Next Article in Journal
Spatial Differentiation of Heavy Metals/Metalloids, Microbial Risk Genes and Soil Microbiota in a Sulfur-Contaminated Landscape
Previous Article in Journal
Molecular Detection and Phylogenetic Analysis of Anaplasma spp. in Ticks Collected from Grasslands near Livestock Farms in Gyeonggi and Gangwon Provinces, Republic of Korea
Previous Article in Special Issue
Clinical Features and Outcomes of Patients with Full Spectrum of COVID-19 Severity and Concomitant Herpesvirus Reactivation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Current Advances and Applications of Animal Models in SARS-CoV-2 Pathogenesis and Vaccine Development

1
Department of Biology, College of Life Sciences, China Jiliang University, Hangzhou 310018, China
2
Department of Biopharmacy, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
*
Authors to whom correspondence should be addressed.
Microorganisms 2025, 13(9), 2009; https://doi.org/10.3390/microorganisms13092009
Submission received: 7 August 2025 / Revised: 25 August 2025 / Accepted: 26 August 2025 / Published: 28 August 2025
(This article belongs to the Collection Advances in SARS-CoV-2 Infection)

Abstract

COVID-19 is the most widespread emerging infectious disease in humans, recently caused by the SARS-CoV-2 virus. Understanding the pathogenesis and development of efficient vaccines is crucial for the prevention and control of this emerging disease. SARS-CoV-2 viruses have widespread hosts, including humans, domesticated/companion animals (cats, dogs), specific farmed animals (minks), specific wildlife (white-tailed deer), and laboratory animal models. Bats are considered the original reservoir, and pangolins may be important intermediate hosts. Suitable animal models play an important role in studying the pathogenicity and evaluation of vaccines and antiviral drugs during the preclinical stage. In this review, we summarized the animal models and potential animal models for the research of SARS-CoV-2 pathogenesis, vaccine and antiviral drugs development, including transgenic mice, cats, hamsters, nonhuman primates, ferrets, and so on. Our summary provides the important information to select the animals used for a specific purpose and facilitates the development of novel vaccines and antivirals to prevent and control COVID-19.

1. Introduction

Global health faces a significant challenge from emerging diseases. Since late 2019, a novel coronavirus named SARS-CoV-2 has emerged to infect humans with severe pneumonia, resulting in death in some cases. As it spread almost all over the world, the World Health Organization (WHO) announced the emergence of a worldwide pandemic. Vaccination and antiviral drugs are two important strategies to prevent and control infectious diseases. As a novel virus, the study of pathogenesis, the development of vaccines, and antiviral drugs is in progress around the world. In this case, animals are critical for these studies mentioned above. In this review, we focused on summarizing the characteristics of the animal models for the research on SARS-CoV-2.

2. Virus and Disease

SARS-CoV-2 belonged to the Coronaviridae family, the Betacoronavirus genus. SARS-CoV-2 carries a non-segmented RNA genome with positive polarity. Coronaviruses contain multiple members distributed broadly among humans, other mammals, and poultry [1], seven of which can infect humans, including HCoV-229E, -OC43, -NL63, -HKU1, SARS-CoV, MERS-CoV, and SARS-CoV-2. HCoV-229E, -HKU1, -NL63, and -OC43 viruses typically cause the common cold in children, the elderly, or immunocompromised people [2]. Both SARS-CoV and MERS-CoV cause zoonotic spillover and cause severe respiratory syndrome with high mortality in humans [3]. SARS-CoV-2 infected humans and multiple animals and caused fever, cough, and severe pneumonia since the end of 2019. It spread all over the world, causing a global pandemic. This virus evolved rapidly from alpha to Omicron and then JN.1/KP.2 [4,5].

3. Target Cells and Receptors

The target cells of SARS-CoV-2 include macrophages, type II pneumocytes in the lungs, absorptive enterocytes in the intestine, and goblet secretory cells in the nasal passages [6]. Co-expression of ACE2 (Angiotensin converting enzyme II) and TMPRSS2 (transmembrane protease, serine 2) TMPRSS2 within these cells enables these target cells to have a high-level binding ability and demonstrates elevated SARS-CoV-2 susceptibility. SARS-CoV-2 hijacks ACE2 on these host cells as its receptor. Spike (S) protein on the viral surface mediates cell attachment and membrane fusion. During this process, the trimeric S protein is activated mainly by TMPRSS2 and cleaved into S1 and S2 proteins. S1 enables viral attachment through the receptor binding domain (RBD)-ACE2 binding, whereas S2 mediates viral-cell membrane fusion [7].
ACE2 is a type I membrane protein, widely expressed on the alveolar monocytes, macrophages, endothelial cells of arteries and veins, mucosal cells of the small intestine, and epithelial cells of the kidney, renal tubules, alveoli, trachea, bronchial branches, and serous glands [8,9]. In addition to the above, its primary physiological function is to facilitate the maturation of angiotensin (Ang) to control vasoconstriction and blood pressure. For membrane trafficking, ACE2 also functions as a chaperone of the amino acid transporter B0AT1 (SLC6A19), which orchestrates neutral AA transport across enterocyte membranes by a Na+-leucine cotransport manner [10]. ACE2, in concert with TMPRSS2 as the main host protease, facilitates the virus entry into target cells. Co-distribution of ACE2 and TMPRSS2 in the respiratory tracts is significantly associated with enhanced cellular permissibility to SARS-CoV-2. The animals, including cats, Syrian hamsters, and white-tailed deer, with the highest level of co-expression of these two host factors, are most susceptible to SARS-CoV-2 infection [11]. ACE2 is the main receptor of SARS-CoV-2 for cell entry. We summarize the key amino acids of ACE2 of different animals interacting with the RBD of the S protein, which may partially interrupt the different susceptibility of multiple animal models (Table 1).
Other receptors and host proteases also exacerbate the pathogenesis of SARS-CoV-2, but their modes of action remain unknown, such as neuropilin-1, furin, and tyrosine-protein kinase receptor UFO (AXL), kidney injury molecule (KIM1), and Toll-like receptor 2/4 (TLR2/4) [12,13,14,15]. ASGR1 and KREMEN1 were two alternative functional receptors for non-ACE2 virus entry and further determined the viral tropism and pathogenesis [16]. CD147 functions as a compensatory entry receptor for SARS-CoV-2 in ACE2-deficient cells, which partially explains why lungs can be widely infected by SARS-CoV-2, although ACE2 is expressed at low levels [17]. Interestingly, leucine-rich repeat-containing protein 15 (LRRC15), which was one TLR-related cell surface receptor, was found to be a novel receptor of the spike protein of SARS-CoV-2; however, it sequesters the infection of this virus [18]. In addition, integrin contributes to the invasion of SARS-CoV-2 into the non-ACE2 respiratory cells to facilitate its infection and spread in the body of the patients [19]. On the other hand, the S mutant also contributes to the expansion of the host range of the SARS-CoV-2 infection. The S N501Y potentially increases the ability of SARS-CoV-2 to infect avian animals, while the S T478I helps SARS-CoV-2 to utilize multiple mammalian ACE2 for infection [20]. Above all, the binding affinity between the RBD of the S protein and ACE2 determines the permissibility of cross-species (Table 1). And S mutations in the RBD region in Omicron variants enhance the affinity of S to ACE2 of multiple animals; therefore, Omicron has a broader host range [21].

4. Animal Models

4.1. Pathogenesis of SARS-CoV-2 in Mouse Models

Mice are the preferred small animal model because of the low cost, high reproduction rate, and ease of handling. However, SARS-CoV-2 does not replicate efficiently in normal wild-type laboratory mice because it cannot use mouse ACE2 as the entry receptor [22,23]. In this case, many teams have made great efforts to develop different strains of mice susceptible to SARS-CoV-2. Therefore, different types of transgenic mice have been developed (Table 2).
The first strategy is to transfer the human Ace2 gene to mice and generate the humanized ACE2 transgenic mice. K18-hACE2 mice are the most popular animal model in the research of pathogenesis, vaccines, and antiviral therapy development. K18-hACE2 mice expressed the human ACE2 under an ectopic cytokeratin promoter. hACE2 knock-in (KI) mice with hACE2 expressed under the native promoter were generated. SARS-CoV-2 caused the respiratory infection but not severe diseases in KI mice [24].
A mouse model has been established by introducing human ACE2 (hACE2) for the SARS-CoV-2 receptor into the genome of ICR mice, and the hACE2 transgenic mice have been demonstrated to support SARS-CoV-2 infection [78]. Sun et al. have generated a mouse model expressing hACE2 under the mouse ACE2 promoter by CRISPR/Cas9 knock-in technology. Human ACE2-transgenic murine models are permissive for SARS-CoV-2 upon intranasal delivery and display interstitial pneumonia after being challenged with the SARS-CoV-2 strain BetaCoV/Wuhan/AMMS01/2020 [79]. However, the SARS-CoV-2 was not lethal to the two transgenic mice above. The other transgenic mice have been generated to express hACE2 under the control of Hfh4 (also known as Foxj1) promoter in C3B6 mice, resulting in lethal SARS-CoV-2 infection [25]. The Hfh4 promoter targets hACE2 expression to respiratory ciliated cells and CNS neurons [26]. Hfh4-driven hACE2 transgenic mice exposed to SARS-CoV-2showed about 40% mortality, probably due to viral invasion of the brain [27]. Although transgenic mice displayed interstitial pneumonia, they were not widely available. It takes time to breed enough mice for high-throughput drug and medical development. For some distinct comorbidities, such as diabetes or obesity, transgenic mice cannot meet these needs completely. In addition, human CD147 transgenic mice could also support the infection of SARS-CoV-2, while wild-type mice did not [17]. The first generation of mouse models susceptible to SARS-CoV-2 was mainly established by ectopic expression of hACE2. Nakandakari-Higa et al. utilized the S82M/F83Y/H353K triple mutations of mACE2 to generate a minimally edited mouse model-Ace2, which is susceptible to multiple SARS-CoV-2 strains, including USA-WA1/2020 and B.1.1.529 (Omicron), benefiting research on the immune responses to sequential infection in mice [28]. Mice with F83Y/H353K mutations in the mouse ACE2 supported the infection of SARS-CoV-2, but the infected mice did not manifest symptoms; in addition, further conditional hybrid CMV-Cre derived Rosa26 hACE2 mice were generated, and these mice were more susceptible to the infection of SARS-CoV-2, with reduced body weight, survival rate, and obvious clinical symptoms. These mouse models facilitate the development of SARS-CoV-2 therapy strategies and the research of long COVID-19 [80]. BA.1/BA.2/BA.5 Omicron sublineages emerged from the end of 2021 and spread globally. These viral strains replicate efficiently in the human upper nasal epithelium, while their pathogenicity in the lungs of the infected K18-hACE2 decreased, but they still caused lung pathological changes in aged mice [81]. These results supported the idea that the old were more susceptible to Omicron than the young. Furthermore, Omicron variant infection in K18-hACE2 caused the brain infection with lymphoid depletion [82]. As SARS-CoV-2 evolves, Omicron variants become weak and poorly permissive to B6.K18-hACE2; however, mice with humanized MHC-I and ACE2 were highly susceptible to infection with Omicron [29]. Another strategy to improve the susceptibility of SARS-CoV-2 is to knock out the antiviral gene in the K18-hACE2 mouse model. The researcher found that interferon-induced transmembrane protein3 (IFITM3) knockout K18-hACE2 (K18-hACE2/IFITM3 KO) mice are more permissive than their parental mice [83]. To avoid the neuroinfection disease resulting from the transgenic K18-hACE2 model, the K18-hACE2 transgene was integrated into the collagen type I alpha chain (COL1A1) locus. The Col1a1-K18-hACE2 mice showed weight loss without neurologic symptoms, and it is suitable for the research of long COVID-19 [30]. Ana-Sosa-Batiz et al. generated a triple gene knock-in mouse model, including hACE2, TMPRSS2, and FCGRT, to evaluate the efficacy of monoclonal antibodies targeting the receptor-binding domain of SARS-CoV-2 and hybrid immunity. They found that the mAbs with long half-life are more effective against Omicron BA.2. Therefore, this triple KI mouse model is a useful tool for the development of mAb therapy against COVID-19 [31].
Replication-defective viruses have been utilized as vectors to carry hACE2 and introduced into BALB/c mice via intranasal inoculation to establish SARS-CoV-2 receptor expression in the lung tissue of mice. Hassan et al. have used replication-defective adenoviruses as the vector to establish SARS-CoV-2 pathogenesis models in conventional murine strains. After being infected with SARS-CoV-2, these mice displayed weight loss and immune cell infiltration. In addition, pneumonia has been observed in these mice when given anti-Ifnar1 antibody [23]. In K18-hACE2 mice, B.1.351/Beta variant was the most pathogenic, while lower viral RNA was detectable in the mice infected with BA.1.1/Omicron, and no symptoms were observed. Among them, B.1.351/Beta and BA.1.1/Omicron can infect the wild-type (WT) mice with transient infection, while B.1 and B.1.617.2/Delta cannot infect them [84]. AAV-hACE2-transduced mice were created by using engineered Adeno-Associated Virus (AAV6.2FF) mediated intratracheal hACE2 gene delivery and SARS-CoV-2 replicates to high titers in the nasal turbinate and lungs, inducing the IgM and IgG antibody response, and modulating cytokine production in the nasal turbinate and lungs [32]. A novel humanized mouse model (hACE2 KI) is generated to study long COVID-19 by investigating the accumulation and longitudinal propagation of the tau protein [85].
A humanized mouse model carrying human lung xenografts was developed and was used to investigate the authentic infection of SARS-CoV-2 and it could replicate efficiently and spread quickly in the infected the lung epithelial cells, suggesting it is a good model to explore the pathogenesis of SARS-CoV-2 and develop antiviral therapy, such as Bafilomycin A1 Ad5-hACE2-transduced NIKO mice (NOD-SCID IL2Rgamma(−/−)) developed SARS-CoV-2-induced pulmonary inflammation with cytokine/chemokine dysregulation, providing a humanized platform for viral pathogenicity research and therapeutic assessment [33]. The tissue-specific mouse model was developed by combining the lung-specific Cre mouse and hACE2 mouse to evaluate the effect of antivirals in vivo and pathogenesis in some specific tissues or organs [86]. Three inducible ACE2 transgenic mice, including AT2, club cells, and ciliated cells, were generated, and SARS-CoV-2 infection caused severe pneumonia in all of these mice. These models allow us to investigate the relationship of specific cell types with the pathogenesis of SARS-CoV-2 [87]. In huCD34(+)-hACE2-NCG mice (hACE2-knock-in immunodeficient NCG reconstituted with human CD34(+) cells), SARS-CoV-2 Beta/Delta variants underwent multi-organ replication—including nasal, pulmonary, and brains—by 72h post-infection., but Omicron strains could not detect in the nasal tissue or brain, these results indicate that antiviral immune response did not result in the lack of neurotropic characteristic of Omicron and we should select the proper SAR-CoV-2 strains and mouse models [34]. NSG mice bearing human lung tissue grafts (NSG-L mice) were found to be susceptible to the infection of SARS-CoV-2. A live virus could be recovered from human lung grafts and other multiple organs, and a human immune response was also observed in these humanized mice [35].
The MVA-vectored prefusion-stabilized SARS-CoV-2 spike vaccine candidate was evaluated in K18-hACE2 mice and in golden Syrian hamsters. The MVA-Spf vaccine candidate induced the production of high-titer antibodies, robust T-cell immunity, and robust challenge protection in both animal models [88]. Walls et al. revealed that neutralizing antibody responses in vaccinated BALB/c mice lacked the breadth and potency against SARS-CoV-2 variants of concern (VOCs) observed in primates or humans across heterologous vaccine platforms. Therefore, we need to be cautious when we analyze the data obtained from the BALB/c mice [89].
The second strategy is to modify the SARS-CoV-2 virus to allow it to replicate in wild-type mice. Therefore, a mouse-adapted SARS-CoV-2 virus has been generated to enable it to infect multiple types of mice. Mouse-adapted SARS-CoV-2 strain has been generated by serial passage in aged mice (9 months) through intranasal inoculation. During serial passages, adaptive mutations have been acquired by SARS-CoV-2, such as N501Y in the RBD of the S protein. The mouse-adapted strain with this mutation has displayed increased infectivity and led to interstitial pneumonia in both young and aged mice [90]. This mutation has been demonstrated to enhance the interaction with the receptor ACE2 and infectivity in humans [91]. Dinnon et al. employed reverse genetics to enhance SARS-CoV-2 spike-mACE2 binding affinity, generating a mouse-adapted chimeric virus (MA-SARS-CoV-2) capable of utilizing murine ACE2 for cellular entry. This recombinant strain exhibited robust replication throughout respiratory tract compartments in BALB/c mice across age groups [27]. In K18-hACE2 mice, mouse-adapted SARS-CoV-2 strain MA10 exhibits compartmentalized pulmonary tropism with accelerated replication kinetics, neurodissemination, and altered pulmonary immune profiles [92]. Serial lung passaging in aged BALB/c and C57BL/6N mice generated mouse-adapted SARS-CoV-2 variants (BMA8, C57MA14). The Q489H RBD mutation confers host adaptation by shifting viral receptor usage from human to murine ACE2 [93]. The mouse-adapted BMA8 strain exhibited age-dependent lethality in BALB/c mice but not C57BL/6N counterparts, with host MHC molecules mediating this pathogenic divergence [93,94]. The mouse-adapted SARS-CoV-2 was lethal to the aged mice due to the activation of TNF signaling [95]. To modify viruses is a fast, relatively simple strategy to meet the requirement for infecting different types of mice, but modification of SARS-CoV-2 must be conducted under a biosafety level 3 laboratory. One strategy is to develop the SARS-CoV-2 pseudovirus that contains the firefly luciferase reporter gene, then assess mAb protection and variant tropism via this model by in vivo bioluminescent imaging [96].
The third strategy is to find special mouse strains that could support the infection and replication of SARS-CoV-2. The severe combined immunodeficient (SCID) mice were found to be susceptible to the SARS-CoV-2 Beta B.1.351 variant, and the viral load is high in the lung on Day 3 post-infection. This animal model did not require the use of mouse-adapted virus strains and transgenic mice, so it is an ideal model to evaluate the effect of antiviral reagents [36]. The North American deer mouse could be infected by SARS-CoV-2, and the California mouse presented clinical symptoms [97,98]. The neonatal mouse model is another candidate model for the study of SARS-CoV-2. Although adult mice did not support the transmission of SARS-CoV-2, a model based on neonatal mice was established, and both the ancestral WA-1 and SARS-CoV-2 variants Alpha (B.1.1.7), Beta (B.1.351), Gamma (P.1), Delta (B.1.617.2), and Omicron sublineages BA.1/BQ.1.1could transmit in this animal model by the contact route [37].

4.2. Hamsters

Syrian/golden hamsters have been demonstrated as a highly susceptible model for SARS-CoV-2 infection. Weight loss and moderate bronchi-interstitial pneumonia have been observed after SARS-CoV-2 infection. High virus load has been detected in nasal mucosa, lower respiratory epithelial cells, lungs, oral swabs, and rectal swabs by intranasal inoculation of SARS-CoV-2 [38,39,40]. SARS-CoV-2 is efficiently transmitted to naive hamsters by direct contact or via aerosols [40]. Zhang et al. have found that SARS-CoV-2 is only transmitted by direct contact but not by the airborne route [41]. Lee et al. established respiratory SARS-CoV-2 infection in hamsters via oral inoculation. Compared to intranasal infection, oral inoculation displayed no clinical signs, lower viral load, and mild inflammation in lungs, but a comparable level of virus shedding, which may be attributed to the replication of SARS-CoV-2 in the intestines [42].
The Chinese hamster is also susceptible to SARS-CoV-2 infection with significant body weight loss, bronchitis, and pneumonia after infection. Virus replicates in the upper and lower respiratory tract of the infected Chinese hamster. Compared to the Syrian hamster model, the Chinese hamster features advantages including augmented pathology, compact physiology, annotated multi-omics, and modular toolkits [43]. However, it remains unclear whether SARS-CoV-2 transmits efficiently in the Chinese hamster.
The investigation of impairment of mucociliary transport (MCT) in Syrian hamsters with SARS-CoV-2 infection indicates that its deficiency could be a key factor in predicting the progression of SARS-CoV-2 pathogenesis [99]. The inoculation volume of SARS-CoV-2 by the intranasal route directly affects the severity of disease in the Syrian hamster model [100]. The experimental infection in aging hamsters with a high-fat diet showed that hypertriglyceridemia, age, and gender are three important risk factors for the severity of COVID-19 [101]. The other group found that white adipose tissue (WAT) dysfunction induced by SARS-CoV-2 contributes to the greater severity in the old hamsters [102]. Further study found that pulmonary intravascular macrophages were recruited into lungs after SARS-CoV-2 infection, which may contribute to the severe lung inflammation in a Syrian hamster model [103].
Since the human adenovirus and influenza A virus infection are normally present in humans, coinfection of HAdV-5 or IAV and SARS-CoV-2 has occurred [104,105]. The experimental coinfection in hamsters and ferrets demonstrates that SARS-CoV-2 coinfection with HAdV-5 or IAV exacerbates pulmonary pathology versus monoinfection, especially when IAV and SARS-CoV-2 coinfection result in more obvious clinical symptoms than mono-infection [106]. Therefore, the development of combined vaccines against the HAdV-5 or IAV and SARS-CoV-2 is urgently needed. Omicron variant infection could not induce a robust, sufficient protective immune response, but a heterologous challenge could enhance the immune response for other variants of SARS-CoV-2, and indicates that the heterologous antigens vaccination should be recommended for the prevention of COVID-19 [107]. In addition, cepharanthine showed the prophylactic and therapeutic effects against SARS-CoV-2 infection in a Syrian hamster model [108]. Similarly, Omicron variants presented weak and mild symptoms in hamsters. One BA.4 omicron isolate, which lacks the three amino acids (aa 141–143) in non-structural protein 1, did not cause all the typical symptoms in the Golden Syrian hamster model, despite replicating similarly with other SARS-CoV-2 isolates [109]. SARS-CoV-2 exposure induced more severe pathology in aging hamsters than in young and humanized ACE2 hamsters, and the humanized ACE2 hamsters died from SARS-CoV-2-related meningoencephalitis, indicating this model is suitable for the evaluation of the effect on the central nervous system (CNS) [110]. As we know, anosmia is a common symptom caused by COVID-19. The study in hamsters demonstrates that this symptom is due to damage to the olfactory epithelium. There is a moderately strong correlation between the level of anosmia and the thickness of the olfactory epithelium; therefore, we can use the food-searching behavioral test in a hamster model for screening the various therapeutics for SARS-CoV-2-related anosmia and the development of effective therapy [111].
COVID-19 led to a part of patients with neurocognitive impairments or neurodegeneration in clinical settings. More and more reports demonstrate that SARS-CoV-2 induces persistent microgliosis with concomitant accumulation of hyperphosphorylated Tau and α-synuclein in distal brain regions post-infection in a Golden Syrian hamster model, which may cause long-term disorders, such as Alzheimer’s or Parkinson’s disease [112,113,114]. The researcher confirmed that COVID-19 leads to pathological bone loss in a hamster model, which would be beneficial for the development of related interventions against pathological bone loss [115].
SARS-CoV-2 infection occurred in both male and female hamsters, inducing pulmonary pathology comparable to human COVID-19 cases. The male hamsters presented more severe symptoms and had higher viral shedding in the lungs than the female hamsters, which was consistent with the difference in patients. These results confirmed that the Syrian hamster is a suitable model for studying the pathogenesis of SARS-CoV-2 [44].
Syrian hamsters are not only suitable for the initial infection of SARS-CoV-2 and variant infections, but they could also be used as an animal model for evaluation of the pathogenesis of SARS-CoV-2 reinfection after recovery from the initial infection, especially males [116]. So hamsters are a good animal model to study long COVID-19.
Biosafety level 3 facilities are limited to operate the live SARS-CoV-2; therefore, development of mimic infection platform of SARS-CoV-2 in vivo is of great value for evaluating the vaccine efficacy and antiviral therapy under Biosafety level 2 environment, luciferase-expressing vesicular stomatitis virus (VSV)-based SARS-CoV-2 pseudotyped virus was created, and the lungs were harvested 24–72 h after inoculating the hamsters and luminescence was measured using an in vivo imaging system (IVIS) to evaluate the vaccine efficacy and antiviral drugs [117]. Based on the benefit of animal awareness, the three “R” principles should also be considered in using the Golden hamsters when we evaluate the efficacy of the SARS-CoV-2 vaccine in vivo, and rehoming of animals used for scientific and educational purposes under specific guidelines is highly recommended [118]. From the genome and transcriptome analysis, the Syrian hamster model is superior to other rodents to study the SARS-CoV-2 infection since it possesses several important genes, including ACE2, with high homology to human counterparts [45] (Table 2).

4.3. Nonhuman Primates (NHP)

4.3.1. Pathogenesis of SARS-CoV-2 in NHPs

Nonhuman primates are often used as the ultimate translational research model due to their close phylogenetic relationship with humans. SARS-CoV-2 infects Old/New World primates, notably rhesus (Macaca mulatta), cynomolgus (M. fascicularis), African green (Chlorocebus sabaeus) monkeys, and marmosets (Callithrix jacchus) [46,47,48,49,50] (Table 2). Macaca mulatta, Macaca fascicularis, and Chlorocebus sabaeus are susceptible to SARS-CoV-2 and displayed similar clinical symptoms and pathological lesions, including respiratory abnormalities, increased temperature, weight loss, pulmonary lesions in chest radiographs/computed tomography imaging, viral shedding, histopathological lesions, thrombocytopenia, Type II pneumocyte hyperplasia, and alveolar septal fibrosis [51,52,53,54]. These three models have also been demonstrated as natural transmission models because SARS-CoV-2 can transmit efficiently in these primates [47,51], which is consistent with the same key amino acids in both the three primate models and humans (Table 1). Callithrix jacchus is not susceptible to SARS-CoV-2. Viral genomes can be detected in nasal swabs, throat swabs, anal swabs, and blood after infection, but no clinical signs have been observed in Callithrix jacchus [48]. From Table 1, we know that several key amino acids of ACE2 in Callithrix jacchus are different from those in humans, which may contribute to this phenotype. However, Thomas et al. have found that Callithrix jacchus developed interstitial pneumonitis, accompanied by bronchiolitis after infection. Viral antigen is predominantly localized to alveolar macrophages and AT1 pneumocytes. Pulmonary tissues, tracheobronchial lymph nodes (TBLN), and myocardium exhibited viral RNA co-detected with inflammatory lesions in a subset of animals. Hepatitis manifested in most subjects. Pathogenicity on some species of nonhuman primates, especially New World monkeys, after SARS-CoV-2 infection remains unknown, such as squirrel monkeys (Saimiri), and mustached tamarins (Saguinus mystax). Collectively, it has been revealed that divergent SARS-CoV-2 susceptibility profiles distinguish Old/New World primates, with Macaca mulatta emerging as the optimal SARS-CoV-2 pathogenesis model. The variants of SARS-CoV-2 Omicron (B.1.1.529) were weaker in rhesus macaques than in hamsters and transgenic BALB/c mice. Rhesus macaques are also a good model for the evaluation of antiviral drugs. Remdesivir could significantly reduce the damage to the lower respiratory tract caused by SARS-CoV-2 infection in rhesus macaques [119]. In addition, black-tailed marmoset (Mico melanurus) was first reported with natural SARS-CoV-2 infection in Brazil in 2022 [120]. SARS-CoV-2 infection induced the aggregation of immune cells, including B cells, T cells, follicular dendritic cells, and CD169 macrophages in the lungs of the infected rhesus macaques, with the presence of Pulmonary lymphoid tissue [121].

4.3.2. Regulatory and Ethical Issues in Using NHPs for SARS-CoV-2 Study

The use of nonhuman primates (NHPs) in SARS-CoV-2 research is subject to the most stringent regulatory and ethical scrutiny within biomedical science. This is due to their high cognitive capacity, social complexity, and phylogenetic proximity to humans, which makes them both valuable for translational research and ethically costly [55]. Ethically, the core principle is the application of the 3Rs (Replacement, Reduction, Refinement), mandating that their use is only justified when no alternative model exists and that protocols are rigorously designed to minimize numbers and suffering through advanced analgesia, social housing, and early humane endpoints regulated by National Research Council. Regulatory oversight requires approval from institutional committees (e.g., IACUC), which conduct a strict harm-benefit analysis to ensure compliance with animal welfare laws like the U.S. Animal Welfare Act and the EU Directive 2010/63/EU. Research with the infectious SARS-CoV-2 virus must be conducted at Biosafety Level 3 (BSL-3) or enhanced BSL-3 (BSL-3+) to add another layer of regulatory scrutiny to protect animal and public health. The acquisition of NHPs for research is tightly controlled. In the U.S., the Centers for Disease Control and Prevention (CDC) regulates the importation of animals to prevent disease transmission. Their source is also critical, with a preference for purpose-bred animals from regulated breeding colonies rather than wild-caught sources, which is often prohibited [56]. Therefore, using NHPs for SARS-CoV-2 research is legally permissible but is one of the most heavily restricted areas of science. It requires navigating a complex web of animal welfare laws, biosafety regulations, and intense ethical scrutiny centered on the 3Rs.

4.4. Transmission in Ferrets

SARS-CoV-2 exhibits cross-species transmission among mammals, including minks, raccoon dogs, felids, ferrets, rodents (hamsters/mice), bats (Rousettus aegyptiacus), and cervids [122]. Ferrets and hamsters have been commonly used for investigating the transmissibility of respiratory viruses that infect humans [123,124] (Table 2). SARS-CoV-2 exhibits robust replication in ferret upper airways. Viruses have been detected in the nasal turbinate, urine, feces, soft palate, saliva, and tonsils of the infected ferrets. Some infected ferrets developed fever and loss of appetite. SARS-CoV-2 leads to interstitial pneumonia and acute bronchiolitis. Histopathological examination has revealed lymphoplasmacytic perivasculitis, vasculitis, and accumulation of macrophages and neutrophils occurred in the alveolar septa, alveolar lumina, bronchial lining, and bronchial lumina [57,58]. However, Schlottau et al. have found that no clinical symptoms were observed in infected ferrets, which may be attributed to the different strains used in these studies [59]. SARS-CoV-2 was transmitted to naive ferrets by direct contact effectively and by indirect contact limitedly [58,59], while the efficient transmission of SARS-CoV-2 via respiratory droplets and direct contact was documented by Richard et al. [60]. These differences among these groups might depend on different conditions of the experiments carried out, such as facilities, and equipment, virus strains, sensitivity of detection methods, sampling time, and individual differences in ferrets. A recent study demonstrated that ferrets are a suitable model for transmission studies and evaluation of the efficacy of vaccines and antiviral drugs by both intranasal and aerosol administration [61]. Higher expression level of two key entry factors, ACE2 and TMPRSS2, in aged ferrets leads to greater susceptibility than that in young ferrets [62].
Although the antigens of SARS-CoV-2 could not be detected in the brains of the infected ferrets with mild respiratory diseases, SARS-CoV-2 still had neurovirulent potential, with evidence of increased microglial activation and decreased astrocytic activation status [125]. One study demonstrated that SARS-CoV-2 could survive on the skin or skin-to-skin transference and then the virus infected a naive host by skin-to-oral or skin-to-intranasal routes in the ferret model [126]. In conclusion, ferrets are a good animal model for transmission, evaluating antiviral drugs or vaccines against SARS-CoV-2.

4.5. Cats

Cats are susceptible to SARS-CoV-2, and younger cats are more susceptible. SARS-CoV-2 can replicate efficiently and transmit via airborne droplets in cats (Table 2). Viral presence was identified in upper respiratory tissues (nasal turbinates, soft palate, tonsils), trachea, lungs, and small intestine following infection [57]. Halfmann et al. found that SARS-CoV-2 could replicate in cats and could transmit efficiently between infected cats and naive cats via airborne droplets, but no symptoms were observed [63]. These results are similar to pandemic situations in humans. Therefore, cats are another ideal model for evaluating the transmissibility of SARS-CoV-2. A linear DNA vaccine candidate elicits high RBD-specific antibody levels and T cell response; these results indicate that cats are a good model for developing a pet vaccine against SARS-CoV-2 [64]. Delta variant (B.1.617.2) of SARS-CoV-2 could cause severe clinical respiratory disease and pulmonary lesions even with a lower amount of SARS-CoV-2 [65]. However, Omicron BA.1.1 presented lower virulence in cats than that of D164G- or Delta variants [66]. Recently, Park et al. also found that cats are not susceptible to the Omicron variants but are susceptible to SARS-CoV-2 infection [127]. The positive antibodies against COVID-19 are prevalent in cats, and the cat-human cross-transmission occurs in a timely manner all over the world [128,129,130].

4.6. Minks

4.6.1. Pathogenesis and Transmission of SARS-CoV-2 in Minks

Minks have been reported to be infected with SARS-CoV-2 from humans on two farms in the Netherlands in April and May 2020 [131]. The studies have demonstrated that the virus might be initially introduced by humans, and then spread to minks, and finally transmitted back to humans, based on using whole-genome sequencing of samples collected from minks and workers of outbreaks on 16 farms [67,68]. American mink experimentally challenged with SARS-CoV-2 exhibited severe acute respiratory disease featuring clinical, radiological, and histological pathology [69]. Shuai et al. demonstrated efficient airborne transmission of SARS-CoV-2 in minks, with robust replication throughout the respiratory tract and severe nasal/pulmonary pathology [70]. The natural SARS-CoV-2 infection causes similar symptoms to those of COVID-19 patients. Immunohistochemistry revealed extensive ACE2 receptor expression in turbinate epithelium, with reduced levels in lower respiratory tract epithelium and alveolar macrophages. Correspondingly, IHC and ISH localized the virus predominantly to nasal turbinate epithelial cells. These results provide expanded knowledge of the pathology and pathogenesis of minks infected with SARS-CoV-2, and minks could be used as a potential animal model to study SARS-CoV-2 infection in humans [71]. Minks presented different manifestations infected by different SARS-CoV-2 clusters and dark-colored males, so minks could be a useful animal model for studying the pathogenesis and transmission of SARS-CoV-2 (Table 2).

4.6.2. Regulatory and Ethical Issues in Using Minks for SARS-CoV-2 Study

The use of minks in SARS-CoV-2 research is subject to strict ethical and regulatory constraints due to their high susceptibility and potential for zoonotic transmission and mink-to-human infection [67,70]. The core ethical framework is the 3Rs principle, mandating the Replacement of minks with alternatives where possible, Reduction in animal numbers through rigorous design, and Refinement of protocols to minimize suffering via enriched housing, analgesia, and early humane endpoints regulated by American Veterinary Medical Association)). AVMA Guidelines for the Euthanasia of Animals. Regulatory oversight requires approval from an IACUC, which performs a harm-benefit analysis to ensure compliance with animal welfare laws. Research must also be conducted under high Biosafety Level (BSL-3) containment to prevent SARS-CoV-2 virus escape, safeguarding both animal welfare and public health [132]. In addition, minks are not typical lab animals. Studies that involve intentional infection of minks are heavily scrutinized by departments of agriculture and environment due to the risk of farm outbreaks and potential establishment of an animal reservoir (USDA). This multi-layered governance ensures that any such research is justified, necessary, and conducted to the highest standards of welfare. In conclusion, conducting SARS-CoV-2 research in minks is a highly regulated activity that sits at the intersection of animal welfare law, biosafety, agricultural security, and profound ethical deliberation.

4.7. Other Animals

Fruit bats support limited-duration SARS-CoV-2 infection, exhibiting viral tropism for respiratory mucosa (nasal, tracheal, pulmonary) and bronchial-associated lymphoid tissue (BALT). SARS-CoV-2 transmits limitedly in contact with bats [59]. Wild Mexican free-tailed bats (Tadarida brasiliensis) can be experimentally infected by SARS-CoV-2 and orally shed moderate amounts of virus for up to 18 days post-infection [72]. Recently, a coronavirus (HKU5-CoV-2) isolated from a patient was found to use the human ACE2 as the receptor to enter the cells. Following experimental challenge, fruit bats develop self-resolving infection where the virus is consistently detected in the nasal epithelium, trachea, lung alveoli, and peribronchial lymphoid tissue [73] (Table 2).
Dogs presented low susceptibility to SARS-CoV-2, and SARS-CoV-2 did not transmit among dogs. Virus RNA can only be detected in rectal swabs in partially infected dogs [57]. However, Alberto-Orlando et al. found that SARS-CoV-2 could transmit from infected owners to cats and dogs by food sharing [133], which is consistent with the national surveillance in the U.S. [97].
Pigs exhibit susceptibility to multiple porcine coronaviruses, including TGEV (transmissible gastroenteritis virus), PEDV (porcine epidemic diarrhea virus), PRCV (porcine respiratory coronavirus), PHEV (porcine hemagglutinating encephalomyelitis virus), SADS-CoV (swine acute diarrhea syndrome coronavirus), and PDCoV (porcine delta coronavirus) [134]. However, pigs and chickens are not susceptible to SARS-CoV-2. The virus could not be detected in swabs and organs. The sera samples were also negative for virus-specific antibody detection [59]. Pigs are a good model for evaluating the safety and immunogenicity of a SARS-CoV-2 RBD fusion heterodimer vaccine candidate, PHH-1V [135]. Pigs expressing human ACE2 (hACE2) exhibit susceptibility to SARS-CoV-2 infection and manifest clinical disease resembling COVID-19 [136].
Tigers and three lions have been reported to be infected with SARS-CoV-2 from a human at the Bronx Zoo, New York, in 2020. The infected animals developed mild, abnormal respiratory signs. SARS-CoV-2 RNA has been detected in respiratory secretions and/or feces from all the infected animals [137]. Three Asiatic lions were found to be infected with the delta mutant (Pango lineage B.1.617.2) of SARS-CoV-2 in India [138]. In addition, contact tracing investigation indicates that lion-to-human transmission occurred in a zoo setting in Indiana, USA [139]. Gorillas were detected as SARS-CoV-2 RNA positive in the Rotterdam Zoo, The Netherlands [140]. Animals from the families Procyonidae and Viverridae were also found to be infected with the SARS-CoV-2 virus in a zoo, indicating that these animals are also a potential model for understanding the characteristics of SARS-CoV-2 [141].
White-tailed deer are susceptible to SARS-CoV-2 infection. Several studies reported that the white-tailed deer presented seroconversion of SARS-CoV-2 infection [74]. SARS-CoV-2 evolves faster than that in humans in free-ranging white-tailed deer, which is a new risk for humans and other livestock [75]. And serological surveillance demonstrates that the positive rate is up to 17.2% [76]. Human-to-deer and deer-to-deer transmission caused the pandemic in white-tailed deer of SARS-CoV-2 [77]. SARS-CoV-2 has been transmitted to domestic mammals, including minks and deer. Viral circulation in these hosts drives species-specific adaptations: minks acquire NSP9_G37E, Spike_F486L/N501T/Y453F, ORF3a_L219V; deer evolve NSP3a_L1035F. Fortunately, these mutations did not advance the ability of human-to-human transmission [142].
In 2020, three snow leopards (Panthera uncia) in a zoo in Kentucky were found to be infected with B.1.2 lineage SARS-CoV-2, closely correlated with human strains. This virus may transmit from humans to snow leopards, and then animal-to-animal transmission occurs [143]. Efficient transmission of SARS-CoV-2 between white-tailed deer has been documented [144,145].
Although small ruminants like sheep and goats are lowly susceptible to SARS-CoV-2, one serological surveillance by ELISA and neutralization assay demonstrated that sheep or goats can be naturally infected with SARS-CoV-2 [146]. SARS-CoV-2 infection in cattle, sheep, goats, and dromedary camels was also found in Oman by serological assay [147].

5. Perspective or How to Choose a Suitable Animal Model for Your Research on SARS-CoV-2

The selection of animal models plays a critical role in the evaluation of the pathogenesis of different SARS-CoV-2 strains, the efficacy of vaccines, and antiviral drugs. SARS-CoV-2 variants, WA1/2020, B.1.617.2/Delta, B.1.1.529/Omicron, and BA5.2/Omicron show different outcomes in different animal models. B.1.617.2 (Delta), WA1, and BA.5.2/Omicron exhibit congruent viral kinetics, pathogenicity, and shedding profiles in hamster models, while B.1.617.2 presented the increased pathogenicity in C57BL/6J mice; therefore, the pathogenesis of SARS-CoV-2 is host-dependent [148]. Combined, the animal models were usually used in the evaluation of the efficacy of the vaccine and the effect of antibodies against SARS-CoV-2. The antibody and antibody cocktails could significantly reduce the viral load in the lungs in a hamster model and a macaque model [149,150]. In addition to the presence of entry receptors, animal body temperature and its fluctuations may play some role in the infection and spreading of SARS-CoV-2 [151].
Future technological progress will position advanced 3D culture models—including pulmonary organoids and microfluidic organ chips—as vital platforms for investigating human respiratory disorders, such as COVID-19. These models are closer to reflecting the real host response induced by SARS-CoV-2 infection in human organs than those in animal models, which is crucial for developing more effective vaccines and drugs [152]. An Ex vivo cell model of the human upper respiratory tract is an alternative to the animal model. One study reported that they infected the ex vivo cell model with Alpha and Omicron BA.1 variants for one month, and they could detect viral RNA throughout the infection [153]. The transgenic cell line models are a good alternative to animals. The researcher generated the transgenic DF1 cell line expressing the human ACE2, TMPRSS2, and SARS-CoV-2, which replicated well in this cell line. In addition, avian DF1 cells are engineered to express feline/golden hamster/caprine ACE2-TMPRSS2 receptor complexes. Pigs or horses support the replication of SARS-CoV-2, consistent with the results in vivo [154]. In April 2025, the FDA and NIH announced plans to reduce and finally eliminate the use of animal experiments in the pharmacy and custom development, replacing them with novel methods, such as tissue chip technology, predictive computational simulations, and next-generation in vitro models.
Alternative models based on human biology offer a more direct and often more ethical path to understanding SARS-CoV-2. These four key complement or replacement strategies could be considered. The first method is to use human organoids, including airway, lung, and intestinal organoids grown from stem cells, to study viral tropism, replication, and host response in specific human tissues. These models recapitulate human disease features better than many standard animal models. The second method is organs-on-chips. For example, we can employ lung-on-a-chip devices to model the human alveolar interface and study SARS-CoV-2 infection, immune cell recruitment, and cytokine storm in a dynamic environment that mimics breathing motions. The third strategy is to use human tissue explants. Specifically, we utilize donated human nasal, bronchial, or lung tissues ex vivo to investigate early infection events and test antiviral drug efficacy in authentic human tissue. The last strategy is to apply advanced in silico models. We can use computational biology to model virus–host protein interactions, predict drug effectiveness, and analyze large-scale human genomic data to identify risk factors. These human-based models complement or replace animal studies by providing faster, more human-relevant data for drug and vaccine candidate screening, and for deciphering the mechanisms of SARS-CoV-2 pathogenesis.

Author Contributions

Conceptualization: L.W. and H.F.; investigation: Y.T., X.W., S.L., R.Y., C.L., Y.Y. and S.X.; resources: Y.H.; writing—original draft preparation: L.W.; writing—review and editing, J.S., Y.H. and H.F.; supervision: L.W.; funding acquisition: L.W. and H.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Zhejiang Provincial Natural Science Foundation of China under Grant No. LQ22C180003, and by the National Natural Science Foundation of China under Grant Nos. 32302904 (For L.W.) and 32172893 (For H.F.), by “Pioneer” and “Leading Goose” R&D Program of Zhejiang under Grant No. 2023C02047 and No. 2025C01138, by Science Foundation of Zhejiang Sci-Tech University (ZSTU) under Grant No. 20042220-Y.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Iwendi, C.; Mahboob, K.; Khalid, Z.; Javed, A.R.; Rizwan, M.; Ghosh, U. Classification of COVID-19 individuals using adaptive neuro-fuzzy inference system. Multimed. Syst. 2021, 28, 1223–1237. [Google Scholar] [CrossRef]
  2. Aleebrahim-Dehkordi, E.; Soveyzi, F.; Deravi, N.; Rabbani, Z.; Saghazadeh, A.; Rezaei, N. Human Coronaviruses SARS-CoV, MERS-CoV, and SARS-CoV-2 in Children. J. Pediatr. Nurs. 2021, 56, 70–79. [Google Scholar] [CrossRef]
  3. Arabi, Y.M.; Balkhy, H.H.; Hayden, F.G.; Bouchama, A.; Luke, T.; Baillie, J.K.; Al-Omari, A.; Hajeer, A.H.; Senga, M.; Denison, M.R.; et al. Middle East Respiratory Syndrome. N. Engl. J. Med. 2017, 376, 584–594. [Google Scholar] [CrossRef]
  4. Li, Q.; Guan, X.; Wu, P.; Wang, X.; Zhou, L.; Tong, Y.; Ren, R.; Leung, K.S.M.; Lau, E.H.Y.; Wong, J.Y.; et al. Early Transmission Dynamics in Wuhan, China, of Novel Coronavirus-Infected Pneumonia. N. Engl. J. Med. 2020, 382, 1199–1207. [Google Scholar] [CrossRef] [PubMed]
  5. Raharinirina, N.A.; Gubela, N.; Bornigen, D.; Smith, M.R.; Oh, D.Y.; Budt, M.; Mache, C.; Schillings, C.; Fuchs, S.; Durrwald, R.; et al. SARS-CoV-2 evolution on a dynamic immune landscape. Nature 2025, 639, 196–204. [Google Scholar] [CrossRef] [PubMed]
  6. Ziegler, C.G.K.; Allon, S.J.; Nyquist, S.K.; Mbano, I.M.; Miao, V.N.; Tzouanas, C.N.; Cao, Y.; Yousif, A.S.; Bals, J.; Hauser, B.M.; et al. SARS-CoV-2 Receptor ACE2 Is an Interferon-Stimulated Gene in Human Airway Epithelial Cells and Is Detected in Specific Cell Subsets across Tissues. Cell 2020, 181, 1016–1035.E19. [Google Scholar] [CrossRef] [PubMed]
  7. Li, F.; Li, W.; Farzan, M.; Harrison, S.C. Structure of SARS coronavirus spike receptor-binding domain complexed with receptor. Science 2005, 309, 1864–1868. [Google Scholar] [CrossRef]
  8. Kuba, K.; Imai, Y.; Rao, S.; Gao, H.; Guo, F.; Guan, B.; Huan, Y.; Yang, P.; Zhang, Y.; Deng, W.; et al. A crucial role of angiotensin converting enzyme 2 (ACE2) in SARS coronavirus-induced lung injury. Nat. Med. 2005, 11, 875–879. [Google Scholar] [CrossRef]
  9. van den Brand, J.M.; Haagmans, B.L.; van Riel, D.; Osterhaus, A.D.; Kuiken, T. The pathology and pathogenesis of experimental severe acute respiratory syndrome and influenza in animal models. J. Comp. Pathol. 2014, 151, 83–112. [Google Scholar] [CrossRef]
  10. Kowalczuk, S.; Broer, A.; Tietze, N.; Vanslambrouck, J.M.; Rasko, J.E.; Broer, S. A protein complex in the brush-border membrane explains a Hartnup disorder allele. FASEB J. 2008, 22, 2880–2887. [Google Scholar] [CrossRef]
  11. Carossino, M.; Izadmehr, S.; Trujillo, J.D.; Gaudreault, N.N.; Dittmar, W.; Morozov, I.; Balasuriya, U.B.R.; Cordon-Cardo, C.; Garcia-Sastre, A.; Richt, J.A. ACE2 and TMPRSS2 distribution in the respiratory tract of different animal species and its correlation with SARS-CoV-2 tissue tropism. Microbiol. Spectr. 2024, 12, e0327023. [Google Scholar] [CrossRef]
  12. Coutard, B.; Valle, C.; de Lamballerie, X.; Canard, B.; Seidah, N.G.; Decroly, E. The spike glycoprotein of the new coronavirus 2019-nCoV contains a furin-like cleavage site absent in CoV of the same clade. Antivir. Res. 2020, 176, 104742. [Google Scholar] [CrossRef]
  13. Cantuti-Castelvetri, L.; Ojha, R.; Pedro, L.D.; Djannatian, M.; Franz, J.; Kuivanen, S.; van der Meer, F.; Kallio, K.; Kaya, T.; Anastasina, M.; et al. Neuropilin-1 facilitates SARS-CoV-2 cell entry and infectivity. Science 2020, 370, 856–860. [Google Scholar] [CrossRef]
  14. Daly, J.L.; Simonetti, B.; Klein, K.; Chen, K.E.; Williamson, M.K.; Anton-Plagaro, C.; Shoemark, D.K.; Simon-Gracia, L.; Bauer, M.; Hollandi, R.; et al. Neuropilin-1 is a host factor for SARS-CoV-2 infection. Science 2020, 370, 861–865. [Google Scholar] [CrossRef] [PubMed]
  15. Landolina, N.; Ricci, B.; Veneziani, I.; Alicata, C.; Mariotti, F.R.; Pelosi, A.; Quatrini, L.; Mortari, E.P.; Carsetti, R.; Vacca, P.; et al. TLR2/4 are novel activating receptors for SARS-CoV-2 spike protein on NK cells. Front. Immunol. 2024, 15, 1368946. [Google Scholar] [CrossRef] [PubMed]
  16. Gu, Y.; Cao, J.; Zhang, X.; Gao, H.; Wang, Y.; Wang, J.; He, J.; Jiang, X.; Zhang, J.; Shen, G.; et al. Receptome profiling identifies KREMEN1 and ASGR1 as alternative functional receptors of SARS-CoV-2. Cell Res. 2022, 32, 24–37. [Google Scholar] [CrossRef] [PubMed]
  17. Wang, K.; Chen, W.; Zhang, Z.; Deng, Y.; Lian, J.Q.; Du, P.; Wei, D.; Zhang, Y.; Sun, X.X.; Gong, L.; et al. CD147-spike protein is a novel route for SARS-CoV-2 infection to host cells. Signal Transduct. Target. Ther. 2020, 5, 283. [Google Scholar] [CrossRef]
  18. Loo, L.; Waller, M.A.; Moreno, C.L.; Cole, A.J.; Stella, A.O.; Pop, O.T.; Jochum, A.K.; Ali, O.H.; Denes, C.E.; Hamoudi, Z.; et al. Fibroblast-expressed LRRC15 is a receptor for SARS-CoV-2 spike and controls antiviral and antifibrotic transcriptional programs. PLoS Biol. 2023, 21, e3001967. [Google Scholar] [CrossRef]
  19. Liu, J.; Lu, F.; Chen, Y.; Plow, E.; Qin, J. Integrin mediates cell entry of the SARS-CoV-2 virus independent of cellular receptor ACE2. J. Biol. Chem. 2022, 298, 101710. [Google Scholar] [CrossRef]
  20. Wang, Q.; Ye, S.B.; Zhou, Z.J.; Li, J.Y.; Lv, J.Z.; Hu, B.; Yuan, S.; Qiu, Y.; Ge, X.Y. Key mutations on spike protein altering ACE2 receptor utilization and potentially expanding host range of emerging SARS-CoV-2 variants. J. Med. Virol. 2023, 95, e28116. [Google Scholar] [CrossRef]
  21. Han, P.; Li, L.; Liu, S.; Wang, Q.; Zhang, D.; Xu, Z.; Han, P.; Li, X.; Peng, Q.; Su, C.; et al. Receptor binding and complex structures of human ACE2 to spike RBD from omicron and delta SARS-CoV-2. Cell 2022, 185, 630–640.e10. [Google Scholar] [CrossRef]
  22. Zhou, P.; Yang, X.L.; Wang, X.G.; Hu, B.; Zhang, L.; Zhang, W.; Si, H.R.; Zhu, Y.; Li, B.; Huang, C.L.; et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature 2020, 579, 270–273. [Google Scholar] [CrossRef]
  23. Hassan, A.O.; Case, J.B.; Winkler, E.S.; Thackray, L.B.; Kafai, N.M.; Bailey, A.L.; McCune, B.T.; Fox, J.M.; Chen, R.E.; Alsoussi, W.B.; et al. A SARS-CoV-2 Infection Model in Mice Demonstrates Protection by Neutralizing Antibodies. Cell 2020, 182, 744–753.e4. [Google Scholar] [CrossRef]
  24. Winkler, E.S.; Chen, R.E.; Alam, F.; Yildiz, S.; Case, J.B.; Uccellini, M.B.; Holtzman, M.J.; Garcia-Sastre, A.; Schotsaert, M.; Diamond, M.S. SARS-CoV-2 Causes Lung Infection without Severe Disease in Human ACE2 Knock-In Mice. J. Virol. 2022, 96, e0151121. [Google Scholar] [CrossRef] [PubMed]
  25. Jiang, R.D.; Liu, M.Q.; Chen, Y.; Shan, C.; Zhou, Y.W.; Shen, X.R.; Li, Q.; Zhang, L.; Zhu, Y.; Si, H.R.; et al. Pathogenesis of SARS-CoV-2 in Transgenic Mice Expressing Human Angiotensin-Converting Enzyme 2. Cell 2020, 182, 50–58.e8. [Google Scholar] [CrossRef] [PubMed]
  26. Hou, Y.J.; Okuda, K.; Edwards, C.E.; Martinez, D.R.; Asakura, T.; Dinnon, K.H., 3rd; Kato, T.; Lee, R.E.; Yount, B.L.; Mascenik, T.M.; et al. SARS-CoV-2 Reverse Genetics Reveals a Variable Infection Gradient in the Respiratory Tract. Cell 2020, 182, 429–446.e14. [Google Scholar] [CrossRef] [PubMed]
  27. Dinnon, K.H., 3rd; Leist, S.R.; Schafer, A.; Edwards, C.E.; Martinez, D.R.; Montgomery, S.A.; West, A.; Yount, B.L., Jr.; Hou, Y.J.; Adams, L.E.; et al. Publisher Correction: A mouse-adapted model of SARS-CoV-2 to test COVID-19 countermeasures. Nature 2021, 590, E22. [Google Scholar] [CrossRef]
  28. Nakandakari-Higa, S.; Parsa, R.; Reis, B.S.; de Carvalho, R.V.H.; Mesin, L.; Hoffmann, H.H.; Bortolatto, J.; Muramatsu, H.; Lin, P.J.C.; Bilate, A.M.; et al. A minimally-edited mouse model for infection with multiple SARS-CoV-2 strains. Front. Immunol. 2022, 13, 1007080. [Google Scholar] [CrossRef]
  29. Le Chevalier, F.; Authie, P.; Chardenoux, S.; Bourgine, M.; Vesin, B.; Cussigh, D.; Sassier, Y.; Fert, I.; Noirat, A.; Nemirov, K.; et al. Mice humanized for MHC and hACE2 with high permissiveness to SARS-CoV-2 omicron replication. Microbes Infect. 2023, 25, 105142. [Google Scholar] [CrossRef]
  30. Pons-Grifols, A.; Tarres-Freixas, F.; Perez, M.; Riveira-Munoz, E.; Raich-Regue, D.; Perez-Zsolt, D.; Munoz-Basagoiti, J.; Tondelli, B.; Pradenas, E.; Izquierdo-Useros, N.; et al. A human-ACE2 knock-in mouse model for SARS-CoV-2 infection recapitulates respiratory disorders but avoids neurological disease associated with the transgenic K18-hACE2 model. mBio 2025, 16, e0072025. [Google Scholar] [CrossRef]
  31. Ana-Sosa-Batiz, F.; Verma, S.K.; Shafee, N.; Miller, R.; Conner, C.; Hastie, K.M.; Timis, J.; Maule, E.; Nguyen, M.N.; Tran, L.; et al. A humanised ACE2, TMPRSS2, and FCGRT mouse model reveals the protective efficacy of anti-receptor binding domain antibodies elicited by SARS-CoV-2 hybrid immunity. EBioMedicine 2025, 113, 105619. [Google Scholar] [CrossRef]
  32. Tailor, N.; Warner, B.M.; Griffin, B.D.; Tierney, K.; Moffat, E.; Frost, K.; Vendramelli, R.; Leung, A.; Willman, M.; Thomas, S.P.; et al. Generation and Characterization of a SARS-CoV-2-Susceptible Mouse Model Using Adeno-Associated Virus (AAV6.2FF)-Mediated Respiratory Delivery of the Human ACE2 Gene. Viruses 2022, 15, 85. [Google Scholar] [CrossRef]
  33. Yong, K.S.M.; Anderson, D.E.; Zheng, A.K.E.; Liu, M.; Tan, S.Y.; Tan, W.W.S.; Chen, Q.; Wang, L.F. Comparison of infection and human immune responses of two SARS-CoV-2 strains in a humanized hACE2 NIKO mouse model. Sci. Rep. 2023, 13, 12484. [Google Scholar] [CrossRef]
  34. Alves, R.; Wang, Y.T.; Mikulski, Z.; McArdle, S.; Shafee, N.; Valentine, K.M.; Miller, R.; Verma, S.K.; Batiz, F.A.S.; Maule, E.; et al. SARS-CoV-2 Omicron (B.1.1.529) shows minimal neurotropism in a double-humanized mouse model. Antivir. Res. 2023, 212, 105580. [Google Scholar] [CrossRef] [PubMed]
  35. Sun, R.; Zhao, Z.; Fu, C.; Wang, Y.; Guo, Z.; Zhang, C.; Liu, L.; Zhang, C.; Shu, C.; He, J.; et al. Humanized mice for investigating SARS-CoV-2 lung infection and associated human immune responses. Eur. J. Immunol. 2022, 52, 1640–1647. [Google Scholar] [CrossRef]
  36. Abdelnabi, R.; Foo, C.S.; Kaptein, S.J.F.; Boudewijns, R.; Vangeel, L.; De Jonghe, S.; Jochmans, D.; Weynand, B.; Neyts, J. A SCID Mouse Model To Evaluate the Efficacy of Antivirals against SARS-CoV-2 Infection. J. Virol. 2022, 96, e0075822. [Google Scholar] [CrossRef] [PubMed]
  37. Rodriguez-Rodriguez, B.A.; Ciabattoni, G.O.; Duerr, R.; Valero-Jimenez, A.M.; Yeung, S.T.; Crosse, K.M.; Schinlever, A.R.; Bernard-Raichon, L.; Rodriguez Galvan, J.; McGrath, M.E.; et al. A neonatal mouse model characterizes transmissibility of SARS-CoV-2 variants and reveals a role for ORF8. Nat. Commun. 2023, 14, 3026. [Google Scholar] [CrossRef] [PubMed]
  38. Rosenke, K.; Meade-White, K.; Letko, M.; Clancy, C.; Hansen, F.; Liu, Y.; Okumura, A.; Tang-Huau, T.L.; Li, R.; Saturday, G.; et al. Defining the Syrian hamster as a highly susceptible preclinical model for SARS-CoV-2 infection. Emerg. Microbes Infect. 2020, 9, 2673–2684. [Google Scholar] [CrossRef]
  39. Imai, M.; Iwatsuki-Horimoto, K.; Hatta, M.; Loeber, S.; Halfmann, P.J.; Nakajima, N.; Watanabe, T.; Ujie, M.; Takahashi, K.; Ito, M.; et al. Syrian hamsters as a small animal model for SARS-CoV-2 infection and countermeasure development. Proc. Natl. Acad. Sci. USA 2020, 117, 16587–16595. [Google Scholar] [CrossRef]
  40. Sia, S.F.; Yan, L.M.; Chin, A.W.H.; Fung, K.; Choy, K.T.; Wong, A.Y.L.; Kaewpreedee, P.; Perera, R.; Poon, L.L.M.; Nicholls, J.M.; et al. Pathogenesis and transmission of SARS-CoV-2 in golden hamsters. Nature 2020, 583, 834–838. [Google Scholar] [CrossRef]
  41. Zhang, C.; Guo, Z.; Li, N.; Cui, H.; Meng, K.; Liu, L.; Zhao, L.; Zhang, S.; Qin, C.; Liu, J.; et al. Impact of Prior Infection on Protection and Transmission of SARS-CoV-2 in Golden Hamsters. bioRxiv 2021. [Google Scholar] [CrossRef]
  42. Lee, A.C.; Zhang, A.J.; Chan, J.F.; Li, C.; Fan, Z.; Liu, F.; Chen, Y.; Liang, R.; Sridhar, S.; Cai, J.P.; et al. Oral SARS-CoV-2 Inoculation Establishes Subclinical Respiratory Infection with Virus Shedding in Golden Syrian Hamsters. Cell Rep. Med. 2020, 1, 100121. [Google Scholar] [CrossRef] [PubMed]
  43. Bertzbach, L.D.; Vladimirova, D.; Dietert, K.; Abdelgawad, A.; Gruber, A.D.; Osterrieder, N.; Trimpert, J. SARS-CoV-2 infection of Chinese hamsters (Cricetulus griseus) reproduces COVID-19 pneumonia in a well-established small animal model. Transbound. Emerg. Dis. 2021, 68, 1075–1079. [Google Scholar] [CrossRef] [PubMed]
  44. Castellan, M.; Zamperin, G.; Franzoni, G.; Foiani, G.; Zorzan, M.; Drzewniokova, P.; Mancin, M.; Brian, I.; Bortolami, A.; Pagliari, M.; et al. Host Response of Syrian Hamster to SARS-CoV-2 Infection including Differences with Humans and between Sexes. Viruses 2023, 15, 428. [Google Scholar] [CrossRef]
  45. Wang, C.; Cheng, Z.; Miao, J.; Xue, X.; Dong, Y.; Zhao, L.; Guo, H.; Wang, J.; Wang, Z.; Lu, S.; et al. Genomic-transcriptomic analysis identifies the Syrian hamster as a superior animal model for human diseases. BMC Genom. 2025, 26, 286. [Google Scholar] [CrossRef]
  46. Rockx, B.; Kuiken, T.; Herfst, S.; Bestebroer, T.; Lamers, M.M.; Oude Munnink, B.B.; de Meulder, D.; van Amerongen, G.; van den Brand, J.; Okba, N.M.A.; et al. Comparative pathogenesis of COVID-19, MERS, and SARS in a nonhuman primate model. Science 2020, 368, 1012–1015. [Google Scholar] [CrossRef]
  47. Johnston, S.C.; Ricks, K.M.; Jay, A.; Raymond, J.L.; Rossi, F.; Zeng, X.; Scruggs, J.; Dyer, D.; Frick, O.; Koehler, J.W.; et al. Development of a coronavirus disease 2019 nonhuman primate model using airborne exposure. PLoS ONE 2021, 16, e0246366. [Google Scholar] [CrossRef]
  48. Lu, S.; Zhao, Y.; Yu, W.; Yang, Y.; Gao, J.; Wang, J.; Kuang, D.; Yang, M.; Yang, J.; Ma, C.; et al. Comparison of SARS-CoV-2 infections among 3 species of non-human primates. bioRxiv 2020. [Google Scholar] [CrossRef]
  49. Goncalves, A.; Maisonnasse, P.; Donati, F.; Albert, M.; Behillil, S.; Contreras, V.; Naninck, T.; Marlin, R.; Solas, C.; Pizzorno, A.; et al. SARS-CoV-2 viral dynamics in non-human primates. PLoS Comput. Biol. 2021, 17, e1008785. [Google Scholar] [CrossRef]
  50. Böszörményi, K.P.; Stammes, M.A.; Fagrouch, Z.C.; Kiemenyi-Kayere, G.; Niphuis, H.; Mortier, D.; van Driel, N.; Nieuwenhuis, I.; Zuiderwijk-Sick, E.; Meijer, L.; et al. Comparison of SARS-CoV-2 infection in two non-human primate species: Rhesus and cynomolgus macaques. bioRxiv 2020. [Google Scholar] [CrossRef]
  51. Lu, S.; Zhao, Y.; Yu, W.; Yang, Y.; Gao, J.; Wang, J.; Kuang, D.; Yang, M.; Yang, J.; Ma, C.; et al. Comparison of nonhuman primates identified the suitable model for COVID-19. Signal Transduct. Target. Ther. 2020, 5, 157. [Google Scholar] [CrossRef]
  52. Munster, V.J.; Feldmann, F.; Williamson, B.N.; van Doremalen, N.; Perez-Perez, L.; Schulz, J.; Meade-White, K.; Okumura, A.; Callison, J.; Brumbaugh, B.; et al. Respiratory disease in rhesus macaques inoculated with SARS-CoV-2. Nature 2020, 585, 268–272. [Google Scholar] [CrossRef]
  53. Hartman, A.L.; Nambulli, S.; McMillen, C.M.; White, A.G.; Tilston-Lunel, N.L.; Albe, J.R.; Cottle, E.; Dunn, M.D.; Frye, L.J.; Gilliland, T.H.; et al. SARS-CoV-2 infection of African green monkeys results in mild respiratory disease discernible by PET/CT imaging and shedding of infectious virus from both respiratory and gastrointestinal tracts. PLoS Pathog. 2020, 16, e1008903. [Google Scholar] [CrossRef] [PubMed]
  54. Woolsey, C.; Borisevich, V.; Prasad, A.N.; Agans, K.N.; Deer, D.J.; Dobias, N.S.; Heymann, J.C.; Foster, S.L.; Levine, C.B.; Medina, L.; et al. Establishment of an African green monkey model for COVID-19 and protection against re-infection. Nat. Immunol. 2021, 22, 86–98. [Google Scholar] [CrossRef] [PubMed]
  55. DeGrazia, D.; Miller, F.G. Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Infection Challenge Experiments in Nonhuman Primates: An Ethical Perspective. Clin. Infect. Dis. 2021, 73, 2121–2125. [Google Scholar] [CrossRef] [PubMed]
  56. CDC. Centers for Disease Control and Prevention. Available online: https://www.cdc.gov/importation/laws-regulations/nonhuman-primates-policy-statements.html (accessed on 25 August 2025).
  57. Shi, J.; Wen, Z.; Zhong, G.; Yang, H.; Wang, C.; Huang, B.; Liu, R.; He, X.; Shuai, L.; Sun, Z.; et al. Susceptibility of ferrets, cats, dogs, and other domesticated animals to SARS-coronavirus 2. Science 2020, 368, 1016–1020. [Google Scholar] [CrossRef]
  58. Kim, Y.I.; Kim, S.G.; Kim, S.M.; Kim, E.H.; Park, S.J.; Yu, K.M.; Chang, J.H.; Kim, E.J.; Lee, S.; Casel, M.A.B.; et al. Infection and Rapid Transmission of SARS-CoV-2 in Ferrets. Cell Host Microbe 2020, 27, 704–709.e702. [Google Scholar] [CrossRef]
  59. Schlottau, K.; Rissmann, M.; Graaf, A.; Schon, J.; Sehl, J.; Wylezich, C.; Hoper, D.; Mettenleiter, T.C.; Balkema-Buschmann, A.; Harder, T.; et al. SARS-CoV-2 in fruit bats, ferrets, pigs, and chickens: An experimental transmission study. Lancet Microbe 2020, 1, e218–e225. [Google Scholar] [CrossRef]
  60. Richard, M.; Kok, A.; de Meulder, D.; Bestebroer, T.M.; Lamers, M.M.; Okba, N.M.A.; Fentener van Vlissingen, M.; Rockx, B.; Haagmans, B.L.; Koopmans, M.P.G.; et al. SARS-CoV-2 is transmitted via contact and via the air between ferrets. Nat. Commun. 2020, 11, 3496. [Google Scholar] [CrossRef]
  61. Zumbrun, E.E.; Zak, S.E.; Lee, E.D.; Bowling, P.A.; Ruiz, S.I.; Zeng, X.; Koehler, J.W.; Delp, K.L.; Bakken, R.R.; Hentschel, S.S.; et al. SARS-CoV-2 Aerosol and Intranasal Exposure Models in Ferrets. Viruses 2023, 15, 2341. [Google Scholar] [CrossRef]
  62. Martins, M.; Fernandes, M.H.V.; Joshi, L.R.; Diel, D.G. Age-Related Susceptibility of Ferrets to SARS-CoV-2 Infection. J. Virol. 2022, 96, e0145521. [Google Scholar] [CrossRef] [PubMed]
  63. Halfmann, P.J.; Hatta, M.; Chiba, S.; Maemura, T.; Fan, S.; Takeda, M.; Kinoshita, N.; Hattori, S.I.; Sakai-Tagawa, Y.; Iwatsuki-Horimoto, K.; et al. Transmission of SARS-CoV-2 in Domestic Cats. N. Engl. J. Med. 2020, 383, 592–594. [Google Scholar] [CrossRef] [PubMed]
  64. Conforti, A.; Sanchez, E.; Salvatori, E.; Lione, L.; Compagnone, M.; Pinto, E.; Palombo, F.; D’Acunto, E.; Muzi, A.; Roscilli, G.; et al. A linear DNA encoding the SARS-CoV-2 receptor binding domain elicits potent immune response and neutralizing antibodies in domestic cats. Mol. Ther. Methods Clin. Dev. 2023, 28, 238–248. [Google Scholar] [CrossRef] [PubMed]
  65. Tamil Selvan, M.; Gunasekara, S.; Xiao, P.; Griffin, K.; Cowan, S.R.; Narayanan, S.; Ramachandran, A.; Hagen, D.E.; Ritchey, J.W.; Rudd, J.M.; et al. SARS-CoV-2 (Delta Variant) Infection Kinetics and Immunopathogenesis in Domestic Cats. Viruses 2022, 14, 1207. [Google Scholar] [CrossRef]
  66. Martins, M.; do Nascimento, G.M.; Nooruzzaman, M.; Yuan, F.; Chen, C.; Caserta, L.C.; Miller, A.D.; Whittaker, G.R.; Fang, Y.; Diel, D.G. The Omicron Variant BA.1.1 Presents a Lower Pathogenicity than B.1 D614G and Delta Variants in a Feline Model of SARS-CoV-2 Infection. J. Virol. 2022, 96, e0096122. [Google Scholar] [CrossRef]
  67. Oude Munnink, B.B.; Sikkema, R.S.; Nieuwenhuijse, D.F.; Molenaar, R.J.; Munger, E.; Molenkamp, R.; van der Spek, A.; Tolsma, P.; Rietveld, A.; Brouwer, M.; et al. Transmission of SARS-CoV-2 on mink farms between humans and mink and back to humans. Science 2021, 371, 172–177. [Google Scholar] [CrossRef]
  68. Sharun, K.; Tiwari, R.; Natesan, S.; Dhama, K. SARS-CoV-2 infection in farmed minks, associated zoonotic concerns, and importance of the One Health approach during the ongoing COVID-19 pandemic. Vet Q 2021, 41, 50–60. [Google Scholar] [CrossRef]
  69. Adney, D.R.; Lovaglio, J.; Schulz, J.E.; Yinda, C.K.; Avanzato, V.A.; Haddock, E.; Port, J.R.; Holbrook, M.G.; Hanley, P.W.; Saturday, G.; et al. Severe acute respiratory disease in American mink experimentally infected with SARS-CoV-2. JCI Insight 2022, 7, e159573. [Google Scholar] [CrossRef]
  70. Shuai, L.; Zhong, G.; Yuan, Q.; Wen, Z.; Wang, C.; He, X.; Liu, R.; Wang, J.; Zhao, Q.; Liu, Y.; et al. Replication, pathogenicity, and transmission of SARS-CoV-2 in minks. Natl. Sci. Rev. 2021, 8, nwaa291. [Google Scholar] [CrossRef]
  71. Ritter, J.M.; Wilson, T.M.; Gary, J.M.; Seixas, J.N.; Martines, R.B.; Bhatnagar, J.; Bollweg, B.C.; Lee, E.; Estetter, L.; Silva-Flannery, L.; et al. Histopathology and localization of SARS-CoV-2 and its host cell entry receptor ACE2 in tissues from naturally infected US-farmed mink (Neovison vison). Vet. Pathol. 2022, 59, 681–695. [Google Scholar] [CrossRef]
  72. Hall, J.S.; Hofmeister, E.; Ip, H.S.; Nashold, S.W.; Leon, A.E.; Malave, C.M.; Falendysz, E.A.; Rocke, T.E.; Carossino, M.; Balasuriya, U.; et al. Experimental Infection of Mexican Free-Tailed Bats (Tadarida brasiliensis) with SARS-CoV-2. mSphere 2023, 8, e0026322. [Google Scholar] [CrossRef] [PubMed]
  73. Boukhvalova, M.S.; Mortensen, E.; Caple, J.; Joseph, J.; Sylla, F.; Kamali, A.; Stylos, D.; Lopez, D.; March, T.; Byrd, K.M.; et al. SARS-CoV-2 infection augments species- and age-specific predispositions in cotton rats. Sci. Rep. 2023, 13, 757. [Google Scholar] [CrossRef] [PubMed]
  74. Roundy, C.M.; Nunez, C.M.; Thomas, L.F.; Auckland, L.D.; Tang, W.; Richison, J.J., 3rd; Green, B.R.; Hilton, C.D.; Cherry, M.J.; Pauvolid-Correa, A.; et al. High Seroprevalence of SARS-CoV-2 in White-Tailed Deer (Odocoileus virginianus) at One of Three Captive Cervid Facilities in Texas. Microbiol. Spectr. 2022, 10, e0057622. [Google Scholar] [CrossRef] [PubMed]
  75. McBride, D.S.; Garushyants, S.K.; Franks, J.; Magee, A.F.; Overend, S.H.; Huey, D.; Williams, A.M.; Faith, S.A.; Kandeil, A.; Trifkovic, S.; et al. Accelerated evolution of SARS-CoV-2 in free-ranging white-tailed deer. Nat. Commun. 2023, 14, 5105. [Google Scholar] [CrossRef]
  76. Boley, P.A.; Dennis, P.M.; Faraone, J.N.; Xu, J.; Liu, M.; Niu, X.; Gibson, S.; Hale, V.; Wang, Q.; Liu, S.L.; et al. SARS-CoV-2 Serological Investigation of White-Tailed Deer in Northeastern Ohio. Viruses 2023, 15, 1603. [Google Scholar] [CrossRef]
  77. Willgert, K.; Didelot, X.; Surendran-Nair, M.; Kuchipudi, S.V.; Ruden, R.M.; Yon, M.; Nissly, R.H.; Vandegrift, K.J.; Nelli, R.K.; Li, L.; et al. Transmission history of SARS-CoV-2 in humans and white-tailed deer. Sci. Rep. 2022, 12, 12094. [Google Scholar] [CrossRef]
  78. Bao, L.; Deng, W.; Huang, B.; Gao, H.; Liu, J.; Ren, L.; Wei, Q.; Yu, P.; Xu, Y.; Qi, F.; et al. The pathogenicity of SARS-CoV-2 in hACE2 transgenic mice. Nature 2020, 583, 830–833. [Google Scholar] [CrossRef]
  79. Sun, S.H.; Chen, Q.; Gu, H.J.; Yang, G.; Wang, Y.X.; Huang, X.Y.; Liu, S.S.; Zhang, N.N.; Li, X.F.; Xiong, R.; et al. A Mouse Model of SARS-CoV-2 Infection and Pathogenesis. Cell Host Microbe 2020, 28, 124–133.e4. [Google Scholar] [CrossRef]
  80. Song, I.W.; Washington, M.; Leynes, C.; Hsu, J.; Rayavara, K.; Bae, Y.; Haelterman, N.; Chen, Y.; Jiang, M.M.; Drelich, A.; et al. Generation of a humanized mAce2 and a conditional hACE2 mouse models permissive to SARS-CoV-2 infection. Mamm. Genome 2024, 35, 113–121. [Google Scholar] [CrossRef]
  81. Shuai, H.; Chan, J.F.; Hu, B.; Chai, Y.; Yoon, C.; Liu, H.; Liu, Y.; Shi, J.; Zhu, T.; Hu, J.C.; et al. The viral fitness and intrinsic pathogenicity of dominant SARS-CoV-2 Omicron sublineages BA.1, BA.2, and BA.5. EBioMedicine 2023, 95, 104753. [Google Scholar] [CrossRef]
  82. Lee, N.Y.; Lee, Y.W.; Hong, S.M.; On, D.; Yoon, G.M.; An, S.H.; Nam, K.T.; Seo, J.Y.; Shin, J.S.; Choi, Y.K.; et al. SARS-CoV-2 Omicron variant causes brain infection with lymphoid depletion in a mouse COVID-19 model. Lab. Anim. Res. 2023, 39, 8. [Google Scholar] [CrossRef]
  83. Kenney, A.D.; Zani, A.; Kawahara, J.; Eddy, A.C.; Wang, X.L.; Mahesh, K.C.; Lu, M.; Thomas, J.; Kohlmeier, J.E.; Suthar, M.S.; et al. Interferon-induced transmembrane protein 3 (IFITM3) limits lethality of SARS-CoV-2 in mice. EMBO Rep. 2023, 24, e56660. [Google Scholar] [CrossRef]
  84. Tarres-Freixas, F.; Trinite, B.; Pons-Grifols, A.; Romero-Durana, M.; Riveira-Munoz, E.; Avila-Nieto, C.; Perez, M.; Garcia-Vidal, E.; Perez-Zsolt, D.; Munoz-Basagoiti, J.; et al. Heterogeneous Infectivity and Pathogenesis of SARS-CoV-2 Variants Beta, Delta and Omicron in Transgenic K18-hACE2 and Wildtype Mice. Front. Microbiol. 2022, 13, 840757. [Google Scholar] [CrossRef] [PubMed]
  85. Choi, C.Y.; Gadhave, K.; Villano, J.; Pekosz, A.; Mao, X.; Jia, H. Generation and characterization of a humanized ACE2 mouse model to study long-term impacts of SARS-CoV-2 infection. J. Med. Virol. 2024, 96, e29349. [Google Scholar] [CrossRef] [PubMed]
  86. Yang, B.; Liu, C.; Ju, X.; Wu, B.; Wang, Z.; Dong, F.; Yu, Y.; Hou, X.; Fang, M.; Gao, F.; et al. A tissue specific-infection mouse model of SARS-CoV-2. Cell Discov. 2023, 9, 43. [Google Scholar] [CrossRef]
  87. Liu, K.; Tang, M.; Xu, W.; Meng, X.; Jin, H.; Han, M.; Pu, J.; Li, Y.; Jiao, F.; Sun, R.; et al. An inducible hACE2 transgenic mouse model recapitulates SARS-CoV-2 infection and pathogenesis in vivo. Proc. Natl. Acad. Sci. USA 2023, 120, e2207210120. [Google Scholar] [CrossRef] [PubMed]
  88. Lorenzo, M.M.; Marin-Lopez, A.; Chiem, K.; Jimenez-Cabello, L.; Ullah, I.; Utrilla-Trigo, S.; Calvo-Pinilla, E.; Lorenzo, G.; Moreno, S.; Ye, C.; et al. Vaccinia Virus Strain MVA Expressing a Prefusion-Stabilized SARS-CoV-2 Spike Glycoprotein Induces Robust Protection and Prevents Brain Infection in Mouse and Hamster Models. Vaccines 2023, 11, 1006. [Google Scholar] [CrossRef] [PubMed]
  89. Walls, A.C.; VanBlargan, L.A.; Wu, K.; Choi, A.; Navarro, M.J.; Lee, D.; Avena, L.; Berrueta, D.M.; Pham, M.N.; Elbashir, S.; et al. Distinct sensitivities to SARS-CoV-2 variants in vaccinated humans and mice. Cell Rep. 2022, 40, 111299. [Google Scholar] [CrossRef]
  90. Gu, H.; Chen, Q.; Yang, G.; He, L.; Fan, H.; Deng, Y.Q.; Wang, Y.; Teng, Y.; Zhao, Z.; Cui, Y.; et al. Adaptation of SARS-CoV-2 in BALB/c mice for testing vaccine efficacy. Science 2020, 369, 1603–1607. [Google Scholar] [CrossRef]
  91. Li, Q.; Wu, J.; Nie, J.; Zhang, L.; Hao, H.; Liu, S.; Zhao, C.; Zhang, Q.; Liu, H.; Nie, L.; et al. The Impact of Mutations in SARS-CoV-2 Spike on Viral Infectivity and Antigenicity. Cell 2020, 182, 1284–1294.e9. [Google Scholar] [CrossRef]
  92. Thieulent, C.J.; Dittmar, W.; Balasuriya, U.B.R.; Crossland, N.A.; Wen, X.; Richt, J.A.; Carossino, M. Mouse-Adapted SARS-CoV-2 MA10 Strain Displays Differential Pulmonary Tropism and Accelerated Viral Replication, Neurodissemination, and Pulmonary Host Responses in K18-hACE2 Mice. mSphere 2023, 8, e0055822. [Google Scholar] [CrossRef]
  93. Yan, F.; Li, E.; Wang, T.; Li, Y.; Liu, J.; Wang, W.; Qin, T.; Su, R.; Pei, H.; Wang, S.; et al. Characterization of Two Heterogeneous Lethal Mouse-Adapted SARS-CoV-2 Variants Recapitulating Representative Aspects of Human COVID-19. Front. Immunol. 2022, 13, 821664. [Google Scholar] [CrossRef]
  94. Qin, T.; Shen, B.; Li, E.; Jin, S.; Luo, R.; Zhang, Y.; Qi, J.; Deng, X.; Shi, Z.; Wang, T.; et al. MHC class I links with severe pathogenicity in C57BL/6N mice infected with SARS-CoV-2/BMA8. Virol. J. 2023, 20, 75. [Google Scholar] [CrossRef]
  95. Bader, S.M.; Cooney, J.P.; Sheerin, D.; Taiaroa, G.; Harty, L.; Davidson, K.C.; Mackiewicz, L.; Dayton, M.; Wilcox, S.; Whitehead, L.; et al. SARS-CoV-2 mouse adaptation selects virulence mutations that cause TNF-driven age-dependent severe disease with human correlates. Proc. Natl. Acad. Sci. USA 2023, 120, e2301689120. [Google Scholar] [CrossRef] [PubMed]
  96. Wu, X.; Fang, N.; Liang, Z.; Nie, J.; Lang, S.; Fan, C.; Liang, C.; Huang, W.; Wang, Y. Development of a Bioluminescent Imaging Mouse Model for SARS-CoV-2 Infection Based on a Pseudovirus System. Vaccines 2023, 11, 1133. [Google Scholar] [CrossRef] [PubMed]
  97. Lewis, J.; Zhan, S.; Vilander, A.C.; Fagre, A.C.; Kiaris, H.; Schountz, T. SARS-CoV-2 infects multiple species of North American deer mice and causes clinical disease in the California mouse. bioRxiv 2022. [Google Scholar] [CrossRef]
  98. Griffin, B.D.; Chan, M.; Tailor, N.; Mendoza, E.J.; Leung, A.; Warner, B.M.; Duggan, A.T.; Moffat, E.; He, S.; Garnett, L.; et al. SARS-CoV-2 infection and transmission in the North American deer mouse. Nat. Commun. 2021, 12, 3612. [Google Scholar] [CrossRef]
  99. Li, Q.; Vijaykumar, K.; Phillips, S.E.; Hussain, S.S.; Huynh, N.V.; Fernandez-Petty, C.M.; Lever, J.E.P.; Foote, J.B.; Ren, J.; Campos-Gomez, J.; et al. Mucociliary transport deficiency and disease progression in Syrian hamsters with SARS-CoV-2 infection. JCI Insight 2023, 8, e163962. [Google Scholar] [CrossRef]
  100. Handley, A.; Ryan, K.A.; Davies, E.R.; Bewley, K.R.; Carnell, O.T.; Challis, A.; Coombes, N.S.; Fotheringham, S.A.; Gooch, K.E.; Charlton, M.; et al. SARS-CoV-2 Disease Severity in the Golden Syrian Hamster Model of Infection Is Related to the Volume of Intranasal Inoculum. Viruses 2023, 15, 748. [Google Scholar] [CrossRef]
  101. Fomin, G.; Tabynov, K.; Islamov, R.; Turebekov, N.; Yessimseit, D.; Yerubaev, T. Cytokine response and damages in the lungs of aging Syrian hamsters on a high-fat diet infected with the SARS-CoV-2 virus. Front. Immunol. 2023, 14, 1223086. [Google Scholar] [CrossRef]
  102. Bogard, G.; Barthelemy, J.; Hantute-Ghesquier, A.; Sencio, V.; Brito-Rodrigues, P.; Seron, K.; Robil, C.; Flourens, A.; Pinet, F.; Eberle, D.; et al. SARS-CoV-2 infection induces persistent adipose tissue damage in aged golden Syrian hamsters. Cell Death Dis. 2023, 14, 75. [Google Scholar] [CrossRef]
  103. Rodrigues, C.R.; Aulakh, G.K.; Kroeker, A.; Kulkarni, S.S.; Lew, J.; Falzarano, D.; Singh, B. Recruitment of pulmonary intravascular macrophages in SARS-CoV-2 infected hamsters. Cell Tissue Res. 2025, 400, 1–15. [Google Scholar] [CrossRef]
  104. Lin, D.; Liu, L.; Zhang, M.; Hu, Y.; Yang, Q.; Guo, J.; Guo, Y.; Dai, Y.; Xu, Y.; Cai, Y.; et al. Co-infections of SARS-CoV-2 with multiple common respiratory pathogens in infected patients. Sci. China Life Sci. 2020, 63, 606–609. [Google Scholar] [CrossRef] [PubMed]
  105. Jiang, S.; Liu, P.; Xiong, G.; Yang, Z.; Wang, M.; Li, Y.; Yu, X.J. Coinfection of SARS-CoV-2 and multiple respiratory pathogens in children. Clin. Chem. Lab. Med. 2020, 58, 1160–1161. [Google Scholar] [CrossRef] [PubMed]
  106. Svyatchenko, V.A.; Ternovoi, V.A.; Lutkovskiy, R.Y.; Protopopova, E.V.; Gudymo, A.S.; Danilchenko, N.V.; Susloparov, I.M.; Kolosova, N.P.; Ryzhikov, A.B.; Taranov, O.S.; et al. Human Adenovirus and Influenza A Virus Exacerbate SARS-CoV-2 Infection in Animal Models. Microorganisms 2023, 11, 180. [Google Scholar] [CrossRef]
  107. Ma, J.; Liu, X.; Zhou, M.; Chen, P.; Chen, R.; Wang, J.; Zhu, H.; Wu, K.; Ye, J.; Zhang, Y.; et al. A Heterologous Challenge Rescues the Attenuated Immunogenicity of SARS-CoV-2 Omicron BA.1 Variant in Syrian Hamster Model. J. Virol. 2023, 97, e0168422. [Google Scholar] [CrossRef]
  108. Uematsu, T.; Takai-Todaka, R.; Haga, K.; Kobayashi, H.; Imajima, M.; Kobayashi, N.; Katayama, K.; Hanaki, H. Pharmacological effect of cepharanthine on SARS-CoV-2-induced disease in a Syrian hamster model. J. Infect. Chemother. 2025, 31, 102505. [Google Scholar] [CrossRef]
  109. Davies, E.R.; Ryan, K.A.; Bewley, K.R.; Coombes, N.S.; Salguero, F.J.; Carnell, O.T.; Biddlecombe, S.; Charlton, M.; Challis, A.; Cross, E.S.; et al. The Omicron Sub-Variant BA.4 Displays a Remarkable Lack of Clinical Signs in a Golden Syrian Hamster Model of SARS-CoV-2 Infection. Viruses 2023, 15, 1133. [Google Scholar] [CrossRef]
  110. Cong, Y.; Lee, J.H.; Perry, D.L.; Cooper, K.; Wang, H.; Dixit, S.; Liu, D.X.; Feuerstein, I.M.; Solomon, J.; Bartos, C.; et al. Longitudinal analyses using 18F-Fluorodeoxyglucose positron emission tomography with computed tomography as a measure of COVID-19 severity in the aged, young, and humanized ACE2 SARS-CoV-2 hamster models. Antivir. Res. 2023, 214, 105605. [Google Scholar] [CrossRef]
  111. Reyna, R.A.; Kishimoto-Urata, M.; Urata, S.; Makishima, T.; Paessler, S.; Maruyama, J. Recovery of anosmia in hamsters infected with SARS-CoV-2 is correlated with repair of the olfactory epithelium. Sci. Rep. 2022, 12, 628. [Google Scholar] [CrossRef]
  112. Rasika, S.; Fernandois, D.; Prevot, V. Sowing SARS-CoV-2 to reap neurodegeneration: A hamster study. EBioMedicine 2022, 80, 104071. [Google Scholar] [CrossRef] [PubMed]
  113. Radhakrishnan, R.K.; Kandasamy, M. SARS-CoV-2-Mediated Neuropathogenesis, Deterioration of Hippocampal Neurogenesis and Dementia. Am. J. Alzheimer's Dis. Other Dement. 2022, 37, 15333175221078418. [Google Scholar] [CrossRef]
  114. Kaufer, C.; Schreiber, C.S.; Hartke, A.S.; Denden, I.; Stanelle-Bertram, S.; Beck, S.; Kouassi, N.M.; Beythien, G.; Becker, K.; Schreiner, T.; et al. Microgliosis and neuronal proteinopathy in brain persist beyond viral clearance in SARS-CoV-2 hamster model. EBioMedicine 2022, 79, 103999. [Google Scholar] [CrossRef]
  115. Qiao, W.; Lau, H.E.; Xie, H.; Poon, V.K.; Chan, C.C.; Chu, H.; Yuan, S.; Yuen, T.T.; Chik, K.K.; Tsang, J.O.; et al. SARS-CoV-2 infection induces inflammatory bone loss in golden Syrian hamsters. Nat. Commun. 2022, 13, 2539. [Google Scholar] [CrossRef]
  116. Plunkard, J.; Mulka, K.; Zhou, R.; Tarwater, P.; Zhong, W.; Lowman, M.; Wong, A.; Pekosz, A.; Villano, J. SARS-CoV-2 Variant Pathogenesis Following Primary Infection and Reinfection in Syrian Hamsters. mBio 2023, 14, e0007823. [Google Scholar] [CrossRef]
  117. Yamada, H.; Sasaki, S.I.; Tani, H.; Somekawa, M.; Kawasuji, H.; Saga, Y.; Yoshida, Y.; Yamamoto, Y.; Hayakawa, Y.; Morinaga, Y. A novel hamster model of SARS-CoV-2 respiratory infection using a pseudotyped virus. Sci. Rep. 2022, 12, 11125. [Google Scholar] [CrossRef]
  118. Strbenc, M.; Kuhar, U.; Lainscek, D.; Orehek, S.; Slavec, B.; Krapez, U.; Malovrh, T.; Majdic, G. Rehoming and Other Refinements and Replacement in Procedures Using Golden Hamsters in SARS-CoV-2 Vaccine Research. Animals 2023, 13, 2616. [Google Scholar] [CrossRef]
  119. Williamson, B.N.; Perez-Perez, L.; Schwarz, B.; Feldmann, F.; Holbrook, M.G.; Singh, M.; Lye, D.S.; Babusis, D.; Subramanian, R.; Haddock, E.; et al. Subcutaneous remdesivir administration prevents interstitial pneumonia in rhesus macaques inoculated with SARS-CoV-2. Antivir. Res. 2022, 198, 105246. [Google Scholar] [CrossRef]
  120. Pereira, A.H.; Vasconcelos, A.L.; Silva, V.L.; Nogueira, B.S.; Silva, A.C.; Pacheco, R.C.; Souza, M.A.; Colodel, E.M.; Ubiali, D.G.; Biondo, A.W.; et al. Natural SARS-CoV-2 Infection in a Free-Ranging Black-Tailed Marmoset (Mico melanurus) from an Urban Area in Mid-West Brazil. J. Comp. Pathol. 2022, 194, 22–27. [Google Scholar] [CrossRef]
  121. Ma, Z.M.; Olstad, K.J.; Van Rompay, K.K.A.; Iyer, S.S.; Miller, C.J.; Reader, J.R. Pulmonary lymphoid tissue induced after SARS-CoV-2 infection in rhesus macaques. Front. Immunol. 2025, 16, 1533050. [Google Scholar] [CrossRef]
  122. Nielsen, S.; Alvarez, J.; Bicout, D.J.; Calistri, P.; Canali, E.; Drewe, J.A.; Garin-Bastuji, B.; Rojas, J.L.G.; Gortázar, C.; Herskin, M.; et al. SARS-CoV-2 in animals: Susceptibility of animal species, risk for animal and public health, monitoring, prevention and control. EFSA J. 2023, 21, e07822. [Google Scholar] [CrossRef]
  123. Stittelaar, K.J.; de Waal, L.; van Amerongen, G.; Veldhuis Kroeze, E.J.; Fraaij, P.L.; van Baalen, C.A.; van Kampen, J.J.; van der Vries, E.; Osterhaus, A.D.; de Swart, R.L. Ferrets as a Novel Animal Model for Studying Human Respiratory Syncytial Virus Infections in Immunocompetent and Immunocompromised Hosts. Viruses 2016, 8, 168. [Google Scholar] [CrossRef]
  124. van den Brand, J.M.; Haagmans, B.L.; Leijten, L.; van Riel, D.; Martina, B.E.; Osterhaus, A.D.; Kuiken, T. Pathology of experimental SARS coronavirus infection in cats and ferrets. Vet. Pathol. 2008, 45, 551–562. [Google Scholar] [CrossRef] [PubMed]
  125. Benavides, F.F.W.; Veldhuis Kroeze, E.J.B.; Leijten, L.; Schmitz, K.S.; van Run, P.; Kuiken, T.; de Vries, R.D.; Bauer, L.; van Riel, D. Neuroinvasive and neurovirulent potential of SARS-CoV-2 in the acute and post-acute phase of intranasally inoculated ferrets. PLoS ONE 2025, 20, e0311449. [Google Scholar] [CrossRef]
  126. Shipley, R.; Seekings, A.H.; Byrne, A.M.P.; Shukla, S.; James, J.; Goharriz, H.; Lean, F.Z.X.; Nunez, A.; Fooks, A.R.; McElhinney, L.M.; et al. SARS-CoV-2 infection and transmission via the skin to oro-nasal route with the production of bioaerosols in the ferret model. J. Gen. Virol. 2024, 105, 002022. [Google Scholar] [CrossRef]
  127. Park, E.S.; Kuroda, Y.; Uda, A.; Kaku, Y.; Okutani, A.; Hotta, A.; Tatemoto, K.; Ishijima, K.; Inoue, Y.; Harada, M.; et al. The comparison of pathogenicity among SARS-CoV-2 variants in domestic cats. Sci. Rep. 2024, 14, 21815. [Google Scholar] [CrossRef]
  128. Karavaizoglu, C.; Suleyman, A.; Dolu, K.O.; Yucel, E.; Demirkale, Z.H.; Ozdemir, C.; Tamay, Z.U. Different Aspects of COVID-19: “Stay at Home” Increased Cat and Mite Sensitivity in Preschool Children. Pediatr. Pulmonol. 2025, 60, e27422. [Google Scholar] [CrossRef]
  129. Wee, J.; Chen, J.; Wei, G.W. Preventing future zoonosis: SARS-CoV-2 mutations enhance human-animal cross-transmission. Comput. Biol. Med. 2024, 182, 109101. [Google Scholar] [CrossRef]
  130. Nilsson, M.G.; Santana Cordeiro, M.C.; Goncalves, A.C.A.; Dos Santos Conzentino, M.; Huergo, L.F.; Vicentini, F.; Reis, J.B.L.; Biondo, A.W.; Kmetiuk, L.B.; da Silva, A.V. High seroprevalence for SARS-CoV-2 infection in dogs: Age as risk factor for infection in shelter and foster home animals. Prev. Vet. Med. 2024, 222, 106094. [Google Scholar] [CrossRef]
  131. Oreshkova, N.; Molenaar, R.J.; Vreman, S.; Harders, F.; Oude Munnink, B.B.; Hakze-van der Honing, R.W.; Gerhards, N.; Tolsma, P.; Bouwstra, R.; Sikkema, R.S.; et al. SARS-CoV-2 infection in farmed minks, the Netherlands, April and May 2020. EuroSurveill 2020, 25, 2001005. [Google Scholar] [CrossRef]
  132. Yeh, K.B.; Tabynov, K.; Parekh, F.K.; Mombo, I.; Parker, K.; Tabynov, K.; Bradrick, S.S.; Tseng, A.S.; Yang, J.R.; Gardiner, L.; et al. Significance of High-Containment Biological Laboratories Performing Work During the COVID-19 Pandemic: Biosafety Level-3 and -4 Labs. Front. Bioeng. Biotechnol. 2021, 9, 720315. [Google Scholar] [CrossRef]
  133. Alberto-Orlando, S.; Calderon, J.L.; Leon-Sosa, A.; Patino, L.; Zambrano-Alvarado, M.N.; Pasquel-Villa, L.D.; Rugel-Gonzalez, D.O.; Flores, D.; Mera, M.D.; Valencia, P.; et al. SARS-CoV-2 transmission from infected owner to household dogs and cats is associated with food sharing. Int. J. Infect. Dis. 2022, 122, 295–299. [Google Scholar] [CrossRef]
  134. Thakor, J.C.; Dinesh, M.; Manikandan, R.; Bindu, S.; Sahoo, M.; Sahoo, D.; Dhawan, M.; Pandey, M.K.; Tiwari, R.; Emran, T.B.; et al. Swine coronaviruses (SCoVs) and their emerging threats to swine population, inter-species transmission, exploring the susceptibility of pigs for SARS-CoV-2 and zoonotic concerns. Vet. Q. 2022, 42, 125–147. [Google Scholar] [CrossRef]
  135. Moros, A.; Prenafeta, A.; Barreiro, A.; Perozo, E.; Fernandez, A.; Canete, M.; Gonzalez, L.; Garriga, C.; Pradenas, E.; Marfil, S.; et al. Immunogenicity and safety in pigs of PHH-1V, a SARS-CoV-2 RBD fusion heterodimer vaccine candidate. Vaccine 2023, 41, 5072–5078. [Google Scholar] [CrossRef]
  136. Chau, L.F.; Lillico, S.; Opriessnig, T.; Blake, R.; Tardy, L.; Lee, C.H.; Maxwell, S.; Warren, C.; Thornton, E.; McLaughlin, C.L.; et al. Human ACE2 transgenic pigs are susceptible to SARS-CoV-2 and develop COVID-19-like disease. Nat. Commun. 2025, 16, 766. [Google Scholar] [CrossRef]
  137. McAloose, D.; Laverack, M.; Wang, L.; Killian, M.L.; Caserta, L.C.; Yuan, F.; Mitchell, P.K.; Queen, K.; Mauldin, M.R.; Cronk, B.D.; et al. From People to Panthera: Natural SARS-CoV-2 Infection in Tigers and Lions at the Bronx Zoo. mBio 2020, 11, e02220-20. [Google Scholar] [CrossRef] [PubMed]
  138. Karikalan, M.; Chander, V.; Mahajan, S.; Deol, P.; Agrawal, R.K.; Nandi, S.; Rai, S.K.; Mathur, A.; Pawde, A.; Singh, K.P.; et al. Natural infection of Delta mutant of SARS-CoV-2 in Asiatic lions of India. Transbound. Emerg. Dis. 2022, 69, 3047–3055. [Google Scholar] [CrossRef]
  139. Siegrist, A.A.; Richardson, K.L.; Ghai, R.R.; Pope, B.; Yeadon, J.; Culp, B.; Behravesh, C.B.; Liu, L.; Brown, J.A.; Boyer, L.V. Probable Transmission of SARS-CoV-2 from African Lion to Zoo Employees, Indiana, USA, 2021. Emerg. Infect. Dis. 2023, 29, 1102–1108. [Google Scholar] [CrossRef] [PubMed]
  140. Dusseldorp, F.; Bruins-van-Sonsbeek, L.G.R.; Buskermolen, M.; Niphuis, H.; Dirven, M.; Whelan, J.; Oude Munnink, B.B.; Koopmans, M.; Fanoy, E.B.; Sikkema, R.S.; et al. SARS-CoV-2 in lions, gorillas and zookeepers in the Rotterdam Zoo, the Netherlands, a One Health investigation, November 2021. Euro Surveill 2023, 28, 2200741. [Google Scholar] [CrossRef] [PubMed]
  141. Allender, M.C.; Adkesson, M.J.; Langan, J.N.; Delk, K.W.; Meehan, T.; Aitken-Palmer, C.; McEntire, M.M.; Killian, M.L.; Torchetti, M.; Morales, S.A.; et al. Multi-species outbreak of SARS-CoV-2 Delta variant in a zoological institution, with the detection in two new families of carnivores. Transbound. Emerg. Dis. 2022, 69, e3060–e3075. [Google Scholar] [CrossRef]
  142. Tan, C.W.; Chia, W.N.; Zhu, F.; Young, B.E.; Chantasrisawad, N.; Hwa, S.H.; Yeoh, A.Y.; Lim, B.L.; Yap, W.C.; Pada, S.; et al. SARS-CoV-2 Omicron variant emerged under immune selection. Nat. Microbiol. 2022, 7, 1756–1761. [Google Scholar] [CrossRef]
  143. Wang, L.F.; Tan, C.W.; Chia, W.N.; Zhu, F.; Young, B.; Chantasrisawad, N.; Hwa, S.H.; Yeoh, A.Y.; Lim, B.L.; Yap, W.C.; et al. Differential escape of neutralizing antibodies by SARS-CoV-2 Omicron and pre-emergent sarbecoviruses. Res. Sq. 2022, rs.3.rs-1362541. [Google Scholar] [CrossRef]
  144. Marques, A.D.; Sherrill-Mix, S.; Everett, J.K.; Adhikari, H.; Reddy, S.; Ellis, J.C.; Zeliff, H.; Greening, S.S.; Cannuscio, C.C.; Strelau, K.M.; et al. Multiple Introductions of SARS-CoV-2 Alpha and Delta Variants into White-Tailed Deer in Pennsylvania. mBio 2022, 13, e0210122. [Google Scholar] [CrossRef] [PubMed]
  145. Martins, M.; Boggiatto, P.M.; Buckley, A.; Cassmann, E.D.; Falkenberg, S.; Caserta, L.C.; Fernandes, M.H.V.; Kanipe, C.; Lager, K.; Palmer, M.V.; et al. From Deer-to-Deer: SARS-CoV-2 is efficiently transmitted and presents broad tissue tropism and replication sites in white-tailed deer. PLoS Pathog. 2022, 18, e1010197. [Google Scholar] [CrossRef]
  146. Fusco, G.; Cardillo, L.; Levante, M.; Brandi, S.; Picazio, G.; Napoletano, M.; Martucciello, A.; Fiorito, F.; De Carlo, E.; de Martinis, C. First serological evidence of SARS-CoV-2 natural infection in small ruminants: Brief report. Vet. Res. Commun. 2023, 47, 1741–1748. [Google Scholar] [CrossRef]
  147. El Masry, I.; Al Makhladi, S.; Al Abdwany, M.; Al Subhi, A.; Eltahir, H.; Cheng, S.; Peiris, M.; Gardner, E.; Von Dobschuetz, S.; Soumare, B.; et al. Serological evidence of SARS-CoV-2 infection in dromedary camels and domestic bovids in Oman. Emerg. Microbes Infect. 2023, 12, 2220577. [Google Scholar] [CrossRef]
  148. Toomer, G.; Burns, W.; Garcia, L.; Henry, G.; Biancofiori, A.; George, A.; Duffy, C.; Chu, J.; Sides, M.; Munoz, M.; et al. Characterization of Three Variants of SARS-CoV-2 In Vivo Shows Host-Dependent Pathogenicity in Hamsters, While Not in K18-hACE2 Mice. Viruses 2022, 14, 2584. [Google Scholar] [CrossRef]
  149. Takamatsu, Y.; Imai, M.; Maeda, K.; Nakajima, N.; Higashi-Kuwata, N.; Iwatsuki-Horimoto, K.; Ito, M.; Kiso, M.; Maemura, T.; Takeda, Y.; et al. Highly Neutralizing COVID-19 Convalescent Plasmas Potently Block SARS-CoV-2 Replication and Pneumonia in Syrian Hamsters. J. Virol. 2022, 96, e0155121. [Google Scholar] [CrossRef]
  150. Takeshita, M.; Fukuyama, H.; Kamada, K.; Matsumoto, T.; Makino-Okamura, C.; Uchikubo-Kamo, T.; Tomabechi, Y.; Hanada, K.; Moriyama, S.; Takahashi, Y.; et al. Potent SARS-CoV-2 neutralizing antibodies with therapeutic effects in two animal models. iScience 2022, 25, 105596. [Google Scholar] [CrossRef]
  151. Klestova, Z. Possible spread of SARS-CoV-2 in domestic and wild animals and body temperature role. Virus Res. 2023, 327, 199066. [Google Scholar] [CrossRef]
  152. Plebani, R.; Bai, H.; Si, L.; Li, J.; Zhang, C.; Romano, M. 3D Lung Tissue Models for Studies on SARS-CoV-2 Pathophysiology and Therapeutics. Int. J. Mol. Sci. 2022, 23, 71. [Google Scholar] [CrossRef]
  153. Mathez, G.; Pillonel, T.; Bertelli, C.; Cagno, V. Alpha and Omicron SARS-CoV-2 Adaptation in an Upper Respiratory Tract Model. Viruses 2022, 15, 13. [Google Scholar] [CrossRef] [PubMed]
  154. Kapczynski, D.R.; Sweeney, R.; Spackman, E.; Pantin-Jackwood, M.; Suarez, D.L. Development of an in vitro model for animal species susceptibility to SARS-CoV-2 replication based on expression of ACE2 and TMPRSS2 in avian cells. Virology 2022, 569, 1–12. [Google Scholar] [CrossRef]
Table 1. Comparison of residues in ACE2 involved in contact with RBD of S protein of SARS-CoV-2.
Table 1. Comparison of residues in ACE2 involved in contact with RBD of S protein of SARS-CoV-2.
Species192427283134353738414245798283325329330353354355357
ChickenD-TFEVREDYELNRFETNKNDR
ChimpanzeeSQTFKHEEDYQLLMYQENKGDR
Chinese hamsterSQTFDQEEDYQLLNYQGNKGDR
Chinese tree shrewTEVFKIEEEHQLQRYQDKKNDR
DogSLTFKYEEEYQLLTYQGNKGDR
Domestic catSLTFKHEEEYQLLTYQENKGDR
Duck (Mallard)D-MFEVREDYELNNFEKNKNDR
FerretSLTFKYEEEYQLHTYEQNKRDR
Golden hamsterSQTFDQEEDYQLLNYQENKGDR
Greater horseshoe batSLKFDSEENHQLLNFENNKGDR
Green monkeySQTFKHEEDYQLLMYQENKGDR
Guinea pigFQTFELKEDYQLLAYQKNKNDR
MinkSLTFKYEEEYQLHTYEQNKHDR
MouseSNTFNQEEDYQLTSFQANHGDR
Norway ratSKSFKQEEDYQLINFPTNHGDR
PangolinSETFKSEEEYQLINYQENKHDR
PigSLTFKLEEDYQLITYQNNKGDR
RabbitSLTFKQEEDYQLLTYQENKGDR
Rhesus monkeySQTFKHEEDYQLLMYQENKGDR
Tree shewTDVFKIEEEYQLQRYQDKNNDR
White-foot mouseSQIFKQEEDYQLLNYQENKGDR
White-tail deerSQTFKHEEDYQLMTYQDNKGDR
White-tufted-ear marmosetSQTFKHEEDHELLTYQENKQDR
HumanSQTFKHEEDYQLLMYQENKGDR
Table 2. Summary of advantages and disadvantages of each model for SARS-CoV-2 study.
Table 2. Summary of advantages and disadvantages of each model for SARS-CoV-2 study.
AnimalsClassificationAdvantagesDisadvantagesRefs.
Transgenic miceK18-hACE2, Hfh4-hACE2,
CD147-hACE2, mACE2-mutant, hMHCI-hACE2,
Colla1-hACE2,
hACE2-TMPRSS2-FCGRT, AAV-hACE2, Ad5-hACE2, hCD34(+)-hACE2-NCG, SCID
Low cost and accessible;
Ease to handle and genetic manipulation; susceptible;
characterize the clinical presentation of COVID-19, including weight loss and interstitial pneumonia or severe pneumonia.
Wild-type not infectious;
Time cost for construction;
Not transmissible;
High viral titers in the brain.
[17,24,25,26,27,28,29,30,31,32,33,34,35,36,37]
HamsterSyrian hamster;
Chinese hamster
Low cost and easy to handle;
Susceptible to all VOCs; transmissible;
Present age and sex bias associated with clinical disease; no or mild clinical disease symptoms.
Virus clears rapidly and is nonlethal.
Related reagents shortage.
[38,39,40,41,42,43,44,45]
Nonhuman PrimatesRhesus, Cynomolgus, African green monkeys, marmosetsSusceptible;
Highly similar to humans;
Mild to moderate clinical signs;
Older age is associated with increased disease severity.
High cost;
Shortage;
Ethical issues;
Almost no severe cases.
[46,47,48,49,50,51,52,53,54,55,56]
FerretFerretHighly susceptible and transmissible;
Mild clinical disease;
Aged ferrets lead to greater susceptibility.
Relatively high cost;
Difficult to obtain specific reagents.
[57,58,59,60,61,62]
CatCatSusceptible;
Highly transmissible.
Lack of standard cats;
Reagent shortage.
[57,63,64,65,66]
MinkMinkHighly susceptible and highly transmissible;
No to mild signs up to strains;
The lab is difficult to handle.[67,68,69,70,71]
OthersFruit bats; dogs; pigs; deer, etc.Susceptible and transmissible (deer);
Permissive (Fruit bats).
Low susceptibility (dogs, pigs);
Shortage of standard animals (bats, deer).
[57,59,72,73,74,75,76,77]
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

Wu, L.; Tao, Y.; Wu, X.; Li, S.; Yang, R.; Li, C.; Yao, Y.; Xu, S.; Shu, J.; He, Y.; et al. Current Advances and Applications of Animal Models in SARS-CoV-2 Pathogenesis and Vaccine Development. Microorganisms 2025, 13, 2009. https://doi.org/10.3390/microorganisms13092009

AMA Style

Wu L, Tao Y, Wu X, Li S, Yang R, Li C, Yao Y, Xu S, Shu J, He Y, et al. Current Advances and Applications of Animal Models in SARS-CoV-2 Pathogenesis and Vaccine Development. Microorganisms. 2025; 13(9):2009. https://doi.org/10.3390/microorganisms13092009

Chicago/Turabian Style

Wu, Li, Yingying Tao, Xing Wu, Shaozhen Li, Rui Yang, Chengying Li, Yao Yao, Shijia Xu, Jianhong Shu, Yulong He, and et al. 2025. "Current Advances and Applications of Animal Models in SARS-CoV-2 Pathogenesis and Vaccine Development" Microorganisms 13, no. 9: 2009. https://doi.org/10.3390/microorganisms13092009

APA Style

Wu, L., Tao, Y., Wu, X., Li, S., Yang, R., Li, C., Yao, Y., Xu, S., Shu, J., He, Y., & Feng, H. (2025). Current Advances and Applications of Animal Models in SARS-CoV-2 Pathogenesis and Vaccine Development. Microorganisms, 13(9), 2009. https://doi.org/10.3390/microorganisms13092009

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