GUIDELINE for Feline bartonellosis

Published: 01/01/2013
Last updated: [acf field='last_updated']
Last reviewed: [acf field='last_reviewed']

The Bartonella species infection in cats guidelines were first published in the J Feline Med Surg 2013, 15: 563-569 by Maria Grazia Pennisi et al. This update was authored by Maria Grazia Pennisi and Fulvio Marsilio.

Key points

  • Bartonella infect humans and domestic animals and some species and subspecies are confirmed or suspected pathogens.
  • Bartonella have a worldwide distribution with higher prevalences in areas where most favorable conditions for arthropod vectors (particularly fleas) exist.
  • Bartonella (B.) henselae is the causative agent of cat scratch disease (CSD) in humans, a self-limiting regional lymphadenopathy.
  • Cats are main reservoir hosts of B. henselae and accidental hosts of other species.
  • The primary role of fleas in the transmission of henselae among cats has been demonstrated.
  • Most cats naturally infected by henselae do not show clinical signs, but some individuals may develop life-threatening cardiovascular diseases and possibly other pathologies associated with generalized lymphadenopathy.
  • Other Bartonella may have pathogenic properties in cats as seen in dogs and humans.
  • Antibodies are not protective and antibody-positive cats may be re-infected.
  • Bartonellosis is diagnosed in symptomatic Bartonella positive cats based on exclusion of other compatible diagnoses, and by assessing the response to antibiotic therapy.
  • No benefit derives from testing healthy cats and humans, except in cases of immunosuppressed people in the home.
  • Strict flea and tick control is the only effective preventive measure.

Agent properties

Bartonella are small (2.0 by 0.5 μm), vector-transmitted Gram-negative intracellular bacteria that are well adapted to one or more mammalian reservoir hosts. Until now, over 22 Bartonella species have been described, but their role as pathogens of humans and domestic animals is the subject of ongoing investigations (Table 1).

Table 1: Species and subspecies of Bartonella that are confirmed or potential human pathogens (Chomel et al., 2006; Molia et al., 2016)

Bartonella spp. Primary reservoir Vector Accidental host
B. bacilliformis Humans Lutzomia verrucarum None
B. quintana Humans Pediculus humanus Cat, dog, monkey
B. elizabethae Rattus norvegicus Xenopsylla cheopis Human, dog
B. grahamii Several species of wild mice Rodent fleas Humans
B. henselae Cat Ctenocephalides felis felis Humans, dog
B. clarridgeiae Cat C. felis Humans, dog
B. koehlerae subsp. koehlertae Cat, lion C. felis Humans
B. koehlerae subsp. boulouisii Mountain lion Fleas? Unknown
B. koehlerae subsp. bothieri Bobcat, cheetah Fleas? Unknown
B. vinsonii subsp. berkhoffii Coyote, dog Ticks? Humans
B. vinsonii subsp. arupensis Peromyscus leucopus Ticks? Fleas? Humans
B. washoensis Spermophilus beecheyii Fleas? Humans, dog
B. asiatica Rabbit Fleas? Humans
B. rochalimae Wild carnivores Fleas Humans

The most common species in both cats and humans is B. henselae, which causes cat scratch disease (CSD) in the latter, as well as other potentially fatal disorders affecting immunocompromised people.

Cats naturally infected with Bartonella usually do not show clinical signs. Given the long-lasting association of B. henselae and domestic cats; there have been adaptations between host and bacterium to facilitate co-existence and minimise pathogenic effects on the mammalian host (Guptill, 2010).

Epidemiology

Prevalence

Bartonella spp. have a worldwide distribution with highest prevalences in areas where conditions are most favourable for arthropod vectors, mainly fleas. In Europe, many studies have been carried out, and the antibody prevalence in cats ranged from 3.6 to 53% (Table 2).

Table 2: Antibody prevalence of Bartonella infection in the feline populations sampled in European countries

Country Number of cats Prevalence (%) Reference
The Netherlands 163 (stray) 52 Bergmans et al., 1997
Austria 96 33 Allerberger et al., 1995
Switzerland 728 8 Glaus et al., 1997
Germany 713 15 Haimerl et al., 1999
245 37.1 Morgenthal et al., 2012
France 64 36 Chomel et al., 1995
94 53 Heller et al., 1997
179 41 Gurfield et al., 2001
Spain 680 23.8 Ayllon et al., 2012
118 78 Álvarez-Fernández et al., 2021
Italy 540 38 Fabbi et al., 2004
55 3.6 Zobba et al., 2009
1300 (stray) 23.1 Brunetti et al., 2013
197 45.7 Persichetti et al., 2018
167 18.0 Morelli et al., 2019
95 29.47 Ebani et al., 2021
Scotland 78 15.3 Bennett et al., 2011
Greece 452 35.4 Kokkinaki et al., 2022

Transmission

Epidemiological evidence and experimental studies have demonstrated the important role of fleas in the transmission of B. henselae and B. clarridgeiae between cats. B. henselae is naturally transmitted among cats by the flea Ctenocephalides felis felis, or by flea faeces.

Bartonella henselae was experimentally transmitted among cats by transferring fleas fed on naturally infected cats to SPF cats, and by intradermal inoculation of excrement collected from fleas fed on B. henselae-infected cats (Chomel et al., 1996). This has demonstrated that both the vector and the cat – through scratches – may transmit the organism. Infection is amplified in the flea hindgut, and B. henselae can persist in the environment in flea faeces for at least nine days (Finkelstein et al., 2002). Using a quantitative real-time PCR, B. henselae DNA was detected in both fleas and their faeces for the entire 12-day life span of the arthropod, starting at 24 hours after the blood meal (Bouhsira et al., 2013).

Bartonella henselae transmission did not occur when infected cats lived together with uninfected cats in a flea-free environment. Transmission consequently does not occur through bites, scratches in the absence of fleas, grooming, or sharing of litter boxes and food dishes. Furthermore, transmission could not be demonstrated between bacteraemic female cats and uninfected males during mating, or to the kittens of infected females either during gestation or in the neonatal period, again in flea-free environments (Guptill et al., 1997).

Blood transfusion represents a risk: cats have been experimentally infected with B. henselae and B. clarridgeiae by intravenous or intramuscular inoculation with infected cat blood (Abbott et al., 1997).

Muz et al. (2021) found during a study on the prevalence of protozoan and microbial pathogens in owned sick cats presented to veterinary clinics in Turkey, B. henselae DNA in 40.1% of 167 blood samples. Furthermore, the authors did not find differences between cats that were less and more than one year of age.

Razgūnaitė et al. (2021) reported Bartonella spp. DNA in 4.9% of Lithuanian cat blood samples (8/163) and from 29.4% (30/102) of fleas collected from the same cats. B. henselae and B. clarridgeiae, were identified in the cats and fleas, with B. henselae to be found more common than B. clarridgeiae.

Three other species, B. koehlaraeB. bovis and B. quintana have been isolated from cat blood, but the modes of transmission and the reservoir potential of these species in felids have not been established. In addition, B. vinsonii subsp. berkhoffii DNA was detected in the blood of a cat (Varanat et al., 2009).

The possible role of several bat fly species (Nycteribiidae) as Bartonella vectors has been studied. It remains a subject of debate, but a reservoir function should be considered in addition to pathogenic, parasitic, or mutualistic interactions (Morse et al., 2012).

Ticks may also act as vectors for transmission among cats, human beings, dogs, and other mammalian hosts: transstadial transmission of B. henselae was demonstrated in Ixodes ricinus (Cottè et al., 2008).

Persichetti et al. (2016) evaluated PCR positivity to vector-borne pathogens in cats and their ectoparasites and they sequenced B. clarridgeiae in PCR products amplified from DNA extracted from two ticks (one Ixodes ventalloi and one Rhipicephalus sanguineus specimen) but not in the blood of the two cats carrying each of them. Similarly, Regier et al. (2017) found B. henselae in Ixodes ricinus ticks collected from one PCR negative cat that was however positive for anti-B. henselae antibodies. Whether this observation implies a role for ticks in the transmission of both Bartonella spp. to cats needs to be resolved.

Mazurek et al. (2019) reported the frequency of the occurrence of Bartonella sppDNA in dogs from households where cats with clinical bartonellosis were kept. The presence of DNA with 99–100% compliance of the nucleotide sequence with the sequence of the Bartonella DNA isolated from cats was demonstrated in the body of 10% of tested dogs. The results indicated that cats serve as a Bartonella reservoir for dogs, and the dogs can play the same role with regard to humans. Furthermore, Mazurek et al. (2020) reported the occurrence of Bartonella spp. in blood by PCR on 672 cats randomly selected from the largest clinics in eastern Poland and a prevalence of 40.48% was found. Interestingly, only the B. henselae DNA was detected.

Pathogenesis

In the infected cat, Bartonella inhabits red blood cells, which are ingested by the flea and survive in its gut. Contaminated flea faeces deposited on the skin end up under the cat’s claws due to grooming. A cat scratch is the common mode of transmission of the organism to other animals, including humans (Chomel et al., 1996).

Chronic bacteraemia mainly occurs in cats under the age of 2 years (Guptill et al., 2004). Young experimentally infected cats maintained relapsing B. henselae or B. clarridgeiae bacteraemia for as long as 454 days (Kordick et al., 1999). Immune system avoidance due to its intracellular location, frequent genetic rearrangements and alteration of outer membrane proteins are considered important for the maintenance of persistent bacteraemia. The location within erythrocytes and vascular endothelial cells is believed to protect Bartonellae from antimicrobial agents. Cats can be re-infected by different strains of Bartonella (Guptill, 2010).

Immunity

Passive immunity

Data concerning passive immunity are lacking.

Active immunity

The antibody response to B. henselae has been investigated for the identification of vaccine candidates. The kinetics in response to B. henselae antigens in chronically infected experimental cats is highly variable in degree and duration (Chomel et al., 1996; Kordick et al., 1999; Yamamoto et al., 2002). The extent of serologic cross-reactivity to other Bartonella species needs to be clarified. Re-infection by a different strain of B. henselae is possible, as supported by the isolation of unrelated bacterial clones from the same cat at different times (Arvand et al., 2008). Antibodies are therefore considered not protective, and Bartonella spp.-seropositive cats may be infected (Fabbi et al., 2004).

Clinical signs

Cats naturally infected with Bartonella spp. usually do not show clinical signs. Both experimental and natural infection studies have tried to establish an association between clinical signs and infection, but a link has not been unequivocally proven. Exposure to infected fleas does not result in clinical signs (Chomel et al., 1996; Bradbury & Lappin, 2009). In some cases of experimental inoculation, a self-limiting febrile disease, transient mild anaemia, localized or generalized lymphadenopathy, mild neurologic signs and reproductive failure have been reported (Kordick et al., 1999). In these animals, pyogranulomatous inflammation was seen in the lung, liver, spleen, kidney, heart (Figure 1) and lymphoid tissue at necropsy (Guptill et al., 1997; Kordick et al., 1999).

Fig 1. Gross and histological findings in two cats from North Carolina shelter that had died after a litter of flea-infested kittens was introduced to the shelter. (a) Coalescent granulomas distributed throughout the myocardium (b) Pyogranulomatous myocarditis in an 8-month-old castrated male cat, which had been co-housed with the flea-infested kittens. Macrophages, with a rare multinucleated giant cell (arrow) are particularly numerous at the upper left of the image. Hematoxylin/eosin stain. Inset: cluster of short bacilli in an inflammatory focus are immunoreactive (brown) for B. henselae-specific monoclonal antibody. Reproduced with permission from Varanat et al., 2012

Fig. 1. Gross and histological findings in two cats from North Carolina shelter that had died after a litter of flea-infested kittens was introduced to the shelter. (a) Coalescent granulomas distributed throughout the myocardium (b) Pyogranulomatous myocarditis in an 8-month-old castrated male cat, which had been co-housed with the flea-infested kittens. Macrophages, with a rare multinucleated giant cell (arrow) are particularly numerous at the upper left of the image. Hematoxylin/eosin stain. Inset: cluster of short bacilli in an inflammatory focus are immunoreactive (brown) for B. henselae-specific monoclonal antibody. Reproduced with permission from Varanat et al., 2012.

The role of Bartonella in causing clinical signs in cats after natural infection is even more unclear. Studies based on antibody detection are of limited value because antibody only proves exposure, but not necessarily active infection. Moreover, there is cross reactivity between different Bartonella species that may or may not cause clinical signs. Because of the high percentage of infected healthy cats in endemic areas, an association between clinical signs and B. henselae infection is not easy to demonstrate.

It has been suggested that Bartonella infection could play a role in chronic gingivostomatitis, (Ueno et al., 1996; Glaus et al., 1997), but the prevalence of antibodies or organisms was not higher in diseased cats than in control populations (Quimby et al., 2008; Dowers et al., 2010; Pennisi et al., 2010; Belgard et al., 2010; Namekata et al., 2010).

Cats positive for both FIV and Bartonella antibodies had an increased risk of lymphadenopathy (Ueno et al., 1996). An association between anti-Bartonella antibodies and urinary tract disease or haematuria has been suggested (Glaus et al., 1997; Breitschwerdt et al., 2005). Pearce et al. (2006) did not find any difference in antibody prevalence between healthy cats and cats with seizures or other neurological conditions. However, a non-controlled retrospective study reported Bartonella DNA in cerebrospinal fluid and confirmed specific antibody production in the CNS of cats with CNS disease (Leibovitz et al., 2008). Despite the EBM grade III of this observation, Bartonella spp. infection should be considered when other compatible diagnoses of CNS disease are excluded in symptomatic cats.

Castel et al. (2019) reported co-infection with B. henselae and Sarcocystis sp. in a 6-year-old male neutered domestic longhair cat with chronic waxing and waning neurologic mild signs and presenting an acute progressive multifocal neurolomuscular syndrome and chorioretinitis. Determining the contribution of each agent and the influence of the co-infection on the pathogenesis of the neuromuscular and ocular lesions was not possible and treatment with fluoroquinolones, trimethoprim sulfadiazine and pyrimethamine was directed at both organisms.  After an initial worsening of clinical signs, a progressive improvement followed by full recovery was observed. As Sarcocystis sp. clinical infection in cats has been associated with immunosuppression, the authors hypothetized that relapsing B. henselae bacteraemia may have decreased the cat’s immune competence and caused a normally quiescent protozoal infection to induce intermittent clinical signs (Castel et al., 2019).

No difference in Bartonella antibody prevalence was found between healthy cats and cats affected by uveitis (Fontenelle et al., 2008), but some had reported evidence of Bartonella spp. exposure in cats with uveitis responsive to drugs considered effective against Bartonella (Lappin and Black, 1999; Ketring et al., 2004). No difference in Bartonella PCR prevalence was found in cats affected by anaemia compared to control cats (Ishak et al., 2007). Prevalence of anti-Bartonella antibodies was lower in cats with fever compared to afebrile controls, but the former had a higher blood DNA positivity approaching statistical significance (Lappin et al., 2009).  Moreover, a unique, identical B. henselae genotype was cultured from blood of three kittens and it was recognised as the causative agent of their cyclic relapsing fever associated with anaemia and neutropenia by excluding other infections and clinical cure with azythromycin treatment (Breitschwerdt et al., 2015).

A study based on serology and culture did not find an association between Bartonella infection and chronic rhinosinusitis (Berryessa et al., 2008). There was also no association found between Bartonella infection and pancreatitis, because cats with normal fPLI values and cats with elevated fPLI values did not show any difference in Bartonella prevalence (Bayliss et al., 2009).

Since 1993, many Bartonella species have been associated with endocarditis in humans and dogs (Breitschwerdt et al., 1995; La Scola and Raoult, 1999; Mc Donald et al., 2004). Some research groups have looked for Bartonella in cats with endocarditis, an uncommon problem in cats. Aortic and fatal mitral valve B. henselae-associated endocarditis was reported in two cats in the USA (Chomel et al., 2003, 2009). Also, B. henselae anterior mitral valve leaflet vegetative endocarditis associated with a grade III to IV systolic heart murmur and signs of aortic embolization (lethargy and weakness in the hind limbs, weak femoral pulses, pelvic pain, increased serum creatine kinase activity) was successfully treated in a cat (Perez et al., 2010), suggesting that Bartonella species may be a cause of blood culture-negative endocarditis, as previously suspected (Malik et al., 1999). Colella et al., (2023) reported B. henselae mitral valve endocarditis associated with multi-resistant Enterococcus faecalis bacteraemia in a cat with prolonged prednisone therapy to treat presumed infiltrative enteropathy.

Myocarditis caused by B. henselae was diagnosed at post-mortem examination in two cats (Varanat et al., 2012) and Joseph et al. (2018) described a clinical case of congestive heart failure with acute onset in a 3 year-old cat from a household fostering stray cats. Echocardiography and electrocardiography respectively evidenced ventricular asymmetrical myocardial thickening with a diffusely mottled hypoechoic echotexture and a left bundle branch block.  After a positive blood PCR test for B. henselae, azythromycin treatment was given for a month and a complete resolution of clinical, ultrasound and electrocardiographic abnormalities were documented.

Lameness and pain during limb palpation were observed in a cat affected by recurrent osteomyelitis and polyarthritis associated with B. vinsonii subsp. berkhoffii infection and bacteraemia (Varanat et al., 2009). Aggressive osteomyelitis causing an incomplete fracture of the radial metaphysis was recently associated with B. henselae bacteraemia in a young cat with generalized peripheral and abdominal lymphadenopathy (Hui et al., 2022). The cat was treated with a combination of doxycycline and pradofloxacin for 6 weeks and clinical and radiographic improvement was documented. A negative blood culture was also obtained as well as evidence of seroreversion one year later (Hui et al., 2022).

In conclusion, most cats naturally infected by B. henselae do not show clinical signs, but some individuals may develop life-threatening cardiovascular diseases and possibly other pathologies associated with generalized lymphadenopathy. Moreover, other Bartonella species, for which cats are accidental hosts, may have pathogenic properties.

Diagnosis

Bartonella laboratory testing is required for feline blood donors, for pet cats belonging to immunosuppressed persons, or when a human Bartonella-related disease is diagnosed in a people that live with cats or have contact with cats.

Cultivation of the bacterium is the gold standard method of diagnosis of Bartonella infection, but because of the high prevalence of infection in healthy cats in endemic areas, a positive culture is not confirmatory of disease, and other compatible diagnoses must be ruled out.

The disease is therefore diagnosed on the basis of exclusion, and by assessing the response to therapy. This ‘ex juvantibus’-inference about disease causation from the observed response to a treatment may apply to uveitis, endocarditis, and multifocal CNS disease, which can all be compatible with feline bartonellosis.

PCR may be used in blood, aqueous humour, cerebrospinal fluid or tissues, and several gene targets have been studied. For rapid laboratory diagnosis, a real-time PCR and pyrosequencing-based algorithm was described that allowed rapid differentiation of at least 11 medically relevant Bartonella spp. within five hours from receipt of the specimens (Buss et al., 2012). Parra et al. (2017) proposed a real time PCR for Bartonella spp. detection to improve PCR sensitivity and to identify species not previously described (). Moreover, in order to detect and classify new Bartonella species from fleas a Matrix-Assisted Laser Desorption/Ionization Time-of-Flight Mass Spectrometry (MALDITOF-MS) has been proposed (El Hamzaoui et al., 2018).

Serology (IFAT or ELISA) is more useful for exclusion than for confirmation, because of the low positive predictive value (39-46%) compared to the good negative predictive value (87-97%; Chomel et al., 1995; Gurfield et al., 2001; Fabbi et al., 2004; Guptill et al., 2004).

Repeated blood cultures are required, or PCR performed on more than one kind of biological sample (blood, lymph node, oral swab; Pennisi et al., 2010; Drummond et al., 2018). A combinational approach with pre-enrichment culture and PCR increases sensitivity (Breitschwerdt et al., 2007).

Treatment

Treatment is recommended for cats living with immunosuppressed persons or in the rare cases where Bartonella has actually caused disease, e.g. endocarditis or myocarditis. Current therapeutic strategies in cats (Table 3) are based on in vitro studies and human bartonellosis.

Table 3: Suggested treatment for Bartonellosis in cats

Drug Dose Duration Reference
Doxycycline 10 mg/kg, PO, q24h 14-28 days Guptill-Yoran, 2012
Azithromycin 10 mg/kg PO q24h (or q48h) 7 days followed by every other day for 6-12 weeks or daily for 3 weeks Ketring et al., 2004; Varanat et al., 2009;
Breitschwerdt et al., 2015;Joseph et al., 2018
Marbofloxacin 5 mg/kg PO q24h 6 weeks Perez et al., 2010
Amoxicillin- clavulanate (in combination with azithromycin) 62.5 mg PO q12h 2 months Varanat et al., 2009
Ciprofloxacin 25 mg/kg PO q24h 15 days Castel et al., 2019
Pradofloxacyn(in combination with doxycycline) 7.5 mg/kg PO q24h 6 weeks Hui et al., 2022

Data from controlled efficacy studies in cats are lacking. A cat affected by recurrent osteomyelitis and polyarthritis associated with B. vinsonii subsp. berkhoffii genotype II infection and bacteraemia recovered after therapy with azithromycin (10 mg/kg PO q48h for three months) and amoxicillin-clavulanate (62.5 mg PO q12 for two months; Varanat et al., 2009).

After natural or experimental infection with B. henselae or B. clarridgeiae, healthy cats have been treated to eliminate bacteraemia (Greene et al., 1996; Regnery et al., 1996; Kordick et al., 1997), and many drugs have been evaluated: doxycycline, amoxicillin, amoxicillin-clavulanate, enrofloxacin, erythromycin, rifampin. Based on these results, clearance of bacteraemia cannot be guaranteed and, in the case of treatment failure, there is the risk of inducing antimicrobial resistance. Treatment of healthy carriers therefore cannot be considered an effective measure for eliminating the zoonotic risk; it is sometimes requested, in human cases of CSD or other Bartonella-related disease in a family member.

Prevention

According to all transmission studies, a strict flea (and tick) control is the only successful preventive measure (Greco et al., 2019). There is no vaccine available against Bartonella infection.

Disease control in specific situations

Strict flea and tick control is the only effective preventive measure.

Zoonotic risk

Cats are the main reservoir for B. henselae, the agent of CSD and other human diseases mainly observed in immunosuppressed persons. Recognised risk factors for bacteraemia in cats are young age, infestation with fleas, outdoor lifestyle and a multicat environment (Chomel et al., 1995; Foley et al., 1998; Gurfield et al., 2001; Guptill et al., 2004; Boulouis et al., 2005).

Sepúlveda-García et al. (2023) studied in households with cats the risk of infection with Bartonella spp. in cats and their owners and estimated the risk factors for Bartonella spp. positivity. Cats younger than one year, non-neutered, sampled at home, and with improper application of tick/flea control products showed a higher risk for Bartonella spp. DNA presence in blood samples. Humans with occupational exposure involving animal contact were more likely to exhibit B. henselae antibody seropositivity in blood. Bartonella spp. DNA was present in at least one cat from 20.4% of the households sampled, but Bartonella DNA was not detected in owners’ blood, inferring that there is a low risk of recent human infection in the studied population.

Clinical signs in humans

CSD in humans is a self-limiting regional lymphadenopathy developing after a primary papular lesion and lasting for a few weeks to several months (Boulouis et al., 2005). Abscessation of the lymph node and systemic signs are occasionally reported. Atypical forms and an expanding spectrum of clinical conditions are being associated with B. henselae infection (Boulouis et al., 2005), such as neuroretinitis, uveitis (Fonollosa et al., 2011), endocarditis (Tsuneoka et al., 2010) and encephalopathy (Samarkos et al., 2018). An unusual CSD case has been reported in a veterinarian affected by persistent fever and back pain after an accidental needle puncture (Lin et al., 2011).

Bacillary angiomatosis (Lange et al., 2009) is one of the most common clinical manifestations in immunocompromised individuals that may be fatal if untreated, whereas immunocompetent persons may experience subclinical Bartonella infection (Massei et al., 2004).

Fig. 2. Immunohistochemical identification of B. henselae in a case of cat scratch disease. Courtesy of Dharam Ramnani, Webpathology.com

There is no benefit of testing asymptomatic cats or people, except in cases of immunosuppressed persons in the home. Infection does not always lead to clinical signs in healthy persons, and many have antibodies (Massei et al., 2004; Mc Gill et al., 2005). Owner education about Bartonella transmission is essential to reduce the zoonotic risk; it is crucial to allow immunosuppressed people to keep their pet cat or to adopt a new one.

Key points to minimise the zoonotic risk (Kaplan et al., 2002; Brunt et al., 2006):

  • Immunosuppressed owners should preferably adopt cats older than 1 year, flea-free, in good health, not from shelters or multicat households, and without contact with cats of unknown health status.
  • A strict flea and tick control should be exercised.
  • Rough play should be avoided, and the cat’s claws kept trimmed.
  • Any wound should promptly be cleaned with soap and water, and medical advice sought.
  • Cats should be kept indoors to avoid exposure to fleas and other possible vectors, but also to prevent other zoonotic risks.

Acknowledgement

ABCD Europe gratefully acknowledges the support of Boehringer Ingelheim (the founding sponsor of the ABCD), Virbac, IDEXX GmbH and MSD Animal Health.

References

Abbott RC, Chomel BB, Kasten RW, Floyd-Hawkins KA, Kikuchi Y, Koehler JE, Pedersen NC (1997): Experimental and natural infection with Bartonella henselae in cats. Comp Immunol Microbiol Infect Dis 20, 41–57.

Allerberger F, Schonbauer M, Zangerle R, Dierich M (1995): Prevalence of antibody to Rochalimaea henselae among Austrian cats. Eur J Ped 154, 165.

Álvarez-Fernández A, Baxarias M, Prandi D, Breitschwerdt EB, Solano-Gallego (2021): Bartonella henselae antibodies in serum and oral fluid specimens from cat. Pathogens 10, 329.

Arvand M, Viezens J, Berghoff J (2008): Prolonged Bartonella henselae bacteremia caused by reinfection in cats. Emerg Infect Dis 14, 152-154.

Ayllon T, Diniz PP, Breitschwerdt EB, Villaescusa A, Rodriguez-Franco F, Sainz A (2012): Vector-borne diseases in client-owned and stray cats from Madrid, Spain. Vector Borne Zoonotic Dis 12, 143-150.

Bayliss DB, Steiner JM, Sucholdolski JS, Radecki SV, Brewer MM, Morris AK, Lappin MR (2009): Serum feline pancreatic lipase immunoreactivity concentration and seroprevalences of antibodies against Toxoplasma gondii and Bartonella species in client-owned cats. J Feline Med Surg 11, 663-667.

Belgard S, Truyen U, Thibault JC, Sauter-Louis C, Hartmann K (2010): Relevance of feline calicivirus, feline immunodeficiency virus, feline leukemia virus, feline herpesvirus and Bartonella henselae in cats with chronic gingivostomatitis. Berl Munch Tierarztl Wochenschr 123, 369-376.

Bennett AD, Gunn-Moore DA, Brewer M, Lappin MR (2011): Prevalence of Bartonella species, haemoplasmas and Toxoplasma gondii in cats in Scotland. J Feline Med Surg 13, 553-557.

Bergmans AMC, de Jong CMA, van Amerongen G, Schot CS, Schouls LM (1997): Prevalence of Bartonella species in domestic cats in The Netherlands. J Clin Microbiol 35, 2256-2261.

Berryessa NA, Johnson LR, Kasten RW, Chomel BB (2008): Microbial culture of blood samples and serologic testing for bartonellosis in cats with chronic rhinosinusitis. J Am Vet Med Assoc 233, 1084-1089.

Bouhsira E, Ferrandez Y, Liu M, Franc M, Bouloius HJ, Biville F (2013): Ctnecephalides felis an in vitro potential vector for five Bartonella species. Comp Immunol Microbiol Inf Dis 36, 105-111.

Boulouis HJ, Chang CC, Henn JB, Kasten RW, Chomel BB (2005): Factors associated with the rapid emergence of zoonotic Bartonella infections. Vet Res 36, 383-410.

Bradbury CA, Lappin MR (2009): Evaluation of topical application of 10% imidacloprid-1% moxidectin to prevent Bartonella henselae transmission from cat fleas. J Am Vet Med Assoc 236, 869-873.

Breitschwerdt E, Broadhurst JJ, Cherry NA (2015): Bartonella henselae as a cause of acute-onset febrile illness in cats. JFMS Open reports, 1-9.

Breitschwerdt EB, Kordick DL, Malarkey DE, Keene B, Hadfield TI, Wilson K (1995): Endocarditis in a dog due to infection with a novel Bartonella subspecies. J Clin Microbiol 33, 154-160.

Breitschwerdt EB, Levine JF, Radulovich S, Hanby SB, Kordick DL, La Perle KMD (2005): Bartonella henselae and Rickettisa seroreactivity in a sick cat population from North Carolina. Int J Appl Res Vet Med 3, 287-302.

Breitschwerdt EB, Maggi RG, Sigmon B, Nicholson WL (2007): Isolation of Bartonella quintana from a woman and a cat following putative bite transmission. J Clin Microbiol 45, 270-272.

Brunetti E, Fabbi M, Ferraioli G, Prati P, Filice C, Sassera D et al (2013): Cat-scratch disease in Northern Italy: atypical clinical manifestations in humans and prevalence of Bartonella infection in cats. Eur J Clin Microbiol Infect Dis 32, 531-534.

Brunt J, Guptill L, Kordick DL, Kudrak S, Lappin MR (2006): Panel report on diagnosis, treatment, and prevention of Bartonella spp. infections. Am Assoc Feline Pract; Academy of Feline Medicine Advisory Panel. J Feline Med Surg 8, 213-226.

Buss SN, Gebhardt LL, Musser KA (2012): Real-time PCR and pyrosequencing for diffentiation of medically relevant Bartonella species. J Microbiol Meth 91, 252-256.

Castel A, Olbyb NJ, Breitschwerdtb EB, Thomas B, Maggi RG, Sheltond GD (2019): Co-infection with Bartonella henselae and Sarcocystis sp. in a 6-year-ol male neutered domestic longhair cat with progressive multifocal neurological signs. Vet Quart 39, 168-173.

Chomel BB, Boulouis H-J, Maruyama S, Breitschwerdt EB (2006): Bartonella spp. in pets and effect on human health. Emerg Inf Dis 12, 389-394.

Chomel BB, Gurfield AN, Boulouis HJ, Kasten RW, Piemont Y (1995): Réservoir félin de l’agent de la maladie des griffes du chat, Bartonella henselae, en région parisienne: resultants préliminaires. Rec Med Vet 171, 841-845.

Chomel BB, Kasten RW, Floyd-Hawkins KA, Chi B, Yamamoto K, Roberts-Wilson J, Gurfield AN, Abbott RC, Pedersen NC, Koehler J (1996): Experimental transmission of Bartonella henselae by the cat flea. J Clin Microbiol 34, 1952–1956.

Chomel BB, Kasten RW, Williams C, Wey AC, Henn JB, Maggi R, Carrasco S, Mazet J, Boulouis HJ, Maillard R, Breitschwerdt EB (2009): Bartonella endocarditis: a pathology shared by animal reservoirs and patients. Ann N Y Acad Sci 1166, 120-126.

Chomel BB, Molia S, Kasten RW, Borgo GM, Stuckey MJ, Maruyama S, Chang CC, Haddad N, Koehler JE (): Isolation of Bartonella henselae and two new bartonella subspecies, Bartonella koehlerae subspecies boulouisii subsp. nov. and Bartonella koehlerae subspecies bothieri subsp. nov. from free-ranging californian mountain lions and bobcats. PLoS One 11 (3), e0148299.

Chomel B, Wey AC, Kasten RW, Stacy BA, Labelle P (2003): Fatal case of endocarditis associated with Bartonella henselae Type I infection in a domestic cat. J Clin Microbiol 41, 5337-5339.

Colella A, Greco G, Cordisco M, Tinelli A, Paradies P (2023): Bartonella henselae mitral valve endocarditis associated with multi-resstant Enterococcus faecalis bacteraemia in a cat. Vet Rec Case Report, 11:e724.

Cottè V, Bonnet S, Le Rhun D, Le Naour E, Chauvin A, Boulouis H-J, Lecuelle B, Lilin T, Vayssier-Taussat M (2008): Transmission of Bartonella henselae by Ixodes ricinus. Emerg Infect Dis 14, 1074–1080.

Dowers KL, Hawley JR, Brewer MM, Morris AK, Radecki SV, Lappin MR (2010): Association of Bartonella species, feline calicivirus, and feline herpesvirus 1 infection with gingivostomatitis in cats. J Feline Med Surg 12, 314-321.

Drummond MR, Lania BG, Diniz PPVP, Gilioli R, Demolin DMR, Scorpio DG, Breitschwerdt EB, Velho PENF (2018): Improvement of Bartonella henselae DNA Detection in Cat Blood Samples by Combining Molecular and Culture Methods. J Clin Microbiol 56, e01732-17.

Ebani VV, Nardoni S, Maestrini M, Perrucci P, Mancianti F (2021): Serological survey on the occurrence of Rickettsia spp., Neospora caninumBartonella henselae and Toxoplasma gondii in cats from Tuscany (Central Italy). Animals 11, 1842.

El Hamzaoui B, Laroche M, Almeras L, Bérenger JM, Raoult D, Parola P (2018): Detection of Bartonella spp. in fleas by MALDI-TOF MS. PLoS Negl Trop Dis 12, e0006189.

Fabbi M, De Giuli L, Tranquillo M, Bragoni R, Casiraghi M, Genchi C (2004): Prevalence of Bartonella henselae in Italian stray cats: evaluation of serology to assess the risk of transmission of Bartonella to humans. J Clin Microbiol 42, 264– 268.

Finkelstein JL, Brown TP, O’Reilly KL, Wedincamp J Jr, Foil LD (2002): Studies on the growth of Bartonella henselae in the cat flea (Siphonaptera: Pulicidae). J Med Entomol 39, 915-919.

Foley JE, Chomel B, Kikuchi Y, Yamamoto K, Pedersen NC (1998): Seroprevalence of Bartonella henselae in cattery cats: association with cattery hygiene and flea infestation. Vet Q 20, 1-5.

Fonollosa A, Galdos M, Artaraz J, Perez-Irezabal J, Martinez-Alday N (2011): Occlusive vasculitis and optic disk neovascularization associated with neuroretinitis. Ocul Immunol Inflamm 19, 62-64.

Fontenelle JP, Powell CC, Hill AE, Radecki SV (2008): Prevalence of serum antibodies against Bartonella species in the serum of cats with or without uveitis. J Feline Med Surg 10, 41-46.

Glaus T, Hofmann-Lehmann R, Greene C, Glaus B, Wolfensberger C, Lutz H (1997): Seroprevalence of Bartonella henselae infection and correlation with disease status in cats in Switzerland. J Clin Microbiol 35, 2883-2885.

Greco G, Brianti E, Buonavoglia C, Carelli G, Pollmeier, Schunack MB, Dowgier G, Capelli G, Dantas-Torres F, Otranto D (2019): Effectiveness of a 10% imidacloprid/4.5% flumethrin polymer matrix collar in reducing the risk of Bartonella spp. infection in privately owned cats. Parasites & Vectors 12, 69.

Greene CE, McDermott M, Jameson PH, Atkins CL, Marks AM (1996): Bartonella henselae infection in cats: evaluation during primary infection, treatment, and rechallenge infection. J Clin Microbiol 34, 1682-1685.

Guptill L (2010): Bartonellosis. Vet Microbiol 140, 347–359.

Guptill L, Slater L, Wu C-C, Lin T-L, Glickman LT, Welch DF, HogenEsch H (1997): Experimental infection of young specific pathogen-free cats with Bartonella henselae. J Infect Dis 176, 206–216.

Guptill L, Wu CC, HogenEsch H, Slater LN, Glickman N, Dunham A, Syme H, Glickman L (2004): Prevalence, risk factors, and genetic diversity of Bartonella henselae infections in pet cats in four regions of the United States. J Clin Microbiol 42, 652-659.

Guptill-Yoran L. (2012): Feline bartonellosis. In 545-552. Infectious diseases of the dog and cat. Ed. C.E. Greene, 4th ed., Elsevier Saunders St. Louis, MO-USA.

Gurfield AN, Boulouis H-J, Chomel BB, Kasten RW, Heller R, Bouillin C, Gandoin C, Thibault D, Chang C-C, Barrat F, Piemont Y (2001): Epidemiology of Bartonella infection in domestic cats in France. Vet Microbiol 80, 185-198.

Haimerl M, Tenter AM, Simon K, Rommel M, Hilger J, Autenrieth IB (1999): Seroprevalence of Bartonella henselae in cats in Germany. J Med Microbiol 48, 849-856.

Heller R, Artois M, Xemar V, de Briel D, Gehin H, Jaulhac B, Monteil H, Piemont Y (1997): Prevalence of Bartonella henselae and Bartonella clarridgeiae in stray cats. J Clin Microbiol 35, 1327-1331.

Hui J, Ryan KA, Rademacher N, Neupane P, Breitschwerdt EB (2022): Osteomyelitis associated wit Bartonella henselae infection in a young cat. J Fel Med Surg Open Rep, 1-5. DOI: 10.1177/20551169221124910

Ishak AM, Radecki S, Lappin MR (2007): Prevalence of Mycoplasma haemofelis, ‘Candidatus Mycoplasma haemominutum’, Bartonella species, Ehrlichia species, and Anaplasma phagocytophilum DNA in the blood of cats with anemia. J Feline Med Surg 9, 1-7.

Joseph JL, Oxford EM, Santilli RA (2018): Transient myocardial thickening in a Bartonella henselae-positive cat. J Vet Cardiol 20, 198-203.

Kaplan JE, Masur H, Holmes KK, USPHS; Infectious Disease Society of America (2002): Guidelines for preventing opportunistic infections among HIV-infected persons. Recommendations of the U.S. Public Health Service and the Infectious Diseases Society of America. MMWR Recomm Rep 51, 1-52.

Ketring KL, Zuckerman EE, Hardy Jr WD (2004): Bartonella: a new etiological agent of feline ocular disease. J Am Animal Hospital Ass 40, 6-12.

Kokkinaki KGG, Saridomichelakis MN, Skampardonis V, Mataragka A, Ikonomopoulos J, Leontides L, Mylonakis ME, Steiner JM, Suchodolski JS, Xenoulis PG (2022): Prevalence and Risk Factors for Bartonella spp. and Haemoplasma Infections in Cats from Greece. Vet Sci 9, 337.

Kordick D, Brown TT, Shin K, Breitschwerdt EB (1999): Clinical and pathologic evaluation of chronic Bartonella henselae or Bartonella clarridgeiae infection in cats. J Clin Microbiol 37, 1536-1547.

Kordick DL, Papich MG, Breitschwerdt EB (1997): Efficacy of enrofloxacin or doxycycline for treatment of Bartonella henselae or Bartonella clarridgeiae infection in cats. Antimicrob Agents Chemother 41, 2448-2455.

Lange D, Oeder C, Waltermann K, Mueller A, Oehme A, Rohrberg R, Marsch W, Fischer M (2009): Bacillary angiomatosis. J Dtsch Dermatol Ges 7, 767-769.

Lappin MR, Black JC (1999): Bartonella spp infection as a possible cause of uveitis in a cat. J Am Vet Med Assoc 214, 1205-1207.

Lappin MR, Breitschwerdt E, Brewer M, Hawley J, Hegarty B (2009): Prevalence of Bartonella species antibodies and Bartonella species DNA in the blood of cats with and without fever. J Feline Med Surg 11, 141-148.

La Scola B, Raoult D. (1999): Culture of Bartonella quintana and Bartonella henselae from human samples: a 5-year experience (1993-1998). J Clin Microbiol 37, 1899-1905.

Leibovitz K, Pearce L, Brewer M, Lappin MR (2008): Bartonella species antibodies and DNA in cerebral spinal fluid of cats with central nervous system disease. J Feline Med Surg 10, 332-337.

Lin JW, Chen CM, Chang CC (2011): Unknown Fever and Back Pain Caused by Bartonella henselae in a Veterinarian After a Needle Puncture: A Case Report and Literature Review. Vector Borne Zoonotic Dis 11, 589-591.

Malik R, Barrs VR, Church DB, Zahn A, Allan GS, Martin P, Wigney DI (1999): Vegetative endocarditis in six cats. J Feline Med Surg 1, 171-180.

Massei F, Messina F, Gori L, Macchia P, Maggiore G. (2004): High prevalence of antibodies to Bartonella henselae among Italian children without evidence of cat scratch disease. Clin Infect Dis 38, 145-148.

Mazurek Ł, Carbonero A, Skrzypczak M, Winiarczyk S, Adaszek Ł (2020): Epizootic situation of feline Bartonella infection in eastern Poland.  J Vet Res 64, 79-83.

Mazurek L, Winiarczyk S, Skrzypczak M, Adaszek L (2019): Cats as a reservoir of Bartonella henselae for dogs. Ann Agr Env Med 26, 669-671.

Mc Donald KA, Chomel BB, Kittlesone MD, Kasten RW, Thomas WP, Pesavento P (2004): A prospective study of canine infective endocarditis in northern California (1999-2001): emergence of Bartonella as a prevalent etiologic agent. J Vet Int Med 18, 56-64.

McGill S, Wesslen L, Hjelm E, Holmberg M, Auvinen M, Berggren K, Grandin-Jarl B, Johnson U, Wikström S, Friman G (2005): Bartonella spp. seroprevalence in healthy Swedish blood donors. Scand J Infect Dis 37, 723-730.

Molia S, Kasten RW, Stuckey MJ, Boulouis HJ, Allen J, Borgo GM, Koehler JE, Chang CC, Chomel BB. (2016): Isolation of Bartonella henselae, Bartonella koehlerae subsp. koehlerae, Bartonella koehlerae subsp. bothieri and a new subspecies of B. koehlerae from free-ranging lions (Panthera leo) from South Africa, cheetahs (Acinonyx jubatus) from Namibia and captive cheetahs from California. Epidemiol Infect 144, 3237-3243.

Morelli S, Crisi PE, Di Cesare A, De Santis F, Barlaam A, Santoprete G, Parrinello C, Palermo S, Mancini P, Traversa D (2019): Exposure of client-owned cats to zoonotic vector-borne pathogens: Clinicpathological alterations and infection risk analysis. Comp Immunol Microbiol Inf Dis 66, 101344.

Morgenthal D, Hamel D, Arndt G, Silaghi C, Pfister K, Kempf VA, et al (2012): Prevalence of haemotropic Mycoplasma spp, Bartonella spp and Anaplasma phagocytophilum in cats in Berlin/Brandenburg (Northeast Germany). Berl Munch Tierarztl Wochenschr 125, 418-427.

Morse SF, Olival KJ, Kosoy M, Billeter S, Patterson BD, Dick CW, et al (2012): Global distribution and genetic diversity of Bartonella in bat flies (Hippoboscoidea, Strebilidae, Nycteribiide). Inf Gen Evol 12, 1717-1723.

Muz MN, Erat S, Mumcuoglu KY (2021): Protozoan and Microbial Pathogens of House Cats in the Province of Tekirdag in Western Turkey. Pathogens 10, 1114.

Namekata DY, Kasten RW, Boman DA, Straub MH, Siperstein-Cook L, Couvelaire K, Chomel BB (2010): Oral shedding of Bartonella in cats: correlation with bacteremia and seropositivity. Vet Microbiol 146, 371-375.

Parra E, Segura F, Tijero J, Pons I, Nogueras MM (2017): Development of a real-time PCR for Bartonella spp. detection, a current emerging microorganism. Molecular and Cellular Probes 32, 55-59.

Pearce LK, Radecki SV, Brewer M, Lappin MR (2006): Prevalence of Bartonella henselae antibodies in serum of cats with and without clinical signs of central nervous system disease. J Feline Med Surg 8, 315-320.

Pennisi MG, La Camera E, Giacobbe L, Orlandella BM, Lentini V, Zummo S, Fera MT (2010): Molecular detection of Bartonella henselae and Bartonella clarridgeiae in clinical samples of pet cats from Southern Italy. Res Vet Sci 88, 379-384.

Pennisi MG, Marsilio F, Hartmann K, Lloret A, Addie D, Bélak S, et al (2013): Bartonella species infection in cats. ABCD guidelines on prevention and management. J Feline Med Surg 15, 563-569.

Perez C, Hummel JB, Keene BW, Maggi RG, Diniz PPVP, Breitschwerdt EB (2010): Successful treatment of Bartonella henselae endocarditis in a cat. J Feline Med Surg 12, 483-486.

Persichetti MF, Pennisi MG, Vullo A, Masucci M, Migliazzo A, Solano-Gallego L (2018): Clinical evaluation of outdoor cats exposed to ectoparasites and associated risk for vector-borne infections in southern Italy. Parasit Vectors 11, 136.

Persichetti MF, Solano-Gallego L, Serrano L, Altet L, Reale S, Masucci M, Pennisi MG (2016): Detection of vector-borne pathogens in cats and their ectoparasites in southern Italy. Parasit Vectors 9, 247.

Quimby J, Elston T, Hawley J, Brewer M, Miller A, Lappin MR (2008): Evaluation of the association of Bartonella species, feline herpesvirus 1, feline calicivirus, feline leukemia virus and feline immunodeficiency virus with chronic feline gingivostomatitis. J Feline Med Surg 10, 66-72.

Razgūnaitė M, Lipatova I, Paulauskas A, Karvelienė B, Riškevičienė V, Radzijevskaja J (2021): Bartonella Infections in Cats and Cat Fleas in Lithuania. Pathogens 10, 1209.

Regier Y, Ballhorn W, Kempf VAJ (2017): Molecular detection of Bartonella henselae in 11 Ixodes ricinus ticks extracted from a single cat. Parasit Vectors 10, 105.

Regnery RL, Rooney JA, Johnson AM, Nesby SL, Manzewitsch P, Beaver K, Olson JG (1996): Experimentally induced Bartonella henselae infections followed by challenge exposure and antimicrobial therapy in cats. Am J Vet Res 57, 1714. Erratum in: Am J Vet Res (1997) 58, 803.

Samarkos M, Antoniadou V, Vaiopoulos AG, Psichogiou M (2018): Encephalopathy in an adult with cat-scratch disease. BMJ Case Rep., bcr-2017-223647.

Sepúlveda-García P, Alabi A, Álvarez K, Rojas L, Mella A, Gonçalves LR, André MR, Machado RZ, Müller A, Monti G (2023) Bartonella spp. In household with cats: Risk factors for infection in cats and human exposure. One Health 16, 100545

Tsuneoka H, Yanagihara M, Otani S, Katayama Y, Fujinami H, Nagafuji H, Asari S, Nojima J, Ichihara K (2010): A first Japanese case of Bartonella henselae-induced endocarditis diagnosed by prolonged culture of a specimen from the excised valve. Diagn Microbiol Infect Dis 68, 174-176.

Ueno H, Hohdatsu T, Muramatsu Y, Koyama H, Morita C (1996): Does coinfection of Bartonella henselae and FIV induce clinical disorders in cats? Microbiol Immunol 40, 617-620.

Varanat M, Broadhurst J, Linder KE, Maggi RG, Breitschwerdt EB (2012): Identification of Bartonella henselae in 2 cats with pyogranulomatous myocarditis and diaphragmatic myositis. Vet Pathol 49, 608-611.

Varanat A, Travis A, Lee W, Maggi RG, Bissett SA, Linder KE, Breitschwerdt EB (2009): Recurrent osteomyelitis in a cat due to infection with Bartonella vinsonii subsp. berkhoffii genotype II. J Vet Intern Med 23, 1273-1277.

Yamamoto K, Chomel BB, Kasten RW, Hew CM, Weber DK, Lee WI (2002): Experimental infection of specific pathogen free (SPF) cats with two different strains of Bartonella henselae type I: a comparative study. Vet Res 33, 669-684.

Zobba R, Chessa G, Mastrandrea S, Pinna Parpaglia ML, Patta C, Masala G (2009): Seriological and molecular detection of Bartonella spp in humans, cats and dogs from Northern Sardinia, Italy. Clin Microbiol Inf 15, suppl 2, 134-135.