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.

Baron S, editor. Medical Microbiology. 4th edition. Galveston (TX): University of Texas Medical Branch at Galveston; 1996.

Cover of Medical Microbiology

Medical Microbiology. 4th edition.

Show details

Chapter 31Bordetella

and .

General Concepts

Clinical Manifestations

Bordetella pertussis causes whooping cough (pertussis), an acute respiratory infection marked by severe, spasmodic coughing episodes during the paroxysmal phase. Leukocytosis with lymphocytosis is also common during this phase of the illness. Dangerous complications are bronchopneumonia and acute encephalopathy. Bordetella parapertussis can cause a milder form of pertussis.

Structure

The bordetellae are small, Gram-negative, aerobic coccobacilli. Bordetella pertussis produces a number of virulence factors, including pertussis toxin, adenylate cyclase toxin, filamentous hemagglutinin, and hemolysin. Agglutinogens and other outer membrane proteins are important antigens.

Classification and Antigenic Types

The genus Bordetella contains the species B pertussis and B parapertussis, which cause pertussis in humans. Other members of the genus are B bronchiseptica, causing respiratory disease in various animals and occasionally in humans, and B avium as well as B hinzii, which cause respiratory disease in poultry and are very rarely found in humans.

Bordetellae are characterized by culture characteristics, biochemical tests, and nucleic acid analysis. Some of them show reversible antigenic modulation under certain culture conditions, and they mutate through several antigenically distinct phases when grown on agar.

Pathogenesis

Transmission is by droplets. The bacteria colonize only ciliated cells of the respiratory mucosa, and they multiply rapidly.

Bacteremia does not occur. The roles of the various toxins in pathogenesis are unclear.

Host Defenses

Infection induces substantial immunity, although the protective antigens have not been identified conclusively. Both nonspecific and specific defenses participate in the response to disease.

Epidemiology

The human respiratory mucosa is the natural habitat for B pertussis and B parapertussis. Transmission is almost always directly from person to person. Patients are most infectious during the early, catarrhal phase of the disease and remain infectious for about 5 weeks. Pertussis is a common and dangerous childhood disease in unvaccinated populations.

Diagnosis

B pertussis can be cultured on modified Bordet-Gengou medium, charcoal-horseblood agar (Regan-Lowe) or grown in supplement Stainer-Scholte broth. Bordetella DNA can also be detected by PCR. Circulating antibodies appear in week 3 of illness and peak in the eighth to tenth week. Antibodies can be demonstrated by an enzyme-linked immunosorbent assay. Detection of specific IgA provides evidence of natural infection.

Control

Treatment with erythromycin does not alter the course of disease, but reduces the infectious period to 5 to 10 days. Inactivated whole-cell vaccines are highly effective, but occasionally cause toxic side effects. Acellular vaccines with fewer side effects have been licensed for booster vaccination and will possibly be also licensed for primary vaccination.

Introduction

Bordetella pertussis was first isolated in pure culture in 1906 by Bordet and Gengou. Today, B pertussis belongs to the genus Bordetella in the family Alcaligenaceae, which contains several species of closely related bacteria with similar morphology. B pertussis and B parapertussis cause whooping cough (pertussis) in humans. Other members of the genus are B bronchiseptica, which causes respiratory disease in various animals and is only occasionally found in humans. Recent additions to the genus are B avium and B hinzii, which both cause respiratory disease in poultry and are very rarely found in humans.

Clinical Manifestations

After an incubation period of 1 to 2 weeks, whooping cough begins with the catarrhal phase (Fig. 31-1). This phase lasts 1 to 2 weeks and is usually characterized by low-grade fever, rhinorrhea, and progressive cough; the patient is highly infectious. The subsequent paroxysmal phase, lasting 2 to 4 weeks, is characterized by severe and spasmodic cough episodes. At the end of the catarrhal phase, a leukocytosis with an absolute and relative lymphocytosis frequently begins, reaching its peak at the height of the paroxysmal stage. At this time, the total blood leukocyte levels may resemble those of leukemia (≥ 100,000/mm3), with 60 to 80 percent being lymphocytes. The convalescent phase, lasting 1 to 3 weeks, is characterized by a continuous decline of the cough before the patient returns to normal. Serious complications, sometimes fatal, are bronchopneumonia and acute encephalopathy, the latter being characterized primarily by convulsions and frequently resulting in death or lifelong brain damage.

Figure 31-1. Pathogenesis of whooping cough.

Figure 31-1

Pathogenesis of whooping cough.

Structure

Bordetella pertussis is a small (approximately 0.8 μm by 0.4 μm), rod-shaped, coccoid, or ovoid Gram-negative bacterium that is encapsulated and does not produce spores. It is a strict aerobe. It is arranged singly or in small groups and is not easily distinguished from Haemophilus species. B pertussis and B parapertussis are nonmotile. Numerous antigens and biologically active structural components have been demonstrated in B pertussis (Fig. 31-2), although their exact chemical structure and location in the bacterial cell are known only in part.

Figure 31-2. Virulence factors of B pertussis.

Figure 31-2

Virulence factors of B pertussis.

Pertussis Toxin

Various immunologic, physiologic, and pharmacologic effects are induced by killed B pertussis cells in experimental animals (e.g., increased sensitivity to histamine and serotonin and active and passive anaphylaxis). Adjuvant activity, leukocytosis, splenomegaly, cell proliferation, hypoglycemia, and hypoproteinemia also occur. Many additional features have been described, including increased sensitivity to factors such as endotoxins, X irradiation, infection, cold stress, pollen extracts, peptone shock, and methacholine; increased resistance to infection; increased capillary permeability; and accelerated production of experimental “allergic” encephalomyelitis.

It is now generally accepted that all those biologic activities are caused by a single active protein produced by B pertussis. To avoid confusion caused by the many different names for this protein, the uniform term pertussis toxin was proposed by Pittman. Pertussis toxin is a protein exotoxin, secreted during in vivo and in vitro growth; it consists of five different subunits, designated S1, S2, S3, S4, and S5. Since the toxin molecule contains two S4 subunits, it is a hexamer. Like many other protein toxins, pertussis toxin consists of an A subunit that carries the biologic activity and a B subunit that binds the complex to the cell membrane. In pertussis toxin, the S 1 subunit constitutes the A protomer and the B oligomer is formed by the remaining five subunits (Fig. 31-3). The toxin binds to cell receptors by two dimers, one consisting of S2 and S4 and the other of S3 and S4. Since glutaraldehyde-inactivated pertussis toxin is capable of adherence, this binding activity evidently has little to do with the various toxic activities of pertussis toxin. The toxin reacts with different cell types, including T lymphocytes, and acts on different cellular regulatory processes. Pertussis toxin is a member of the family of ADP-ribosylating bacterial toxins. The S1 subunit of pertussis toxin ADP-ribosylates the Cys352 of protein Gi (GTP-binding protein), as well as the corresponding cysteine of protein Gα and of transducin. Although pertussis toxin is synthesized solely by B pertussis, both B parapertussis and B bronchiseptica possess genes for pertussis toxin without expressing them. Bordetella parapertussis expresses pertussis toxin when the toxin gene from the B pertussis chromosome is introduced into B parapertussis.

Figure 31-3. Binding of pertussis toxin to cell membranes.

Figure 31-3

Binding of pertussis toxin to cell membranes.

Like many other bacteria, B pertussis possesses hemagglutinating activity, expressed as its capacity to agglutinate red cells from geese, chickens, and other animals. Pertussis toxin is one of the hemagglutinins, whereas another component with hemagglutinating activity is called filamentous hemagglutinin. This component appears as fine filaments, about 2 nm in diameter and 40 to 100 nm in length. Like pertussis toxin, it has hemagglutinating activity as well as the ability to effect the adherence of B pertussis to cilia by its lectin-like binding to lactose-containing moieties.

Heat-Labile Toxin

The heat-labile toxin of Bordetella is a proteinaceous dermonecrotic toxin with a molecular weight of about 100,000, localized in the protoplasm. This toxin produces strong vasoconstrictive effects, which are probably important during the initial phase of pertussis by their action on the respiratory tract. Thus, heat-labile toxin, in association with tracheal cytotoxin and lipopolysaccharide, possibly causes tissue damage in the respiratory tract.

Adenylate Cyclase Toxin

Adenylate cyclase toxin is a protein toxin that penetrates the host cell, is activated by host cell calmodulin, and increases intracellular CAMP massively. The increase of CAMP, which is rather short-lived as in contrast to the action of pertussis toxin, is associated with the inhibition of phagocytic cell oxidative responses and natural killer cell (NK) activity.

Tracheal Cytotoxin

Tracheal cytotoxin, which is chemically related to peptidoglycan, destroys the ciliated cell population of a hamster trachea in 60 to 96 hours.

Lipopolysaccharide

The heat-stable Bordetella lipooligosaccharide (LOS) endotoxin is similar in structure, chemical composition, and biologic activity to other endotoxins produced by Gram-negative bacteria. Endotoxin from B pertussis, of which two types can be distinguished, is serologically different from corresponding preparations from B parapertussis and B bronchiseptica. It is remarkable that heat-labile toxin, adenylate cyclase toxin, tracheal cytotoxin, and LPS are formed by the three Bordetella species, whereas pertussis toxin is produced solely by B pertussis.

Agglutinogens

The agglutinogens are surface antigens responsible for agglutination of the bacterial cells in the presence of their corresponding antibodies. To date, 14 different agglutinogens (AGG 1 through AGG 14) have been distinguished. AGG1 is specific for B pertussis, and is associated with lipooligosaccharide. AGG 14 is thought to be specific for B parapertussis. The AGGs 2 and 3 (formerly 2, 3, 4, 6) are associated with different types of fimbriae of B pertussis, which may also be true for the AGGs 8, 9 and 10 of B parapertussis.

Outer Membrane Proteins

At least four different outer membrane protein structures are distinguished on B pertussis; they are designated OMP 15, OMP 18, OMP 69, and OMP 91. They are believed to be protective antigens.

Classification and Antigenic Types

The genus Bordetella contains species of serologically related bacteria with similar morphology, size, and staining reactions. B pertussis and B parapertussis are genomically extremely closely related. Other members of the genus are B bronchiseptica, which by DNA-DNA and DNA-rRNA hybridization is also closely related. Recent additions to the genus are B avium (formerly designated Alcaligenes faecalis) and B hinzii (formerly designated A faecalis type II), which cause respiratory disease in poultry and are very rarely found in humans.

Bordetella pertussis was first isolated in pure culture in 1906 and was long considered the sole agent of whooping cough. Later studies revealed that this disease also can be caused in a mild form by B parapertussis and occasionally by B bronchiseptica. A phenomenon of B pertussis organisms is their variation during growth on agar plates: the antigenically competent, smooth, virulent form (phase I) can mutate to the antigenically incomplete, nonvirulent, rough form (phase IV). This change is associated with a loss of capacity to synthesize pertussis toxin, filamentous hemagglutinin, heat-labile toxin, adenylate cyclase toxin, agglutinogens, and certain outer membrane proteins. There are also two intermediate forms, called phases II and III.

In addition to this spontaneous phase variation, B pertussis undergoes antigenic modulation in response to changes in environmental conditions, such as growth at low temperatures or on agar plates with high concentrations of MgSO4 or nicotinic acid. Bordetella pertussis organisms grown under such conditions are avirulent and are characterized by the loss of the capacity to synthesize the numerous toxic factors and other structural components. Both phase variation and antigenic modulation are reversible and also occur in B parapertussis and B bronchiseptica. Both phenomena are under the control of a single genetic locus. The virulent strains are therefore designated Bvg+, and the avirulent strains Bvg. Phase variation has been observed in vivo. Another type of serotype variation in B pertussis—the loss of one or more agglutinogens—occurs independently of phase variation.

Pathogenesis

The agent of whooping cough is transmitted primarily via droplets. Infection results in colonization and rapid multiplication of the bacteria on the mucous membranes of the respiratory tract. Bacteremia does not occur. Electron microscopic studies have demonstrated that phase I strains of B pertussis adhere only to the tuft of ciliated cells in the mucosa of the human respiratory tract; no attachment to nonciliated cells was observed. Convincing experimental data indicate that the adherence of B pertussis to human cilia is effected by a synergistic action of pertussis toxin and filamentous hemagglutinin, each acting as a bivalent bridge between the bacterium and the ciliary receptor (Fig. 31-4).

Figure 31-4. Synergy between pertussis toxin and the filamentous hemagglutinin in binding to ciliated respiratory epithelial cells.

Figure 31-4

Synergy between pertussis toxin and the filamentous hemagglutinin in binding to ciliated respiratory epithelial cells.

Studies of numerous B pertussis toxins and their corresponding biologic activities have yielded plausible explanations for many of the symptoms of whooping cough. These include, for example, the frequent occurrence of absolute lymphocytosis (an unusual phenomenon in bacterial infections), hypoglycemia, and the adjuvant effect of pertussis toxin on the immune response to unrelated antigens. The finding that phase I isolates of B bronchiseptica produce almost complete ciliostasis within 3 hours in ciliated epithelial cell outgrowths from canine tracheal explants may be explained by the action of adenylate cyclase toxin and tracheal cytotoxin. The same toxins evidently inhibit the phagocytic activities of the host. In humans, an initial local peribronchial lymphoid hyperplasia occurs, accompanied or followed by necrotizing inflammation and leukocyte infiltration in parts of the larynx, trachea, and bronchi. Usually, peribronchiolitis and variable patterns of atelectasis and emphysema also develop.

To date, there is no plausible explanation for the development of the paroxysmal coughing syndrome characteristic of pertussis. According to Pittman, pertussis is mediated by pertussis toxin and is characterized by a two-stage process—infection (colonization) and disease—thus resembling other bacterial toxicoses such as diphtheria, tetanus, and cholera. This fascinating idea can be accepted only if it is clearly demonstrated that pertussis toxin causes paroxysmal coughing. Such a demonstration is lacking. Moreover, paroxysmal coughing occurs in infections with B parapertussis, which does not synthesize pertussis toxin. On the other hand, an additional infection with B pertussis cannot be excluded in such cases. There is no convincing explanation for the acute encephalopathy sometimes observed in pertussis. Research into the pathogenetic mechanisms of pertussis are hampered by the lack of an adequate animal model showing the characteristic paroxysmal coughing syndrome and by the limited opportunity to perform direct studies of the respiratory tract of babies and children.

Host Defenses

A case of whooping cough confers substantial immunity, which usually lasts for many years. Second infections of adults, usually with atypical symptoms and thus not regularly diagnosed as pertussis, may be more frequent than previously assumed. Immunity acquired after infection with B pertussis does not protect against the other Bordetella species. Pertussis toxin is assumed to be one essential protective immunogen, but numerous findings indicate that other components, such as filamentous hemagglutinin, heat-labile toxin, agglutinogens, outer membrane proteins, and adenylate cyclase toxin, may also contribute to immunity after infection or vaccination. The immunogenicity of the substances may be significantly increased by the presence of pertussis toxin. This synergism indicates that pertussis toxin could function as an adjuvant to a variety of protective antigens of B pertussis. The defense mechanisms are both nonspecific (local inflammation, increase in macrophage activity, and production of interferon) and specific (proliferation of B and T cells). The basis of immunity in whooping cough is, however, incompletely understood. A role of circulating antibody in immunity is indicated by the correlation between protection of human vaccinees and their serum agglutinin titers. However, effective immunity does not necessarily depend on the presence of serum agglutinins, and immunity to whooping cough may therefore be mediated essentially by cellular mechanisms. This cell-mediated immunity may be considered the crucial carrier of long-term immunity, and titers of specific humoral antibodies may diminish over the years. This may be the reason why infants usually do not benefit significantly from maternal antibody.

Epidemiology

The mucous membranes of the human respiratory tract are the natural habitat for B pertussis and B parapertussis. Although B pertussis can survive outside the body for a few days and so may be transmitted by contaminated objects, most infections occur after direct contact with diseased persons—specifically, by inhalation of bacteria-bearing droplets expelled in cough spray. The patient is most infectious during the early catarrhal phase, when clinical symptoms are relatively mild and noncharacteristic (Fig. 31-5). Subclinical cases may have similar epidemiologic significance. Healthy carriers of B pertussis or B parapertussis are assumed to play no significant epidemiologic role. The natural habitat of B bronchiseptica is the respiratory tract of smaller animals such as rabbits, cats, and dogs. Therefore, human infections with B bronchiseptica are extremely rare and occur only after close contact with carrier animals.

Figure 31-5. Relationship of B pertussis to the developing antibody response during whooping cough.

Figure 31-5

Relationship of B pertussis to the developing antibody response during whooping cough.

Whooping cough, a highly communicable, worldwide infection, was once common and dangerous, killing many thousands of children per year. Widespread vaccination has caused a continuous decrease in incidence and mortality over the years, but large numbers of patients still die in countries where vaccination is inadequate. Whooping cough is mainly an infection of infants and children, although susceptibility is general. The disease is especially dangerous in the first 6 months of life. Neither season nor climate seems to affect the morbidity rate.

Diagnosis

Bordetellae can be cultured from nasopharyngeal swabs or nasopharyngeal secretions. The sensitivity of the method depends mainly on the technique of taking the swabs or secretions. Swabs (one for each nostril) should be introduced deeply into the nose as to reach the nasopharynx. Swabs should be made of dacron or calcium alginate, and they should be transported in half strength charcoal blood agar. Secretions should be from the nasopharynx using a suction device with a mucus trap. Nasopharyngeal secretions should be immediately plated onto Regan-Lowe medium, which has replaced Bordet-Gengou medium as the medium of choice. The transportation time for both materials should be kept as short as possible. For culture isolation, Bordet-Gengou agar containing blood, potato extract, and glycerol remains one of the effective means, although minor modifications regarding blood concentrations and addition of penicillin and nicotinamide have been recommended. For routine use, charcoal-blood agar (REGAN-LOWE medium) is most widely used. A (2,6-O-dimethyl)-b-cyclodextrin supplemented STAINER-SCHOLTE broth can be used as an enrichment medium. The Bordetella species do not need factors X and V (NAD+ and hemin).

Bordetella pertussis usually grows after 3 to 4 days of incubation at 37° C. The small, transparent colonies are indistinguishable from those of B bronchiseptica, but usually are smaller than those of B parapertussis. All three species produce hemolysis. Biochemically they are relatively inert and do not ferment carbohydrates or produce H2S and indole. An important characteristic of B parapertussis is its capacity to produce brown pigmentation on blood-free peptone agar. B pertussis and B parapertussis can be distinguished by certain biochemical and culture characteristics (Table 31-1) in addition to slide agglutination with specific antisera. B bronchiseptica as well as B avium and B hinzii can be differentiated by conventional methods for typing gram-negative nonfermenting rods (such as API-NE).

Table 31-1. Differential Characteristics of B pertussis and B parapertussis.

Table 31-1

Differential Characteristics of B pertussis and B parapertussis.

Detection of B pertussis and B parapertussis DNA by PCR has been described, and various primers and detection methods were applied. Ongoing studies will define the role of a standardized PCR method in pertussis diagnostics.

Circulating antibodies, appearing as late as week 3 of illness and reaching their maximum at weeks 8 to 10, have been demonstrated by agglutination and complement fixation tests. The agglutination test is applied mainly in epidemiologic studies. Although no direct relationship has been shown between the agglutinin concentration and the degree of protection, high agglutinin titers (>1:320) are assumed to correlate with protection from disease. Modern serologic techniques, such as enzyme-linked immunosorbent assay (ELISA), have been used to detect IgG, IgM, IgA, and IgE antibodies to both whole B pertussis cells and certain isolated components. In accordance with other serologic methods, seroconversion could be observed only 2 to 4 weeks after the onset of the disease (Fig. 31-5). The detection of specific IgA and IgM antibodies, however, is indicative of recent infection and is useful for the differential diagnosis of pertussiform syndromes of longer duration. IgA antibodies to pertussis toxin and FHA (filamentous hemagglutinin are found mainly after natural infection; the same is true of secretory IgA in nasopharyngeal secretions, which usually appears during week 2 or 3 of illness. Unfortunately, infants do not regularly produce IgA antibody before 6 months of age. Specific IgM antibodies may be used in infants as an indicator of acute infection.

Control

Although B pertussis is susceptible in vitro to several antibiotics, such as tetracycline, erythromycin, and chloramphenicol, the efficacy of these drugs in patients during the paroxysmal phase is not convincing. Treatment with erythromycin, which is usually considered the antibiotic of choice, will eliminate viable B pertussis organisms from the respiratory tract within a few days. The treatment, however, has no influence on the course of the disease. Human hyperimmune pertussis globulin is still used occasionally, but no reliable data support its efficacy. Further treatment is symptomatic.

Susceptible children (unimmunized children without a history of whooping cough) should have no contact with pertussis patients during the first 4 weeks of illness, although such isolation is often difficult. A patient treated with erythromycin may be contagious for only 5 to 10 days. Exposed immunized children younger than 4 years are given booster doses of pertussis vaccine. Exposed unimmunized children are given erythromycin for 10 days after contact is discontinued or after the patient ceases to be contagious.

Pertussis vaccine is produced from smooth forms (phase I) of the bacteria as a killed whole-cell vaccine. In the United States, pertussis vaccination of infants and children is recommended. Owing to a relatively mild course of disease and to occasional neurologic complications after vaccination, it has been argued by numerous pediatricians that general vaccination with the whole-cell vaccine is no longer justified.

Acellular pertussis vaccines have been developed, and were licensed in Japan since 1981 for children (older than two years), and also have been used in infants since 1990. These vaccines are composed very differently and contain various amounts of structural components from the bacteria. Components available for vaccine production include pertussis toxin (which is detoxified), filamentous hemagglutinin, a 69 kDa outer-membrane protein called pertactin, and fimbrial antigens 2 and 3. Some of these vaccines have been licensed in the U.S. for booster vaccinations since 1991. Recent data suggest that after primary vaccinations of infants these vaccines can convey similar levels of protection as the whole-cell vaccine. Thus, acellular vaccines have also been licensed in some European countries for primary vaccination.

References

  1. Alouf JE, Freer JH: Sourcebook of bacterial protein toxins. Academic Press, London, 1991 .
  2. Arai H, Munoz J. Crystallization of pertussigen from Bordetella pertussis. Infect Immun. 1981;31:495. [PMC free article: PMC351809] [PubMed: 6260667]
  3. Bemis DA, Kennedy JR. An improved system for studying of Bordetella bronchiseptica on the ciliary activity of canine tracheal epithelial cells. J Infect Dis. 1981;144:349. [PubMed: 7288215]
  4. Bordet J, Gengou U. Le microbe de la coqueluche. Ann Inst Pasteur. 1906;20:731.
  5. Finger H, Heymer B, Hof H. et al. Ueber Struktur und biologische Aktivitat von Bordetella pertussis Endotoxin. Zentralbl Bakteriol Mikrobiol Hyg 1 Abt OrigA. 1976;235:56. [PubMed: 185848]
  6. Finger H, Wirsing von Koenig CH: Enhancement and suppression of immune responsiveness by bacteria, bacterial products and extracts. p. 16. In Zschiesche W (ed): Immune Modulation by Infectious Agents. Gustav Fischer Verlag, Jena, 1987 .
  7. Finger H, Wirsing von Koenig CH. Serological diagnosis of whooping cough. Dev Biol Stand. 1985;61:331. [PubMed: 2872121]
  8. Goldman WE, Klapper DG, Basemann JB. Detection, isolation and analysis of a released Bordetella pertussis product toxic to cultured tracheal cells. Infect Immun. 1982;36:782. [PMC free article: PMC351298] [PubMed: 6177637]
  9. Goodman YE, Wort AJ, Jackson FL. Enzyme-linked immunosorbent assay for detection of pertussis immunoglobulin A in nasopharyngeal secretions as an indicator of recent infection. J Clin Microbiol. 1981;13:286. [PMC free article: PMC273779] [PubMed: 6259201]
  10. Kersters K, Hinz K-H, Hertle A. et al. Bordetella avium sp. nov., isolated from the respiratory tract of turkeys and other birds. Int J Syst Bacteriol. 1984;34:56.
  11. Meade BD, Mink CM, Manclark CR: Serodiagnosis of pertussis, p. 322. In: Manclark CR: Proc. 6th Intl Symp Pertussis, DHHS (FDA) Publication No. 90-1164; Bethesda, MD, 1990 .
  12. Meade BD, Bollen A. Recommendations for use of the polymerase chain reaction in the diagnosis of Bordetella pertussis infections. J Med Microbiol. 1994;41:51–55. [PubMed: 7911841]
  13. Monack D, Munoz JJ, Peacock MG. et al. Expression of pertussis toxin correlates with pathogenesis in Bordetella species. J Infect Dis. 1989;159:205. [PubMed: 2464653]
  14. Munoz JJ, Bergman RK: Bordetella pertussis. Vol. 4. Marcel Dekker, New York, 1977 .
  15. Pittman M. Pertussis toxin: the cause of the harmful effects and prolonged immunity in whooping cough. A hypothesis. Rev Infect Dis. 1979;1:402. [PubMed: 233166]
  16. Pittman M. The concept of pertussis as a toxin-mediated disease. Pediatr Infect Dis J. 1984;3:467. [PubMed: 6093069]
  17. Robinson A, Duggleby CJ, Gorringe AR et al: Antigenic variation in Bordetella pertussis. p. 147. In Birbeck TH, Penn CW (eds): Antigenic Variation and Infectious Diseases. Society for General Microbiology, IRL Press, Oxford, 1986 .
  18. Vandamme P, Hommez J, Vancanneyt M, Monsieurs M, Hoste B, Cookson BT, Wirsing von Konig CH, Kersters K, Blackall PJ. Bordetella hinzii sp. nov. isolated from poultry and humans. Int J Syst Bact. 1995;45:37–45. [PubMed: 7857806]
  19. Wardlaw AC, Parton R: Pathogenesis and Immunity in Pertussis. John Wiley & Sons, New York, 1988 .
  20. Weiss AA, Falkow S. Genetic analysis of phase change in Bordetella pertussis. Infect Immun. 1984;43:263. [PMC free article: PMC263420] [PubMed: 6317569]
  21. Wirsing von Koenig CH, Tacken A, Finger H. Use of supplemented Stainer-Scholte broth for the isolation of Bordetella pertussis from clinical material. J Clin Microbiol. 1988;26:2558. [PMC free article: PMC266945] [PubMed: 2906641]
Copyright © 1996, The University of Texas Medical Branch at Galveston.
Bookshelf ID: NBK7813PMID: 21413270

Views

  • PubReader
  • Print View
  • Cite this Page

More on the Subject in Bookshelf

Related Items in Bookshelf

Related information

  • PMC
    PubMed Central citations
  • PubMed
    Links to PubMed

Similar articles in PubMed

See reviews...See all...

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...