1,593
Views
0
CrossRef citations to date
0
Altmetric
Research Article

First detection of Omicron variant BA.4.1 lineage in dogs, Chile

ORCID Icon, , , ORCID Icon, , & ORCID Icon show all
Pages 1-10 | Received 09 Aug 2023, Accepted 16 Dec 2023, Published online: 04 Jan 2024

Abstract

SARS-CoV-2’s rapid global spread caused the declaration of COVID-19 as a pandemic in March 2020. Alongside humans, domestic dogs and cats are also susceptible to infection. However, limited reports on pet infections in Chile prompted a comprehensive study to address this knowledge gap. Between March 2021 and March 2023, the study assessed 65 pets (26 dogs and 39 cats) from 33 COVID-19+ households alongside 700 nasal swabs from animals in households with unknown COVID-19 status. Using RT-PCR, nasal, fecal, and environmental samples were analyzed for the virus. In COVID-19+ households, 6.06% tested positive for SARS-CoV-2, belonging to 3 dogs, indicating human-to-pet transmission. Pets from households with unknown COVID-19 status tested negative for the virus. We obtained 2 SARS-CoV-2 genomes from animals, that belonged to Omicron BA.4.1 variant, marking the first report of pets infected with this lineage globally. Phylogenetic analysis showed these sequences clustered with human sequences collected in Chile during the same period when the BA.4.1 variant was prevalent in the country. The prevalence of SARS-CoV-2 in Chilean pets was relatively low, likely due to the country’s high human vaccination rate. Our study highlights the importance of upholding and strengthening human vaccination strategies to mitigate the risk of interspecies transmission. It underscores the critical role of the One Health approach in addressing emerging zoonotic diseases, calling for further research on infection dynamics and risk factors for a comprehensive understanding.

Introduction

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the COVID-19 pandemic. This enveloped, single-stranded, positive polarity RNA virus belongs to the Coronaviridae family, subfamily Coronavirinae, genus Betacoronavirus (Gorbalenya et al. Citation2020). Notably, this classification is shared with SARS-CoV-1 and MERS coronavirus, both of which have zoonotic origins (Lee and Hsueh Citation2020). The genome of SARS-CoV-2 is approximately 30 kb in size, encoding various structural and non-structural proteins, with each playing a distinct role in viral infectivity and transmission. The structural proteins include spike (S), envelope (E), membrane (M), and nucleocapsid (N) proteins (Durmaz et al. Citation2020; Mittal et al. Citation2020; Almehdi et al. Citation2021).

The S protein is a transmembrane glycoprotein, serves as a primary target for neutralizing antibodies due to its receptor-binding function via the receptor-binding domain (RBD) (Astuti and Ysrafil Citation2020; Luan et al. Citation2020). On the other hand, the N protein, highly immunogenic and abundantly expressed during infection, plays a vital role in RNA transcription, replication, and packaging of the viral genome into virions, while the M and E proteins are essential for virus assembly (Siracusano et al. Citation2020).

SARS-CoV-2 has demonstrated the ability to infect at least 18 different animal species, encompassing companion animals, captive, farmed, and wild animals (Cui et al. Citation2022). Among these, the list includes pets such as dogs, cats, ferrets, and hamsters, captive animals including tigers, lions, snow leopards, cougars, lynxes, fishing cats, binturongs, hyenas, otters, coatimundis, hippos, and gorillas, farmed animals such as minks, and wild animals including deer, wild otters, feral minks, and feral cats (Cui et al. Citation2022).

Surveillance of these cases is of paramount importance to gain a better understanding of the susceptible animal populations and the potential risks associated with the emergence of new hosts and reservoirs. Such reservoirs could serve as potential hiding places for the virus to undergo mutations and possibly re-emerge as new variants that may pose a risk to the human population (CDC Citation2021). Vigilant monitoring and research in this area are crucial for public health preparedness and to mitigate potential future threats.

Serological prevalence studies have revealed that human-to-companion animal transmission of SARS-CoV-2 in dogs and cats mostly results in cases without clinical signs or mild respiratory signs. However, in a smaller number of cases, these animals can develop disease and show manifestations similar to those observed in humans, including respiratory clinical signs and more severe outcomes, especially when comorbidities are present (Ferasin et al. Citation2021; Hosie et al. Citation2021; Keller et al. Citation2021; Teixeira et al. Citation2022). Furthermore, the animal detection of the virus in homes with COVID-19 positive individuals has shown varied rates of positivity in pet samples through RT-PCR, ranging from 2.6% to 33% (Sailleau et al. Citation2020; Calvet et al. Citation2021; Fuentealba et al. Citation2021; Jairak et al. Citation2021; Ruiz-Arrondo et al. Citation2021).

Global reports of infection cases in dogs and cats are on the rise, as well as surveillance studies focusing on seroprevalence; however, data for South America remains limited. Currently, there are only a few reports of SARS-CoV-2 infections in pets from countries such as Argentina, Brazil, Colombia, Ecuador, Peru, Uruguay, and Chile (Fuentealba et al. Citation2021; Neira et al. Citation2021; Schiaffino et al. Citation2021; Agopian et al. Citation2022; Alberto-Orlando et al. Citation2022; Rivero et al. Citation2022; Panzera et al. Citation2023). One notable report was published by Neira et al. Citation2021, detailing a viral detection study conducted in Chile in 2020.

These findings emphasize the importance of monitoring and understanding the potential for SARS-CoV-2 transmission between humans and their companion animals. To contribute to this field, we present a broader study focusing on viral detection in pets from homes under quarantine due to COVID-19. Our investigation spans from the initiation of the human vaccination program in Chile to the end of the pandemic. This research aligns with the position of the WOAH (World Organization for Animal Health), which emphasizes the need for epidemiological investigations of SARS-CoV-2 transmission to and from animals exposed to COVID-19+ patients, targeted surveillance of potential hosts, and continuous monitoring of SARS-CoV-2 mutations in animals. Such efforts are crucial to comprehending viral evolution, the emergence of new variants, and the potential risks they may pose to public health (WOAH Citation2022).

Materials and methods

Design of study and sample collection

Between March 2021 and March 2023, a total of 33 households with at least one confirmed SARS-CoV-2 positive person, as determined by RT-qPCR or antigen test, were recruited for viral detection in this study. As an exclusion criterion, the houses had to have at least one veterinarian in the household, since this person was in charge of taking animal samples during their quarantine period. To conduct the investigation, nasal/oral swabs, fecal samples, and/or environmental samples (food and water bowls gauze) were collected from each animal residing in the COVID19+ households over a period of five consecutive days, starting from the confirmation of the owner’s diagnosis. Nasal/oral swab samples were taken with nasopharyngeal swabs, stool samples corresponded to a fresh environmental stool sample, and environmental samples were taken with sterile gauze soaked in viral transport medium rubbed into dishes of water and/or animal food. All samples were placed in a viral transport medium and appropriately labeled according to individual and date. The study included 26 dogs and 39 cats, comprising 34 females and 31 males, with an age range of 2 months to 15 years. If any animal tested positive for SARS-CoV-2 within the initial five-day sampling period, the sampling was extended for an additional five days. Furthermore, to establish the infection rate in the general animal population, 700 nasal swabs were obtained from animals visiting veterinary clinics. These animals were from households with unknown COVID-19 history and visited the clinics for various reasons, including illness, routine check-ups, or sterilization procedures. The sampling was performed but not limited to the Región Metropolitana de Santiago (Metropolitan Region), Chile. The COVID19+ households corresponded to 32 from the metropolitan region and 1 from Los Rios region. The samples of unknown COVID19 status corresponded to 18 animals from Magallanes and Chilean Antarctic region, 69 animals from Bío-Bío region and 613 animals from the Metropolitan region (Figure S1 and ). In this analysis, no information on the socioeconomic aspect of the households was collected.

Table 1. Positive RT-PCR samples for SARS-CoV-2 during investigation in 33 COVID-19+ households in Chile, 2021-2023.

To ensure ethical considerations, all pet owners provided informed consent, and veterinary clinicians collected all samples following the guidelines set by the bioethics committee of the Facultad de Ciencias Veterinarias y Pecuarias of the Universidad de Chile (FAVET, UCHILE) by veterinary clinics (code 21438 – VET – UCH).

SARS-CoV-2 detection

All samples were tested by real-time RT-qPCR at the Animal Virology Lab at FAVET, UCHILE. Briefly, RNA was purified using Chomczynski-phenol solution (Winkler, BM-1755, Chile). Nasal/oral swab and environmental swab samples were extracted directly from the viral transport solution containing the sample. For fecal samples, a 1:10 dilution of the sample, homogenization, and centrifugation at 2500 g for 10 min were previously carried out. After this step, the previously described RNA extraction was carried out. After this, RT-PCR was carried out to identify a portion of the ORFb1 gene, highly conserved in Sarbecovirus, using the primers Forward: 5′-TGGGGYTTTACRGGTAACCT-3′, Reverse: 5′-AACRCGCTTAACAAAGCACTC-3′, and 5′-TAGTTGTGATGCWATCATGACTAG-3′ as probe, according to Chu et al. Citation2020, with a plasmid that contains part of the ORF1b region as positive control. This analysis was performed on a BioRad CFX96 thermal cycler. Samples were considered positive when presented Ct value < 40. Whole genome sequencing was attempted in all positive samples.

Sequencing and phylogenetic analysis

Positive samples were derived to be sequenced using the ARTIC SARS-CoV-2 ONT (Oxford Nanopore Technologies) sequencing protocol at the Molecular Virology Laboratory at Pontificia Universidad Católica de Chile. The pipeline ARTIC V3/V4 whole-genome amplicon-based sequencing in a minIONTM Sequencer (Oxford Nanopore Technologies) were employed. Briefly, RNA extraction, cDNA synthesis and multiple PCR were done according to Tyson et al. Citation2020. Then, libraries were performed according to the manufacturer’s instructions using the Native barcodes (EXP-NBD196, and SQK-LSK109). FAST reads were evaluated to determine the quality of the sequencing and then SARS-CoV-2 genomes were assembled even using the modified ARTIC network bioinformatics pipeline (v1.2.1), at https://github.com/artic-network/fieldbioinformatics/issues/59. The clade and lineage were identified according to the Nextstrain and Pangolin nomenclature (Aksamentov et al. Citation2021; O’Toole et al. Citation2021). Finally, complete genomes (>95% of coverage) were uploaded to GISAID (Elbe and Buckland-Merrett Citation2017). Once this information was obtained, the sequences to build the SARS-CoV-2 phylogenetic tree were obtained from a blast search of each sequence obtained, in NCBI and Epicov platforms. Other sequences collected previously from Chilean animals were also included (Neira et al. Citation2021). Sequence alignment was performed using MUSCLE (3.7) on the CIPRES platform (Miller et al. Citation2010). The phylogenetic tree was built according to the Randomized Accelerated Maximum Likelihood (RAxML) (Stamatakis Citation2014) from Geneious Prime software (Biomatters Citation2023).

SARS-CoV-2 ELISA

After a minimum of 14 days from initiating the household sampling, serum samples were collected from each animal in the COVID19+ households. The serum samples were tested for SARS-CoV-2 antibodies using the ID Screen® SARS-CoV-2 Double Antigen Multi-species kit (Innovative Diagnostics, Grabels, France) which uses a purified recombinant N antigen covering the wells. Once the samples to be tested and controls are added, anti-SARS-CoV-2 antibodies form an antibody-antigen complex. The peroxidase-labeled purified N protein recombinant antigen conjugate then binds to the free fab of bound anti-SARS-CoV-2 antibodies. This reaction is read at 450 nm and evaluated in S/P%. Values equal to or less than 50% were considered negative, values greater than 50% and less than 60% were considered doubtful, and values equal to or greater than 60% were considered positive, as indicated by the manufacturer. The testing procedure followed the manufacturer’s instructions meticulously. It is crucial to emphasize that cats and dogs can be susceptible to other coronaviruses such as feline coronavirus (FCoV), canine enteric coronavirus (CeCoV), and canine respiratory coronavirus (CRCoV), which lack cross-reactivity with SARS-CoV-2 (Chen et al. Citation2020; Michelitsch et al. Citation2020; Patterson et al. Citation2020; Stevanovic et al. Citation2021).

Results

Three dogs from two households (6.06%) tested positive for SARS-CoV-2 by RT-PCR. Additionally, SARS-CoV-2 RNA was detected in environmental samples from three households, with only one household testing positive for both animal and environmental samples. A summary of the positive results can be found in and , and all results of COVID19+ households in Table S1.

Table 2. Positive ELISA samples for SARS-CoV-2 during investigation in 33 COVID-19+ households in Chile, 2021-2023.

Among the three dogs positive for RT-PCR, the first is Dog N° 15, a 2-year-old female poodle that does not share the home with other pets. She showed no signs of SARS-CoV-2 infection and tested positive in April 2022, five days after her owner tested positive. However, the animal tested negative to ELISA.

The other two positive dogs belonged to the same household: Dog N° 21, a 7-year-old male giant schnauzer, and Dog N° 22, a 5-year-old male English bulldog. Both showed no signs of SARS-CoV-2 infection and tested positive in July 2022. Dog N° 22 tested negative to ELISA, and Dog N° 21 could not be tested as it passed away. Unfortunately, we were unable to collect additional data and perform a necropsy as the pet owner declined to continue participating in the study. From the detected 3 positive dogs, one of them was positive for a single day, the second was detected positive on two consecutive days, while the third remained positive for a total of 5 days.

In addition to the positive dogs, a 1-year-old female feline (Cat N° 1) and a 7-year-old male canine (Dog N° 5), belonging to two different households, tested positive for SARS-CoV-2 antibodies by ELISA but not by RT-qPCR (). Overall, antibodies against SARS-CoV-2 were detected in 1 out of 21 cats (4.76%) and 1 out of 17 dogs (5.88%).

The environment-positive samples consisted of gauzes from feeding bowls. One of the samples corresponded to dog N° 15, who was also detected positive for SARS-CoV-2 by RT-qPCR. Another sample was collected from a 9-year-old cat (Cat N° 29) that lives in a household without other pets, and the third sample was from a household with two male dogs, 5 and 7 years old (Dog N° 23 and Dog N° 24). The owners of these households tested positive 5-7 days before the sampling, respectively.

The overall results showed Ct values ranging from 24.6 to 37.5 for the animal samples, while the environmental samples exhibited Ct values ranging from 30.3 to 37.8 (). On the other hand, all 700 samples obtained from animals in households with unknown COVID-19 status tested negative for SARS-CoV-2 by RT-qPCR.

Out of the total positive samples, four complete SARS-CoV-2 genomes were obtained (Table S3). Two genomes were collected from Dog N° 15, corresponding to one oral swab and one environmental sample. Another genome was obtained from a nasal swab from Dog N° 22, and the last genome was from an environmental sample associated with Cat N° 29. All of these genomes belong into the variant of concern Omicron (B.1.1.529 + BA.*), pangolin BA.4.1 lineage. The sequences obtained are over 29.6 kb in length and have a coverage of more than 213x. They have been submitted to GISAID with the following accession numbers: EPI_ISL_14981147, EPI_ISL_14981148, EPI_ISL_14981149, and EPI_ISL_14981150. The closest sequences found in public repositories, GenBank, and GISAID, to the genomes reported were directly related to human cases in Chile. These sequences were obtained from individuals sampled in the same city (Santiago, Chile) during the same collection year.

The phylogeny reveals that all the sequences obtained in this study belong to the same cluster, which also includes sequences collected from three unrelated humans in the same city and during the same timeframe (). It’s worth noting that the older sequences from Chilean cats in 2020 belonged to a different cluster (not shown). Importantly, no other SARS-CoV-2 sequences from animals worldwide demonstrated close genetic relatedness to the sequences obtained in this study.

Figure 1. Phylogenetic tree of SARS-CoV2 by using the complete genome. The final dataset included 53 genomes. Chilean sequences are highlighted in red. The animal sequences are depicted with a red star.

Figure 1. Phylogenetic tree of SARS-CoV2 by using the complete genome. The final dataset included 53 genomes. Chilean sequences are highlighted in red. The animal sequences are depicted with a red star.

Discussion

During the period from March 2021 to March 2023, our study focused on investigating the presence of SARS-CoV-2 in pets from households with COVID-19 positive individuals. Additionally, we examined the general animal population for the presence of the virus. Through this research, we aimed to gain insights into the potential transmission of SARS-CoV-2 between humans and their companion animals, as well as the occurrence of the virus in animals within the broader community.

The obtained results in our study provide confirmation of SARS-CoV-2 infection in pets in Chile, consistent with previous findings reported worldwide. Analyzing the positive RT-PCR SARS-CoV-2 results for animals in Chile during the study period (in COVID19+ households), we observed a general pet positivity rate of 4.62%. This rate is lower compared to the detection rate reported in Chile in 2020 by Neira et al. (Citation2021), who found 11% (3 out of 27 animals) positive for RT-qPCR SARS-CoV-2. Is important to remark, that this study is observational, and the cases were selected conveniently due to the difficulties in recruiting volunteers during the pandemic. As a result, they do not represent a specific neighborhood or social status. Similar difficulties were observed in other investigations around the world.

The lower positivity rate obtained in our research aligns more closely with several studies conducted in 2020, which initially reported very low values of viral detection in animals, ranging between 0% and 5.55% (Chen et al. Citation2020; Patterson et al. Citation2020; Sailleau et al. Citation2020; Sit et al. Citation2020; Dias et al. Citation2021; Fuentealba et al. Citation2021; Goryoka et al. Citation2021; Hosie et al. Citation2021; Klaus et al. Citation2021; Michael et al. Citation2021; Mohebali et al. Citation2022; Stranieri et al. Citation2022).

In contrast, higher detection rates were reported in Brazil, with 27.58% in dogs and 40% in cats (Calvet et al. Citation2021), in Texas (USA), with 17.6% in cats (Hamer et al. Citation2021), and in China, with 13.33% in dogs (Sit et al. Citation2020). Our study contributes valuable data to the understanding of SARS-CoV-2 infection in pets in Chile and provides valuable insights into the variation of detection rates among different geographic regions.

Indeed, studies conducted later in the pandemic have revealed a significant increase in infection rates within COVID19+ households, particularly in felines, with positivity rates ranging from 8% to 35.6% (Alberto-Orlando et al. Citation2022; Goletic et al. Citation2022; Meisner et al. Citation2022; Akhtardanesh et al. Citation2023; Bae et al. Citation2023; Hamdy et al. Citation2023; Kuhlmeier et al. Citation2023). In dogs, a similar trend is observed, albeit with lower values, varying from 0% to 5% (Meisner et al. Citation2022; Akhtardanesh et al. Citation2023), and increasing up to 26.2% (Alberto-Orlando et al. Citation2022; Goletic et al. Citation2022; Molini et al. Citation2022; Bae et al. Citation2023; Hamdy et al. Citation2023; Kuhlmeier et al. Citation2023).

The lower index detected in our study in Chile becomes more pronounced when compared with specific indices reported for other countries in Latin America, such as 24% in Ecuador (Alberto-Orlando et al. Citation2022), and 30.77% in Brazil (Calvet et al. Citation2021). Moreover, considering the previous report of 11% in Chile during 2020 (Neira et al. Citation2021), it is evident that the infection rates have increased over time. These results offsite of Chile could be explained gradual relaxation of control measures associated with COVID-19 cases, which declined in inverse relation to the levels of human vaccination. This relaxation may have facilitated a higher risk of human-to-pet transmission within the same households, making pet contagion more likely. Another possible explanation of the variability of the occurrence of SARS-CoV-2 around the world could be related to the diagnostic method implemented. Most of the studies used real-time RT-PCR, but the targeting gene and the extraction method vary between them, which may implicate differences in sensibility (Table S4).

In contrast to other regions, Chile maintained stricter restricting measures for an extended period and swiftly implemented human vaccination protocols. As a result, Chile stands out as one of the leading democratic countries with remarkably high vaccination rates, a substantial number of vaccine doses administered, a large population of fully vaccinated individuals, and widespread booster administrations. This information is publicly available at Our World in Data (Mathieu et al. Citation2020). In detail, in Chile, the level of human vaccination has been remarkable, with 93.3% of the population fully vaccinated and 88.9% receiving a booster dose to date, as part of an ongoing vaccination campaign (Mathieu et al. Citation2020). The vaccination has been associated with decreased viral shedding time and viral load in human populations (Ergoren et al. 2023; Jung et al. Citation2022). It may have contributed to the lower detection rate of SARS-CoV-2 in pets observed in our study, where a lower overall positivity rate was found in comparison to previous reports from Chile and other countries.

We found a higher positivity rate in dogs compared to cats, which is in contrast to the early pandemic findings suggesting a higher infection rate in cats (Hamer et al. Citation2021; Krafft et al. Citation2021). In Chile, a previous report indicated a 17.6% positivity rate in cats and 0% in dogs using RT-qPCR SARS-CoV-2 (Neira et al. Citation2021). Is well recognized that cats had greater susceptibility of SARS-CoV-2 infection compared to dogs (Bosco-Lauth et al. Citation2020; Shi et al. Citation2020). Therefore, these unexpected results should be interpreted with caution, as the number of cases evaluated in our study was limited to 33. This small sample size may not be sufficient to accurately determine positive rates and could only be seen as an additional observation. It’s important to acknowledge that the limited number of COVID19+ cases visited was due to restrictions and concerns for people’s safety during the pandemic, which made it challenging to conduct extensive data collection. Therefore, further studies with larger sample sizes and a more comprehensive approach are necessary to gain a clearer understanding of the prevalence of SARS-CoV-2 in pets from COVID19+ households.

In addition, no SARS-CoV-2 was detected in animals from households with unknown COVID19 status, aligning with research in Turkey (Kadi et al. Citation2023). The single sampling process without repetitions might have led to an underestimation of the infection in animals in both studies. Similarly, antibodies against SARS-CoV-2 were detected in only 1 cat (4.76%) and 1 dog (5.88%), which is in line with similar findings reported in Portugal and Spain (5% to 7.06%) (Barroso-Arévalo et al. Citation2022; Oliveira et al. Citation2022). However, the relatively low rate of seropositivity in cats stands out when compared to other regions. Interestingly, the animals detected as positive by RT-PCR in our study did not exhibit antibodies against the virus after the short viral excretion period, indicating a possible lack of significant infection that triggered the development of immunity (Happi et al. Citation2023). This situation might be attributed to the animals being initially infected with a low viral load, which could explain the limited immune response.

Other factors related to the variation of the occurrence between countries could include the period of sampling, the variant in circulation during the sampling, the number of households visited, socioeconomic differences, and the time of sampling after human diagnosis. Most of this variability is most likely related to the difficulties of recruitment during the pandemic.

All animal sequences obtained in this study belong to the Omicron variant of concern (B.1.1.529 +BA.*), specifically, the Pangolin lineage BA.4.1. This lineage, BA.4.1, was observed between February 2022 and March 2023 and showed a worldwide distribution. Notably, it was highly frequent in Chilean humans during the same period (Hadfield et al. Citation2018). The BA.4.1 lineage is a derivative of the BA.4 lineage, which initially originated in Africa and was associated with the fifth wave of infections (Tegally et al. Citation2022). Currently, there are 68,288 sequenced viruses belonging to the BA.4.1 lineage, with the animal sequences from our study being the only ones reported so far. This highlights the significance of our findings in understanding the transmission and spread of this particular lineage, especially in the context of its presence in animals.

It’s possible that the detected variant is related to the obtained results. The Omicron variant presents 60 unique mutations and has been reported as less severe in humans compared to Delta. Within the Omicron subvariants, BA.4/5 has been reported to be more transmissible and infectious than other Omicron lineages, which could be attributed to its L452R mutation (Pandit and Matthews Citation2023). This raises the question of whether this mutation could confer an advantage in infecting companion animals, specifically dogs, which could explain the lack of detection in felines in this research, despite substantial evidence of greater susceptibility in cats. Currently, there are few reports on the Omicron variant in pets, and none on the BA.4/5 subvariants. Therefore, there is a pressing need to maintain surveillance of this situation and conduct specific studies to analyze the potential impact of the BA subvariant mutations on animal susceptibility.

It is widely recognized that SARS-CoV-2 can infect animals, and there is substantial evidence of infections with various variants other than BA.4.1(Saied and Metwally Citation2022; Pandit and Matthews Citation2023). Interestingly, the USDA (US) reported that so far, most of the WHO variants include the wildtype, alpha, epsilon, delta, iota, gamma, mu, and ómicron. Notably, the most frequently detected variant in animals has been the delta variant (USDA APHIS Citation2020). In general, the virus does not transmit well between animals in natural conditions, with the exception of farmed minks (Fenollar et al. Citation2021) and free-range white-tailed deer (Feng et al. Citation2023). In fact, SARS-CoV-2 has been successfully disseminated in free-range white-tailed deer, which may serve as a reservoir of extinct VOC (Caserta et al. Citation2023).

While our study focuses on the BA.4.1 lineage, it is essential to acknowledge the broader context of viral transmission to animals and the potential involvement of different variants in infecting and spreading among them. The coexistence of multiple viral lineages in animals warrants ongoing monitoring and surveillance to better understand the dynamics of interspecies transmission and its implications for public health. In addition, the phylogenetic study indicates that the sequences obtained belong to the human variants of SARS-CoV-2 circulating in the human population during the infection period analyzed. This detection corresponds to its presence in the human population during the year 2022 in Chile, reaching its peak during June 2022 (AN de DM Innovación y Desarrollo (ANDID) Citation2022), the late autumn season in Chile, consequently with low temperatures as a risk factor for infection in pets, previously published by Murphy et al. Citation2022. Although the two dogs infected with this variant did not show clinical signs, it is necessary to maintain surveillance under the One Health approach, to early detect mutations or the establishment of new reservoirs that may put public health at risk.

This study confirms the infection of animals by SARS-CoV-2 in Chile in COVID19+ households. However, the values detected are within the lowest range of those detected around the world. This situation can probably be explained by high level of human vaccination in this country. Although more studies are required to confirm this hypothesis, this research suggests this as an additional reason to maintain and strengthen vaccination plans among the human population, under the One Health approach.

Ethics statement

Sampling of animals for this study was approved by Comité Institucional De Cuidado Y Uso De Animales (Cicua), Faculty of Veterinary and Animal Sciences, Universidad de Chile, code 21438 – VET – UCH. Additionally, informed consent was obtained from all pet owners involved in the study.

Supplemental material

Supplemental Material

Download MS Excel (16.3 KB)

Acknowledgments

We gratefully acknowledge the veterinary clinics responsible for obtaining the specimens. Thanks to Valentina Valdés, Patricio Espinoza, Bárbara Brito and Gabriela Muñoz for helping in sample collection and processing at Animal Virology Lab, Animal Virology Laboratory, Faculty of Veterinary and Animal Sciences, Universidad de Chile (FAVET). Thanks to the submitting laboratories for generating the genetic sequences and metadata and sharing via GISAID Initiative, acknowledgment data retrieved at https://doi.org/10.55876/gis8.231120 to and supplementary information.

Disclosure statement

The authors report there are no competing interests to declare.

Data availability statement

The data that support the findings of this study are available in supplementary information, NCBI and Epicov platforms or from the corresponding author upon reasonable request.

Additional information

Funding

This study was partially funded by the Animal Virology Laboratory, Faculty of Veterinary and Animal Sciences, Universidad de Chile; Program Fondecyt N 1211517; the Center for Research in Influenza Pathogenesis (CRIP); a National Institute of Allergy and Infectious Diseases – Funded Center of Excellence in Influenza Research and Response (CEIRR) N75N93021C00014, and ANID Programa Beca Doctorado Nacional N° 21212316/2021, to B.A.

References

  • Agopian RG, da Luz SCG, Zebral AGB, de Sousa GF, de Oliveira IAV, Lima LS, Sechi MA, de Oliveira MC, Rudiniski VF, Brandespim DF, et al. 2022. First reported cases of SARS-CoV-2 infection in pets in São Paulo, Brazil. Vet World. 15(11):2593–2596. doi: 10.14202/vetworld.2022.2593-2596.
  • Akhtardanesh B, Jajarmi M, Shojaee M, Salajegheh Tazerji S, Khalili Mahani M, Hajipour P, Gharieb R. 2023. Molecular screening of SARS-CoV-2 in dogs and cats from households with infected owners diagnosed with COVID-19 during Delta and Omicron variant waves in Iran. Vet Med Sci. 9(1):82–90. doi: 10.1002/vms3.1036.
  • Aksamentov I, Roemer C, Hodcroft EB, Neher RA. 2021. Nextclade: clade assignment, mutation calling and quality control for viral genomes. JOSS. 6(67):3773. doi: 10.21105/joss.03773.
  • Alberto-Orlando S, Calderon JL, Leon-Sosa A, Patiño L, Zambrano-Alvarado MN, Pasquel-Villa LD, Rugel-Gonzalez DO, Flores D, Mera MD, Valencia P, et al. 2022. SARS-CoV-2 transmission from infected owner to household dogs and cats is associated with food sharing. Int J Infect Dis. 122:295–299. doi: 10.1016/j.ijid.2022.05.049.
  • Almehdi AM, Khoder G, Alchakee AS, Alsayyid AT, Sarg NH, Soliman SSM. 2021. SARS-CoV-2 spike protein: pathogenesis, vaccines, and potential therapies. Infection. 49(5):855–876. doi: 10.1007/s15010-021-01677-8.
  • AN de DM Innovación y Desarrollo (ANDID). 2022. Vigilancia Genómica SARS-Cov2 ISP [Internet]. [accessed 2023 June 10]. https://vigilancia.ispch.gob.cl/app/varcovid.
  • Astuti I, Ysrafil Y. 2020. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2): an overview of viral structure and host response. Diabetes Metab Syndr. 14(4):407–412. doi: 10.1016/j.dsx.2020.04.020.
  • Bae J, Ro C, Kang Y, Ga E, Na W, Song D. 2023. Human-to-animal transmission of SARS-CoV-2, South Korea, 2021. Emerg Infect Dis. 29(5):1066–1067. doi: 10.3201/eid2905.221359.
  • Barroso-Arévalo S, Barneto A, Ramos ÁM, Rivera B, Sánchez R, Sánchez-Morales L, Pérez-Sancho M, Buendía A, Ferreras E, Ortiz-Menéndez JC, et al. 2022. Large-scale study on virological and serological prevalence of SARS-CoV-2 in cats and dogs in Spain. Transbound Emerg Dis. 69(4):e759–e774. doi: 10.1111/tbed.14366.
  • Biomatters. 2023. Geneious version 2023.1 [Internet]. https://www.geneious.com.
  • Bosco-Lauth AM, Hartwig AE, Porter SM, Gordy PW, Nehring M, Byas AD, VandeWoude S, Ragan IK, Maison RM, Bowen RA. 2020. Experimental infection of domestic dogs and cats with SARS-CoV-2: pathogenesis, transmission, and response to reexposure in cats. Proc Natl Acad Sci U S A. 117(42):26382–26388. doi: 10.1073/pnas.2013102117.
  • Calvet GA, Pereira SA, Ogrzewalska M, Pauvolid-Corrêa A, Resende PC, Tassinari WdS, Costa AdP, Keidel LO, Rocha A d, Silva M d, et al. 2021. Investigation of SARS-CoV-2 infection in dogs and cats of humans diagnosed with COVID-19 in Rio de Janeiro, Brazil. PLoS One. 16(4):e0250853. doi: 10.1371/journal.pone.0250853.
  • Caserta LC, Martins M, Butt SL, Hollingshead NA, Covaleda LM, Ahmed S, Everts MRR, Schuler KL, Diel DG. 2023. White-tailed deer (Odocoileus virginianus) may serve as a wildlife reservoir for nearly extinct SARS-CoV-2 variants of concern. Proc Natl Acad Sci U S A. 120(6):e2215067120. doi: 10.1073/pnas.2215067120.
  • CDC. 2021. Coronavirus Disease 2019. Centers for Disease Control and Prevention [Internet]. [accessed 2023 April 10]. https://www.cdc.gov/media/releases/2021/s1103-one-health.html.
  • Chen J, Huang C, Zhang Y, Zhang S, Jin M. 2020. Severe acute respiratory syndrome coronavirus 2-specific antibodies in pets in Wuhan, China. J Infect. 81(3):e68–e69. doi: 10.1016/j.jinf.2020.06.045.
  • Chu DKW, Pan Y, Cheng SMS, Hui KPY, Krishnan P, Liu Y, Ng DYM, Wan CKC, Yang P, Wang Q, et al. 2020. Molecular diagnosis of a novel coronavirus (2019-nCoV) causing an outbreak of pneumonia. Clin Chem. 66(4):549–555. doi: 10.1093/clinchem/hvaa029.
  • Cui S, Liu Y, Zhao J, Peng X, Lu G, Shi W, Pan Y, Zhang D, Yang P, Wang Q. 2022. An updated review on SARS-CoV-2 infection in animals. Viruses. 14(7):1527. doi: 10.3390/v14071527.
  • Dias HG, Resck MEB, Caldas GC, Resck AF, Silva N d, Santos A d, Sousa TdC, Ogrzewalska MH, Siqueira MM, Pauvolid-Corrêa A, et al. 2021. Neutralizing antibodies for SARS-CoV-2 in stray animals from Rio de Janeiro, Brazil. PLoS One. 16(3):e0248578. doi: 10.1371/journal.pone.0248578.
  • Durmaz B, Abdulmajed O, Durmaz R. 2020. Mutations observed in the SARS-CoV-2 spike glycoprotein and their effects in the interaction of virus with ACE-2 receptor. Medeni Med J. 35(3):253–260. doi: 10.5222/MMJ.2020.98048.
  • Elbe S, Buckland-Merrett G. 2017. Data, disease and diplomacy: GISAID’s innovative contribution to global health. Glob Chall. 1(1):33–46. doi: 10.1002/gch2.1018.
  • Feng A, Bevins S, Chandler J, DeLiberto TJ, Ghai R, Lantz K, Lenoch J, Retchless A, Shriner S, Tang CY, et al. 2023. Transmission of SARS-CoV-2 in free-ranging white-tailed deer in the United States. Nat Commun. 14(1):4078. doi: 10.1038/s41467-023-39782-x.
  • Fenollar F, Mediannikov O, Maurin M, Devaux C, Colson P, Levasseur A, Fournier P-E, Raoult D. 2021. Mink, SARS-CoV-2, and the human-animal interface. Front Microbiol. 12:663815. doi: 10.3389/fmicb.2021.663815.
  • Ferasin L, Fritz M, Ferasin H, Becquart P, Corbet S, Ar Gouilh M, Legros V, Leroy EM. 2021. Infection with SARS-CoV-2 variant B.1.1.7 detected in a group of dogs and cats with suspected myocarditis. Vet Rec. 189(9):e944. doi: 10.1002/vetr.944.
  • Fuentealba NA, Moré G, Bravi ME, Unzaga JM, De Felice L, Salina M, Viegas M, Nabaes Jodar MS, Valinotto LE, Rivero FD, et al. 2021. First detection and molecular analysis of SARS-CoV-2 from a naturally infected cat from Argentina. Vet Microbiol. 260:109179. doi: 10.1016/j.vetmic.2021.109179.
  • Goletic S, Goletic T, Softic A, Zahirovic A, Rukavina D, Kavazovic A, Omeragic J, Umihanic S, Hukic M. 2022. The evidence of SARS-CoV-2 human-to-pets transmission in household settings in bosnia and Herzegovina. Front Genet. 13:839205. doi: 10.3389/fgene.2022.839205.
  • Gorbalenya AE, Baker SC, Baric RS, de Groot RJ, Drosten C, Gulyaeva AA, Haagmans BL, Lauber C, Leontovich AM, Neuman BW, et al. 2020. The species Severe acute respiratory syndrome-related coronavirus: classifying 2019-nCoV and naming it SARS-CoV-2. Nat Microbiol. 5(4):536–544. doi: 10.1038/s41564-020-0695-z.
  • Goryoka GW, Cossaboom CM, Gharpure R, Dawson P, Tansey C, Rossow J, Mrotz V, Rooney J, Torchetti M, Loiacono CM, et al. 2021. One health investigation of SARS-CoV-2 infection and seropositivity among pets in households with confirmed human COVID-19 Cases—Utah and Wisconsin, 2020. Viruses. 13(9):1813. doi: 10.3390/v13091813.
  • Hadfield J, Megill C, Bell SM, Huddleston J, Potter B, Callender C, Sagulenko P, Bedford T, Neher RA. 2018. Nextstrain: real-time tracking of pathogen evolution. Bioinformatics. 34(23):4121–4123. doi: 10.1093/bioinformatics/bty407.
  • Hamdy ME, El-Deeb AH, Hagag NM, Shahein MA, Liyanage NPM, Shalaan M, Hussein HA. 2023. SARS-CoV-2 infection of companion animals in Egypt and its risk of spillover. Vet Med Sci. 9(1):13–24. doi: 10.1002/vms3.1029.
  • Hamer SA, Pauvolid-Corrêa A, Zecca IB, Davila E, Auckland LD, Roundy CM, Tang W, Torchetti MK, Killian ML, Jenkins-Moore M, et al. 2021. SARS-CoV-2 infections and viral isolations among serially tested cats and dogs in households with infected owners in Texas, USA. Viruses. 13(5):938. doi: 10.3390/v13050938.
  • Happi AN, Ayinla AO, Ogunsanya OA, Sijuwola AE, Saibu FM, Akano K, George UE, Sopeju AE, Rabinowitz PM, Ojo KK, et al. 2023. Detection of SARS-CoV-2 in terrestrial animals in Southern Nigeria: potential cases of reverse zoonosis. Viruses. 15(5):1187. doi: 10.3390/v15051187.
  • Hosie MJ, Epifano I, Herder V, Orton RJ, Stevenson A, Johnson N, MacDonald E, Dunbar D, McDonald M, Howie F, et al. 2021. Detection of SARS-CoV-2 in respiratory samples from cats in the UK associated with human-to-cat transmission. Vet Rec. 188(8):e247. doi: 10.1002/vetr.247.
  • Jairak W, Charoenkul K, Chamsai E, Udom K, Chaiyawong S, Bunpapong N, Boonyapisitsopa S, Tantilertcharoen R, Techakriengkrai N, Surachetpong S, et al. 2021. First cases of SARS-CoV-2 infection in dogs and cats in Thailand. Transboundary and Emerging Diseases. DOI: 10.1111/TBED.14383
  • Jung J, Kim JY, Park H, Park S, Lim JS, Lim SY, Bae S, Lim YJ, Kim EO, Kim J, et al. 2022. Transmission and Infectious SARS-CoV-2 Shedding Kinetics in Vaccinated and Unvaccinated Individuals. JAMA Network Open 5, e2213606. DOI: 10.1001/JAMANETWORKOPEN.2022.13606.
  • Kadi H, Kurucay HN, Elhag AE, Dogan F, Yildirim S, Tutuncu H, Muftuoglu B, Tamer C, Okur Gumusova S, Yazici Z, et al. 2023. A one-year extensive molecular survey on SARSCoV-2 in companion animals of Turkey shows a lack of evidence for viral circulation in pet dogs and cats. Veterinary and Animal Science, 19, 100280. doi: 10.1016/j.vas.2022.100280
  • Keller M, Hagag IT, Balzer J, Beyer K, Kersebohm JC, Sadeghi B, Wernike K, Höper D, Wylezich C, Beer M, et al. 2021. Detection of SARS-CoV-2 variant B.1.1.7 in a cat in Germany. Res Vet Sci. 140:229–232. doi: 10.1016/j.rvsc.2021.09.008.
  • Klaus J, Zini E, Hartmann K, Egberink H, Kipar A, Bergmann M, Palizzotto C, Zhao S, Rossi F, Franco V, et al. 2021. SARS-CoV-2 infection in dogs and cats from southern germany and northern italy during the first wave of the COVID-19 pandemic. Viruses. 13(8):1453. doi: 10.3390/v13081453.
  • Krafft E, Denolly S, Boson B, Angelloz-Pessey S, Levaltier S, Nesi N, Corbet S, Leterrier B, Fritz M, Leroy EM, et al. 2021. Report of one-year prospective surveillance of SARS-CoV-2 in dogs and cats in france with various exposure risks: confirmation of a low prevalence of shedding, detection and complete sequencing of an alpha variant in a cat. Viruses. 13(9):1759. doi: 10.3390/v13091759.
  • Kuhlmeier E, Chan T, Meli ML, Willi B, Wolfensberger A, Reitt K, Hüttl J, Jones S, Tyson G, Hosie MJ, et al. 2023. A risk factor analysis of SARS-CoV-2 infection in animals in COVID-19-affected households. Viruses. 15(3):731. doi: 10.3390/v15030731.
  • Lee P-I, Hsueh P-R. 2020. Emerging threats from zoonotic coronaviruses-from SARS and MERS to 2019-nCoV. J Microbiol Immunol Infect. 53(3):365–367. doi: 10.1016/j.jmii.2020.02.001.
  • Luan J, Lu Y, Jin X, Zhang L. 2020. Spike protein recognition of mammalian ACE2 predicts the host range and an optimized ACE2 for SARS-CoV-2 infection. Biochem Biophys Res Commun. 526(1):165–169. doi: 10.1016/j.bbrc.2020.03.047.
  • Mathieu E, Ritchie H, Rodés-Guirao L, Appel C, Giattino C, Hasell J, Macdonald B, Dattani S, Beltekian D, Ortiz-Ospina E, et al. 2020. Coronavirus Pandemic (COVID-19). [On Line resource]. https://ourworldindata.org/explorers/coronavirus-data-explorer?zoomToSelection=true&time=2021-07-17..2023-02-08&uniformYAxis=0&country=CHL∼OWID_WRL&pickerSort=asc&pickerMetric=location&Metric=Vaccine+doses%2C+people+vaccinated%2C+and+booster+doses&Interval=Cumulative&Relative+to+Population=true&Color+by+test+positivity=false. [Accessed 22-07-2022].
  • Meisner J, Baszler TV, Kuehl KE, Ramirez V, Baines A, Frisbie LA, Lofgren ET, Avila D d, Wolking RM, Bradway DS, et al. 2022. Household Transmission of SARS-CoV-2 from Humans to Pets, Washington and Idaho, USA. Emerg Infect Dis. 28(12):2425–2434. doi: 10.3201/eid2812.220215.
  • Michael HT, Waterhouse T, Estrada M, Seguin MA. 2021. Frequency of respiratory pathogens and SARS-CoV-2 in canine and feline samples submitted for respiratory testing in early 2020. J Small Anim Pract. 62(5):336–342. doi: 10.1111/jsap.13300.
  • Michelitsch A, Hoffmann D, Wernike K, Beer M. 2020. Occurrence of Antibodies against SARS-CoV-2 in the Domestic Cat Population of Germany. Vaccines (Basel). 8(4):772. doi: 10.3390/vaccines8040772.
  • Miller MA, Pfeiffer W, Schwartz T. 2010. Creating the CIPRES Science Gateway for inference of large phylogenetic trees [Internet]. New Orleans LA. [accessed 2023 April 10]. https://www.phylo.org/portal2/home.action.
  • Mittal A, Manjunath K, Ranjan RK, Kaushik S, Kumar S, Verma V. 2020. COVID-19 pandemic: insights into structure, function, and hACE2 receptor recognition by SARS-CoV-2. PLoS Pathog. 16(8):e1008762. doi: 10.1371/journal.ppat.1008762.
  • Mohebali M, Hassanpour G, Zainali M, Gouya MM, Khayatzadeh S, Parsaei M, Sarafraz N, Hassanzadeh M, Azarm A, Salehi-Vaziri M, et al. 2022. SARS-CoV-2 in domestic cats (Felis catus) in the northwest of Iran: evidence for SARS-CoV-2 circulating between human and cats. Virus Res. 310:198673. doi: 10.1016/j.virusres.2022.198673.
  • Molini U, Coetzee LM, Engelbrecht T, de Villiers L, de Villiers M, Mangone I, Curini V, Khaiseb S, Ancora M, Cammà C, et al. 2022. SARS-CoV-2 in namibian dogs. Vaccines (Basel). 10(12):2134. doi: 10.3390/vaccines10122134.
  • Murphy H, Sanchez S, Ahmed S, Rahman M, Di D, Dileepan M, Heinrich D, Liang Y, Ly H. 2022. SARS-CoV-2 in companion animals: do levels of SARS-CoV-2 seroconversion in pets correlate with those of pet’s owners and with protection against subsequent SARS-CoV-2 infection? Virulence. 13(1):1216–1220. doi: 10.1080/21505594.2022.2098922.
  • Neira V, Brito B, Agüero B, Berrios F, Valdés V, Gutierrez A, Ariyama N, Espinoza P, Retamal P, Holmes EC, et al. 2021. A household case evidences shorter shedding of SARS-CoV-2 in naturally infected cats compared to their human owners. Emerg Microbes Infect. 10(1):376–383. doi: 10.1080/22221751.2020.1863132.
  • O’Toole Á, Scher E, Underwood A, Jackson B, Hill V, McCrone JT, Colquhoun R, Ruis C, Abu-Dahab K, Taylor B, et al. 2021. Assignment of epidemiological lineages in an emerging pandemic using the pangolin tool. Virus Evol. 7(2):veab064. doi: 10.1093/ve/veab064.
  • Oliveira A, Pereira MA, Mateus TL, Mesquita JR, Vala H. 2022. Seroprevalence of SARS-CoV-2 in client-owned cats from Portugal. Vet Sci. 9(7):363. doi: 10.3390/vetsci9070363.
  • Pandit R, Matthews QL. 2023. A SARS-CoV-2: companion animal transmission and variants classification. Pathogens. 12(6):775. doi: 10.3390/pathogens12060775.
  • Panzera Y, Mirazo S, Baz M, Techera C, Grecco S, Cancela F, Fuques E, Condon E, Calleros L, Camilo N, et al. 2023. Detection and genome characterisation of SARS-CoV-2 P.6 lineage in dogs and cats living with Uruguayan COVID-19 patients. Mem Inst Oswaldo Cruz. 117:e220177. doi: 10.1590/0074-02760220177.
  • Patterson EI, Elia G, Grassi A, Giordano A, Desario C, Medardo M, Smith SL, Anderson ER, Prince T, Patterson GT, et al. 2020. Evidence of exposure to SARS-CoV-2 in cats and dogs from households in Italy. Nat Commun. 11(1):6231. doi: 10.1038/s41467-020-20097-0.
  • Rivero R, Garay E, Botero Y, Serrano-Coll H, Gastelbondo B, Muñoz M, Ballesteros N, Castañeda S, Patiño LH, Ramirez JD, et al. 2022. Human-to-dog transmission of SARS-CoV-2, Colombia. Sci Rep. 12(1):7880. doi: 10.1038/s41598-022-11847-9.
  • Ruiz-arrondo I, Portillo A, Palomar AM, Santibáñez S, Santibáñez P, Cervera C, Oteo JA. 2021. Detection of SARS-CoV-2 in pets living with COVID-19 owners diagnosed during the COVID-19 lockdown in Spain: A case of an asymptomatic cat with SARS-CoV-2 in Europe. Transboundary and Emerging Diseases. 68, 973–976, doi: 10.1111/TBED.13803
  • Saied AA, Metwally AA. 2022. SARS-CoV-2 variants of concerns in animals: an unmonitored rising health threat. Virusdisease. 33(4):466–476. doi: 10.1007/s13337-022-00794-8.
  • Sailleau C, Dumarest M, Vanhomwegen J, Delaplace M, Caro V, Kwasiborski A, Hourdel V, Chevaillier P, Barbarino A, Comtet L, et al. 2020. First detection and genome sequencing of SARS-CoV-2 in an infected cat in France. Transbound Emerg Dis. 67(6):2324–2328. doi: 10.1111/tbed.13659.
  • Schiaffino F, Ferradas C, Jara LM, Salvatierra G, Dávila-Barclay A, Sanchez-Carrion C, Ulloa A, Mascaro L, Pajuelo MJ, Guevara Sarmiento L, et al. 2021. First detection and genome sequencing of SARS-CoV-2 lambda (C.37) variant in symptomatic domestic cats in Lima, Peru. Front Vet Sci. 8:737350. doi: 10.3389/fvets.2021.737350.
  • Shi J, Wen Z, Zhong G, Yang H, Wang C, Huang B, Liu R, He X, Shuai L, Sun Z, et al. 2020. Susceptibility of ferrets, cats, dogs, and other domesticated animals to SARS–coronavirus 2. Science. 368(6494):1016–1020. doi: 10.1126/science.abb7015.
  • Siracusano G, Pastori C, Lopalco L. 2020. Humoral immune responses in COVID-19 patients: a window on the state of the art. Front Immunol. 11:1049. doi: 10.3389/fimmu.2020.01049.
  • Sit THC, Brackman CJ, Ip SM, Tam KWS, Law PYT, To EMW, Yu VYT, Sims LD, Tsang DNC, Chu DKW, et al. 2020. Infection of dogs with SARS-CoV-2. Nature. 586(7831):776–778. doi: 10.1038/s41586-020-2334-5.
  • Stamatakis A. 2014. RAxML version 8: a tool for phylogenetic analysis and post-analysis of large phylogenies. Bioinformatics. 30(9):1312–1313. doi: 10.1093/bioinformatics/btu033.
  • Stevanovic V, Tabain I, Vilibic-Cavlek T, Mauric Maljkovic M, Benvin I, Hruskar Z, Kovac S, Smit I, Miletic G, Hadina S, et al. 2021. The emergence of SARS-CoV-2 within the dog population in croatia: host factors and clinical outcome. Viruses. 13(8):1430. doi: 10.3390/v13081430.
  • Stranieri A, Lauzi S, Giordano A, Galimberti L, Ratti G, Decaro N, Brioschi F, Lelli D, Gabba S, Amarachi NL, et al. 2022. Absence of SARS-CoV-2 RNA and anti-SARS-CoV-2 antibodies in stray cats. Transbound Emerg Dis. 69(4):2089–2095. doi: 10.1111/tbed.14200.
  • Tegally H, Moir M, Everatt J, Giovanetti M, Scheepers C, Wilkinson E, Subramoney K, Makatini Z, Moyo S, Amoako DG, et al. 2022. Emergence of SARS-CoV-2 omicron lineages BA.4 and BA.5 in South Africa. Nat Med. 28(9):1785–1790. doi: 10.1038/s41591-022-01911-2.
  • Teixeira AIP, de Brito RN, Gontijo CC, Romero GAS, Ramalho WM, Haddad R, Noronha EF, de Araújo WN. 2022. The role of pets in SARS-CoV-2 transmission: an exploratory analysis. Infection [Internet]. 51(2):455–458. doi: 10.1007/s15010-022-01860-5.
  • Tyson JR, James P, Stoddart D, Sparks N, Wickenhagen A, Hall G, Choi JH, Lapointe H, Kamelian K, Smith AD, et al. 2020. Improvements to the ARTIC multiplex PCR method for SARS-CoV-2 genome sequencing using nanopore bioRxiv.:20200904283077. doi: 10.1101/2020.09.04.283077.
  • USDA APHIS. 2020. USDA APHIS | Confirmed cases of SARS-CoV-2 in Animals in the United States [Internet]. [accessed 2020 June 20]. https://www.aphis.usda.gov/aphis/ourfocus/animalhealth/sa_one_health/sars-cov-2-animals-us.
  • WOAH. 2022. Statement from the advisory group on SARS-CoV-2 evolution in animals [Internet]. [accessed 2023 May 22]. https://www.woah.org/app/uploads/2022/01/statement-agve-omicron.pdf.