Next Article in Journal
Dynamic and Postural Changes in Forelimb Amputee Dogs: A Pilot Study
Previous Article in Journal
Classification of Behaviour in Conventional and Slow-Growing Strains of Broiler Chickens Using Tri-Axial Accelerometers
Previous Article in Special Issue
Seroprevalence and Risk Factors Associated with Chlamydia abortus Infection in Sheep and Goats in North-Western Italy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Case Report

Outbreak of Chlamydia psittaci Infection in a Commercial Psittacine Breeding Aviary in Argentina

by
María Belén Riccio
1,*,
Jorge Pablo García
1,
María Laura Chiapparrone
2,
Juliana Cantón
2,
Claudio Cacciato
2,
Javier Anibal Origlia
3,
María Estela Cadario
4,
Santiago Sain Diab
5 and
Francisco Alejandro Uzal
6
1
Servicio de Diagnóstico Veterinario FCV Tandil, Facultad de Ciencias Veterinarias, Universidad Nacional del Centro de la Provincia de Buenos Aires, Tandil B7000GHG, Argentina
2
Laboratorio de Microbiología Clínica y Experimental, Centro de Investigación Veterinaria de Tandil (CIVETAN) (UNCPBA-CICPBA-CONICET), Facultad de Ciencias Veterinarias, Universidad Nacional del Centro de la Provincia de Buenos Aires, Tandil B7000GHG, Argentina
3
Cátedra de Patología de Aves y Pilíferos, Facultad de Ciencias Veterinarias, Universidad Nacional de La Plata, La Plata B1900BVB, Argentina
4
INEI-ANLIS «Dr. Carlos G. Malbrán», Ciudad Autónoma de Buenos Aires, Buenos Aires B1282AFF, Argentina
5
Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
6
California Animal Health and Food Safety Laboratory, School of Veterinary Medicine, University of California Davis, San Bernardino, CA 92408, USA
*
Author to whom correspondence should be addressed.
Animals 2024, 14(13), 1959; https://doi.org/10.3390/ani14131959
Submission received: 30 May 2024 / Revised: 27 June 2024 / Accepted: 28 June 2024 / Published: 2 July 2024
(This article belongs to the Special Issue Chlamydial Diseases in Animals)

Abstract

:

Simple Summary

Chlamydiosis, caused by the bacterium Chlamydia psittaci, commonly affects psittacine birds and can spread to humans. It is highly contagious among birds, manifesting in various form with clinical signs like respiratory distress, lethargy, and diarrhea. In humans, psittacosis presents with flu-like symptoms, including fever, headache, and respiratory difficulty; it can be severe or, if left untreated, fatal. The potential transmission of psittacosis from infected birds to humans, primarily through the inhalation of contaminated dust or aerosols, possesses a threat to human health. This article describes an outbreak of chlamydiosis in a commercial psittacine breeding aviary in Buenos Aires, Argentina, in 2021. The outbreak led to the deaths of different bird species, all showing weight loss, diarrhea, and respiratory distress. Necropsies revealed severe internal conditions, and histological examination confirmed the infection. The diagnosis was verified through immunohistochemistry and molecular techniques, identifying genotypes A and B of Chlamydia psitacci.

Abstract

Chlamydiosis, caused by Chlamydia psittaci is a bacterial infection found in at least 465 species of birds worldwide. It is highly contagious among birds and can spread to humans. In birds, the disease can manifest itself in acute, subacute, and chronic forms with signs including anorexia, diarrhea, lethargy, weight loss, or, occasionally, mucopurulent or serous oculonasal discharge. This article describes an outbreak of chlamydiosis that occurred in a commercial psittacine breeding aviary in 2021 in Buenos Aires province, Argentina. In total, 16 juvenile blue-fronted parrots, more than 60 blue-fronted parrot chicks, and 2 adult macaws died during the outbreak. In all cases, clinical signs were weight loss, diarrhea, yellowish green excrement, and respiratory distress. The necropsy of four juvenile blue-fronted parrots, two blue-fronted parrot chicks, and two adult macaws revealed cachexia, hepatomegaly, splenomegaly, splenic petechial hemorrhages, ascites, pulmonary edema, and hydropericardium. Histologically, multifocal lymphoplasmacytic and heterophilic airsaculitis, multifocal lymphoplasmacytic and necrotizing hepatitis with intracytoplasmic elementary bodies, multifocal necro-heterophilic hepatitis, multifocal lymphoplasmacytic nephritis, and diffuse heterophilic pneumonia were found. A presumptive diagnosis was established based on gross and microscopic lesions, and it was confirmed using immunohistochemistry and polymerase chain reactions. The sequencing and phylogenetic analysis of the ompA gene revealed genotype A and B of Chlamydia psittaci.

1. Introduction

Chlamydial infections caused by widespread obligate intracellular Gram-negative bacteria from the Chlamydiaceae family are associated with a wide spectrum of clinical symptoms in livestock, companion animals, wildlife, and various exotic species including several birds species [1,2,3]. In addition, some chlamydial species, such as Chlamydia abortus and Chlamydia psittaci, are capable of causing zoonotic infections [4,5].
In humans, Chlamydia psittaci (C. psitacci) causes a severe disease, known as psittacosis, which, in most cases, can lead to pneumonia and significant mortality if left untreated [6,7,8,9]. Psittacosisis is a significant threat to public health and is considered an occupational zoonosis of pet shop owners, zookeepers, veterinarians, abattoir workers, bird breeders, and pet bird enthusiasts [2,10,11].
At least 465 bird species belonging to 30 orders can have chlamydiosis. However, psittacidae (lories, parakeets, parrots, and cockatoos) and columbiformes (pigeons) are the most frequently affected. Primary reservoirs for zoonotic psittacosis are psittacine pets, as well as wild and racing pigeons [12].
C. psittaci replicates using a biphasic developmental cycle within cytoplasmic inclusion vacuoles of eukaryotic cells. This cycle involves two morphologically different forms, as follows: the infectious extracellular form, termed the elementary body (EB), and the reproductive reticulated body (RB). The EB enters a host cell through receptor-mediated endocytosis. This process, which lasts several hours, results in the conversion of the EB into a reticulate body (RB). The RB divides via binary fission to produce more EBs. This cycle continues for several hours after infection, at which time the host cell lyses, releasing several hundred EBs, RBs, and intermediates [13].
Chlamydiosis causes a range of clinical signs in birds, including respiratory difficulty, diarrhea, and lethargy, and it can also lead to chronic or asymptomatic infections. Some infected birds may appear healthy, but shed the organism intermittently. Shedding may be exacerbated by internal factors such as the bird’s immune status, in addition to external factors such as stress, reproductive activities, relocation, shipping, crowding, and cold temperature. The severity of clinical signs varies significantly, influenced by the species and age of the birds, as well as the specific strains of the pathogen involved [2].
In psittacine birds, gross lesions include splenomegaly, hepatomegaly, enteritis, sinusitis, air sacculitis, pericarditis, and conjunctivitis. Microscopic lesions of chlamydiosis in birds include necrotizing changes in several organs associated with chlamydial intracytoplasmic elementary bodies and the infiltration of heterophils, macrophages, and plasma cells [14,15,16].
Chlamydia psittaci infection in birds has been reported previously in many countries [1,17,18,19,20]. In recent years, there has been a global increase in the number of reported cases of avian chlamydiosis in both birds and humans [9,21,22,23].
The analysis of C. psittaci ompA gene sequences is crucial for genotype determination [24]. In the studied psittacidae population, the circulating genotypes are identified as A and B. Initially, C. psittaci was categorized into nine genotypes—A-F, E/B, M56, and WC, with the later proposal of eight new genotypes [1,24]. Genotype A is prevalent among psittacine birds, but is also found in turkeys, ducks, pigeons, and Passeriformes. Genotype B is endemic in pigeons but can infect various bird species including chickens, turkeys, ducks, Psittacidae, and Passeriformes. Waterfowl are commonly infected with genotype C and E/B strains, while turkeys can harbor genotypes D and C. Genotype E is found in ducks, pigeons, ostriches, and rheas, and genotype F is in psittacine and turkey isolates [1,25,26,27].
This article describes a large outbreak of psittacosis in a commercial breeding aviary in Buenos Aires, Argentina, and emphasizes the need to monitor and quickly identify the disease to minimize its impact in birds and prevent the spread to people at risk.

2. Materials and Methods

2.1. Case Presentation

The outbreak occurred between January and April 2021 at a new facility of a commercial psittacine breeding aviary in Buenos Aires province, Argentina. This aviary specializes in the conservation, breeding, and export of blue-fronted Amazon parrots. It also acts as a rescue center, receiving and rehabilitating parrots and macaws that have been confiscated from the illegal trade. The population of psittacine birds included blue-fronted parrots (Amazona aestiva), red-and-green macaws (Ara chloropterus), and scarlet macaws (Ara macao). Previously, they were in other facilities with outdoor cages. The new aviary has a ground floor with the breeding and quarantine area, and the first floor houses the juvenile and adult birds in indoor cages. These cages are 7.5 m long, 1.5 m wide, and 2.4 m high. The parrots have ad libitum access to fruit, vegetable, and seeds. Water is provided ad libitum in bowls. Excess food is removed daily, the floors are dry cleaned, the trays are washed, and fresh water is replenished.
During January and April 2021, 63 out of 120 few-days-old blue-fronted parrot chicks, 16 out of 100 juvenile blue-fronted parrots, 1 1.5-year-old red-and-green macaw (Ara chloropterus), and a similarly aged juvenile scarlet macaw (Ara macao) died.
The affected birds exhibited lethargy, refusal to eat, regurgitation, depression, cachexia, anorexia, respiratory distress, diarrhea, and yellowish green excrements. Despite treatment, most birds with these symptoms died quickly.
The mortality rate of the outbreak was 52.5% in blue-fronted parrot chicks, 16% in juvenile blue-fronted parrots, and 25% in green-and-red macaws. The only scarlet macaw specimen also died.
After confirming the diagnosis of chlamydiosis in mid-April, a new therapeutic approach consisting of the oral administration of oxytetracycline at a rate of 300 mg/kg in food, per animal, every 24 h for 45 days was started.
To evaluate the effectiveness of this treatment, a triple mucosal swab from conjunctiva, choanae, and cloaca obtained from two adult blue-fronted parrots previously tested positive for C. psitacci were collected and submitted for Chlamydia spp. real-time PCR analysis.

2.2. Necropsy and Ancillary Tests

Necropsies were performed on four juvenile blue-fronted parrots (P1, P2, P3, P4), two blue-fronted parrot chicks (C1, C2), one red-and-green macaw, and one scarlet macaw. All birds were weighed. Sterile organ samples, including lung, liver, bursa of Fabricius, air sac, and laryngeal swabs, were analyzed for Trichomonas and Salmonella sp. from P1 and C1. Additionally, Salmonella cultures were performed on liver samples from all birds, except for macaws. Lung and liver samples from all birds, excluding macaws, were submitted for fungal culture. Selected organs, such as liver, lung, air sacs, spleen, intestine, and kidneys, were processed for histopathology. Triple mucosal swabs from the conjunctiva and choanae, or pooled organ samples from liver, lung, and spleen, were submitted for real-time PCR to detect C. psittaci, Psittacid Herpesvirus, and Avian Polyomavirus. Paraffin-embedded spleen and kidney samples from P2, P3, and a scarlet macaw were submitted for immunohistochemistry (IHC).

2.3. Microbiology

For the detection of aerobic and microaerophilic mesophile bacteria, the organ and swab samples submitted from juvenile and chick blue-fronted parrots were cultured in two culture media—tryptone soy agar supplemented with 10% bovine blood, under aerobic and microaerophilic (5% O2, 10% CO2, and 85% N2) conditions for 24 and 48 h, and MacConkey agar at 37 °C, under aerobic conditions, for 24 h. The samples were cultured in Selenite broth at 37 °C and 42 °C as a pre-enrichment medium and were sub-cultured at 24 h and 48 h in Salmonella–Shigella agar, and were incubated for 24 h at 37 °C, all of this in aerobic conditions. The isolations were identified through colonies and bacterial morphologies, Gram-stain, and biochemical tests [13,28].
For Trichomonas sp., the samples were cultured in Diamond (TYM) medium at 37 °C in aerobic conditions and were observed every 24 h for 7 days. For fungi, the samples were cultured in Sabouraud agar at 25 °C in aerobic conditions and were observed every 24 h for 10 days.
The antibiotic susceptibility test was performed using the disc diffusion method (Kirby Bauer) according to Clinical and Laboratory Standards Institute guidelines (CLSI, 2020).

2.4. Histopathology

The organs submitted (liver, spleen, air sacs, heart, intestine, and kidneys) from all birds were fixed in a 10% neutral buffered formalin solution. Tissues were embedded in paraffin, sectioned at 4 μm, mounted on glass slides, and stained with hematoxylin and eosin (H&E) using the routine method. Selected kidney sections from the red-and-green macaw were stained with the Gimenez method.

2.5. Inmunohistochemistry

Selected sections of liver and spleen from the scarlet macaw, and the spleen, liver, and kidney from parrots P2 and P4 were processed using immunohistochemistry (IHC) following standard operating procedures at the California Animal Health and Food Safety Laboratory, University of California, Davis, USA. Briefly, paraffin-embedded tissues were sectioned at 4 mm and were mounted on charged slides, deparaffinized, and hydrated, quenching endogenous peroxidase with 3% hydrogen peroxide. Slides were then placed in 0.4% acidulated pepsin for antigen retrieval for 15 min in a 37 °C water bath and were subsequently rinsed in running lab-grade water for 3–5 min and then buffer solution (TBS-Tween, Saint Louis, MI, USA) for 3–10 min. The primary antibody (ViroStat #1681 (Le Pontet, Vaucluse, France), Chlamydia pecorum-LPS (genus specific, cross-reactive with Chlamydia pneumonia and Chlamydia psittaci), and non-specific reagent were applied for 30–60 min. Sections were rinsed in buffer solution, and a secondary antibody (Dako Envision + K4001, Santa Clara, CA, USA) was applied for 30 min. After rinsing in buffer solution again, a chromogen (AEC, ready-to-use, Dako K3464) was applied to the sections for 10–20 min. Slides were then rinsed in lab-grade water and were subsequently placed in Mayer’s hematoxylin for 1–4 min. After rinsing with lab-grade water again, slides were blued in Scott’s tap water for 1–2 min. Slides were finally rinsed in running lab-grade water and coverslips with aqueous mounting media were applied. All incubations were at room temperature.

2.6. Chlamydia Isolation Procedure

The isolation of Chlamydia in VERO cell cultures was attempted for some of the samples stored in transport medium, following previously described procedures under biosafety level 3 precautions [29].

2.7. Polymerase Chain Reaction

Fresh and frozen tissues (liver, spleen, and lung) and trimucosal swabs were subjected to DNA extraction. Approximately 25 µg of the homogenated pool of organs from the necropsied birds were used for DNA extraction. Each swab was resuspended in 600 µL of phosphate-buffered saline (PBS) and were homogenized using a vortex; DNA extraction was carried out from 200 µL of each sample. For both types of samples, the High Pure PCR Template Preparation Kit (Roche, Mannheim, Germany) was used according to the manufacturer’s instructions. Extracted DNA was stored at −20 °C until use.
For the detection of C. psittaci, real-time PCR was performed for the ompA gene target, as described by Pantchev et al. 2009 [30]. Those samples with a CT bellow 38 were considered positive. Specific real-time PCR was also performed for the detection of Psittacid Herpesvirus and Avian Polyomavirus [31]. Endpoint PCR was performed on the samples that were positive for C. psittaci real-time PCR for the partial amplification of the ompA gene (approximately 1050 bp) [24]. Positive and negative control samples were included in each PCR reaction. The endpoint PCR products were analyzed using electrophoresis in 2% agarose gels stained with Syber Safe DNA gel Stain (Invitrogen, Waltham, MA, USA) and a 100 bp DNA Ladder (Genbiotech, Buenos Aires, Argentina) as a marker, and they were visualized under ultraviolet light. PCR products were purified with DNA Clean & Concentrator (Zymo Research, Tustin, CA, USA) and were sent for sequencing using Sanger technology.
For the alignment and obtaining of the consensus sequences, the SeqAssem program was used. Consensus sequences were compared using BLASTn 2.2.19 with other fragments of the Chlamydia spp. ompA gene obtained from GenBank. The dendrogram was constructed using the Tree Explorer module of the MEGA7 program, employing the Neighbor-joining method with the p-distance parameter. Statistical support was determined through nonparametric bootstrapping with 1000 pseudo-replications [32].
The scheme developed by Pannekoek et al. [33] targeting seven housekeeping genes (gatA, oppA, hflX, gidA, enoA, hemN, and fumC) was used for Multi-Locus Sequence Typing (MLST) genotyping. Genes were amplified and sequenced using primers for the C. psittaci scheme available on the Chlamydiales PUBMLST database (http://pubmist.org/chlamidiales, accessed on 26 April 2024) and following the protocols described by Jelocnik et al. [34] and Mattman et al. [25]. The PCR products’ visualization, purification, and sequencing were performed as describe above for endpoint PCR. The SeqAssem program was used for the alignment and obtaining of the consensus sequences. The allele designation for each aligned sequence and the allelic profile to determine sequence type (ST) was obtained using the “sequence query” and “Search by combination of allele” options, respectively, from the Chlamydiales PubMLST database (http://pubmist.org/chlamidiales, accessed on 26 April 2024) [34]. The MLST sequences generated were deposited in the previously mentioned database.

3. Results

3.1. Necropsy

Grossly, all birds were in poor nutritional condition, with no fat reserves, prominent keels, and atrophy of the pectoral muscles. The liver of all birds, with the exception of chick blue-fronted parrots, was enlarged and the surface cross section had multifocal, irregular, variably sized, tan-to-yellow, non-raised areas of discoloration. In both macaws, the spleen was enlarged, pale, and mottled by multiple dark red foci. The pericardium of P 3 was opaque and the pericardial sac was filled with a moderate amount of yellow liquid (0.5–1 mL). The coelomic cavity of P2 was filled with 2 to 3 mL of a cloudy yellow liquid. In addition, congested and edematous lungs were observed in all birds (Figure 1).

3.2. Microbiology

Cultures for Trichomonas sp. and fungi were negative in all birds tested.
Pseudomonas aeruginosa was isolated and identified from all the samples of P1, lung of P2, and lung of C1. Salmonella sp. was isolated from the liver of both blue-fronted parrot chicks, as well as from the laryngeal swab of C2. In addition to Pseudomonas and Salmonella isolated from the samples of both nestlings, Escherichia coli, Klebsiella sp., and Proteus sp. were also isolated.

3.3. Histology

The main microscopic findings are summarized in Table 1. Hepatitis, splenitis, air sacculitis, pulmonary congestion, and edema were the main findings in all animals examined (Figure 2). In addition, nephritis and pneumonia were found in C1 and green-and-red macaws.
Figure 2 shows the microscopic and immunohistochemical findings.

3.4. Immunohistochemistry

IHC for Chlamydia pecorum cross-reacting with Chlamydia spp. was positive on the liver, kidney, and spleen sections of two juvenile blue-fronted parrots (P2 and P4) and the scarlet macaw tested.

3.5. Chlamydia Isolation

Isolation was obtained only from samples from the scarlet macaw that had been adequately preserved prior to processing. Identification of the strain was carried out with the specific real-time PCR of C. psittaci.

3.6. Polymerase Chain Reaction

From swab samples of cloaca, choanae, and conjunctiva, as well as of the pooled organ samples (liver, spleen, and lung) of juvenile blue-fronted parrots (P2, P3, and P4) and blue-fronted parrot chicks (C1 and C2), bacterial nucleic acid belonging to C. psittaci DNA was detected. In conjunctiva, choanae, and cloacal swab samples, as well as liver, spleen, and lung pool samples of both macaws, bacterial nucleic acid belonging to C. psittaci was detected. No positive results were shown for Psittacine Herpesvirus or Avian Polyomavirus in the tested samples.
The partial ompA gene could be amplified and sequenced directly from the clinical specimens obtained from the swab samples of cloaca, choanae, and conjunctiva of juvenile blue-fronted parrots (P2 and P3), blue-fronted parrot chicks (C1 and C2), pooled organ samples (liver, spleen, and lung) from juvenile blue-fronted parrots (P2–P4), blue-fronted parrot chicks (C1 and C2), and of the isolate obtained from the scarlet macaw. All sequences obtained from the blue-fronted parrots were identical to each other and were grouped together with the genotype A reference strains. Moreover, the sequence obtained from the scarlet macaw (Genbank accession number PP706175) was similar to the reference genotype B sequence (Figure 3).
The MLST analysis was carried out only on the isolated strain from the scarlet macaw and this showed that it corresponded to ST27.
The mucosal swab samples obtained after the treatment of two juvenile blue-fronted parrots gave a negative result.

4. Discussion

Chlamydia psittaci is an important zoonotic pathogen that could infect a wide range of hosts, although the most common hosts of C. psittaci are birds, especially parrots. Our results are consistent with findings reported by other authors [12,35,36].
After reviewing the literature available from PubMed, Google Scholar, and Web of Science, we could not find any reports of an outbreak of avian chlamydiosis in a commercial psittacine breeding aviary in Argentina. However, many publications indicate the detection of C. psittaci in various provinces of Argentina, in both bird and human samples [37,38,39,40].
C. psittaci transmission occurs primarily by close contact when the agent is shed in nasal discharges and feces. fecal shedding is intermittent and it is exacerbated when animals are stressed. During natural infection, virulence of the strain, infection dose, and host immune status greatly affect bacterial excretion [41].
In all of the Chlamydia-positive birds included in this study, clinical signs compatible with chlamydiosis [41,42] were observed and submitted to necropsy. All birds analyzed were emaciated which, along with the presence of hepatic and splenic lesions, is consistent with chlamydiosis, as previously reported [2,42,43]. For the differential diagnosis, psittacid Herpesvirus 1 (Pacheco´s parrot disease) and Avian Polyomavirus infection should be considered. In this study, PCR for both diseases was negative in all birds analyzed. Regarding the isolation of Salmonella and Pseudomona aeruginosa, we believe that the co-infection may have contributed to increased mortality. However, only 37% of the birds analyzed had a co-infection, which suggests that Chlamydia infection can produce a high mortality rate on its own.
The presumptive diagnosis of chlamydiosis is based on gross and microscopic lesions and confirmed using PCR [44,45] and/or immunohistochemistry. The isolation of C. psittaci from a clinical specimen is a very good confirmatory test but is not frequently performed by veterinary diagnostic laboratories because of its technical difficulty [46]. In addition, culturing is hampered by limited sensitivity, previous antibiotic use, and strict biosafety regulations [47]. We were able to perform the isolation from a pooled sample of organs (liver, spleen, and lung) of a scarlet macaw that was refrigerated in SPG medium. Unfortunately, the rest of the samples were frozen at −20 °C without preservation medium prior to being sent to the laboratory, so their culture was not performed. From the isolation, it was possible to obtain DNA in good concentration and quality, which allowed the MLST characterization of the strain.
Using the genotyping strategy based on the ompA gene, we were able to determine that two genotypes were involved in the outbreak studied by us. We were able to determine that the samples from blue-fronted parrots had genotype A. This genotype is often involved in outbreaks of avian chlamydiosis with high mortality in psittacines [2], as was in our case. On the other hand, we found that genotype B in the scarlet macaw was associated with severe clinical signs and lesions. Previously, this genotype was found in an African gray parrot (Psittacus erithacus erithacus) kept as a pet that presented avian chlamydiosis [27]. Furthermore, in Argentina, the circulation of these two genotypes has been confirmed in previous studies [38,48].
Strains of C. psittaci had not been previously characterized using MLST in Argentina; in our study, we were able to determine that the strain isolated from the scarlet macaw corresponded to ST27. This ST has been found in pigeons, passerines, parrots, penguins, and horses in North America, Europe, Australia, and New Zealand. In Australia, it has been associated with abortions in horses, which provides evidence of potential host-switching and spillover between different hosts of this ST [24,25,49].
The epidemiological characterization of animal and human samples provides information on prevalent genotypes in human infections, the likely avian sources, and aids in surveillance and outbreak management [47]. Psittaciformes are a major source of the Chlamydia responsible for psittacosis in humans [36]. Human psittacosis ranges from mild to severe systemic illness, with genotype A considered to be the most virulent and closely associated with human cases and psittacine birds [48,50]. However, genotype B, mainly associated with pigeons, also poses zoonotic transmission risks [36,47]. There is little information in Argentina demonstrating the genotypes involved in cases of zoonotic transmission. Cadario et al. [48] found genotype A in two cases of human psittacosis and managed to determine the same genotype in the psittacidae considered as the source of infection.
During the outbreak, a worker displayed symptoms suggestive of psittacosis, but amidst the COVID-19 pandemic, was diagnosed with SARS-CoV-2 based solely on clinical presentation without confirmatory tests. The clinical features of SARS-CoV-2 and C. psittaci infection can overlap, posing challenges for accurate differential diagnosis [51,52]. Co-infections of SARS-CoV-2 and Chlamydia spp. have also been documented [51,52]. The worker was eventually treated with doxycycline after an X-ray showed a lung lesion compatible with bacterial pneumonia, but unfortunately no samples were obtained to make a confirmatory diagnosis.
Although the exact point of entry of Chlamydia into the aviary remains unknown, we speculate that the source of infection was the introduction of a different sick bird, which could explain the independent introduction of the pathogens, as the aviary also serves as a rescue center for confiscated birds. The proximity of the breeding and quarantine areas may have increased the risk of transmission. Additionally, we speculate that the detection of both genotypes, genotype A and B, may be due to the birds contracting the infection when the aviary was located at its previous facilities, where the cages were kept outdoors. The area where the aviary is located is a rural and rugged area with an abundant presence of wildlife. Among the wild birds, the monk parakeet (Myiopsitta monachus), the burrowing parrot (Cyanoliseus patagonus) and various species of pigeons (Columba livia, Zenaida auriculata, Patagioenas picazuro), in addition to a wide variety of passerines, are very abundant in the place, and free-living wild birds can act as reservoirs and shedders of C. psittaci and other chlamydial agents [17,24,25]. It is well known that the main sources of C. psittaci genotype A are psittacidae, and that of genotype B are pigeons. Furthermore, passerines are also recognized as possible hosts for both genotypes [2].
Regarding treatment, oxytetracycline was administered orally through food. Although we are aware of the disadvantages of this route of administration, it was chosen to reduce the stress associated with individual parenteral treatments. The effectiveness of using oxytetracycline as the antibiotic of choice during the outbreak has been previously demonstrated by other authors [53,54]. Administering drugs through food or drinking water reduces the stress associated with individual handling and injections, which can be particularly challenging and distressing for birds, but it is important to remember that acutely ill birds that stop eating and drinking are less likely to recover.

5. Conclusions

The present study is the first report of an outbreak of chlamydiosis in a commercial psittacine breeding aviary from Buenos Aires, Argentina. The pathogen, predominantly affecting birds, particularly parrots, demonstrates zoonotic potential. Zoonotic transmission, particularly involving genotypes A and B, emphasizes the importance of surveillance and diagnostic efforts. Understanding the prevalent genotypes in regions like Argentina is crucial for effective public health interventions and outbreak management. In conclusion, this analysis highlights the complex epidemiology of C. psittaci infections, emphasizing the critical role of surveillance, genotype characterization, and differential diagnosis in mitigating the disease’s impact on both avian and human health.

Author Contributions

M.B.R. performed the necropsy, obtained the samples, and contributed to the conception and design of the study; they contributed to the acquisition, analysis, and interpretation of the data; drafted the manuscript; critically revised the manuscript; gave final approval; and agreed to be accountable for all aspects of the work in ensuring that questions relating to the accuracy or integrity of any part of the work are appropriately investigated and resolved. J.P.G. performed the necropsy and histopathology study; contributed to the conception and design of the study; critically revised the manuscript; and gave final approval. M.L.C., J.C. and C.C. conducted microbiological studies and contributed to the acquisition, analysis, and interpretation of data, and critically revised the manuscript. J.A.O. and M.E.C. conducted the isolation procedures, molecular tests, and sequencing; critically revised the manuscript; and gave final approval. F.A.U. contributed to the Gimenez stain and immunohistochemistry; critically revised the manuscript; and gave final approval. S.S.D. served as a consultant on this case and contributed to drafting and critically revising the manuscript before submission. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The sequences generated in this study are available in GenBank under Accession Numbers PP751030, PP751031, PP751032, PP751033, and PP706175. The MLST sequences generated in this study are deposited under the ID 5164 in PubMLST/Chlamydiales (https://pubmlst.org/chlamydiales/, accessed on 10 May 2024 and 26 April 2024 respectively).

Acknowledgments

The authors would like to sincerely acknowledge Nicolás Porfilio and Rubén Aranda for enabling us to conduct the study and publish the results.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Stokes, H.S.; Berg, M.L.; Bennett, A.T.D. A Review of Chlamydial infections in wild birds. Pathogens 2021, 10, 948. [Google Scholar] [CrossRef] [PubMed]
  2. Ravichandran, K.; Anbazhagan, S.; Karthik, K.; Angappan, M.; Dhayananth, B. A comprehensive review on avian Chlamydiosis: A neglected zoonotic disease. Trop. Anim. Health Prod. 2021, 53, 414. [Google Scholar] [CrossRef] [PubMed]
  3. Jenkins, C.; Jelocnik, M.; Onizawa, E.; McNally, J.; Coilparampil, R.; Pinczowski, P.; Bogema, D.; Westermann, T. Chlamydia pecorum ovine abortion: Associations between maternal infection and perinatal mortality. Pathogens 2021, 10, 1367. [Google Scholar] [CrossRef] [PubMed]
  4. Marti, H.; Jelocnik, M. Animal Chlamydiae: A concern for human and veterinary medicine. Pathogens 2022, 11, 364. [Google Scholar] [CrossRef] [PubMed]
  5. Vanrompay, D.; Harkinezhad, T.; van de Walle, M.; Beeckman, D.; van Droogenbroeck, C.; Verminnen, K.; Leten, R.; Martel, A.; Cauwerts, K. Chlamydophila psittaci transmission from pet birds to humans. Emerg. Infect. Dis. 2007, 13, 1108–1110. [Google Scholar] [CrossRef] [PubMed]
  6. Branley, J.M.; Weston, K.M.; England, J.; Dwyer, D.E.; Sorrell, T.C. Clinical features of endemic community-acquired Psittacosis. New Microbes New Infect. 2014, 2, 7–12. [Google Scholar] [CrossRef] [PubMed]
  7. Telfer, B.L.; Moberley, S.A.; Hort, K.P.; Branley, J.M.; Dwyer, D.E.; Muscatello, D.J.; Correll, P.K.; England, J.; McAnulty, J.M. Probable Psittacosis outbreak linked to wild birds. Emerg. Infect. Dis. 2005, 11, 391–397. [Google Scholar] [CrossRef] [PubMed]
  8. Balsamo, G.; Maxted, A.M.; Midla, J.W.; Murphy, J.M.; Wohrle, R.; Edling, T.M.; Fish, P.H.; Flammer, K.; Hyde, D.; Kutty, P.K.; et al. Compendium of measures to control Chlamydia psittaci infection among humans (Psittacosis) and pet birds (Avian Chlamydiosis), 2017. J. Avian Med. Surg. 2017, 31, 262–282. [Google Scholar] [CrossRef] [PubMed]
  9. Dai, N.; Li, Q.; Geng, J.; Guo, W.; Yan, W. Severe pneumonia caused by Chlamydia psittaci: Report of two cases and literature review. J. Infect. Dev. Ctries. 2022, 16, 1101–1112. [Google Scholar] [CrossRef]
  10. Kalmar, I.D.; Dicxk, V.; Dossche, L.; Vanrompay, D. Zoonotic infection with Chlamydia psittaci at an avian refuge centre. Vet. J. 2014, 199, 300–302. [Google Scholar] [CrossRef]
  11. Szymańska-Czerwińska, M.; Niemczuk, K. Avian Chlamydiosis zoonotic disease. Vector-Borne Zoonotic Dis. 2016, 16, 1–3. [Google Scholar] [CrossRef]
  12. Kaleta, E.F.; Taday, E.M.A. Avian host range of Chlamydophila spp. based on isolation, antigen detection and serology. Avian Pathol. J. WVPA 2003, 32, 435–461. [Google Scholar] [CrossRef] [PubMed]
  13. Markey, B.K. Clinical Veterinary Microbiology, 2nd ed.; Elsevier: Edinburgh, Scotland, 2013; ISBN 978-0-7234-3237-1. [Google Scholar]
  14. Suwa, T.; Touchi, A.; Hirai, K.; Itakura, C. Pathological studies on Chlamydiosis in parakeets (Psittacula krameri manillensis). Avian Pathol. J. WVPA 1990, 19, 355–369. [Google Scholar] [CrossRef] [PubMed]
  15. Vanrompay, D.; Ducatelle, R.; Haesbrouck, F. Pathology of experimental chlamydiosis in turkeys. Vlaams Diergeneeskd. Tijdschr. 1995, 64, 19–24. [Google Scholar]
  16. Van Buuren, C.E.; Dorrestein, G.M.; Van Dijk, J.E. Chlamydia psittaci infections in birds: A review on the pathogenesis and histopathological features. Vet. Q. 1994, 16, 38–41. [Google Scholar] [CrossRef] [PubMed]
  17. Burt, S.A.; Röring, R.E.; Heijne, M. Chlamydia psittaci and C. avium in feral pigeon (Columba livia domestica) droppings in two cities in the Netherlands. Vet. Q. 2018, 38, 63–66. [Google Scholar] [CrossRef] [PubMed]
  18. Kowalczyk, K.; Wójcik-Fatla, A. Chlamydia psittaci in faecal samples of feral pigeons (Columba livia forma urbana) in urban areas of Lublin City, Poland. Curr. Microbiol. 2022, 79, 367. [Google Scholar] [CrossRef] [PubMed]
  19. Perez-Sancho, M.; García-Seco, T.; Porrero, C.; García, N.; Gomez-Barrero, S.; Cámara, J.M.; Domínguez, L.; Álvarez, J. A Ten-year-surveillance program of zoonotic pathogens in feral pigeons in the city of Madrid (2005–2014): The importance of a systematic pest control. Res. Vet. Sci. 2020, 128, 293–298. [Google Scholar] [CrossRef] [PubMed]
  20. Sukon, P.; Nam, N.H.; Kittipreeya, P.; Sara-In, A.; Wawilai, P.; Inchuai, R.; Weerakhun, S. Global Prevalence of Chlamydial Infections in Birds: A systematic review and meta-analysis. Prev. Vet. Med. 2021, 192, 105370. [Google Scholar] [CrossRef] [PubMed]
  21. Chen, X.; Cao, K.; Wei, Y.; Qian, Y.; Liang, J.; Dong, D.; Tang, J.; Zhu, Z.; Gu, Q.; Yu, W. Metagenomic next-generation sequencing in the diagnosis of severe pneumonias caused by Chlamydia psittaci. Infection 2020, 48, 535–542. [Google Scholar] [CrossRef]
  22. Rybarczyk, J.; Versteele, C.; Lernout, T.; Vanrompay, D. Human psittacosis: A review with emphasis on surveillance in Belgium. Acta Clin. Belg. 2020, 75, 42–48. [Google Scholar] [CrossRef] [PubMed]
  23. Villagra, S.M. Brote de psitacosis durante la pandemia de COVID-19 en la provincia de santa Fe. Serie de casoS. Rev. Argent. Med. 2022, 10, 139–142. [Google Scholar]
  24. Szymańska-Czerwińska, M.; Mitura, A.; Niemczuk, K.; Zaręba, K.; Jodełko, A.; Pluta, A.; Scharf, S.; Vitek, B.; Aaziz, R.; Vorimore, F.; et al. Dissemination and genetic diversity of Chlamydial agents in polish wildfowl: Isolation and molecular characterisation of avian Chlamydia abortus strains. PLoS ONE 2017, 12, e0174599. [Google Scholar] [CrossRef] [PubMed]
  25. Mattmann, P.; Marti, H.; Borel, N.; Jelocnik, M.; Albini, S.; Vogler, B.R. Chlamydiaceae in wild, feral and domestic pigeons in Switzerland and insight into population dynamics by Chlamydia psittaci multilocus sequence typing. PLoS ONE 2019, 14, e0226088. [Google Scholar] [CrossRef] [PubMed]
  26. Origlia, J.A.; Cadario, M.E.; Frutos, M.C.; Lopez, N.F.; Corva, S.; Unzaga, M.F.; Piscopo, M.V.; Cuffini, C.; Petruccelli, M.A. Detection and molecular characterization of Chlamydia psittaci and Chlamydia abortus in psittacine pet birds in Buenos Aires Province, Argentina. Rev. Argent. Microbiol. 2019, 51, 130–135. [Google Scholar] [CrossRef] [PubMed]
  27. Harkinezhad, T.; Verminnen, K.; Van Droogenbroeck, C.; Vanrompay, D. Chlamydophila psittaci Genotype E/B transmission from african grey parrots to humans. J. Med. Microbiol. 2007, 56, 1097–1100. [Google Scholar] [CrossRef] [PubMed]
  28. Barrow, G.I.; Feltham, R.K.A. Cowan and Steel’s Manual for the Identification of Medical Bacteria, 3rd ed.; Cambridge University Press: Cambridge, UK, 1993; ISBN 978-0-521-54328-6. [Google Scholar]
  29. Laroucau, K.; Vorimore, F.; Aaziz, R.; Solmonson, L.; Hsia, R.C.; Bavoil, P.M.; Fach, P.; Hölzer, M.; Wuenschmann, A.; Sachse, K. Chlamydia buteonis, a new Chlamydia species isolated from a red-shouldered hawk. Syst. Appl. Microbiol. 2019, 42, 125997. [Google Scholar] [CrossRef]
  30. Pantchev, A.; Sting, R.; Bauerfeind, R.; Tyczka, J.; Sachse, K. New Real-Time PCR Tests for species-specific detection of Chlamydophila psittaci and Chlamydophila abortus from tissue samples. Vet. J. 2009, 181, 145–150. [Google Scholar] [CrossRef] [PubMed]
  31. Katoh, H.; Ohya, K.; Fukushi, H. Development of Novel Real-Time PCR Assays for detecting DNA virus infections in Psittaciform birds. J. Virol. Methods 2008, 154, 92–98. [Google Scholar] [CrossRef]
  32. Kumar, S.; Stecher, G.; Tamura, K. MEGA7: Molecular evolutionary genetics analysis version 7.0 for Bigger Datasets. Mol. Biol. Evol. 2016, 33, 1870–1874. [Google Scholar] [CrossRef]
  33. Pannekoek, Y.; Dickx, V.; Beeckman, D.S.A.; Jolley, K.A.; Keijzers, W.C.; Vretou, E.; Maiden, M.C.J.; Vanrompay, D.; van der Ende, A. Multi locus sequence typing of Chlamydia reveals an association between Chlamydia psittaci genotypes and host species. PLoS ONE 2010, 5, e14179. [Google Scholar] [CrossRef] [PubMed]
  34. Jelocnik, M.; Polkinghorne, A.; Pannekoek, Y. Multilocus Sequence typing (MLST) of Chlamydiales. Methods Mol. Biol. 2019, 2042, 69–86. [Google Scholar] [CrossRef]
  35. Hopkins, J.; Alsop, J.; Varga, C.; Pasma, T.; Jekel, P.; Rishi, L.; Hirji, M.; Filejski, C. Investigation and management of Psittacosis in a public aviary: A One Health approach. Can. Commun. Dis. Rep. 2016, 42, 112–116. [Google Scholar] [CrossRef] [PubMed]
  36. Hogerwerf, L.; Roof, I.; de Jong, M.J.K.; Dijkstra, F.; van der Hoek, W. Animal sources for zoonotic transmission of Psittacosis: A systematic review. BMC Infect. Dis. 2020, 20, 192. [Google Scholar] [CrossRef]
  37. Maza, Y.; Chaparro, M.; Argañaráz, C.; Genero, S. Brote de Psitacosis en la localidad de Fontana (Chaco, Argentina) Durante Enero de 2014. Rev. Vet. 2016, 27, 45–47. [Google Scholar] [CrossRef]
  38. Mariño, B.; Sciabarrasi, A.; Imhoberdoff, P.; Talamé, M.P.; Frutos, M.C.; Mosmann, J.; Cuffini, C. Detección de Chlamydia psittaci en muestras de pichones de loro hablador (Amazona aestiva) incautadas en Santa Fe, Argentina. Rev. Fac. Cienc. Médicas Córdoba 2022, 79. Available online: https://revistas.unc.edu.ar/index.php/med/article/view/39023 (accessed on 30 May 2024).
  39. de Chazal, L.E. Brote de psitacosis en San Miguel de Tucumán y Gran San Miguel. Rev. Med. Tucumán 2007, 12, 30–34. [Google Scholar]
  40. Frutos, M.C.; Monetti, M.; Kiguen, X.; Venezuela, F.; Ré, V.; Cuffini, C. Genotyping of C. psittaci in central area of Argentina. Diagn. Microbiol. Infect. Dis. 2012, 74, 320–322. [Google Scholar] [CrossRef]
  41. Beeckman, D.S.A.; Vanrompay, D.C.G. Zoonotic Chlamydophila psittaci infections from a clinical perspective. Clin. Microbiol. Infect. 2009, 15, 11–17. [Google Scholar] [CrossRef] [PubMed]
  42. Andersen, A.A.; Vanrompay, D. Avian Chlamydiosis. In Diseases of Poultry; Blackwell: Hoboken, NJ, USA, 2008; pp. 971–986. ISBN 978-0-8138-0718-8. [Google Scholar]
  43. Muroni, G.; Pinna, L.; Serra, E.; Chisu, V.; Mandas, D.; Coccollone, A.; Liciardi, M.; Masala, G. A Chlamydia psittaci outbreak in Psittacine birds in Sardinia, Italy. Int. J. Environ. Res. Public. Health 2022, 19, 14204. [Google Scholar] [CrossRef]
  44. Vanrompay, D.; Butaye, P. Characterization of avian Chlamydia psittaci strains using Ompl restriction mapping and serovar-specific monoclonal antibodies. Res. Microbiol. 1997, 148, 327–333. [Google Scholar] [CrossRef] [PubMed]
  45. Ehricht, R.; Slickers, P.; Goellner, S.; Hotzel, H.; Sachse, K. Optimized DNA Microarray assay allows detection and genotyping of single PCR-amplifiable target copies. Mol. Cell. Probes 2006, 20, 60–63. [Google Scholar] [CrossRef] [PubMed]
  46. Smith, K.A.; Bradley, K.K.; Stobierski, M.G.; Tengelsen, L.A. Compendium of measures to control Chlamydophila psittaci (Formerly Chlamydia psittaci) infection among humans (Psittacosis) and pet birds, 2005. J. Am. Vet. Med. Assoc. 2005, 226, 532–539. [Google Scholar] [CrossRef] [PubMed]
  47. Heddema, E.R.; Van Hannen, E.J.; Bongaerts, M.; Dijkstra, F.; Ten Hove, R.J.; De Wever, B.; Vanrompay, D. Typing of Chlamydia psittaci to monitor epidemiology of Psittacosis and aid disease control in the Netherlands, 2008 to 2013. Eurosurveillance 2015, 20, 21026. [Google Scholar] [CrossRef] [PubMed]
  48. Cadario, M.E.; Frutos, M.C.; Arias, M.B.; Origlia, J.A.; Zelaya, V.; Madariaga, M.J.; Lara, C.S.; Ré, V.; Cuffini, C.G. Epidemiological and molecular characteristics of Chlamydia psittaci from 8 human cases of Psittacosis and 4 related birds in Argentina. Rev. Argent. Microbiol. 2017, 49, 323–327. [Google Scholar] [CrossRef] [PubMed]
  49. Kasimov, V.; White, R.T.; Foxwell, J.; Jenkins, C.; Gedye, K.; Pannekoek, Y.; Jelocnik, M. Whole-Genome sequencing of Chlamydia psittaci from Australasian avian hosts: A Genomics Approach to a Pathogen That Still Ruffles Feathers. Microb. Genom. 2023, 9, 001072. [Google Scholar] [CrossRef] [PubMed]
  50. Harkinezhad, T.; Geens, T.; Vanrompay, D. Chlamydophila psittaci infections in birds: A Review with emphasis on zoonotic consequences. Vet. Microbiol. 2009, 135, 68–77. [Google Scholar] [CrossRef] [PubMed]
  51. Yin, Q.; Li, Y.; Pan, H.; Hui, T.; Yu, Z.; Wu, H.; Zhang, D.; Zheng, W.; Wang, S.; Zhou, Z.; et al. Atypical pneumonia caused by Chlamydia psittaci during the COVID-19 pandemic. Int. J. Infect. Dis. 2022, 122, 622–627. [Google Scholar] [CrossRef] [PubMed]
  52. Zhang, A.; Liang, J.; Lao, X.; Xia, X.; Liang, J. Pneumonia caused by Chlamydia psittaci and SARS-cov-2 Coinfection Diagnosed using metagenomic next-generation sequencing: A case report. Int. Med. Case Rep. J. 2024, 17, 187–194. [Google Scholar] [CrossRef]
  53. Bommana, S.; Polkinghorne, A. Mini Review: Antimicrobial control of Chlamydial infections in animals: Current practices and issues. Front. Microbiol. 2019, 10, 113. [Google Scholar] [CrossRef]
  54. Prohl, A.; Lohr, M.; Ostermann, C.; Liebler-Tenorio, E.; Berndt, A.; Schroedl, W.; Rothe, M.; Schubert, E.; Sachse, K.; Reinhold, P. Enrofloxacin and macrolides alone or in combination with rifampicin as antimicrobial treatment in a bovine model of acute Chlamydia psittaci infection. PLoS ONE 2015, 10, e0119736. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Post-mortem lesions of organs collected from birds affected by Chlamydia psittaci in a commercial psittacine breeding aviary in Argentina. (A) Marbled surface of the liver with white, irregular, yellowish flat spots of variable size. Juvenile blue-fronted parrot. (B) Marbled cross section of the liver from a red-and-green macaw. (C) Enlarged, pale, and mottled spleen from scarlet macaw. (D) The lung is edematous and moderately congested.
Figure 1. Post-mortem lesions of organs collected from birds affected by Chlamydia psittaci in a commercial psittacine breeding aviary in Argentina. (A) Marbled surface of the liver with white, irregular, yellowish flat spots of variable size. Juvenile blue-fronted parrot. (B) Marbled cross section of the liver from a red-and-green macaw. (C) Enlarged, pale, and mottled spleen from scarlet macaw. (D) The lung is edematous and moderately congested.
Animals 14 01959 g001
Figure 2. Microscopic lesions of organs collected from birds affected by Chlamydia psittaci in a commercial psittacine breeding aviary in Argentina (A) Air sac: Lymphoplasmacytic and necrotizing multifocal airsacculitis 20X H&E. (B) Liver: Intracytoplasmic elementary bodies in the cytoplasm of hepatocytes and mononuclear cells (black arrows) 40X H&E. (C) Liver: elementary bodies in the cytoplasm of hepatocytes, Gimenez stain (Black arrows). (D) Positive Chlamydia spp. IHC on liver.
Figure 2. Microscopic lesions of organs collected from birds affected by Chlamydia psittaci in a commercial psittacine breeding aviary in Argentina (A) Air sac: Lymphoplasmacytic and necrotizing multifocal airsacculitis 20X H&E. (B) Liver: Intracytoplasmic elementary bodies in the cytoplasm of hepatocytes and mononuclear cells (black arrows) 40X H&E. (C) Liver: elementary bodies in the cytoplasm of hepatocytes, Gimenez stain (Black arrows). (D) Positive Chlamydia spp. IHC on liver.
Animals 14 01959 g002
Figure 3. Neighbor-joining dendrogram based on comparison of the ompA gene fragment (about 1030 bp) of Chlamydia psittaci. Representative sequences from the different C. psittaci genotypes were included. Samples belonging to this study were pooled organs from juvenile blue-fronted parrots (P2, P3, and P4), pooled trimucosal swabs from blue-fronted parrot chicks (C1 and C2), pooled trimucosal swabs from juvenile blue-fronted parrots (P2 and P3), pooled organs from blue-fronted parrot chicks (C1 and C2), and pooled organs from a scarlet macaw. The GenBank accession numbers were PP751030, PP751031, PP751032, PP751033, and PP706175, respectively. The evolutionary distances were computed using the Kimura 2-parameter method and are in the units of the number of base substitutions per site. Numbers above branches are Bootstrap values as a percentage of 1000 pseudo replicates. C. caviae GPIC was used as an outgroup. The scale bar shows the sequence diversity percentage [32].
Figure 3. Neighbor-joining dendrogram based on comparison of the ompA gene fragment (about 1030 bp) of Chlamydia psittaci. Representative sequences from the different C. psittaci genotypes were included. Samples belonging to this study were pooled organs from juvenile blue-fronted parrots (P2, P3, and P4), pooled trimucosal swabs from blue-fronted parrot chicks (C1 and C2), pooled trimucosal swabs from juvenile blue-fronted parrots (P2 and P3), pooled organs from blue-fronted parrot chicks (C1 and C2), and pooled organs from a scarlet macaw. The GenBank accession numbers were PP751030, PP751031, PP751032, PP751033, and PP706175, respectively. The evolutionary distances were computed using the Kimura 2-parameter method and are in the units of the number of base substitutions per site. Numbers above branches are Bootstrap values as a percentage of 1000 pseudo replicates. C. caviae GPIC was used as an outgroup. The scale bar shows the sequence diversity percentage [32].
Animals 14 01959 g003
Table 1. Histology results for samples collected from parrots, chicks, and macaws.
Table 1. Histology results for samples collected from parrots, chicks, and macaws.
Lesion ObservedP1P2P3P4C 1C 2Green-and-Red MacawScarlet Macaw
Multifocal lymphoplasmacytic, heterophilic, and necrotizing splenitis with intracytoplasmic elementary bodies
YesYesYesYesYesYesYesYes
Pulmonary congestion with edemaYesYesYesYesYesYesYesYes
Multifocal, lymphoplasmacytic, and necrotizing hepatitis with intracytoplasmic elementary bodies
NoYesNoNoYesNoNoYes
Multifocal necro-heterophilic hepatitisNoNoNoYesNoYesNoNo
Multifocal lymphoplasmacytic, heterophilic airsacculitisYesYesNoNoNoYesYesYes
Multifocal lymphoplasmacytic nephritisNoNoNoNoNoNoYesNo
Heterophilic pneumonia NoNoNoNoYesNoNoNo
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Riccio, M.B.; García, J.P.; Chiapparrone, M.L.; Cantón, J.; Cacciato, C.; Origlia, J.A.; Cadario, M.E.; Diab, S.S.; Uzal, F.A. Outbreak of Chlamydia psittaci Infection in a Commercial Psittacine Breeding Aviary in Argentina. Animals 2024, 14, 1959. https://doi.org/10.3390/ani14131959

AMA Style

Riccio MB, García JP, Chiapparrone ML, Cantón J, Cacciato C, Origlia JA, Cadario ME, Diab SS, Uzal FA. Outbreak of Chlamydia psittaci Infection in a Commercial Psittacine Breeding Aviary in Argentina. Animals. 2024; 14(13):1959. https://doi.org/10.3390/ani14131959

Chicago/Turabian Style

Riccio, María Belén, Jorge Pablo García, María Laura Chiapparrone, Juliana Cantón, Claudio Cacciato, Javier Anibal Origlia, María Estela Cadario, Santiago Sain Diab, and Francisco Alejandro Uzal. 2024. "Outbreak of Chlamydia psittaci Infection in a Commercial Psittacine Breeding Aviary in Argentina" Animals 14, no. 13: 1959. https://doi.org/10.3390/ani14131959

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop