You are on page 1of 11

J. Comp. Path. 2013, Vol. 148, 115e125 Available online at www.sciencedirect.

com

www.elsevier.com/locate/jcpa
INFECTIOUS DISEASE: REVIEW ARTICLE

Kochs Postulates and the Pathogenesis of


Comparative Infectious Disease Causation
Associated with Bartonella species
E. B. Breitschwerdt*, K. L. Linder*, M. J. Day, R. G. Maggi*,
B. B. Chomel and V. A. J. Kempfx
*Intracellular Pathogens Research Laboratory, Center for Comparative Medicine and Translational Research, College of
Veterinary Medicine, North Carolina State University, Raleigh, NC, USA, School of Veterinary Sciences, University of
Bristol, Langford, Bristol BS40 5DU, UK, Department of Population Health and Reproduction, School of Medicine,
University of California, Davis, CA 95616, USA and x Institut fur Medizinische Mikrobiologie und Krankenhaushygiene,
Universitatsklinikum, Goethe-Universitat, Paul Ehrlich Str. 40,
60596 Frankfurt am Main, Germany

Summary
In his homage to Lucretius (Georgica), Vergil is credited with stating: Felix qui potuit rerum cognoscere cau-
sas (Fortunate is he who knows the causes of things). Based on numerous commentaries and publications it is
obvious that clinicians, diagnosticians and biomedical research scientists continue to struggle with disease cau-
sation, particularly in the assessment of the pathogenic role of stealth pathogens that produce persistent in-
fections in the host. Bartonella species, because of their evolutionary ability to induce persistent intravascular
infections, present substantial challenges for researchers attempting to clarify the ability of these stealth bacte-
ria to cause disease. By studying the comparative biological and pathological behaviour of microbes across
mammalian genera, researchers might be able more rapidly to advance medical science and, subsequently, pa-
tient care by undertaking focused research efforts involving a single mammalian species or by attempting to
recapitulate a complex disease in an rodent model. Therefore, in an effort to further assist in the establishment
of disease causation by stealth pathogens, we use recent research observations involving the genus Bartonella to
propose an additional postulate of comparative infectious disease causation to Kochs postulates.
2013 Elsevier Ltd. All rights reserved.

Keywords: disease; infection; pathology; Kochs postulates

Contents

Persistent Infection and Complex Disease Causation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 116


Bartonella spp. Infection and Vasoproliferative Pathology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 117
Bartonella spp. Infection and Granulomatous Inflammation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 119
Bartonella spp. Infection and Endocarditis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 120
Bartonella spp. and Chronic Intravascular Infection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 120
Concluding Remarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 121
Conflict of Interest Statement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 122
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 122

Correspondence to: E.B. Breitschwerdt (e-mail: ed_breitschwerdt@ncsu.


edu).

0021-9975/$ - see front matter 2013 Elsevier Ltd. All rights reserved.
http://dx.doi.org/10.1016/j.jcpa.2012.12.003
116 E.B. Breitschwerdt et al.

Persistent Infection and Complex Disease proposed including (1) a nucleic acid sequence be-
Causation longing to the putative pathogen should be present
in most cases of an infectious disease. Microbial nu-
Based on his work related to anthrax and the detec- cleic acids should be found preferentially in those or-
tion of the tubercle bacillus, Robert Koch proposed gans or gross anatomical sites known to be diseased
a set of postulates for establishing disease causation and not in those organs that lack pathology, (2) fewer,
by an infectious agent on March 24, 1882, in Berlin, or no, copy numbers of pathogen-associated nucleic
Germany (Loeffler, 1884; Tabrah, 2011). With strong acid sequences should occur in hosts or tissues without
support from the scientific community, these postu- disease, (3) with resolution of disease, the copy num-
lates ultimately became the foundation for establish- ber of pathogen-associated nucleic acid sequences
ing the pathogenic role of a microorganism in should decrease or become undetectable. With clini-
a disease process. The original postulates included cal relapse, the opposite should occur, (4) when se-
(1) the organism must be regularly associated with quence detection predates disease or sequence copy
the disease and its characteristic lesions, (2) the organ- number correlates with severity of disease or pathol-
ism must be isolated from the diseased host and grown ogy, the sequenceedisease association is more likely
in culture and (3) the disease must be reproduced to be a causal relationship, (5) the nature of the mi-
when a pure culture of the organism is introduced croorganism inferred from the available sequence
into a healthy susceptible host. Subsequently, a fourth should be consistent with the known biological char-
postulate was added by Friedrich August Loeffler, acteristics of that group of organisms, (6) tissueese-
a German physician: (4) the same organism must be quence correlates should be sought at the cellular
reisolated from the experimentally infected host level and efforts should be made to demonstrate spe-
(Tabrah, 2011). cific in-situ hybridization of the microbial sequence
In 1988, in recognition of the advances in technol- to areas of tissue pathology and to visible microorgan-
ogy related to microbial identification and classifica- isms or to areas where microorganisms are presumed
tion, Falkow proposed an alternative set of to be located and (7) these sequence-based forms of
molecular Kochs postulates, which could be applied evidence for microbial causation should be reproduc-
to microbial studies designed to examine the role of ible. Fredericks and Relman concluded that the
specific genes and their products in the pathogenesis power of Kochs postulates comes not from their rigid
of infection and disease (Falkow, 1988). These postu- application, but from the spirit of scientific rigour that
lates included (1) the phenotype or property under in- they foster and that proof of disease causation rests on
vestigation should be associated with pathogenic the concordance of scientific evidence, with Kochs
members of a genus or pathogenic strains of a species, postulates serving as guidelines for collecting the evi-
(2) specific inactivation of the gene(s) associated with dence (Fredericks and Relman, 1996). When reflect-
the suspected virulence trait should lead to a measur- ing on the molecular postulates years later, Stanley
able loss in pathogenicity or virulence, or the gene(s) Falkow emphasized that there is still no microbiolog-
associated with the suspected virulence trait should ical consensus as to what definitively constitutes
be isolated by molecular methods. Specific inactiva- a pathogen (Falkow, 2004). With the advent of data
tion or deletion of the gene(s) should lead to loss of from the human microbiome project, it is becoming
function, (3) reversion or allelic replacement of the obvious that in addition to host and environmental
mutated gene should lead to restoration of pathoge- factors, complex interactions among microbial popu-
nicity, or the replacement of the modified gene(s) for lations may ultimately contribute to complexities of
its allelic counterpart in the strain of origin should chronic disease expression.
lead to loss of function and loss of pathogenicity or In 2002, Jacomo et al. suggested that Kochs obser-
loss of virulence. Restoration of pathogenicity should vation that the blood should be free of bacteria would
accompany the reintroduction of the wild type not be applicable for establishing disease causation for
gene(s). Subsequently, in 1996, Fredericks and Rel- the genus Bartonella (Jacomo et al., 2002). In 2004,
man proposed molecular Kochs postulates based pri- Merrell and Falkow proposed that clinicians, micro-
marily on sequence-based identification of microbial biologists and research scientists divide pathogenic in-
pathogens (Fredericks and Relman, 1996). fectious agents into two major categories: frontal
In conjunction with an enhanced understanding of pathogens and stealth pathogens (Merrell and
the complexity of microbial pathogenesis, particu- Falkow, 2004). Frontal pathogens typically induce
larly as occurs in association with fastidious or uncul- an acute infection, while stealth pathogens are char-
turable organisms, polymicrobial infections and acterized by persistent infections for protracted pe-
chronic or persistent infections, the number of postu- riods of time. Factors that differentiate between
lates continued to increase. Seven postulates were stealth and frontal pathogens are summarized in
Kochs Postulates and Comparative Bartonellosis 117

Table 1. Causation associated with frontal pathogens arbitrary, the strength of this type of association
can in most instances be established using the original would be supported by the fact that random detection
Kochs postulates. In contrast, establishing causation of microbial DNA of a specific organism in a defined
for stealth pathogens, particularly in the context of pathological lesion obtained from different animal
chronic disease causation, is more likely to be success- genera, occurring spontaneously in nature at different
ful when the more recently proposed molecular time points, would have a low probability of being an
Kochs postulates are applied to investigations of dis- unrelated occurrence. We suggest that the new postu-
ease pathogenesis. In addition to considering the at- late be referred to as the postulate of comparative in-
tributes of frontal versus stealth pathogens, this fectious disease causation. Examples involving
important and insightful discussion focused on two Bartonella will be given below to illustrate the poten-
specific stealth pathogens, Helicobacter pylori and Barto- tial application of this suggested addition to Kochs
nella henselae (Merrell and Falkow, 2004). postulates. It is important to acknowledge that some
The aim of the present review is to extend previous stealth pathogens may not induce infection in three
discussions of disease causation induced by stealth or more mammalian genera with the ultimate induc-
bacterial pathogens, using members of the genus Bar- tion of a defined pathological lesion and, as such,
tonella as contemporary microbial examples of bacte- other postulates or other approaches would be neces-
ria that can potentially induce chronic intravascular sary to infer disease causation for those organisms.
infection and complex disease expression. These
Gram-negative alpha proteobacteria cause long last- Bartonella spp. Infection and
ing intra-erythrocytic and endotheliotropic infections
Vasoproliferative Pathology
in reservoir hosts and chronic relapsing bacteraemias
following opportunistic infections of non-reservoir In North America prior to the 1990s, Bartonella spp.
hosts (Chomel et al., 2009a; Breitschwerdt et al., were not known to infect man or animals. If it were
2010a). Importantly, we wish to emphasize the com- not for the acquired immunodeficiency syndrome
parative medical importance and pathogenic rele- (AIDS) epidemic and the recognition of two proto-
vance of combining clinical, microbiological and typical vasoproliferative lesions in this population of
pathological observations across animal genera and immunocompromised patients, it is likely that much
species in order to elucidate the capacity of a specific of the contemporary literature about the genus Barto-
microorganism (or a combination of microorganisms nella, as pathogens of increasing microbiological rele-
in the context of co-infections) to cause similar disease vance in human and veterinary medicine, would not
manifestations or pathology in more than one mam- exist. Early in the 1990s, Bartonella quintana and B. hen-
malian species. Based on recent research observations selae were implicated as the cause of bacillary angio-
derived from studies involving the genus Bartonella, we matosis (BA), a vasoproliferative lesion of the skin,
propose that the following postulate be added to which subsequently was described most often in im-
Kochs postulates in order to assist in the establish- munocompromised patients infected with the human
ment of disease causation by stealth pathogens: causa- immunodeficiency virus (HIV) (Koehler and
tion can be established if the same infectious agent (or Tappero, 1993). Recently, BA was reported for the
combination of agents) is isolated or organism-specific first time in an iatrogenically-immunosuppressed
DNA sequences are amplified from a naturally occur- dog infected with Bartonella vinsonii subsp. berkhoffii
ring pathological entity found in at least three differ- (Yager et al., 2010). In Fig. 1, vasoproliferation asso-
ent mammalian genera. Although the selection of ciated with B. henselae infection in a human patient is
a minimum of three different mammalian genera is compared with a similar histopathological lesion in
a dog, only in association with B. vinsonii subsp. ber-
khoffii infection. This observation was of comparative
Table 1
microbiological relevance in the context of disease
Features that facilitate differentiation of frontal versus
stealth pathogens pathogenesis, as subsequent studies documented the
ability of B. vinsonii subsp. berkhoffii to induce the pro-
Frontal Stealth duction of vascular endothelial growth factor
Incubation Short (hours to days) Long (months to years) (VEGF) (Beerlage et al., 2012), as previously de-
Symptoms Acute Chronic scribed for B. quintana and B. henselae in human pa-
Immunity Sterilizing Non-sterilizing tients. Interestingly, immunosuppression apparently
Transmission Direct Indirect contributed to the subsequent development of BA in
Replication Rapid Slow
Carrier state Uncommon Common
this dog, as has been documented for HIV-infected
BA patients. On a comparative pathological basis,
Adapted from Merrell and Falkow (2004). B. henselae is the only member of the genus that has
118 E.B. Breitschwerdt et al.

___________________________________________
EXAMPLES

Bartonella Vasoproliferation Granulomatous Endocarditis


inflammation

(a) B. henselae (c) Human bacillary (e) Human cat (g) Human
angiomatosis scratch disease endocarditis

(b) B. vinsonii subsp. (d) Canine (f) Canine granulomatous (h) Canine
berkhoffii angiomatosis lymphadenitis endocarditis

Fig. 1. Examples of Bartonella spp.-associated vasoproliferation, granulomatous inflammation and endocarditis. The top images show ex-
amples from human patients associated with B. henselae infection, while the bottom images show similar histopathological lesions in
dogs associated with B. vinsonii subsp. berkhoffii infection. Images are reproduced with permission from (a) Beerlage et al., 2011, (b)
Breitschwerdt et al., 1995, (c) Kempf et al., 2001b, (d) Yager et al., 2010, (e) Kempf et al., 2001a, (f) Pappalardo et al., 2000,
(g) Albrich et al., 2004 and (h) Breitschwerdt et al., 1999.

been associated with peliosis hepatis (PH; a vasoproli- thelioma (EHE) (Emamaullee et al., 2010) in human
ferative lesion in the liver characterized by the pres- patients and haemangiopericytoma in dogs (Hatva
ence of irregular blood-filled spaces within the et al., 1996). In 2009, B. vinsonii subsp. berkhoffii geno-
parenchyma) in a human patient or a dog (Koehler type II was isolated from a boy with EHE and from
et al., 1997; Kitchell et al., 2000). Therefore, DNA of a dog with a haemangiopericytoma (Breitschwerdt
the same Bartonella species has been amplified from et al., 2009). Subsequently, infection with B. henselae
dogs and people, both with clinically similar and his- and Bartonella koehlerae was documented in patients
topathologically confirmed vasoproliferative lesions with EHE from Australia and England (Mascarelli
(PH and BA). et al., 2011) and DNA of B. henselae, B. vinsonii subsp.
Additionally, in the context of Bartonella spp. and berkhoffii or both organisms was amplified and se-
vascular endothelial cell proliferation, the following quenced from haemangiopericytomas obtained from
observations provide additional support for the adop- dogs, a horse and a red wolf (Beerlage et al., 2012).
tion of a postulate of comparative infectious disease Thus, intravascular infection with one or more Barto-
causation. A substantial body of literature now sup- nella spp. has been documented in EHE patients from
ports the ability of B. henselae, B. quintana, Bartonella ba- three continents and in haemangiopericytomas from
cilliformis and B. vinsonii subsp. berkhoffii to induce the three hosts, suggesting that persistent intravascular
production of VEGF and resultant vasoproliferation infection with selected Bartonella spp. might contrib-
of endothelial cells (Kempf et al., 2001b, 2005; ute to the development of these highly unusual and
Resto-Ruiz et al., 2002; Dehio, 2005; Berrich et al., uncommon vascular tumours in immunocompetent
2011; Beerlage et al., 2012). In the context of naturally people and in animals, respectively. On a comparative
occurring cancers, increased expression of VEGF has microbiological and pathological basis, B. henselae,
been documented in association with several vascular B. koehlerae or B. vinsonii subsp. berkhoffii DNA has re-
tumours including haemangiosarcoma in dogs cently been amplified and sequenced (Beerlage et al.,
(Yonemaru et al., 2006), epithelioid haemangioendo- 2012) from formalin-fixed and paraffin wax-
Kochs Postulates and Comparative Bartonellosis 119

embedded tissues from cats (Fuji et al., 2005) and mangiopericytoma and haemangiosarcoma in dogs
a steer (Breshears and Johnson, 2008) with systemic and systemic reactive angiomatosis in cats. If the asso-
reactive angiomatosis. This rare disease is character- ciation between infection with a Bartonella spp. and
ized pathologically by the proliferation of vessels haemangiosarcoma is confirmed as causal, the devel-
throughout numerous tissues within the animals opment of a vaccine to prevent Bartonella spp. infec-
body. tions might decrease or eliminate one of the more
Collectively, the above comparative microbiologi- common and serious malignancies affecting dogs,
cal and pathological findings served as the basis for analogous to the papillomavirus vaccine that was re-
a retrospective study designed to determine if Barto- cently introduced to decrease the prevalence of cervi-
nella spp. might play a role in the pathogenesis of ca- cal cancer in woman (Jenkins et al., 2012). In
nine haemangiosarcoma (a malignant tumour of summary and on a comparative microbiological and
vascular endothelial cells that generally arises in ei- pathological basis, Bartonella spp. may contribute to
ther the spleen or the right atrium of the heart), one the development of naturally occurring vasoprolifera-
of the most common cancers affecting dogs tive tumours in cats, dogs, horses and people, thereby
(Varanat et al., 2011). Using surgically obtained providing molecular support for the proposed Kochs
formalin-fixed and paraffin wax-embedded tissue bi- postulate of comparative infectious disease causation.
opsy samples from 50 dogs with splenic haemangio-
sarcoma, a statistically higher prevalence of Bartonella spp. Infection and Granulomatous
Bartonella spp. DNA was found (26%), compared
Inflammation
with DNA from haemotropic Mycoplasma spp. (2%)
or Babesia spp. (6%) (Varanat et al., 2011). In addi- Historically, granulomatous inflammation (a tissue
tion, Bartonella spp. DNA was statistically more prev- inflammatory response dominated by macrophages
alent in the spleens of dogs with haemangiosarcoma and multinucleated macrophage giant cells) has
(26%), compared with the spleens from dogs with long been associated with lymphadenopathy in pa-
lymphoid nodular hyperplasia (10%). Bartonella tients with classical cat scratch disease (CSD)
spp. DNA was not amplified from the spleens of spe- (Kempf et al., 2001a; Chomel et al., 2009a;
cific pathogen-free research dogs. Although Bartonella Breitschwerdt et al., 2010a). Additionally, granulo-
spp. DNA was amplified and sequenced from only matous hepatitis and/or splenitis (historically referred
26% of the haemangiosarcoma tissues tested, numer- to as atypical CSD) have been reported in a subset of
ous factors could be responsible for the relatively low patients, particularly children, infected with B. hense-
overall percentage of splenic haemangiosarcomas lae (Gerber et al., 2002; Giladi et al., 2005; Psarros
from which organism-specific DNA sequences were et al., 2012; VanderHeyden et al., 2012). Granuloma-
obtained. For example, these tumours are often mas- tous lymphadenitis caused by Bartonella alsatica infec-
sive and for technical reasons only a small quantity of tion has also been reported in patients with
tissue was available for DNA extraction and polymer- generalized lymphadenopathy (Angelakis et al.,
ase chain reaction (PCR) amplification, prolonged 2008). Rabbits are the reservoir hosts for this Barto-
formalin fixation may have compromised PCR am- nella species and infection can seemingly be acquired
plification due to DNA cross-linking and DNA degra- through contact while hunting or butchering wild
dation, organisms may not have been distributed rabbits. As reviewed by Breitschwerdt et al. (2010a),
uniformly throughout the tumour, or infection with granulomatous lymphadenitis, hepatitis, panniculitis,
a Bartonella spp. may have induced a genetic mutation rhinitis, meningitis and encephalitis have been de-
resulting in neoplastic transformation. Alternatively, scribed in dogs infected naturally with B. henselae or
documentation of Bartonella spp. DNA in dogs with B. vinsonii subsp. berkhoffii (Gillespie et al., 2003;
splenic haemangiosarcoma may reflect preferential Mellor et al., 2006; Cross et al., 2008). Moreover, gran-
localization of these intravascular bacteria to a neo- ulomatous osteomyelitis, an atypical manifestation of
plastic spleen because of defective immune surveil- CSD in human patients infected with B. henselae
lance and, therefore, the bacteria did not contribute (Giladi et al., 2005) occurred in a cat infected with
to tumourigenesis.Although causation cannot be es- B. vinsonii subsp. berkhoffii (Varanat et al., 2009). In
tablished by a geographically limited, retrospective Fig. 1, granulomatous lymphadenitis associated
study, the comparative microbiological evidence out- with B. henselae infection in a human patient is com-
lined above implicates a potential pathogenic role for pared with a similar histopathological lesion in
Bartonella spp. through the induction of hypoxia in- a dog, only in association with B. vinsonii subsp. ber-
ducible factor resulting in overproduction of VEGF khoffii infection. Thus, in the context of granuloma-
(Kempf et al., 2005) in immunocompetent animals tous inflammation, the same pathological response
and people, including EHE in human patients, hae- has occurred in cats, dogs and human patients when
120 E.B. Breitschwerdt et al.

infected naturally with a Bartonella spp., thereby sup- a dog, only in association with B. vinsonii subsp. ber-
porting the proposed Kochs postulate of comparative khoffii infection. Thus, in the context of research obser-
infectious disease causation. vations related to endocarditis, cats, cows, dogs and
human patients, when infected with a Bartonella
spp., have developed a very similar life-threatening
Bartonella spp. Infection and Endocarditis
pathology involving infected heart valves. Therefore,
In 1993, for the first time in the medical literature, hu- in support of the proposed postulate of comparative
man endocarditis was associated with different Barto- infectious disease causation, infection with a Bartonella
nella species (B. quintana, B. elizabethae and B. henselae) spp. has induced histologically similar pathology in
in three separate patients (Chomel et al., 2009b). four different mammalian genera.
Shortly thereafter, B. vinsonii subsp. berkhoffii became
the first Bartonella spp. to be isolated from dogs with Bartonella spp. and Chronic Intravascular
endocarditis (Kordick et al., 1996; Breitschwerdt
Infection
et al., 1999). Subsequently, this subspecies was added
to the list of Bartonella species that cause endocarditis Historically and excluding Carrions disease caused
in human patients (Roux et al., 2000). All known Bar- by B. bacilliformis for which chronic bacteraemia has
tonella spp. (currently at least 32 named or candidatus been documented in people for over 100 years, infec-
species) are highly fastidious bacteria, which histori- tions associated with Bartonella spp. have for the most
cally has contributed to microbiological failure to iso- part been relegated to three distinct medical entities:
late them by blood culture from animal or human CSD, BA and endocarditis (Dehio, 2005; Varanat
patients with chronic bacteraemia, endocarditis or et al., 2009; Jenkins et al., 2012). With the exception
other forms of pathology. Following the initial de- of endocarditis, BA and PH in immunocompromised
scriptions of Bartonella spp. endocarditis, comparative patients, Bartonella spp. bacteraemia was historically
infectious disease research over the ensuing two de- considered self-limiting and of brief duration in im-
cades implicated numerous members of this genus as munocompetent patients. Because CSD was consid-
frequent causes of culture-negative endocarditis in ered self-limiting in all patients, antibiotic treatment
both dogs and people (Avidor et al., 2004; Fenollar historically has not been recommended. In contrast,
et al., 2005; Houpikian and Raoult, 2005; Fournier chronic bacteraemia is often documented among the
et al., 2010). As reviewed by Chomel et al. (2009b), numerous clinically normal animal reservoir hosts
five of the nine Bartonella species that have been asso- that are naturally infected by a spectrum of arthropod
ciated with canine endocarditis have also been associ- vectors (Billeter et al., 2008). Persistent infections have
ated with human endocarditis. From a comparative been described previously in domestic and wild ani-
microbiological perspective, many of the same Barto- mal species including, but not limited to, cats, cattle,
nella spp. cause endocarditis in both dogs and in peo- dogs and naturally- and experimentally-infected ro-
ple and from a comparative pathophysiological dents (Kordick and Breitschwerdt, 1997, 1998;
perspective, infection in both mammalian species Cherry et al., 2009; Bai et al., 2011). With the develop-
preferentially involves the aortic valve, for reasons ment of novel insect cell culture-based isolation ap-
that remain unclear (MacDonald et al., 2004; proaches (Duncan et al., 2007; Riess et al., 2008), it
Houpikian and Raoult, 2005; Henn et al., 2009; is increasingly clear that bacteraemia, potentially
Chomel et al., 2009b; Fournier et al., 2010). Although spanning decades in duration, can persist in immuno-
cats are considered the primary reservoir host for B. competent people (Breitschwerdt et al., 2007, 2008,
henselae, this Bartonella species has also been implicated 2010b,c, 2011; Maggi et al., 2011). Among other pub-
as a cause of aortic valve endocarditis in cats (Perez lished examples, documentation of infection with B.
et al., 2010). Similarly, Bartonella bovis, for which cattle henselae and B. vinsonii subsp. berkhoffii in a mother,
are the primary reservoir hosts, has been reported to her 10-year-old son and a twin daughter who died
cause endocarditis in older dairy cattle (Maillard shortly after childbirth supports both persistent infec-
et al., 2007). Pre-existing heart valve pathology, ther- tion in the mother and son and the likelihood of peri-
apeutic suppression of the immune system or immune natal infection of the children (Breitschwerdt et al.,
senescence with ageing appear to be common risk fac- 2010b). Transplacental transmission of Bartonella
tors among animals and people for the development of spp. occurs commonly in naturally- and experimen-
Bartonella endocarditis (Fenollar et al., 2005; tally infected rodents (Bai et al., 2011) and may be
Houpikian and Raoult, 2005; Varanat et al., 2009; a source of transmission for other animals. Therefore,
Henn et al., 2009; Fournier et al., 2010). In Fig. 1, en- on a comparative medical basis, chronic bacteraemia
docarditis associated with B. henselae infection in a hu- appears to be another pathophysiological finding that
man patient is compared with the endocarditis in is associated with disease causation in animals and
Kochs Postulates and Comparative Bartonellosis 121

Table 2 vascular infection in a young dog infected with B. vin-


Comparative pathological and haematological sonii subsp. berkhoffii. As described above, persistent
abnormalities associated with canine and human
bartonellosis
bacteraemia with a spectrum of Bartonella spp. was
a well-documented phenomenon in multiple, natu-
Abnormality Dog Man rally infected animal species before research efforts
PH + +
to document persistent bacteraemia in human pa-
BA + + tients became a consideration.
Endocarditis + +
Myocarditis + +
Granulomatous Concluding Remarks
Lymphadenitis + +
Hepatitis + + It is very clear that the original and the more recently
Panniculitis + + proposed molecular Kochs postulates have been his-
Anterior uveitis + + torically useful, particularly when attempting to con-
Encephalitis + + firm that a disease is caused by a single infectious
Thrombocytopenia + +
agent that induces an acute versus a chronic infection.
Haemolytic anaemia + +
Causation is established more readily when an organ-
Causation has not been clearly established for all of these entities in ei- ism has a brief incubation period, when there is a read-
ther species. Details are provided in Chomel et al., 2009a, b; ily defined source of exposure and when the pathogen
Breitschwerdt et al., 2010a. induces an acute illness (microbiological features of
frontal pathogens). Unfortunately, as was acknowl-
people infected with Bartonella spp. Additionally, in edged by Koch and others, application of the original
the context of disease causation, the symptoms and four postulates has serious limitations when attempt-
pathology reported by chronically bacteraemic im- ing to attribute disease causation to stealth pathogens
munocompetent patients are in many instances simi- that can induce chronic, slowly progressive disease
lar to the clinical signs and pathological lesions found manifestations in an animal or human patient. In ad-
in dogs naturally infected with the same Bartonella spe- dition, Kochs postulates do not allow researchers to
cies (Table 2). Although causation has not been vali- readily address naturally occurring environmental,
dated scientifically for each of these entities (by the nutritional, genetic and other medically relevant fac-
principles of evidence-based medicine), evolving tors that influence disease causation and do not con-
comparative medical research observations increas- sider the pathogenic complexities induced by
ingly support a primary or cofactor role for Bartonella sequential or simultaneous infection with more than
spp. in the pathogenesis of a spectrum of chronic dis- one pathogenic microorganism. As approximately
eases in man and animals (Henn et al., 2005; 70% of emerging infectious diseases are zoonotic in
Goodman and Breitschwerdt, 2005; Breitschwerdt nature (Taylor et al., 2001), understanding the com-
et al., 2007, 2008, 2010a,b,c, 2011; Diniz et al., 2009; parative biological and pathological behaviour of
Maggi et al., 2011, 2012). Table 3 provides evidence a specific infectious agent across different animal
of persistent intravascular infection with B. henselae hosts, particularly human patients, pet cats and
in a human patient with seizures and a prior history dogs that share the same environment, might provide
of CSD. Table 4 provides evidence of persistent intra- useful indicators of chronic disease causation (Day

Table 3
Serological, PCR and culture results for a 23-year-old woman with progressive neurological dysfunction, seizures and
persistent B. henselae infection

Date of sample and IFA titre PCR result after


sample type
B. henselae B. quintana B. vinsonii subsp. Direct extraction BAPGM enrichment Blood agar plate isolate
berkhoffii culture

26/05/2005, blood 256 128 256 B. henselae (H1)* e e


27/06/2005, blood 256 64 128 B. henselae (H1)* B. henselae (H1)* B. henselae (H1)*
20/09/2005, blood 256 128 128 e e e
10/02/2006, CSF NT NT NT e B. henselae (H1)* e
31/08/2006, blood 64 64 64 e e e

Data from Breitschwerdt et al., 2008. CSF, cerebrospinal fluid; IFA, indirect fluorescent antibody; BAPGM, Bartonella alpha proteobacteria growth
medium.
*
16S-23S ITS DNA sequencing results.
122 E.B. Breitschwerdt et al.

Table 4
Sequential lysis centrifugation blood culture and IFA results for a 7-month-old neutered female mixed breed dog with
persistent B. vinsonii subsp. berkhoffii bacteraemia

Date of sample Culture results Colony counts/ml IFA titre

B. henselae B. clarridgeiae B. vinsonii subsp. berkhoffii

19/11/1995 + (Bvb) >1000 128 <16 512


25/02/1996 + (Bvb) 369 64 <16 256
24/03/1996 + (Bvb) 164 32 <16 256
21/04/1996 + (Bvb) 154 16 <16 128
22/06/1996 + (Bvb) 357 16 <16 128
24/08/1996 + (Bvb) 12 <16 <16 64
21/10/1996 e 0 <16 <16 64
18/12/1996 + (Bvb) 6 16 <16 64
30/01/1997* + (Bvb) 1 16 <16 64
21/03/1997 e 0 16 <16 32

Data from Kordick and Breitschwerdt (1998). Bvb, B. vinsonii subsp. berkhoffii; IFA, indirect fluorescent antibody.
*
After collection of blood for culture, the dog received doxycycline hyclate for 28 days.

et al., 2012). This is an important concept with respect infecting infectious agents in patients with chronic,
to the mounting importance of the global One complex disease expression.
Health paradigm that proposes much closer integra-
tion of human and veterinary medicine (Jones et al., Conflict of Interest Statement
2008; People, Pathogens and Our Planet Volume 1,
2010). It is essential that cross-species infectious In conjunction with Dr. S. Sontakke and North Car-
agents are investigated in a collaborative fashion by olina State University, Dr. Breitschwerdt holds US
integrated teams of medical, veterinary medical and Patent No. 7,115,385; Media and Methods for culti-
other public health officials and that appropriate re- vation of microorganisms, which was issued October
sources are allocated to permit this approach to be 3, 2006. He is the chief scientific officer for Galaxy Di-
successful. Although the present review is focused on agnostics, a newly formed company that provides di-
organisms of the genus Bartonella, there are numerous agnostic testing for the detection of Bartonella spp.
other frontal pathogens (e.g. influenza virus, infection in animals and in human patient samples.
methicillin-resistant Staphylococcus aureus, SARS coro- Dr. R. Maggi has led efforts to optimize the BAPGM
navirus and monkey pox) and stealth pathogens platform and is the Scientific Technical Adviser and
(e.g. Anaplasma phagocytophilum, Borrelia burgdorferi, Laboratory Director for Galaxy Diagnostics. All
Coxiella burnetii, Ehrlichia spp. and Toxoplasma gondii), other authors report no conflicts of interest.
shared between man and small companion animals,
for which a co-ordinated One Health approach to in- References
vestigation and application of our proposed Kochs
Albrich WC, Kraft C, Fisk T, Albrecht H (2004) A me-
postulate of comparative disease causation would be chanic with a bad valve: blood-culture-negative endo-
of benefit to animal and human health. carditis. Lancet Infectious Diseases, 4, 777e784.
In conclusion, we propose the addition of a fifth Angelakis E, Lepidi H, Canel A, Rispal P, Perraudeau F
Kochs postulate to address comparative disease cau- et al. (2008) Human case of Bartonella alsatica lymphade-
sation where the same infectious agent is isolated or nitis. Emerging Infectious Diseases, 14, 1951e1953.
DNA sequences are found in at least three mamma- Avidor B, Graidy M, Efrat G, Leibowitz C, Shapira G et al.
lian genera. In the examples provided in this manu- (2004) Bartonella koehlerae, a new cat-associated agent of
script, this postulate has been achieved for Bartonella culture-negative human endocarditis. Journal of Clinical
spp. and vasoproliferative lesions in cats, dogs, a horse, Microbiology, 42, 3462e3468.
people and a steer, granulomatous inflammatory le- Bai Y, Calisher CH, Kosoy MY, Root JJ, Doty JB (2011)
Persistent infection or successive reinfection of deer
sions in cats, dogs and human patients, endocarditis
mice with Bartonella vinsonii subsp. arupensis. Applied and
in cats, cows, dogs and human patients and chronic Environmental Microbiology, 77, 1728e1731.
bacteraemia in cats, dogs, people and numerous ro- Beerlage C, ORourke F, Kempf VAJ (2011) Bartonella hen-
dent species. The authors hope that the postulate of selae verbindet Infektionsforschung mit Blut-
comparative infectious disease causation can be gefawachstum. Forschung Frankfurt, 2, 26e29.
used by other infectious disease researchers to assist Beerlage C, Varanat M, Linder K, Maggi RG, Colley J
with efforts to define the role of specific or co- et al. (2012) Bartonella vinsonii subsp. berkhofffii and
Kochs Postulates and Comparative Bartonellosis 123

Bartonella henselae as potential causes of proliferative vas- henselae in the blood of beef cattle. Veterinary Microbiology,
cular disease in animals. Medical Microbiology and Immu- 135, 308e312.
nology, 201, 319e326. Chomel BB, Boulouis HJ, Breitschwerdt EB, Kasten RW,
Berrich M, Kieda C, Grillon C, Monteil M, Lamerant N Vayssier-Taussat M et al. (2009a) Ecological fitness
et al. (2011) Differential effects of Bartonella henselae on and strategies of adaptation of Bartonella species to their
human and feline macro- and micro-vascular endothe- hosts and vectors. Veterinary Research, 40, 29.
lial cells. PLoS One, 6, e20204. Chomel BB, Kasten RW, Williams C, Wey AC, Henn JB
Billeter SA, Levy MG, Chomel BB, Breitschwerdt EB et al. (2009b) Bartonella endocarditis: a pathology shared
(2008) Vector transmission of Bartonella species with em- by animal reservoirs and patients. Annals of the New York
phasis on the potential for tick transmission. Medical and Academy of Sciences, 1166, 120e126.
Veterinary Entomology, 22, 1e15. Cross JR, Rossmeisl JH, Maggi RG, Breitschwerdt EB,
Breitschwerdt EB, Atkins CE, Brown TT, Kordick DL, Duncan RB (2008) Bartonella-associated meningoradi-
Snyder PS (1999) Bartonella vinsonii subsp. berkhoffii and culoneuritis and dermatitis or panniculitis in 3 dogs.
related members of the alpha subdivision of the Proteo- Journal of Veterinary Internal Medicine, 22, 674e678.
bacteria in dogs with cardiac arrhythmias, endocarditis, Day MJ, Breitschwerdt EB, Cleaveland S, Karkare U,
or myocarditis. Journal of Clinical Microbiology, 37, Khanna C et al. (2012) Surveillance of zoonotic infec-
3618e3626. tious disease transmitted by small companion animals.
Breitschwerdt EB, Kordick DL, Malarkey DE, Keene B, Emerging Infectious Diseases, http://dx.doi.org/10.3201/
Hadfield TL et al. (1995) Endocarditis in a dog due to eid1812.120664. 2012 Dec [date cited].
infection with a novel Bartonella subspecies. Journal of Dehio C (2005) Bartonella e host cell interactions and vascular
Clinical Microbiology, 33, 154e160. tumor formation. Nature Reviews Microbiology, 3, 621e631.
Breitschwerdt EB, Maggi RG, Chomel BB, Lappin MR Diniz PP, Wood M, Maggi RG, Sontakke S, Stepnik M
(2010a) Bartonellosis: an emerging infectious disease of et al. (2009) Co-isolation of Bartonella henselae and Barto-
zoonotic importance to animals and human beings. nella vinsonii subsp. berkhoffii from blood, joint and subcu-
Journal of Veterinary Emergency and Critical Care, 20, 8e30. taneous seroma fluids from two naturally infected dogs.
Breitschwerdt EB, Maggi RG, Duncan AW, Nicholson WL, Veterinary Microbiology, 138, 368e372.
Hegarty BC et al. (2007) Bartonella species in blood of im- Duncan AW, Maggi RG, Breitschwerdt EB (2007) A com-
munocompetent persons with animal and arthropod bined approach for the enhanced detection and isolation
contact. Emerging Infectious Diseases, 13, 938e941. of Bartonella species in dog blood samples: pre-
Breitschwerdt EB, Maggi RG, Farmer P, Mascarelli PE enrichment liquid culture followed by PCR and subcul-
(2010b) Molecular evidence of perinatal transmission ture onto agar plates. Journal of Microbiological Methods,
of Bartonella vinsonii subsp. berkhoffii and Bartonella henselae 69, 273e281.
to a child. Journal of Clinical Microbiology, 48, Emamaullee JA, Edgar R, Toso C, Thiesen A, Bain V et al.
2289e2293. (2010) Vascular endothelial growth factor expression in
Breitschwerdt EB, Maggi RG, Lantos PM, Woods CW, hepatic epithelioid hemangioendothelioma: implica-
Hegarty BC et al. (2010c) Bartonella vinsonii subsp. ber- tions for treatment and surgical management. Liver
khoffii and Bartonella henselae bacteremia in a father and Transplantation, 16, 191e197.
daughter with neurological disease. Parasites and Vectors, Falkow S (1988) Molecular Kochs postulates applied to
3, 29e37. microbial pathogenicity. Reviews of Infectious Diseases,
Breitschwerdt EB, Maggi RG, Nicholson WL, Cherry NA, 10(Suppl. 2), S274eS276.
Woods CW (2008) Bartonella sp. bacteremia in patients Falkow S (2004) Molecular Kochs postulates applied to
with neurological and neurocognitive dysfunction. Jour- bacterial pathogenicity e a personal recollection 15
nal of Clinical Microbiology, 46, 2856e2861. years later. Nature Reviews Microbiology, 2, 67e72.
Breitschwerdt EB, Maggi RG, Varanat M, Linder KE, Fenollar F, Sire S, Raoult D (2005) Bartonella vinsonii subsp.
Weinberg G (2009) Isolation of Bartonella vinsonii subsp. arupensis as an agent of blood culture-negative endocarditis
berkhoffii genotype II from a boy with epithelioid heman- in a human. Journal of Clinical Microbiology, 43, 945e947.
gioendothelioma and a dog with hemangiopericytoma. Fournier PE, Thuny F, Richet H, Lepidi H, Casalta JP
Journal of Clinical Microbiology, 47, 1957e1960. et al. (2010) Comprehensive diagnostic strategy for
Breitschwerdt EB, Mascarelli PE, Schweickert LA, blood culture-negative endocarditis: a prospective study
Maggi RG, Hegarty BC et al. (2011) Hallucinations, of 819 new cases. Clinical Infectious Diseases, 51, 131e140.
sensory neuropathy, and peripheral visual deficits in Fredericks DN, Relman DA (1996) Sequence-based identi-
a young woman infected with Bartonella koehlerae. Journal fication of microbial pathogens: a reconsideration of
of Clinical Microbiology, 49, 3415e3417. Kochs postulates. Clinical Microbiology Reviews, 9, 18e33.
Breshears MA, Johnson BJ (2008) Systemic reactive Fuji RN, Patton KM, Steinbach TJ, Schulman FY,
angioendotheliomatosis-like syndrome in a steer pre- Bradley GA et al. (2005) Feline systemic reactive an-
sumed to be persistently infected with bovine viral diar- gioendotheliomatosis: eight cases and literature review.
rhea virus. Veterinary Pathology, 45, 645e649. Veterinary Pathology, 42, 608e617.
Cherry NA, Maggi RG, Cannedy AL, Breitschwerdt EB Gerber JE, Johnson JE, Scott MA, Madhusudhan KT
(2009) PCR detection of Bartonella bovis and Bartonella (2002) Fatal meningitis and encephalitis due to
124 E.B. Breitschwerdt et al.

Bartonella henselae bacteria. Journal of Forensic Sciences, 47, Koehler JE, Sanchez MA, Garrido CS, Whitfeld MJ,
640e664. Chen FM et al. (1997) Molecular epidemiology of Barto-
Giladi M, Maman E, Paran D, Bickels J, Comaneshter D nella infections in patients with bacillary angiomatosis-
et al. (2005) Cat-scratch disease-associated arthropathy. peliosis. New England Journal of Medicine, 337, 1876e1883.
Arthritis and Rheumatism, 52, 3611e3617. Koehler JE, Tappero JW (1993) Bacillary angiomatosis
Gillespie TN, Washabau RJ, Goldschmidt MH, and bacillary peliosis in patients infected with human
Cullen JM, Rogala AR et al. (2003) Detection of Barto- immunodeficiency virus. Clinical Infectious Diseases, 17,
nella henselae and Bartonella clarridgeiae DNA in hepatic 612e624.
specimens from two dogs with hepatic disease. Journal Kordick DL, Breitschwerdt EB (1997) Relapsing bacteremia
of the American Veterinary Medical Association, 222, 47e51. following blood transmission of Bartonella henselae in cats.
Goodman RA, Breitschwerdt EB (2005) Clinicopathologic American Journal of Veterinary Research, 58, 492e497.
findings in dogs seroreactive to Bartonella henselae anti- Kordick DL, Breitschwerdt EB (1998) Persistent infection
gens. American Journal of Veterinary Research, 66, of pets within a household with three Bartonella species.
2060e2064. Emerging Infectious Diseases, 4, 325e328.
Hatva E, Bohling T, Jaaskelainen J, Persico MG, Haltia M Kordick DL, Swaminathan B, Greene CE, Wilson KH,
et al. (1996) Vascular growth factors and receptors in Whitney AM et al. (1996) Bartonella vinsonii subsp. berkhof-
capillary hemangioblastomas and hemangiopericyto- fii subsp. nov., isolated from dogs; Bartonella vinsonii subsp.
mas. American Journal of Pathology, 148, 763e775. vinsonii; and amended description of Bartonella vinsonii. In-
Henn JB, Gabriel MW, Kasten RW, Brown RN, ternational Journal of Systemic Bacteriology, 46, 704e709.
Koehler JE et al. (2009) Infective endocarditis in a dog Loeffler F (1884) Mitt. Aus dem Kaiserl. Gesundheitsamte, 2,
and the phylogenetic relationship of the associated Bar- 421e499.
tonella rochalimae strain with isolates from dogs, gray MacDonald KA, Chomel BB, Kittleson MD, Kasten RW,
foxes, and a human. Journal of Clinical Microbiology, 47, Thomas WP et al. (2004) A prospective study of canine
787e790. infective endocarditis in northern California (1999-
Henn JB, Liu CH, Kasten RW, VanHorn BA, Beckett LA 2001): emergence of Bartonella as a prevalent etiologic
et al. (2005) Seroprevalence of antibodies against Barto- agent. Journal of Veterinary Internal Medicine, 18, 56e64.
nella species and evaluation of risk factors and clinical Maggi RG, Mascarelli PE, Pultorak EL, Hegarty BC,
signs associated with seropositivity in dogs. American Bradley JM et al. (2011) Bartonella spp. bacteremia in
Journal of Veterinary Research, 66, 688e694. high-risk immunocompetent patients. Diagnostic Micro-
Houpikian P, Raoult D (2005) Blood culture-negative en- biology and Infectious Disease, 71, 430e437.
docarditis in a reference center: etiologic diagnosis of Maggi RG, Mozayeni RB, Hegarty BC, Bradley JM,
348 cases. Medicine (Baltimore), 84, 162e173. Breitschwerdt EB (2012) Bartonella spp. bacteremia in
Jacomo V, Kelly PJ, Raoult D (2002) Natural history of rheumatic patients from Lyme disease-endemic region.
Bartonella infections (an exception to Kochs postulate). Emerging Infectious Diseases, 18, 783e791.
Clinical and Diagnostic Laboratory Immunology, 9, 8e18. Maillard R, Petit E, Chomel B, Lacroux C, Schelcher F
Jenkins M, Chiriva-Internati M, Mirandola L, Tonroy C, (2007) Endocarditis in cattle caused by Bartonella bovis.
Tedjarati SS et al. (2012) Perspective for prophylaxis Emerging Infectious Diseases, 13, 1383e1385.
and treatment of cervical cancer: an immunological ap- Mascarelli PE, Iredell JR, Maggi RG, Weinberg G,
proach. International Reviews of Immunology, 31, 3e21. Breitschwerdt EB (2011) Bartonella species bacteremia
Jones KE, Patel NG, Levy MA, Storeygard A, Balk D et al. in two patients with epithelioid hemangioendothelioma.
(2008) Global trends in emerging infectious diseases. Journal of Clinical Microbiology, 49, 4006e4012.
Nature, 451, 990e994. Mellor PJ, Fetz K, Maggi RG, Haugland S, Dunning M
Kempf VA, Lebiedziejewski M, Alitalo K, Walzlein JH, et al. (2006) Alpha1-proteinase inhibitor deficiency
Ehehalt U et al. (2005) Activation of hypoxia- and Bartonella infection in association with panniculitis,
inducible factor-1 in bacillary angiomatosis: evidence polyarthritis, and meningitis in a dog. Journal of Veteri-
for a role of hypoxia-inducible factor-1 in bacterial infec- nary Internal Medicine, 20, 1023e1028.
tions. Circulation, 111, 1054e1062. Merrell DS, Falkow S (2004) Frontal and stealth attack strat-
Kempf VA, Petzold H, Autenrieth IB (2001a) Cat scratch egies in microbial pathogenesis. Nature, 430, 250e256.
disease due to Bartonella henselae infection mimicking pa- Pappalardo BL, Brown T, Gookin JL, Morrill CL,
rotid malignancy. European Journal of Clinical Microbiology Breitschwerdt EB (2000) Granulomatous disease associ-
and Infectious Diseases, 20, 732e733. ated with Bartonella infection in 2 dogs. Journal of Veteri-
Kempf VA, Volkmann B, Schaller M, Sander CA, nary Internal Medicine, 14, 37e42.
Alitalo K et al. (2001b) Evidence of a leading role for People, Pathogens and Our Planet. Vol. 1. (2010) Towards
VEGF in Bartonella henselae-induced endothelial cell pro- a One Health Approach to Controlling Zoonotic Disease. World
liferations. Cellular Microbiology, 3, 623e632. Bank Report 50833-GLB. International Bank for Re-
Kitchell BE, Fan TM, Kordick D, Breitschwerdt EB, construction and Development/World Bank, 1818 H
Wollenberg G et al. (2000) Peliosis hepatis in a dog in- Street NW, Washington DC. p. 20433.
fected with Bartonella henselae. Journal of the American Vet- Perez C, Hummel JB, Keene BW, Maggi RG, Diniz PP
erinary Medical Association, 216, 519e523. et al. (2010) Successful treatment of Bartonella henselae
Kochs Postulates and Comparative Bartonellosis 125

endocarditis in a cat. Journal of Feline Medicine and Sur- VanderHeyden TR, Yong SL, Breitschwerdt EB,
gery, 12, 483e486. Maggi RG, Mihalik AR et al. (2012) Granulomatous
Psarros G, Riddell J 4th, Gandhi T, Kauffman CA, hepatitis due to Bartonella henselae infection in an immu-
Cinti SK (2012) Bartonella henselae infections in solid or- nocompetent patient. BMC Infectious Diseases, 12, 17.
gan transplant recipients: report of 5 cases and review of Varanat M, Maggi RG, Linder KE, Breitschwerdt EB
the literature. Medicine (Baltimore), 91, 111e121. (2011) Molecular prevalence of Bartonella, Babesia and
Resto-Ruiz SI, Schmiederer M, Sweger D, Newton C, hemotropic Mycoplasma sp. in dogs with splenic disease.
Klein TW et al. (2002) Induction of a potential para- Journal of Veterinary Internal Medicine, 25, 1284e1291.
crine angiogenic loop between human THP-1 macro- Varanat M, Travis A, Lee W, Maggi RG, Bissett SA et al.
phages and human microvascular endothelial cells (2009) Recurrent osteomyelitis in a cat due to infection
during Bartonella henselae infection. Infection and Immunity, with Bartonella vinsonii subsp. berkhoffii genotype II. Jour-
70, 4564e4570. nal of Veterinary Internal Medicine, 23, 1273e1277.
Riess T, Dietrich F, Schmidt KV, Kaiser PO, Schwarz H Yager JA, Best SJ, Maggi RG, Varanat M, Znajda N et al.
et al. (2008) Analysis of a novel insect cell culture (2010) Bacillary angiomatosis in an immunosuppressed
medium-based growth medium for Bartonella species. dog. Veterinary Dermatology, 21, 420e428.
Applied and Environmental Microbiology, 74, 5224e5227. Yonemaru K, Sakai H, Murakami M, Yanai T, Masegi T
Roux V, Eykyn SJ, Wyllie S, Raoult D (2000) Bartonella vin- (2006) Expression of vascular endothelial growth factor,
sonii subsp. berkhoffii as an agent of afebrile blood culture- basic fibroblast growth factor, and their receptors (flt-1,
negative endocarditis in a human. Journal of Clinical Mi- flk-1, and flg-1) in canine vascular tumors. Veterinary Pa-
crobiology, 38, 1698e1700. thology, 43, 971e980.
Tabrah FL (2011) Kochs postulates, carnivorous cows,
and tuberculosis today. Hawaii Medical Journal, 70,
144e148.
Taylor LH, Latham SM, Woolhouse ME (2001) Risk fac-
tors for human disease emergence. Philosophical Transac-
tions of the Royal Society of London, 356, 983e989. Received,
Accepted, January 5th, 2013 
September 27th, 2012

You might also like