Coxiella burnetii: the query fever bacterium

12 downloads 0 Views 620KB Size Report
contains sugars such as L-virenose, dihydrohydroxy- streptose and ..... [9] Weisburg, W.G., Dobson, M.E., Samuel, J.E., Dasch, G.A.,. Mallavia, L.P., Baca, O., ...
FEMS Microbiology Reviews 19 (1997) 209^217

Coxiella burnetii

: the `query' fever bacterium

A model of immune subversion by a strictly intracellular microorganism Jean-Louis Mege, Max Maurin, Christian Capo, Didier Raoult *

Uniteè des Rickettsies, CNRS UPRESA 6020, Faculteè de Meèdecine, 27 Bd Jean Moulin, 13385 Marseille Cedex 05, France Received 20 February 1996 ; revised 8 November 1996 ; accepted 10 December 1996

Abstract Although substantial progress occurred in the knowledge of

Coxiella burnetii

during the past years, the pathophysiology of Q

fever is still obscure. Emerging evidence from clinical investigations suggested that certain disorders of cell-mediated immunity play a pivotal role in Q fever and especially in its chronic form. This review analyses the potential stategies that

C. burnetii

, a

strictly intracellular pathogen, use to divert microbicidal mechanisms of macrophages and to depress protective T-cell mediated immunity. The role of monocytes in the induction of Q fever is specifically discussed.

Keywords: Coxiella burnetii

; Q fever ; Macrophage ; Immunosuppression ; Cytokine

Contents 1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

209

2. What kind of bacterium is

210

3. How does

Coxiella burnetii

? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

Coxiella burnetii C. burnetii Coxiella burnetii Coxiella burnetii . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . C. burnetii divert microbicidal mechanisms ?

. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

211

in acidic vacuoles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

211

targeting . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

212

depress T cell-mediated immunity ? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

213

3.1. Survival of

3.2. Cell receptors and

3.3. Immunomodulation by 4. How does

212

5. Monocytes and Q fever endocarditis : a hypothesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

214

References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

215

1. Introduction

rium, is the etiologic agent of Q fever, a zoonosis

Coxiella burnetii

, an obligate intracellular bacte-

of worldwide distribution [1]. The precise prevalence of

C. burnetii

infections in humans is unknown as its

clinical picture is non-speci¢c and diagnosis is usu* Corresponding author. Tel : +33 (4) 91 83 43 75 ; fax : +33 (4) 91 83 03 90 ; e-mail : [email protected]

ally based on serology. In Southern France, the prevalence of acute Q fever is 50 per 100,000 inhabitants.

0168-6445 / 97 / $32.00 ß 1997 Federation of European Microbiological Societies. Published by Elsevier Science B.V.

PII

S0168-6445(96)00030-7

210

J.-L. Mege et al. / FEMS Microbiology Reviews 19 (1997) 209^217

Large outbreaks of Q fever have been also reported in Switzerland, Great Britain and Canada [2]. Q fever is characterized by two major evolutive forms with distinct prognosis. The clinical expression of the acute form is wide with acute febrile illness, pneumonia, hepatitis and meningoencephalitis. During acute infection, doughnut granulomas consisting of macrophages and lymphocytes have been found in liver and bone marrow biopsy specimens [3] whereas the alveolar exsudates of C. burnetii-induced pneumonia were composed of histiocytes [4]. All these lesions result from an e¤cient cell-mediated immune response limiting bacterial multiplication and leading to a favorable prognosis. On the other hand, the chronic form of Q fever may occur several months to years after acute infection and is usually expressed as an endocarditis with severe prognosis and more rarely as vascular infection. Q fever endocarditis is the main cause of culture-negative endocarditis, and represents approximatively 10% of all diagnosed endocarditis cases. In situ studies showed many microorganisms without granulomatous formation in heart valves [4]. Bone infection or chronic hepatitis may also indicate chronic Q fever. Clinical and epidemiological investigations demonstrated that the development of chronic Q fever is often associated with immunosuppression caused by Human Immunode¢ciency Virus (HIV) infection, cancer, lymphoma, chronic renal failure or pregnancy [5^8]. Hence, it is likely that some disorders of cell-mediated immunity play a pivotal role in chronic infection. Three main questions will be evoked in this review. First, what kind of bacterium is C. burnetii ? Second, how does C. burnetii divert microbicidal mechanisms and escape immune response? Third, how does C. burnetii cause endocarditis? 2. What kind of bacterium is

Coxiella burnetii ?

C. burnetii, a Gram-negative bacterium, is the only species of the genus Coxiella. It was initially classi¢ed in the Rickettsiaceae family and Rickettsiale order because of its obligate intracellular life and because it is transmitted by ticks. More recently, molecular biology techniques including 16S rRNA

gene sequence analysis have placed C. burnetii, Wolbachia persica and Legionella pneumophila in the

gamma

subdivision

of

Proteobacteria,

and

Rickettsia, Ehrlichia and Bartonella spp. in the alpha group [9,10]. Whereas C. burnetii and L. pneumophila

survive in soil and water and are most often transmitted to humans by aerosols, only C. burnetii is a strictly intracellular bacterium. The reservoirs of C. burnetii may be multiple. Its transmission to humans by tick bite is rare. Pets, speci¢cally pregnant females at the time of delivery, may be responsible for infection by C. burnetii since it displays high tropism for the female reproductive system, thus causing abortions. Humans are also infected by inhalation of infected fomites or, less frequently, after ingestion of contaminated dairy products such as milk [11]. Envelopes of C. burnetii contain outer membrane proteins, lipopolysaccharide (LPS) and A-1-gamma type peptidoglycan. Mutational variation in the LPS is linked to shifts in antigenicity and virulence, termed `phase variation'. Phase I C. burnetii are recovered from infected humans and animals, and are characterized by smooth-type LPS and high virulence. Conversely, phase II C. burnetii are obtained after serial passages in embryonated eggs or cell cultures, and express rough-type LPS and dramatically reduced virulence. In addition, the sugar composition of each LPS is quite di¡erent. LPS in phase I contains sugars such as L-virenose, dihydrohydroxystreptose and galactosamine uronyl-K-(1^6)-glucosamine which are lacking in phase II LPS [12,13]. Although phenotype reversion upon passage of phase II microorganisms through laboratory animals had been suspected, it is now evident that there is a selection for few phase I remaining in uncloned phase II bacteria [14]. In addition, C. burnetii undergoes a complex intracellular cycle leading to the formation of variants of di¡erent size and to sporulation under speci¢c environmental conditions. Although few genetic studies have been conducted, it is known that the genome of C. burnetii contains about 1600 kb [15]. Only a few chromosomal genes have been cloned and expressed in Escherichia coli : the gltA citrate synthase gene [16], the superoxide dismutase gene [17], the genes coding for the heat shock proteins htpA and htpB [18], and a gene coding for a 27 kDa surface antigen [19]. Base composition and codon usage have been analyzed to com-

J.-L. Mege et al. / FEMS Microbiology Reviews 19 (1997) 209^217 pare C. burnetii with other microorganisms. A codon bias is probable in C. burnetii since an inordinate use of U or A in the ¢rst and the third codons has been described. In contrast to the previous theory concerning codon usage, C. burnetii has a preference for A or U in the ¢rst and the third positions regardless the nucleotide in the second position [20]. In addition, three plasmids in di¡erent strains of C. burnetii have been described: QpH1, QpRS, and QpDG [21]. QpH1 and QpRS plasmids were tentatively associated with the onset of acute and chronic diseases respectively. The Nine Mile and Henzerling strains, recovered from ticks and humans su¡ering from acute disease, contained the QpH1 plasmid and were considered as the reference strains for acute Q fever. The Priscilla and Q212 strains, recovered from a goat chronically infected and from a patient with endocarditis, contained the QpRS plasmid and were considered as reference strains for chronic Q fever [22]. More recent investigations, using an enlarged panel of C. burnetii strains which were mainly obtained from infected patients, did not however con¢rm such hypothesis [23]. It is likely that these plasmids remain cryptic.

3. How does Coxiella mechanisms?

211

burnetii

divert microbicidal

3.1. Survival of C. burnetii in acidic vacuoles The maintenance of C. burnetii infection depends on the multiplication of the pathogen inside an acidic vacuole and the avoidance of microbicidal activities in host macrophages. In contrast to other intracellular bacteria such as Legionella or Mycobacteria, the adaptation of C. burnetii to the intracellular environment is a prerequisite for its survival and its multiplication [1]. The acidic environment allows the penetration of nutrients necessary for the metabolism of C. burnetii [24]. It also protects C. burnetii from antibiotic treatment since the activity of antibiotics is altered by acidic pH. This may account for Q fever relapses despite antibacterial treatment. When lysosomotropic agents such as chloroquine which alkalinize the phagolysosomal compartment were combined with doxycycline, macrophage microbicidal machinery towards C. burnetii was stimulated [25]. Several features characterize C. burnetii parasitism

Fig. 1. Intracellular fate of Coxiella burnetii. The virulent bacteria in phase I (CB I) are recognized by the LRI-IAP complex on human monocytes but are poorly phagocytozed. The avirulent bacteria in phase II (CB II) also bind CR3 of phagocytes accounting for an e¤cient phagocytosis. The survival and the multiplication of Coxiella burnetii is observed in mature phagolysosomes, i.e., expressing membrane proton ATPase and lysosomal markers LAMP1 and LAMP2, and containing lysosomal enzymes. Golgi vesicles are not engaged in the intracellular fate of C. burnetii. It is probable that the acidic pH of phagolysosomes protects C. burnetii of the activity of antibiotics. Oxygen scavengers expressed by C. burnetii impair the oxidative response, a major antimicrobial defense of macrophages.

J.-L. Mege et al. / FEMS Microbiology Reviews 19 (1997) 209^217

212

sistent infection of ¢broblastic cell lines such as

Legionella pneumophila or Mycobacterium tuberculosis [32]. The interaction of C. burnetii with

Vero, L929 and HEL cells and myeloid cells such

human monocytes and macrophages involves anoth-

as P388D1 cells. But, in vivo, its multiplication is

er class of integrins. The binding of

apparently restricted to cells belonging to the mono-

human monocytes is mediated by the complex con-

nuclear phagocyte system [26]. Second, infected mur-

sisting of LRI (Leukocyte Response Integrin,

ine macrophage-like cells show a single vacuole ¢lled

and IAP (Integrin Associated Protein) [33]. While

with microorganisms, which is able to fuse with ly-

phase I (virulent form) which is weakly internalized

sosomes. Vacuoles containing

only engages the integrin complex,

(Fig. 1). First,

C. burnetii can establish in vitro per-

C. burnetii ful¢lled the

spp.,

C. burnetii

KvL3)

C. burnetii L2

to

in

criteria of mature phagolysosomes including the se-

phase II (avirulent form) also bind

integrins,

questration of £uid phase markers, the expression of

thus accounting for its more e¤cient phagocytosis

proton ATPase and lysosomal glycoproteins LAMP-

(manuscript in preparation). It is tempting to spec-

1 and LAMP-2, and lysosomal enzymes such as

ulate that LRI and IAP are required for the adhesion

cathepsin D and acid phosphatase. In addition, these

of

vacuoles do not interact with Golgi-derived vesicles

C. burnetii to several cell types but the fate of C. burnetii in monocytes and macrophages is in£uenced

containing

by the engagement of speci¢c coreceptors (Fig. 1).

newly

synthesized

sphingolipids

Third, phagolysosomes containing

C. burnetii

[27]. have

a pH of 5.2 [26,27]. It has been suggested that the

3.3. Immunomodulation by Coxiella burnetii

acidi¢cation of a maturing phagosome depends upon the delivery to the phagosome of the vacuolar proton

The subversion of macrophage microbicidal mech-

C. burnetii to survive in acidic

pump which is present in early and late endosomes

anisms is required for

[28]. This ATPase is lacking in phagosomes contain-

vacuoles. First of all, limiting the microbicidal e¡ects

ing mycobacteria, known to inhibit phagosome-ly-

of reactive oxygen intermediates is a strategy used by

some fusion and to survive in alkaline environment

occasionally intracellular pathogens such as

[29].

Leishmania spp. and C. burnetii share the prop-

Mycobacteria spp. and Legionella spp. [34,35]. C. burnetii

erty of surviving and multiplying in acidic phagoly-

possesses oxygen scavengers such as superoxide dis-

sosomes. While particles such as yeasts,

mutase and catalase [36] which can blunt the oxida-

L glucan and

C. burnetii did not induce superoxide

latex beads were easily transferred to vacuoles con-

C. burnetii in CHO cells, the e¤ciency of the

tive response.

taining

by itself and did not depress agonist-induced super-

vectorial transfer was dampened in cells infected with

oxide generation in human monocytes (unpublished

Leishmania amazonensis [30]. Although C. burnetii and L. amazonensis were con¢ned within the same

data). Second, the decreased generation of macrophage cytotoxic cytokines such as Tumor Necrosis

K is bene¢cial for the survival of intra-

vacuoles in double infected cells, it is likely that the

Factor (TNF)

speci¢c behavior of

cellular pathogens. For instance, lipoarabinomannan

C. burnetii

depends on cellular

from virulent isolates of

events upstream from phagosome formation.

3.2. Cell receptors and Coxiella burnetii targeting During the past few years, it has become evident

gers TNF

K

M. tuberculosis poorly trig-

production whereas that from avirulent

isolates markedly increases TNF

burnetii

K

release [37].

C.

exhibits a distinct behavior. Virulent phase

I bacteria highly stimulated the synthesis and the

K

that numerous microorganisms use speci¢c eucary-

secretion of TNF

otic receptors such as integrins to invade host cells.

avirulent phase II bacteria were less e¤cient (manu-

Integrins

script

are

heterodimeric

glycoproteins

whose

in

in human monocytes whereas

preparation).

Cytokines

such

as

K

TNF

C. burnetii,

binding speci¢city is generated by varied combina-

probably favor the ingestion of phase I

tions of 14

which is poorly internalized, by upregulating adhe-

K

subunits and 8

L

subunits identi¢ed

to date [31] (Fig. 1). One of these receptors, CR3 (

KM L2)

is known to be one of the most important

sion

receptors

Moreover, TNF

on

K

monocytes

and

macrophages.

may also trigger pathophysiologi-

phagocyte receptors involved in the binding and the

cal responses associated with acute infection such as

uptake of bacteria and parasites such as

the development of granulomas [3]. Third, mainte-

Leishmania

J.-L. Mege et al. / FEMS Microbiology Reviews 19 (1997) 209^217 nance of C. burnetii infection probably involves either the depression of speci¢c T cell responses and/or the down-modulation of macrophage responsiveness. The decrease in cell-mediated immunity by ionizing radiations, corticosteroids, or parturition modi¢es the natural resistance of mice to C. burnetii [38]. Athymic mice became chronically infected by bacteria in phase I whereas euthymic mice were able to kill them [39]. The onset of chronic Q fever is largely determined by the immunological status of human recipients. Several sporadic cases of Q fever have been reported in patients immunocompromised by Crohn's disease, alcoholism, or immunosuppressive treatment [5,40]. Q fever also occurred as a complication of hematologic and solid neoplasms [41]. A higher percentage of HIV-infected individuals were positive for Q fever antibodies as compared with blood donors [8]. It is probable that antigen-presenting defect of monocytes and dendritic cells, an initial event in HIV infection [42], favors the persistence of C. burnetii in macrophages without the development of protective immune response. In addition, Q fever is reactivated during pregnancy [43,44], suggesting that the fetalplacental unit secretes Th2 cytokines to divert the maternal Th1-mediated response [45]. On the other hand, the down-modulation of macrophage responses to lymphokines may create a protected environment for the pathogen. The priming of microbicidal functions of macrophages by interferon (IFN) Q [46] limits the multiplication of intracellular parasites and bacteria including C. burnetii [47,48]. However, we recently found that monocytes infected with C. burnetii cannot be primed by IFNQ for oxidative metabolism (unpublished data). This de¢ciency is reminiscent of defective IFNQ-mediated priming for cytotoxicity observed in macrophages from A/J mice which are highly susceptible to C. burnetii infection [49]. 4. How does C. immunity?

burnetii depress T cell-mediated

In addition to subverted microbicidal functions of macrophages, the survival of C. burnetii requires the impairment of protective T cell responses. Di¡erent strategies may be used: down-modulation of Th1

213

lymphokine production, alteration of antigen processing and/or presentation, and induction of suppressor factors. Ine¤cient antigen presentation and down-modulation of MHC Class II molecules have been reported in M. tuberculosis-infected monocytes [50] and may be operative in C. burnetii infection. They may result from inhibition of the intracellular tra¤c of MHC Class II molecules or peptides and/or secretion of cytokines depressing antigen-presenting activity, such as interleukin (IL)-10. Moreover, leishmanial or mycobacterial infection down-modulated the expression of B7 and ICAM-1 antigens [51] which are cosignal molecules necessary for T cell activation [52]. To date, it is not possible to establish either hypothesis. The mechanisms of antigen-dependent suppression are more documented. Peripheral blood lymphocytes (PBL) from patients who have convalesced from acute Q fever and patients with active Q fever hepatitis manifested a marked proliferative response when cultured in vitro with C. burnetii antigen [53]. Similarly, a high percentage of individuals exposed to low infection risk exhibited speci¢c lymphoproliferative response early after vaccination with formalin-inactivated C. burnetii [54,55]. A large percentage of antigen-stimulated PBL secreted IFNQ [56]. In contrast, PBL from patients with Q fever endocarditis did not proliferate in the presence of C. burnetii antigen while their response to Candida albicans antigen or mitogens was maintained [57]. This suppression is not mediated by CD8‡ T cells since their removal did not restore lymphoproliferation, in contrast to lepromatous leprosis [57]; it seems that prostaglandins (PG) depress T-cell mediated immunity against C. burnetii. High amounts of PGE2 were produced in response to C. burnetii by peripheral blood mononuclear cells (PBMC) from patients with endocarditis; PBL proliferation was restored by indomethacine [57]. Cytokines such as Transforming Growth Factor (TGF) L and IL-10 are produced in excess by PBMC and monocytes from patients with Q fever endocarditis, and we showed that the increase in IL-10 production was associated with the occurrence of Q fever relapses [58]. These mediators may also be involved in the down-modulation of speci¢c T cell responses. We also found that T cell compartment may be a¡ected in chronic Q fever. Indeed, these patients exhibited CD4‡ T cell lym-

J.-L. Mege et al./FEMS Microbiology Reviews 19 (1997) 209^217

214

Coxiella burnetii

Fig. 2. A model for Q fever endocarditis. stimulated the secretion of proin£ammatory cytokines such as TNFK, known to up-regulate adhesion molecules, possibly integrins and selectins. The interaction between infected macrophages and endothelium is favored by this up-regulation of adhesion molecules and chemokine synthesis, leading to the colonization of valvular tissues. On the other hand, the infection is characterized by a defective cell-mediated immunity probably driven by a cytokine such as IL-10. The resulting impaired production of IFNQ by lymphocytes limits the microbicidal response of macrophages and then favors the intracellular survival of .

C. burnetii C. burnetii

phopenia related to the activity of the disease. The study of CD45RO and CD45RA expression by CD4‡ T cells provided evidence that lymphopenia preferentially a¡ected unprimed lymphocytes [59]. The induction of suppressive mediators by monocytes may result from the action of speci¢c bacterial determinants. Indeed, lipoarabinomannan isolated from and inhibited T cell proliferation responses, the expression of IL-2 gene in human T cells and the IFNQ-mediated activation of macrophages [60]. Growing monocytes and T cells from vaccinated individuals with phase I led to residual production of IFNQ whereas phase II bacteria stimulated strong IFNQ production. Pretreatment of phase I LPS with periodate, which yields an arti¢cial phase II LPS, restored IFNQ production [56]. In addition, treating phase I with trichloroacetic acid yielded a soluble antigen consisting of LPS and proteins, and a residue with low amounts of LPS. While the residue clearly elicited lympho-proliferative response, the soluble antigen was weakly e¡ective [61]. Hence, LPS probably promotes the suppression of IFNQ production by

M. tuberculosis

M. leprae

C. burnetii C. burnetii

PBL from patients with Q fever endocarditis by masking bacterial determinants critical for lymphokine production. Indeed, the extended structure of phase I LPS might sterically block the access of speci¢c antibodies to surface proteins [62]. 5. Monocytes and Q fever endocarditis: a hypothesis

Q fever endocarditis is one of the rare endocardites caused by an intracellular pathogen; most of the infections of endocardial heart surfaces are caused by extracellular microorganisms, such as streptococci or staphylococci [63]. This endocarditis is atypical because it is associated with rare vegetations and negative blood culture. These cause diagnosis delay, accounting for the especially severe prognosis [2]. Clearly, the occurrence of Q fever endocarditis does not depend on the sub-type of isolate (see Section 2). It is now accepted that the onset of Q fever endocarditis requires previous valvular lesions and/or defective cell-mediated immunity [5]. induced valvular endocar-

C.

burnetii

C. burnetii

J.-L. Mege et al. / FEMS Microbiology Reviews 19 (1997) 209^217 ditis in mice treated by cyclophosphamide but not in immunocompetent mice without preinjury of cardiac valves [64]. Clinical investigations of chronic Q fever have revealed suppression of speci¢c T cell responses [57] and activation of circulating monocytes, assessed by the release of in£ammatory cytokines [65] (Fig. 2). The strictly intracellular behavior of C. burnetii, excluding a direct colonization of valvular tissues, implies its vectorization by monocytes. Nevertheless, the infection of circulating monocytes cannot account for their activation and their speci¢c targeting since few monocytes from patients with endocarditis harbor the pathogen. Since the interaction between these few infected cells and valvular tissue would be rare, additional factors are probably required for valvular colonization. As it is established that immune complexes are constantly found in chronic Q fever [2], we hypothesize that immune complexes play a crucial role in the initiation of C. burnetii endocarditis. They would elicit the release of eicosanoids and cytokines by monocytes [66] and suppress protective cell-mediated immunity via the induction of IL-10 [67]. In the context of Q fever, once circulating monocytes are activated by immune complexes, they release TNFK and IL-1L which upregulate the expression of cell adhesion molecules including L integrins. Monocytes can then ¢rmly attach to modi¢ed valvular tissue. Its modi¢cation may result in the release of chemoattractants, such as L chemokines, which may favor the tra¤c of monocytes, thus creating the conditions for the targeting of the minority of monocytes containing C. burnetii. Our hypothesis will now be tested in dynamic conditions with infected monocytes and modi¢ed endothelium.

References [1] Baca, O.G. and Paretsky, D. (1983) Q fever and Coxiella burnetii : a model for host-parasite interaction. Microbiol. Rev. 47, 127^149. [2] Raoult, D. and Marrie, T. (1995) Q fever. State of the art. Clin. Infect. Dis. 20, 489^496. [3] Khavkin, T. (1990) Experimental studies of the infectious process in Q fever. In: Q Fever. The Disease (Marrie T.J., Ed.), Vol. 1, pp. 71^106. CRC Press, Boca Raton, FL. [4] Sawyer, L.A., Fishbein, D.B. and McDade, J.E. (1987) Q fever: current concepts. Rev. Infect. Dis. 9, 935^946.

215

[5] Raoult, D. (1990) Host factors in the severity of Q fever. Ann. N.Y. Acad. Sci. 590, 33^38. [6] Brouqui, P., Tissot Dupont, H., Drancourt, M., Berland, Y., Etienne, J., Leport, C., Goldstein, C., Massip, P., Micoud, M., Bertrand, A. and Raoult, D. (1993) Chronic Q fever: 92 cases from France including 27 without endocarditis. Arch. Int. Med. 153, 642^648. [7] Raoult, D., Levy, P.Y., Harle, J.R., Etienne, J., Massip, P., Goldstein, F., Micoud, M., Beytout, J., Gallais, H., Remy, G. and Capron, J.P. (1990) Chronic Q fever: diagnosis and follow-up. Ann. N.Y. Acad. Sci. 590, 51^60. [8] Raoult, D., Levy, P.Y., Tissot Dupont, H., Chicheportiche, C., Tamalet, C., Gastaut, J.A. and Salducci, J. (1993) Q fever and HIV infection. AIDS 7, 81^86. [9] Weisburg, W.G., Dobson, M.E., Samuel, J.E., Dasch, G.A., Mallavia, L.P., Baca, O., Mandelco, L., Sechrest, J.E., Weiss, E. and Woese, C.R. (1989) Phylogenetic diversity of the Rickettsiae. J. Bacteriol. 171, 4202^4206. [10] Stein, A., Saunders, N.A., Taylor, A.G. and Raoult, D. (1993) Phylogenic homogenicity of Coxiella strain as determined by 16S ribosomal RNA sequencing. FEMS Microbiol. Lett. 113, 339^344. [11] Lang, G.H. (1990) Coxiellosis (Q fever) in animals. In: Q Fever. The Disease (Marrie T.J., Ed.), Vol. 1, pp. 23^48. CRC Press, Boca Raton, FL. [12] Amano, K.I., Williams, J.C., Missler, S.R. and Reinhold, V.N. (1987) Structure and biological relationships of Coxiella burnetii lipopolysaccharides. J. Biol. Chem. 262,4740^4747. [13] Hackstadt, T. (1990) The role of lipopolysaccharides in the virulence of Coxiella burnetii. Ann. N.Y. Acad. Sci. 590, 27^ 32. [14] Hackstadt, T. (1996) Biosafety concerns and Coxiella burnetii. Trends Microbiol. 4, 341^342. [15] Myers, W.F., Baca, O.G. and Wisseman, C.L. (1980) Genome size of the Rickettsia Coxiella burnetii. J. Bacteriol. 144, 460^ 461. [16] Heinzen, R.A. and Mallavia, L.P. (1987) Cloning and functional expression of the Coxiella burnetii citrate synthase gene in Escherichia coli. Infect. Immun. 55, 848^855. [17] Heinzen, R.A., Frazier, M.E. and Mallavia, L.P. (1992) Coxiella burnetii superoxide dismutase gene: cloning, sequencing, and expression in Escherichia coli. Infect. Immun. 60, 3814^ 3823. [18] Vodkin, M.H. and Williams, J.C. (1988) A heat shock operon in Coxiella burnetii produces a major antigen homologous to a protein to both Mycobacteria and Escherichia coli. J. Bacteriol. 170, 1227^1234. [19] Hendrix, L.R., Samuel, J.E. and Mallavia, L.P. (1990) Identi¢cation and cloning of a 27-kDa Coxiella burnetii immunoreactive protein. Ann. N.Y. Acad. Sci. 590, 534^540. [20] Minnick, M.F., Heinzen, R.A., Frazier, M.E. and Mallavia, L.P. (1990) Characterization and expression of cbbE' gene of Coxiella burnetii. J. Gen. Microbiol. 136, 1099^1107. [21] Samuel, J.E., Frazier, M.E., Kahn, M.L., Thomashow, L.S. and Mallavia, L.P. (1983) Isolation and characterization of a plasmid from phase I Coxiella burnetii. Infect. Immun. 41, 488^493.

J.-L. Mege et al. / FEMS Microbiology Reviews 19 (1997) 209^217

216

[22] Hendrix, L.R., Samuel, J.E. and Mallavia, L.P. (1991) Di¡erentiation of

Coxiella burnetii

isolates by analysis of restriction-

endonuclease-digested DNA separated by SDS-PAGE. J. Gen.

[23] Stein, A. and Raoult, D. (1993) Lack of pathotype speci¢c

Coxiella burnetii

. Microb. Pathog. 15, 177^185.

[24] Hackstadt, T. and Williams, J.C. (1981) Biochemical stratagem for obligate parasitism of eukaryotic cells by

burnetii

Coxiella

[39] Kishimoto,

Cox-

. J. Gen. Microbiol. 132, 1415^1422. R.A,

Rozmiarek,

H.

and

Larson,

E.W.

[25] Maurin, M., Benoliel, A.M., Bongrand, P. and Raoult, D. (1992) Phagolysosomal alkalinisation and the bactericidal ef-

Coxiella burnetii

mice. Infect. Immun. 22, 69^71. [40] Heard, S.R., Ronalds, C.J. and Hearth, R.B. (1985)

burnetii

(1978)

paradigm. J. Infect.

11, 15^18.

(1992)

D.,

(1983) Lysosomal response of a murine macrophage-like cell

Coxiella burnetii

Brouqui,

Acute and

P.,

Marchon,

chronic

Q

fever

B.

in

and

Gastaut,

patients

with

J.A.

cancer.

Clin. Infect. Dis. 14, 127^130. [42] Helbert, M.R., L'Age-Stehr, J. and Mitchison, N.A. (1993) Antigen

Dis. 166, 1097^1102. [26] Akporiaye, E.T., Rowatt, J.D., Aragon, A.A. and Baca, O.G.

Coxiella

infection in immunocompromised hosts. J. Infect. Dis.

[41] Raoult,

. Proc. Natl. Acad. Sci. USA 78, 3240^3244.

fect of antibiotics : the

iella burnetii

Experimental Q fever infection in congenitally athymic nude

Microbiol. 137, 269^276.

gene in human

model to explain persistent infection of host cells with

presentation,

loss

of

immunological

memory

and

AIDS. Immunol. Today 14, 340^344. [43] Riechman,

N.,

Raz,

R.,

Keysary,

A.,

Goldwasser,

R.

and

. Infect. Immun.

Flatan, E. (1988) Chronic Q fever and severe thrombocytope-

[27] Heinzen, R.A., Scidmore M.A., Rockley D.D. and Hackstadt,

[44] Raoult, D. and Stein, A. (1994) Q fever during pregnancy, a

T. (1996). Di¡erential interaction with endocytic and exocytic

risk for women, foetuses and obstetricians. N. Engl. J. Med.

line persistently infected with

nia in a pregnant woman. Am. J. Med. 85, 253^254.

40, 1155^1162.

pathways

burnetii

distinguish

parasitophorous

Chlamydia trachomati

and

vacuoles

Coxiella

s. Infect. Immun. 64, 796^

809. [28] Clemens, D.L. (1996) Characterization of the

tuberculosis

of

Mycobacterium

Leishmania

and

Mycobacterium

T.G.,

Lin,

H.,

Guilbert,

L.

and

Mossman,

T.

(1993) Bidirectional cytokine interactions in the maternal-fetal relationship : is successful pregnancy a Th2 phenomenon ? Immunol. Today 14, 353^356.

phagosome. Trends Microbiol. 4, 113^118.

[29] Russel, D.G. (1994) Intracellular parasitism : the contrasting styles of

330, 371. [45] Wegmann,

. Bailliere's Clin. In-

[46] Nathan, C.F., Murray, H.W., Wiebe, M.E. and Rubin, B.Y. (1983). Identi¢cation of interferon-

Q

as the lymphokine that

activates human macrophage oxidative metabolism and anti-

fect. Dis. 1, 227^243. [30] Veras, P.S.T., De Chastellier, C., Moreau, M.F., Villiers, V.,

microbial activity. J. Exp. Med. 158, 670^689.

Thibon, M., Mattei, D. and Rabinovitch, M. (1994) Fusion

[47] Horwitz, M.A. and Silverstein, S.C. (1981) Activated human

between large phagocytic vesicles : targeting of yeast and other

monocytes inhibit the intracellular multiplication of Legion-

particulates

to

phagolysosomes

Coxiella burnetii

or

the

that

protozoan

shelter

the

bacterium

Leishmania amazonensis

in Chinese hamster ovary cells. J. Cell Sci. 107, 3065^3076. [31] Hynes, R.O. (1992) Integrins : versatility, modulation and sig-

of

microorganisms

by

integrin

receptors.

[33] Brown, E., Hooper, L., Ho, T. and Gresham, H. (1990) Integrin-associated protein : a 50-kD plasma membrane antigen physically and functionally associated with integrin. J. Cell

[34] Orme, I.M. (1994) Experimental models of chronic mycobac-

Coxiella burnetii

in mouse ¢bro-

to

phase

I

Coxiella burnetii

.

J.

Gen.

Microbiol.

133,

691^

[50] Gercken,

[35] Jacob T., Escallier, J.C., Sanguedolce, M.V., Chicheportiche, C., Bongrand, P., Capo, C. and Mege, J.L. (1994)

Legionella

inhibits superoxide generation in human mono-

K

and

L

protein kinase C

Pryjma,

J.,

Ernst,

M.

sis

and

Flad,

H.D.

(1994)

Mycobacterium tuberculo-

-infected monocytes. Infect. Immun. 62, 3472^3478.

and

P.M.

antigen

(1994)

Leishmaniasis :

presentation.

immunology,

Bailliere's

Clin.

persistence

Infect.

Dis.

1,

[52] Linsley, P.S. and Ledbetter, J.A. (1993) The role of the CD28 receptor during T cell responses to antigen. Annu. Rev. Immunol. 11, 191^212. [53] Koster, F.T., Williams, J.C. and Goodwin, J.S. (1985) Cellular immunity in Q fever : modulation of responsiveness by a sup-

isotypes. J. Leuk. Biol. 55, 310^312. [36] Akporiaye, E.T. and Baca, O.G. (1983) Superoxide production and superoxide dismutase and catalase activities in

J.,

Defective antigen presentation by

351^366.

terial diseases. Bailliere's Clin. Infect. Dis. 1, 367^384.

iella burnetii

inhibits growth of

mal models in Q fever : pathological response of inbred mice

[51] Kaye,

Biol. 111, 2785^2794.

cytes via the down-modulation of

Q

blasts. Infect. Immun. 45, 781^783.

700.

Trends Microbiol. 2, 10^14.

pneumophila

feron-

[49] Scott, G.H., Williams, J.C. and Stephenson, E.H. (1987) Ani-

naling in cell adhesion. Cell 69, 11^25. [32] Isberg, R.R. and Tran Van Nhieu, G. (1994) Binding and internalization

naires' disease bacteria. J. Exp. Med. 154, 1618^1635. [48] Turco, J., Thompson, H.A. and Winkler, H.H. (1984) Inter-

Cox-

. J. Bacteriol. 154, 520^523.

[37] Chatterjee, D., Roberts, A.D., Lowell, K., Brennan, P.J. and Orme, I.M. (1992) Structural basis of capacity of lipoarabinomannan to induce secretion of tumor necrosis factor. Infect. Immun. 60, 1249^1253. [38] Roman, M.J., Coriz, P.D and Baca, O.G. (1986) A proposed

pressor T cell-monocyte circuit. J. Immunol. 135, 1067^1072. [54] Ascher, M.S., Williams, J.C. and Berman, M.A. (1983) Dermal granulomatous hypersensitivity in Q fever : comparative studies of the granulomatous potential of whole cells of

iella burnetii

Cox-

phase I and subfractions. Infect. Immun. 42, 887^

889. [55] Izzo,

A.A.,

Marmion,

B.P.

and

Worswick,

D.A.

(1988)

Markers of cell-mediated immunity after vaccination with an

J.-L. Mege et al. / FEMS Microbiology Reviews 19 (1997) 209^217 inactivated,

whole-cell

Q

fever

vaccine.

J.

Infect.

Dis. 157,

[62] Hackstadt, T. (1988) Steric hindrance of antibody binding to surface proteins of

781^789. [56] Izzo, A.A. and Marmion, B.P. (1993) Variations in interferon-

Coxiella burnetii

217

Coxiella burnetii

by phase I lipolysacchar-

ide. Infect. Immun. 56, 802^807.

antigens with lympho-

[63] Sheld, W.M. and Sande, M.A. (1995) Endocarditis and intra-

cytes from vaccinated or naturally infected subjects. Clin. Exp.

vascular infections. In : Principles and Practice of Infectious

Immunol. 94, 507^515.

Diseases

gamma responses to

[57] Koster, F.T., Williams, J.C. and Goodwin, J.S. (1985) Cellular immunity in Q fever : speci¢c lymphocyte unresponsiveness in

[58] Capo, C., Za¡ran, Y., Zugun, F., Houpikian, P., Raoult, D. and Mege, J.L. (1996) Production of interleukin-10 and transgrowth

factor

L

by peripheral

blood mononuclear

cells in Q fever endocarditis. Infect. Immun. 64, 4143^4147. [59] Sabatier,

F.,

Dignat-George,

F.,

Mege,

Raoult, D. and Sampol, J. (1997) CD4

‡

J.L.,

Brunet,

C.,

T cell lymphopenia

in Q fever endocarditis. Clin. Diag. Lab. Immunol. 4, 89^92. [60] Ilangumaran,

S.,

Arni,

S.,

Poincelet,

M.,

G.L.,

Bennett

J.E.

and

Dolin

R.,

Eds),

[64] Atzpodien, E., Baumga ë rtner, W., Artlet, A. and Thiele, D. (1994) Valvular endocarditis occurs as a part of a dissemi-

Q fever endocarditis. J. Infect. Dis. 152, 1283^1289.

forming

(Mandell

pp. 740^783. Churchill Livingstone, New York.

Theler,

J.M.,

nated

Coxiella burnetii

infection

in

immunocompromised

d

Balb/cJ (H-2 ) mice infected with the Nine Mile isolate of

C. burnetii

. J. Infect. Dis. 170, 223^226.

[65] Capo, C., Zugun, F., Stein, A., Tardei, G., Lepidi, H., Raoult,

K and IL-1L in

D. and Mege, J.L. (1996) Upregulation of TNF

Q fever endocarditis. Infect. Immun. 64, 1638^1642. [66] Debets, J.M.H., Van der Linden, C.J., Dieteren, I.E.M., Leeuwenberg,

J.F.M.

and

Buurman,

W.A.

(1988)

Fc

receptor

Brennan, P.J., Din, N.U. and Hoessli, D.C. (1995) Integration

cross-linking induces rapid secretion of tumor necrosis factor

of mycobacterial lipoarabinomannans into glycosylphosphati-

(cachectin) by human peripheral blood monocytes. J. Immu-

dylinositol-rich

domains

of

lymphomonocytic

cell

plasma

[61] Lukacova, M., Brezina, R., Schramek, S. and (1989) Chemical composition of phase I

nol. 141, 1197^1201. [67] Tripp, C.S., Beckerman, K.P. and Unanue, E.R. (1995) Im-

membrane. J. Immunol. 155, 1334^1342. Pastorek, J.

mune complexes inhibit antimicrobial responses through inter-

solu-

leukin-10 production. E¡ects in severe combined immunode-

Coxiella burnetii

ble antigen prepared by trichloracetic acid extraction. Acta

¢cient mice during

Virol. 33, 75^80.

1634.

Listeria

infection. J. Clin. Invest. 95, 1628^