: PMC2691636] [PubMed: 8977373]
  • Nikulin M, Reijula K, Jarvis BB, Veijalainen P, Hintikka EL. 1997. Effects of intranasal exposure to spores of Stachybotrys atra in mice. Fundamental and Applied Toxicology 35(2):182–188. [PubMed: 9038239]
  • Norbäck D, Björnsson E, Janson C, Palmgren U, Boman G. 1999. Current asthma and biochemical signs of inflammation in relation to building dampness in dwellings. International Journal of Tuberculosis and Lung Disease 3(5):368– 376. [PubMed: 10331724]
  • Norman PS, Ohman JL Jr, Long AA, Creticos PS, Gefter MA, Shaked Z, Wood RA, Eggleston PA, Hafner KB, Rao P, Lichtenstein LM, Jones NH, Nicodemus CF. 1996. Treatment of cat allergy with T-cell reactive peptides. American Journal of Respiratory and Critical Care Medicine 154(6 Pt 1):1623–1628. [PubMed: 8970345]
  • Nowak D, Heinrich J, Jorres R, Wassmer G, Berger J, Beck E, Boczor S, Claussen M, Wichmann HE, Magnussen H. 1996. Prevalence of respiratory symptoms, bronchial hyperresponsiveness and atopy among adults: West and East Germany. European Respiratory Journal 9(12):2541–2552. [PubMed: 8980967]
  • O'Hollaren MT, Yunginger JW, Offord KP, Somers MJ, O'Connell EJ, Ballard DJ, Sachs MI. 1991. Exposure to an aeroallergen as a possible precipitating factor in respiratory arrest in young patients with asthma. New England Journal of Medicine 324(6):359–363. [PubMed: 1987459]
  • O'Neil CE, Horner WE, Reed MA, Lopez M., Lehrer SB. 1990. Evaluation of basidiomycete and deuteromycete (Fungi imperfecti) extracts for shared allergenic determinants. Clinical and Experimental Allergy 20(5):533–538. [PubMed: 2253084]
  • Ordaz VA, Castaneda CB, Campos CL, Rodriguez VM, Saenz JG, Rios PC. 1998. Asthmatic exacerbations and environmental pollen concentration in La Comarca Lagunera (Mexico). Revista Alergia Mexico 45(4):106–111. [PubMed: 9796384]
  • Ormstad H, Johansen BV, Gaarder PI. 1998. Airborne house dust particles and diesel exhaust particles as allergen carriers. Clinical and Experimental Allergy 28(6):702–708. [PubMed: 9677134]
  • Orta JC, Navarro AM, Bartolome B, Delgado J, Martinez J, Sanchez MC, Martinez A, Valverdu A, Conde J, Palacios R. 1998. Comparative allergenic study of Tetranychus urticae from different sources. Journal of Investigational Allergology and Clinical Immunology 8(3):149–154. [PubMed: 9684187]
  • Park JH. 1999. Endotoxin in the home: exposure assessment and health effects. Environmental Health. Harvard University School of Public Health, Boston, MA.
  • Pasanen AL, Niininen M, Kalliokoski P, Nevalainen A, Jantunen MJ. 1992. Airborne Cladosporidium and other fungi in damp versus reference residences. Atmospheric Environment 26B(1):121–124.
  • Pattemore PK, Johnston SL, Bardin PG. 1992. Viruses as precipitants of asthma symptoms. I. Epidemiology. Clinical and Experimental Allergy 22(3):325–336. [PMC free article: PMC7162032] [PubMed: 1586873]
  • Peat JK, Toelle BG, Gray EJ, Haby MM, Belousova E, Mellis CM, Woolcock AJ. 1995. Prevalence and severity of childhood asthma and allergic sensitisation in seven climatic regions of New South Wales. Medical Journal of Australia 163(1):22–26 [see comments]. [PubMed: 7609683]
  • Peat JK, Tovey E, Toelle BG, Haby MM, Gray EJ, Mahmic A, Woolcock AJ. 1996. House dust mite allergens. A major risk factor for childhood asthma in Australia. American Journal of Respiratory and Critical Care Medicine 153(1):141–146. [PubMed: 8542107]
  • Perzanowski MS, Sporik R, Squillace SP, Gelber LE, Call R, Carter M, Platts-Mills TA. 1998. Association of sensitization to Alternaria allergens with asthma among school-age children. Journal of Allergy and Clinical Immunology 101(5):626–632. [PubMed: 9600499]
  • Piacentini GL, Martinati L, Mingoni S, Boner AL. 1996. Influence of allergen avoidance on the eosinophil phase of airway inflammation in children with allergic asthma. Journal of Allergy and Clinical Immunology 97(5):1079–1084. [PubMed: 8626985]
  • Platts-Mills TA, Tovey ER, Mitchell EB, Moszoro H, Nock P, Wilkins SR. 1982. Reduction of bronchial hyperreactivity during prolonged allergen avoidance. Lancet 2(8300):675–678. [PubMed: 6126624]
  • Platts-Mills TA, Longbottom J, Edwards J, Cockroft A, Wilkins S. 1987. Occupational asthma and rhinitis related to laboratory rats: serum IgG and IgE antibodies to the rat urinary allergen. Journal of Allergy and Clinical Immunology 79(3):505–515. [PubMed: 3819230]
  • Platts-Mills TA, de Weck AL. 1988. Dust mite allergens and asthma: a worldwide problem. Bulletin of the World Health Organization 66(6):769–780. [PMC free article: PMC2491145] [PubMed: 3069235]
  • Platts-Mills TA, Vervloet D, Thomas WR, Aalberse RC, Chapman MD. 1997. Indoor allergens and asthma: report of the Third International Workshop, Cuenca, Spain. Journal of Allergy and Clinical Immunology 100(6 Pt 1):S2– S24. [PubMed: 9438476]
  • Pollart SM, Mullins DE, Vailes LD, Hayden ML, Platts-Mills TA, Sutherland WM, Chapman MD. 1991. a. Identification, quantitation, and purification of cockroach allergens using monoclonal antibodies. Journal of Allergy and Clinical Immunology 87(2):511–521. [PubMed: 1993811]
  • Pollart SM, Smith TF, Morris EC, Gerber LE, Platts-Mills TA. 1991. b. Environmental exposure to cockroach allergens: analysis with monoclonal antibody-based enzyme immunoassays. Journal of Allergy and Clinical Immunology 87(2):505–510. [PubMed: 1993810]
  • Potekhin SA, Loseva OI, Tiktopulo EI, Dobritsa AP. 1999. Transition state of the rate-limiting step of heat denaturation of Cry3A delta-endotoxin. Biochemistry 38(13):4121–4127. [PubMed: 10194327]
  • Prahl P, Roed-petersen J. 1979. Type I allergy from cows in veterinary surgeons. Contact Dermatitis 5(1):33–38. [PubMed: 154386]
  • Prescott SL, Macaubas C, Smallacombe T, Holt BJ, Sly PD, Loh R, Holt PG. 1998. Reciprocal age-related patterns of allergen-specific T-cell immunity in normal vs. atopic infants. Clinical and Experimental Allergy 5(Supplement):39–44; discussion50–51. [PubMed: 9988446]
  • Pugin J, Schurer-Maly CC, Leturcq D, Moriarty A, Ulevitch RJ, Tobias PS. 1993. Lipopolysaccharide activation of human endothelial and epithelial cells is mediated by lipopolysaccharide-binding protein and soluble CD14. Proceedings of the National Academy of Sciences of the United States of America 90(7):2744–2748. [PMC free article: PMC46172] [PubMed: 7681988]
  • Pullan CR, Hey EN. 1982. Wheezing, asthma, and pulmonary dysfunction 10 years after infection with respiratory syncytial virus in infancy. British Medical Journal (Clinical Research Edition) 284(6330):1665–1669. [PMC free article: PMC1498624] [PubMed: 6805648]
  • Rains N, Siebers R, Crane J, Fitzharris P. 1999. House dust mite allergen (Der p I) accumulation on new synthetic and feather pillows [see comments]. Clinical and Experimental Allergy 29(2):182–185. [PubMed: 10051721]
  • Rakes GP, Arruda E, Ingram JM, Hoover GE, Zambrano JC, Hayden FG, Platts-Mills TAE, Heymann PW. 1999. Rhinovirus and respiratory syncytial virus in wheezing children requiring emergency care. American Journal of Respiratory and Critical Care Medicine 159(3):785–790. [PubMed: 10051251]
  • Rao CY. 1999. A quantitative risk assessment for Stachybotrys chartarum exposure. D. Sc. Thesis, Department of Environmental Health, Harvard School of Public Health, Boston. 152 pages.
  • Rautalahi M, Terho EO, Vohlonen I, Husman K. 1987. Atopic sensitization of dairy farmers to work-related and common allergens. European Journal of Respiratory Diseases 71(Supplement 152):155–164. [PubMed: 3478214]
  • Rautiala S, Reponen T, Nevalainen A, Husman T, Kalliokoski P. 1998. Control of exposure to airborne viable microorganisms during remediation of moldy buildings; report of three case studies. American Industrial Hygiene Association Journal 59(7):455–460. [PubMed: 9697292]
  • Richard JL, Thurston JR. 1975. Effect of aflatoxin on phagocytosis of Aspergillus fumigatus spores by rabbit alveolar macrophages. Applied Microbiology 30(1):44–47. [PMC free article: PMC187111] [PubMed: 1096823]
  • Richards G, Kolbe J, Fenwick J, Rea H. 1995. The effects of privet exposure on asthma morbidity. New Zealand Medical Journal 108(996):96–99. [PubMed: 7715885]
  • Rietschel ET, Brade H. 1992. Bacterial endotoxins. Scientific American 267(2): 55–61. [PubMed: 1641625]
  • Robinson D, Hamid Q, Bentley A, Ying S, Kay AB, Durham SR. 1993. Activation of CD4+ T cells, increased TH2-type cytokine mRNA expression, and eosinophil recruitment in bronchoalveolar lavage after allergen inhalation challenge in patients with atopic asthma. Journal of Allergy and Clinical Immunology 92(2):313–324. [PubMed: 8349942]
  • Romagnani S. 1992. Human TH1 and TH2 subsets: regulation of differentiation and role in protection and immunopathology. International Archives of Allergy Immunology 98(4):279–285. [PubMed: 1422257]
  • Roman M, Calhoun WJ, Hinton KL, Avendano LF, Simon V, Escobar AM, Gaggero A, Diaz PV. 1997. Respiratory syncytial virus infection in infants is associated with predominant Th-2-like response. American Journal of Respiratory and Critical Care Medicine 156(1):190–195. [PubMed: 9230746]
  • Ronmark E, Lundback B, Jonsson E, Platts-Mills T. 1998. Asthma, type-1 allergy and related conditions in 7-and 8-year-old children in northern Sweden: prevalence rates and risk factor pattern. Respiratory Medicine 92(2):316–324. [PubMed: 9616532]
  • Rosas I, McCartney HA, Payne RW, Calderon C, Lacey J, Chapela R, Ruiz-Velazco S. 1998. Analysis of the relationships between environmental factors (aeroallergens, air pollution, and weather) and asthma emergency admissions to a hospital in Mexico City. Allergy 53(4):394–401. [PubMed: 9574882]
  • Rosenstreich DL, Eggleston P, Kattan M, Baker D, Slavin RG, Gergen P, Mitchell H, McNiff-Mortimer K, Lynn H, Ownby D, Malveaux F. 1997. The role of cockroach allergy and exposure to cockroach allergen in causing morbidity among inner-city children with asthma. New England Journal of Medicine 336(19):1356–1363. [Comment in N Engl J Med 1997. 336(19):1382–1384 and 337(11):791–792.] [PubMed: 9134876]
  • Rylander R, Haglind P, Lundholm M, Mattsby I, Stenqvist K. 1978. Humidifier fever and endotoxin exposure. Clinical Allergy 8(5):511–516. [PubMed: 709796]
  • Rylander R, Morey P. 1982. Airborne endotoxin in industries processing vegetable fibers. American Industrial Hygiene Association Journal 43(11):811–812. [PubMed: 7168439]
  • Saito K, Nishijima M, Ohno N, Nagi N, Yadomae T, Miyazaki T. 1992. Activation of complement and Limulus coagulation system by an alkali-soluble glucan isolated from Omphalia lapidescens and its less branched derivatives. Chemical and Pharmaceutical Bulletin 40:1227–1230.
  • Sakaguchi M, Inouye S, Miyazawa H, Kamimura H, Kimura M, Yamazaki S. 1990. a. Evaluation of countermeasures for reduction of mouse airborne allergens. Laboratory Animal Science 40(6):613–615. [PubMed: 2172625]
  • Sakaguchi M, Inouye S, Yasueda H, Irie T, Yoshizawa S, Shida T. 1990. b. Measurement of allergens associated with dust mite allergy. II. Concentrations of airborne mite allergens (Der I and Der II) in the house. International Archives of Allergy and Applied Immunology 90(2):190–193. [PubMed: 2583857]
  • Sakamoto T, Ito K, Yamada M, Iguchi H, Ueda M, Matsuda Y, Torii S. 1990. Allergenicity of the osmophilic fungus Aspergillus restrictus evaluated by skin prick test and radioallergosorbent test [Japanese]. Arerugi—Japanese Journal of Allergology 39(11):1492–1498. [PubMed: 2288495]
  • Sakurai T, Ohno N, Yadomae T. 1994. Changes in immune mediators in mouse lung produced by administration of soluble (1→3)-beta-D-glucan. Biological and Pharmaceutical Bulletin 17(5):617–622. [PubMed: 7920419]
  • Sanchez H, Bush RK. 1994. Complete sequence of a cDNA encoding an Alternaria allergen. Journal of Allergy and Clinical Immunology 93:208 [abstract].
  • Santiago-Schwarz F, McHugh DM, Fleit HB. 1989. Functional analysis of monocyte-macrophages derived from nonadherent cord blood progenitor cells: correlation with the ontogeny of cell surface proteins. Journal of Leukocyte Biology 46(3):230–238. [PubMed: 2527285]
  • Saraf A, Larsson L, Burge H, Milton D. 1997. Quantification of ergosterol and 3-hydroxy fatty acids in settled house dust by gas chromatography mass spectrometry—comparison with fungal culture and determination of endotoxin by a Limulus amebocyte lysate assay. Applied and Environmental Microbiology 63:2554–2559. [PMC free article: PMC168553] [PubMed: 9212406]
  • Saraf A, Park JH, Milton DK, Larsson L. 1999. Use of quadrupole GC-MS and iontrap GC-MSMS for determining 3-hydroxy fatty acids in settled house dust: relation to endotoxin activity. Journal of Environmental Monitoring 2:163–168. [PubMed: 11529095]
  • Sarpong SB, Hamilton RG, Eggleston PA, Adkinson NF Jr. 1996. a. Socioeconomic status and race as risk factors for cockroach allergen exposure and sensitization in children with asthma. Journal of Allergy and Clinical Immunology 97(6):1393–1401. [PubMed: 8648037]
  • Sarpong SB, Wood RA, Eggleston PA. 1996. b. Short-term effect of extermination and cleaning on cockroach allergen Bla g II in settled dust. Annals of Allergy, Asthma, and Immunology 76(3):257–260. [PubMed: 8634880]
  • Sarpong SB, Wood RA, Karrison T, Eggleston PA. 1997. Cockroach allergen (Bla g 1) in school dust. Journal of Allergy and Clinical Immunology 99(4):486–492. [PubMed: 9111492]
  • Sarpong SB, Karrison T. 1998. a. Season of birth and cockroach allergen sensitization in children with asthma. Journal of Allergy and Clinical Immunology 101(4 Pt 1):566–568. [PubMed: 9564817]
  • Sarpong SB, Karrison T. 1998. b. Skin test reactivity to indoor allergens as a marker of asthma severity in children with asthma. Annals of Allergy, Asthma, and Immunology 80(4):303–308. [PubMed: 9564978]
  • Sarpong SB, Han Y. 1999. A threshold of cockroach allergen (Bla g II) exposure and sensitization. American Journal of Respiratory and Critical Care Medicine 156(3):A128.
  • Sastre J, Ibanez MD, Lombardero M, Laso MT, Lehrer S. 1996. Allergy to cockroaches in patients with asthma and rhinitis in an urban area (Madrid). Allergy 51(8):582–586. [PubMed: 8874663]
  • Schappi GF, Suphioglu C, Taylor PE, Knox RB. 1997. Concentrations of the major birch tree allergen Bet v I in pollen and respirable fine particles in the atmosphere. Journal of Allergy and Clinical Immunology 100(5):656–661. [PubMed: 9389296]
  • Schenkelberger V, Freitag M, Altmeyer P. 1998. Ficus benjamina—-the hidden allergen in the house [German]. Hautarzt 49(1):2–5. [PubMed: 9522185]
  • Schmidt M, Zargari A, Holt P, Lindbom L, Hellman U, Whitley P, Van Der ploey I, Harfast B, Scheynius A. 1997. The complete cDNA sequence and expression of the first major allergenic protein of Malasseziu furfur, Mal f I. European Journal of Biochemistry 246(1):181–185. [PubMed: 9210481]
  • Schou C. 1993. Defining allergens of mammalian origin. Clinical and Experimental Allergy 23(1):7–14. [PubMed: 8439823]
  • Schou C, Lind P, Fernandez-Caldas E, Lockey RF, Lowenstein H. 1990. Identification and purification of an important cross-reactive allergen from American (Periplaneta americana) and German (Blattella germanica) cockroach. Journal of Allergy and Clinical Immunology 86(6 Pt 1):935–946. [PubMed: 2262648]
  • Schulz O, Sewell HF, Shakib F. 1998. Proteolytic cleavage of CD25, the alpha subunit of the human T cell interleukin 2 receptor, by Der p I, a major mite allergen with cysteine protease activity. Journal of Experimental Medicine 187(2):271–275. [PMC free article: PMC2212095] [PubMed: 9432986]
  • Schumacher MJ, Tait BD, Holmes MC. 1981. Allergy to murine antigens in a biological research institute. Journal of Allergy and Clinical Immunology 68(4):310–318. [PubMed: 7288048]
  • Schwandner R, Dziarski R, Wesche H, Rothe M, Kirschning CJ. 1999. Peptidoglycan- and lipoteichoic acid-induced cell activation is mediated by toll-like receptor 2. Journal of Biological Chemistry 274(25):17406–17409. [PubMed: 10364168]
  • Scott R, Pullan CR, Scott M, McQuillin J. 1984. Cell-mediated immunity in respiratory syncytial virus disease. Journal of Medical Virology 13(1):105–114. [PubMed: 6693860]
  • Seabury J, Becker B, Salvaggio J. 1976. Home humidifier thermophilic actinomycete isolates. Journal of Allergy and Clinical Immunology 57(2):174–176. [PubMed: 1078519]
  • Sears MR, Herbison GP, Holdaway MD, Hewitt CJ, Flannery EM, Silva PA. 1989. The relative risks of sensitivity to grass pollen, house dust mite, and cat dander in the development of childhood asthma. Clinical and Experimental Allergy 19(4):419–424. [PubMed: 2758355]
  • Seggev JS, Lis I, Siman-Tov R, Gutman R, Abu-Samara H, Schey G, Naot Y. 1986. Mycoplasma pneumoniae is a frequent cause of exacerbation of bronchial asthma in adults. Annals of Allergy 57(4):263–265. [PubMed: 3094410]
  • Shahan TA, Sorenson WG, Paulauskis JD, Morey R, Lewis DM. 1998. Concentration- and time-dependent upregulation and release of the cytokines MIP-2, KC, TNF, and MIP-1alpha in rat alveolar macrophages by fungal spores implicated in airway inflammation. American Journal of Respiratory Cell and Molecular Biology 18(3):435–440. [PubMed: 9490662]
  • Shaheen SO. 1995. Changing patterns of childhood infection and the rise in allergic disease. Clinical and Experimental Allergy 25(11):1034–1037. [PubMed: 8581834]
  • Shaver JR, Zangrilli JG, Cho SK, Cirelli RA, Pollice M, Hastie AT, Fish JE, Peters SP. 1997. Kinetics of the development and recovery of the lung from IgE-mediated inflammation: dissociation of pulmonary eosinophilia, lung injury, and eosinophil-active cytokines. American Journal of Respiratory and Critical Care Medicine 155(2):442–448. [PubMed: 9032176]
  • Shen HD, Liaw SF, Lin WL, Ro LH, Yang HL, Han SH. 1995. Molecular cloning of cDNA encoding for the 68 kDA allergen of Penicillium notatum using MoAbs. Clinical and Experimental Allergy 25(4):350–356. [PubMed: 7600381]
  • Shen HD, Au LC, Lin WL, Liaw SF, Tsai JJ, Han SH. 1997. Molecular cloning and expression of Penicillium citrinum allergen with sequence homology and antigenic crossreactivity to a hsp 70 human heat shock protein. Clinical and Experimental Allergy 27(6):682–690. [PubMed: 9208190]
  • Shen HD, Lin WL, Tam MF, Wang SR, Tsai JJ, Chou H, Han SH. 1998. Alkaline serine proteinase: a major allergen of Aspergillus oryzae and its cross-reactivity with Penicillium citrinum. International Archives of Allergy and Immunology 116(1):29–35. [PubMed: 9623506]
  • Shulaner FA. 1970. Sensitivity to the cockroach in three groups of allergic children. Pediatrics 45(3):465–466. [PubMed: 5442917]
  • Sicherer SH, Wood RA, Eggleston PA. 1997. Determinants of airway responses to cat allergen: comparison of environmental challenge to quantitative nasal and bronchial allergen challenge. Journal of Allergy and Clinical Immunology 99(6 Pt 1):798–805. [PubMed: 9215248]
  • Sietsma JH, Wessels JG. 1981. Solubility of (1 leads to 3)-beta-D/(1 leads to 6)-beta-D-glucan in fungal walls: importance of presumed linkage between glucan and chitin. Journal of General Microbiology 125(Pt 1):209–212. [PubMed: 6460846]
  • Sigsgaard T, Hjort C, Omland O, Miller MR, Pedersen OF. 1996. Skin prick test to house dust mite, asthma and hyperreactivity in a cohort of young rurals. European Respiratory Journal 9(Supplement):379s.
  • Sigurs N, Bjarnason R, Sigurbergsson F, Kjellman B, Bjorksten B. 1995. Asthma and immunoglobulin E antibodies after respiratory syncytial virus bronchiolitis: a prospective cohort study with matched controls. Pediatrics 95(4):500–505. [PubMed: 7700748]
  • Skjonsberg OH, Clench-Aas J, Leegaard J, Skarpaas IJ, Giaever P, Bartonova A, Moseng J. 1995. Prevalence of bronchial asthma in schoolchildren in Oslo, Norway. Comparison of data obtained in 1993 and 1981. Allergy 50(10): 806–810. [PubMed: 8607562]
  • Slezak JA, Persky VW, Kviz FJ, Ramakrishnan V, Byers C. 1998. Asthma prevalence and risk factors in selected Head Start sites in Chicago. Journal of Asthma 35(2):203–212. [PubMed: 9576146]
  • Smith JM, Disney ME, Williams JD, Goels ZA. 1969. Clinical significance of skin reactions to mite extracts in children with asthma. British Medical Journal 1(659):723–726. [PMC free article: PMC1983509] [PubMed: 5786760]
  • Snapper CM, Pecanha LM, Levine AD, and Mond JJ. 1991. IgE class switching is critically dependent upon the nature of the B cell activator, in addition to the presence of IL-4. Journal of Immunology 147(4):1163–1170. [PubMed: 1714474]
  • Solomon KR., Kurt-Jones EA, Saladino RA, Stack AM, Dunn IF, Ferretti M, Golenbock D, Fleisher GR, Finberg RW. 1998. Heterotrimeric G proteins physically associated with the lipopolysaccharide receptor CD14 modulate both in vivo and in vitro responses to lipopolysaccharide. Journal of Clinical Investigations 102(11):2019–2027. [PMC free article: PMC509155] [PubMed: 9835628]
  • Solomon WR, Burge HA, Boise JR. 1980. Exclusion of particulate allergens by window air conditioners. Journal of Allergy and Clinical Immunology 65(4):305–308. [PubMed: 7358946]
  • Solomon WR, Burge HA, Muilenberg ML. 1983. Allergen carriage by atmospheric aerosol. I. Ragweed pollen determinants in smaller micronic fractions. Journal of Allergy and Clinical Immunology 72(5 Pt 1):443–447. [PubMed: 6195205]
  • Sonesson HRA, Zähringer U, Grimmecke HD, Westphal O, Rietschel ET. 1994. Bacterial Endotoxin: chemical structure and biological activity. In: Endotoxin and the Lungs. Brigham K., editor. , Ed. New York: Marcel Dekker, Inc.
  • Sorenson WG, Simpson J, Castranova V. 1985. Toxicity of the mycotoxin patulin for rat alveolar macrophages. Environmental Research 38(2):407–416. [PubMed: 2415351]
  • Sorenson WG, Simpson J. 1986. Toxicity of penicillic acid for rat alveolar macrophages in vitro. Environmental Research 41(2):505–513. [PubMed: 2430790]
  • Sorenson WG, Gerberick GF, Lewis DM, Castranova V. 1986. Toxicity of mycotoxins for the rat pulmonary macrophage in vitro. Environmental Health Perspectives 66:45–53. [PMC free article: PMC1474366] [PubMed: 2423320]
  • Soto-Quiros M, Gutierrez I, Calvo N, Araya C, Karlberg J, Hanson LA, Belin L. 1998. Allergen sensitization of asthmatic and nonasthmatic schoolchildren in Costa Rica. Allergy 53(12):1141–1147. [PubMed: 9930589]
  • Sporik R, Holgate ST, Platts-Mills TA, Cogswell JJ. 1990. Exposure to house-dust mite allergen (Der p I) and the development of asthma in childhood. A prospective study. New England Journal of Medicine 323(8):502–507. [Comment in: N Engl J Med 1991. 324(5):337–338.] [PubMed: 2377175]
  • Sporik R, Ingram JM, Price W, Sussman JH, Honsinger RW, Platts-Mills TA. 1995. Association of asthma with serum IgE and skin test reactivity to allergens among children living at high altitude. Tickling the dragon's breath. American Journal of Respiratory and Critical Care Medicine 151(5):1388–1392. [Comment in: Am J Respir Crit Care Med 1997. 155(2):769–770.] [PubMed: 7735590]
  • Squillace SP, Sporik RB, Rakes G, Couture N, Lawrence A, Merriam S, Zhang J, Platts-Mills TA. 1997. Sensitization to dust mites as a dominant risk factor for asthma among adolescents living in central Virginia. Multiple regression analysis of a population-based study. American Journal of Respiratory and Critical Care Medicine 156(6):1760–1764. [PubMed: 9412552]
  • Staib F. 1992. Pathogenic fungi in human dwellings. Mycoses 35(11–12):289–292. [PubMed: 1302800]
  • Staib F. 1996. Fungi in the home and hospital environment [German]. Mycoses 39(Suppl 1):26–29. [PubMed: 8767265]
  • Staib F, Folkens U, Tompak B, Abel T, Thiel D. 1978. a. A comparative study of antigens of Aspergillus fumigatus isolates from patients and soil of ornamental plants in the immunodiffusion test. Zentralblatt für Bakteriologie, Parasitenkunde, Infektionskrankheiten und Hygiene—Erste Abteilung Originale—Reihe A: Medizinische Mikrobiologie und Parasitologie 242(1): 93–99. [PubMed: 83753]
  • Staib F, Tompak B, Thiel D, Blisse A. 1978. b. Aspergillus fumigatus and Aspergillus niger in two potted ornamental plants, cactus (Epiphyllum truncatum) and clivia (Clivia miniata). Biological and epidemiological aspects. Mycopathologia 66(1– 2):27–30. [PubMed: 375093]
  • Staib F, Mishra SK, Blisse A. 1980. Interaction between Aspergilli and Streptomycetes in the soil of potted indoor plants: a preliminary report (contribution to the epidemiology of human aspergillosis). Mycopathologia 70(1):9–12. [PubMed: 6769046]
  • Stewart GA, Thompson PJ. 1996. The biochemistry of common aeroallergens. Clinical and Experimental Allergy 26(9):1020–1044. [Comment in: Clin Exp Allergy 1997. 27(6):714–715.] [PubMed: 8889258]
  • Strachan DP. 1988. Damp housing and childhood asthma: validation of reporting of symptoms. British Medical Journal 297(6658):1223–1226. [PMC free article: PMC1834724] [PubMed: 3145060]
  • Strachan DP, Carey IM. 1995. Home environment and severe asthma in adolescence: a population based case-control study. British Medical Journal 311(7012):1053–1056. [PMC free article: PMC2551362] [PubMed: 7580660]
  • Strand V, Rak S, Svartengren M, Bylin G. 1997. Nitrogen dioxide exposure enhances asthmatic reaction to inhaled allergen in subjects with asthma. American Journal of Respiratory and Critical Care Medicine 155(3):881–887. [PubMed: 9117021]
  • Strand V, Svartengren M, Rak S, Barck C, Bylin G. 1998. Repeated exposure to an ambient level of NO2 enhances asthmatic response to a nonsymptomatic allergen dose. European Respiratory Journal 12(1):6–12. [PubMed: 9701406]
  • Su HJ, Rotnitzky A, Burge HA, Spengler JD. 1992. Examination of fungi in domestic interiors by using factor analysis: correlations and associations with home factors. Applied and Environmental Microbiology 58:181–186. [PMC free article: PMC195189] [PubMed: 1539973]
  • Subiza J, Cabrera M, Valdivieso R, Subiza JL, Jerez M, Jimenez JA, Narganes MJ, Subiza E. 1994. Seasonal asthma caused by airborne Platanus pollen. Clinical and Experimental Allergy 24(12):1123–1129. [PubMed: 7889425]
  • Suda T, Sato A, Ida M, Gemma H, Hayakawa H, Chida K. 1995. Hypersensitivity pneumonitis associated with home ultrasonic humidifiers. Chest 107(3): 711–717. [PubMed: 7874942]
  • Sue MA, Gordon EH, Freund LH. 1993. Utility of additional skin testing in “nonallergic” asthma. Annals of Allergy 68(5):395–397. [PubMed: 1586003]
  • Sugawara S, Arakaki R, Rikiishi H, Takada H. 1999. Lipoteichoic acid acts as an antagonist and an agonist of lipopolysaccharide on human gingival fibroblasts and monocytes in a CD14-dependent manner. Infection and Immunology 67(4):1623–1632. [PMC free article: PMC96505] [PubMed: 10084995]
  • Suliaman FA, Holmes WF, Kwick S, Khouri F, Ratard R. 1997. Pattern of immediate type hypersensitivity reactions in the Eastern Province, Saudi Arabia. Annals of Allergy 78(4):415–418. [PubMed: 9109711]
  • Summerbell RC, Krajden S, Kane J. 1989. Potted plants in hospitals as reservoirs of pathogenic fungi. Mycopathologia 106(1):13–22. [PubMed: 2671744]
  • Suoniemi I, Bjorksten F, Haahtela T. 1981. Dependence of immediate hypersensitivity in the adolescent period on factors encountered in infancy. Allergy 36(4):263–268. [PubMed: 7316100]
  • Suphioglu C. 1998. Thunderstorm asthma due to grass pollen. International Archives of Allergy and Immunology 116(4):253–260. [Published erratum appears in Int Arch Allergy Immunol 1999. 119(2):37.] [PubMed: 9693274]
  • Sutton P, Waring O, Mullbacher A. 1996. Exacerbation of invasive aspergillosis by the immunosuppressive fungal metabolite, gliotoxin. Immunology and Cell Biology 74(4):318–322. [PubMed: 8872181]
  • Svanes C, Jarvis D, Chinn S, Burney P. 1999. Childhood environment and adult atopy: results from the European Community Respiratory Health Survey. Journal of Allergy and Clinical Immunology 103(3 Pt 1):415–420. [PubMed: 10069874]
  • Svartengren M, Falk R, Linnman L, Philipson K, Camner P. 1987. Deposition of large particles in human lung. Experimental Lung Research 12(1):75–88. [PubMed: 3102217]
  • Swanson MC, Agarwal MK, Reed CE. 1985. An immunochemical approach to indoor aeroallergen quantitation with a new volumetric air sampler: studies with mite, roach, cat, mouse, and guinea pig antigens. Journal of Allergy and Clinical Immunology 76(5):724–729. [PubMed: 4056257]
  • Szantho A, Osvath P, Horvath Z, Novak EK, Kujalek E. 1992. Study of mold allergy in asthmatic children in Hungary. Journal of Investigational Allergology and Clinical Immunology 2(2):84–90. [PubMed: 1342888]
  • Takahashi T. 1997. Airborne fungal colony-forming units in outdoor and indoor environments in Yokohama, Japan. Mycopathologia 139(1):23–33. [PubMed: 9511234]
  • Tariq SM, Matthews SM, Stevens M, Hakim EA. 1996. Sensitization to Alternaria and Cladosporium by the age of 4 years. Clinical and Experimental Allergy 26(7):794–798. [PubMed: 8842553]
  • Tauer-Reich I, Fruhmann G, Czuppon AB, Baur X. 1994. Allergens causing bird fancier's asthma. Allergy 49(6):448–453. [PubMed: 8074264]
  • Terleckyj B, Axler DA. 1987. Quantitative neutralization assay of fungicidal activity of disinfectants. Antimicrobial Agents and Chemotherapy 31(5):794– 798. [PMC free article: PMC174835] [PubMed: 3300541]
  • Tovey ER, Chapman MD, Platts-Mills TA. 1981. a. Mite faeces are a major source of house dust allergens. Nature 289(5798):592–593. [PubMed: 7464922]
  • Tovey ER, Chapman MD, Wells CW, Platts-Mills TA. 1981. b. The distribution of dust mite allergen in the houses of patients with asthma. American Review of Respiratory Disease 124(5):630–635. [PubMed: 7305119]
  • Tovey E, Marks G. 1999. Methods and effectiveness of environmental control. Journal of Allergy and Clinical Immunology 103(2 Pt 1):179–191. [PubMed: 9949306]
  • Travis J, Whitworth T, Matheson N, Bagarozzi D Jr. 1996. Proteinases from pollen and pests. Acta Biochimica Polonica 43(3):411–417. [PubMed: 8922023]
  • Twiggs JT, Agarwal MK, Dahlberg MJ, Yunginger JW. 1982. Immunochemical measurement of airborne mouse allergens in a laboratory animal facility. Journal of Allergy and Clinical Immunology 69(6):522–526. [PubMed: 7076993]
  • Tyndall RL, Lehman E, Bowman EK, Milton DK, Barbaree J. 1995. Home humidifiers as a potential source of exposure to microbial pathogens, endotoxins and allergens. Indoor Air 5:171–178.
  • Ulevitch RJ. 1999. Endotoxin opens the tollgates to innate immunity. Nature Medicine 5(2):144–145. [PubMed: 9930855]
  • Valovirta E, Koivikko A, Vanto T, Viander M, Ingeman L. 1984. Immunotherapy in allergy to dog: a double-blind clinical study. Annals of Allergy 53(1):85–88. [PubMed: 6742528]
  • van der Heide S, van Aalderen WM, Kauffman HF, Dubois AE, de Monchy JG. 1999. Clinical effects of air cleaners in homes of asthmatic children sensitized to pet allergens. Journal of Allergy and Clinical Immunology 104(2 Pt 1): 447–451. [PubMed: 10452769]
  • Vanto T, Viander M, Koivikko A. 1980. Skin prick test in the diagnosis of dog dander allergy: a comparison of different extracts with clinical history, provocation tests and RAST. Clinical Allergy 10(2):121–132. [PubMed: 7389067]
  • Vanto T, Koivikko A. 1983. Dog hypersensitivity in asthmatic children. A clinical study with special reference to the relationship between the exposure to dogs and the occurrence of hypersensitivity symptoms. Acta Paediatrica Scandinavica 72(4):571–575. [PubMed: 6624434]
  • Vaughan JW, McLaughlin TE, Perzanowski MS, Platts-Mills TA. 1999. Evaluation of materials used for bedding encasement: effect of pore size in blocking cat and dust mite allergens. Journal of Allergy and Clinical Immunology 103(2 Pt 1):227–231. [PubMed: 9949312]
  • Venables KM, Tee RD, Hawkins ER, Gordon DJ, Wale CJ, Farrer NM, Lam TH, Baxter PJ, Newman Taylor AJ. 1988. Laboratory animal allergy in a pharmaceutical company. British Journal of Industrial Medicine 45(10): 660–666. [PMC free article: PMC1009674] [PubMed: 3264181]
  • Venables KM, Allitt U, Collier CG, Emberlin J, Greig JB, Hardaker PJ, Highham JH, Laing-Morton T, Maynard RL, Murray V, Strachan D, Tee RD. 1997. Thunderstorm-related asthma—the epidemic of 24/25 June 1994. Clinical and Experimental Allergy 27(7):725–736. [PubMed: 9249264]
  • Verhoeff AP, van Wijnen JH, Brunekreef B, Fischer P, van Reenen-Hoekstra ES, Samson RA. 1992. Presence of viable mould propagules in indoor air in relation to house damp and outdoor air. Allergy 47(2 Pt 1):83–91. [PubMed: 1632482]
  • Verma J, Sridhara S, Singh BP, Gangal SV. 1995. Studies on shared antigenic/ allergenic components among fungi. Allergy 50(10):811–816. [PubMed: 8607563]
  • von Hertzen L, Töyrylä M, Gimishanov A, Bloigu A, Leinonen M, Saikku P, Haahtela T. 1999. Asthma, atopy and Chlamydia pneumoniae antibodies in adults. Clinical and Experimental Allergy 29(4):522–528. [PubMed: 10202367]
  • von Mutius E, Martinez FD, Fritzsch C, Nicolai T, Reitmeir P, Thiemann HH. 1994. Skin test reactivity and number of siblings [see comments]. British Medical Journal 308(6930):692–695. [PMC free article: PMC2539417] [PubMed: 8142793]
  • Voorhorst R, Spieksma FThM, Varekamp N. 1969. House dust mite atopy and the house dust mite Dermatophagoides pteronyssinus (Troussart, 1897). Leiden: Stafleu's Scientific Publishing Co.
  • Walls AF, Longbottom JL. 1985. Comparison of rat fur, urine, saliva, and other rat allergen extracts by skin testing, RAST, and RAST inhibition. Journal of Allergy and Clinical Immunology 75(2):242–251. [PubMed: 3968335]
  • Walters M, Milton DK, Larsson L, Ford T. 1994. Airborne environmental endotoxin: a cross-validation of sampling and analysis techniques. Applied and Environmental Microbiology 60(3):996–1005. [PMC free article: PMC201423] [PubMed: 8161191]
  • Wang JY, Chen WY. 1992. Inhalant allergens in asthmatic children in Taiwan: comparison evaluation of skin testing, radioallergosorbent test and multiple allergosorbent chemiluminescent assay for specific IgE. Journal of the Formosan Medical Association 91(12):1127–1132. [PubMed: 1363631]
  • Waring P, Beaver J. 1996. Gliotoxin and related epipolythiodioxopiperazines. General Pharmacology 27(8):1311–1316. [PubMed: 9304400]
  • Waring P, Khan T, Sjaarda A. 1997. Apoptosis induced by gliotoxin is preceded by phosphorylation of histone H3 and enhanced sensitivity of chromatin to nuclease digestion. Journal of Biological Chemistry 272(29):17929–17936. [PubMed: 9218417]
  • Warner JA. 1992. Environmental allergen exposure in homes and schools [editorial]. Clinical and Experimental Allergy 22(12):1044–1045. [PubMed: 1486531]
  • Warner JA, Little SA, Pollock I, Longbottom JL, Warner JO. 1991. The influence of exposure to house dust mite on sensitization in asthma. Pediatric Allergy and Immunology 1:79–86.
  • Warner JA, Jones AC, Miles EA, Colwell BM, Warner JO. 1997. Prenatal origins of asthma and allergy. Ciba Foundation Symposium 206:220–208; discussion 228–232. [PubMed: 9257015]
  • Weatherstone KB, Rich EA. 1989. Tumor necrosis factor/cachectin and interleukin-1 secretion by cord blood monocytes from premature and term neonates. Pediatric Research 25(4):342–346. [PubMed: 2786182]
  • Welliver RC, Wong DT, Sun M, Middleton E Jr, Vaughan RS, Ogra PL. 1981. The development of respiratory syncytial virus-specific IgE and the release of histamine in nasopharyngeal secretions after infection. New England Journal of Medicine 305(15):841–846. [PubMed: 6168908]
  • Welliver RC, Sun M, Rinaldo D, Ogra PL. 1986. Predictive value of respiratory syncytial virus–specific IgE responses for recurrent wheezing following bronchiolitis. Journal of Pediatrics 109(5):776–780. [PubMed: 3772657]
  • Welliver RC, Duffy L. 1993. The relationship of RSV-specific immunoglobulin E antibody responses in infancy, recurrent wheezing, and pulmonary function at age 7–8 years. Pediatric Pulmonology 15(1):19–27. [PubMed: 8419894]
  • Wierenga EA, Snoek M, de Groot C, Chretien I, Bos JD, Jansen HM, Kapsenberg ML. 1990. Evidence for compartmentalization of functional subsets of CD2+ T lymphocytes in atopic patients. Journal of Immunology 144(12):4651–4656. [PubMed: 1972164]
  • Williams LW, Reinfried PA, Brenner RJ. 1999. Cockroach extermination does not rapidly reduce allergen in settled dust. Journal of Allergy and Clinical Immunology 104(3 Pt 1):702–703. [PubMed: 10482850]
  • Williamson IJ, Martin CJ, McGill G, Monie RD, Fennerty AG. 1997. Damp housing and asthma: a case-control study. Thorax 52(3):229–234. [PMC free article: PMC1758502] [PubMed: 9093337]
  • Wilson NW, Robinson NP, Hogan MB. 1999. Cockroach and other inhalant allergies in infantile asthma. Annals of Allergy, Asthma, and Immunology 83(1):27–30. [PubMed: 10437813]
  • Witteman AM, Stapel SO, Sjamsoedin DH, Jansen HM, Aalberse RC, van der Zee JS. 1996. Fel d I-specific IgG antibodies induced by natural exposure have blocking activity in skin tests. International Archives of Allergy and Immunology 109(4):369–375. [PubMed: 8634522]
  • Wood RA, Chapman MD, Adkinson NF Jr, Eggleston PA. 1989. The effect of cat removal on allergen content in household-dust samples. Journal of Allergy and Clinical Immunology 83(4):730–734. [PubMed: 2708734]
  • Wood RA, Johnson EF, Van Natta ML, Chen PH, Eggleston PA. 1998. A placebo-controlled trial of a HEPA air cleaner in the treatment of cat allergy. American Journal of Respiratory and Critical Care Medicine 158(1):115–120. [PubMed: 9655716]
  • Woodcock A, Custovic A. 1998. Role of the indoor environment in determining the severity of asthma. Thorax 53 (Suppl 2):S47–51. [PMC free article: PMC1765899] [PubMed: 10193348]
  • Wright SD. 1999. Toll, a new piece in the puzzle of innate immunity. Journal of Experimental Medicine 189(4):605–609. [PMC free article: PMC2192938] [PubMed: 9989974]
  • Wright SD, Ramos RA, Tobias PS, Ulevitch RJ, Mathison JC. 1990. CD14, a receptor for complexes of lipopolysaccharide (LPS) and LPS binding protein. Science 249(4975):1431–1433. [PubMed: 1698311]
  • Wuthrich B, Schindler C, Leuenberger P, Ackermann-Liebrich U. 1995. Prevalence of atopy and pollinosis in the adult population of Switzerland (SAPALDIA study). Swiss Study on Air Pollution and Lung Diseases in Adults. International Archives of Allergy and Immunology 106(2):149–156. [PubMed: 7819743]
  • Yang RB, Mark MR, Gray A, Huang A, Xie MH, Zhang M, Goddard A, Wood WI, Gurney AL, Godowski PJ. 1998. Toll-like receptor–2 mediates lipopoly-saccharide-induced cellular signaling. Nature 395(6699):284–288. [PubMed: 9751057]
  • Yano T, Ichikawa Y, Komatu S, Arai S, Oizumi K. 1994. Association of Mycoplasma pneumoniae antigen with initial onset of bronchial asthma. American Journal of Respiratory and Critical Care Medicine 149(5):1348–1353. [PubMed: 8173777]
  • Yman L, Brandt R, Ponterius G. 1973. Serum albumin—an important allergen in dog epithelia extract. International Archives of Allergy 44:358–368. [PubMed: 4708776]
  • Young RP, Hart BJ, Merrett TG, Read AF, Hopkins JM. 1992. House dust mite sensitivity: interaction of genetics and allergen dosage. Clinical and Experimental Allergy 22:205–211. [PubMed: 1571813]
  • Young RP, Dekker JW, Wordsworth BP, Schou C, Pile KD, Matthiesen F, Rosenberg WM, Bell JI, Hopkin JM, Cookson WO. 1993. HLA and HLA-DP genotypes and immunoglobulin E responses to common major allergens. Clinical and Experimental Allergy 24:431–439. [PubMed: 8087654]
  • Yunginger JW, Jones RT, Nesheim ME, Geller M. 1980. Studies on Alternaria allergens. III. Isolation of a major allergenic fraction (ALT-I). Journal of Allergy and Clinical Immunology 66(2):138–147. [PubMed: 7190582]
  • Zhang K, Petty HR. 1994. Influence of polysaccharides on neutrophil function: Specific antagonists suggest a model for cooperative saccharide-associated inhibition of immune complex-triggered superoxide production. Journal of Cellular Biochemistry 56(2):225–235. [PubMed: 7829584]
  • Zielonka TM, Charpin D, Berbis P, Luciani P, Casanova D, Vervloet D. 1994. Effects of castration and testosterone on Fel d I production by sebaceous glands of male cats: I—Immunological assessment [see comments]. Clinical and Experimental Allergy 24(12):1169–1173. [PubMed: 7889432]
  • Zimmer S, Pollard V, Marshall GD, Garofalo RP, Traber D, Prough D, Herndon DN. 1996. The 1996 Moyer Award. Effects of endotoxin on the TH1/TH2 response in humans. Journal of Burn Care and Rehabilitation 17(6 Pt 1):491– 496. [PubMed: 8951535]
Copyright 2000 by the National Academy of Sciences. All rights reserved.
Bookshelf ID: NBK224482

Views

Related information

  • PMC
    PubMed Central citations
  • PubMed
    Links to PubMed

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...