Agricultural Reviews

  • Chief EditorPradeep K. Sharma

  • Print ISSN 0253-1496

  • Online ISSN 0976-0741

  • NAAS Rating 4.84

Frequency :
Quarterly (March, June, September & December)
Indexing Services :
AGRICOLA, Google Scholar, CrossRef, CAB Abstracting Journals, Chemical Abstracts, Indian Science Abstracts, EBSCO Indexing Services, Index Copernicus

Comparative Microscopic Pathology of SARS-COVID 19 Infection in Human and Corona Virus Infection in Animals: A Review

N. Pazhanivel1,*
1Department of Veterinary Pathology, Madras Veterinary College, Tamil Nadu Veterinary and Animal Sciences University, Chennai-600 007, Tamil Nadu, India.
COVID 19is also known as Coronavirus disease 2019 and is an extremely contagious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which leads to producing great public health importance worldwide as well as in animals. At present COVID 19 is spreading throughout the world. The microscopical lesions are caused by covid19 is mainly affects upper respiratory tract and coronavirus infection also affects bronchiolar epithelial cells and type II pneumocyte of respiratory tract. Here we review the microscopical lesions caused by SARS COVID 19 in human and corona virus infection in animals is described for the benefit of the public.
COVID-19 is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and is a highly contagious disease of human beings (Wu et al., 2020; Zhou et al., 2020; Zhu et al., 2020). The most common symptoms are fever, cough, headache, gastrointestinal symptoms, liver injury, olfactory and gustatory dysfunctions (Lechien et al., 2020; Lin et al., 2020; Zhang et al., 2020). Computerized tomography (CT) imaging features of the chest are the ground glass opacities in bilateral multiple lobular, consolidation, adjacent pleura thickening and combined linear opacities (Xu et al., 2020). COVID-19 has been considered the sixth public health emergency of international concern (PHEIC) by the World Health Organization (WHO) {Eurosurveillance Editorial Team, 2020).
       
Coronaviruses (CoVs) are belonging to a large group of positive-sense RNA, enveloped viruses is producing a variety of diseases in various mammalian and avian hosts, including humans, cattle, pigs, chickens, cats, mice and many other species (Fehr et al., 2015).
       
The probable natural reservoir host of SARS-CoV-2 was a bat and in this whole-genome was highly similar to a bat coronavirus RaTG13 along with a genome sequence identity of 96.2% (Zhou et al., 2020). SARS-CoV is transmitted to humans through masked palm civets and the previous Middle East respiratory syndrome coronavirus (MERS-CoV) is transmitted dromedary camels (Alagaili et al., 2014). Pangolin might be the potential intermediate host for SARS-CoV-2 due to its multiple lineages of pangolin coronavirus were like SARS-CoV-2 (Lam et al., 2020; Zhang et al., 2020).  The present cases in the human population are mostly occurring along with co-morbid conditions of diabetes, hypertension and heart disease are susceptible to this virus (Wang et al., 2020). SARS-CoV-2 pathogenesis is highly complex with multiple factors leading to severe lung injury followed by virus dissemination to several organs (Jiang and Christine 2007).
       
COVID-19 infection caused aggressive inflammatory reaction due to release of more pro-inflammatory cytokines leads to hyperactive host immune response to the SARS-CoV-2 virus (Dina et al., 2020). SARS-CoV-2 virus is having a spike glycoprotein on the envelope which can bind to specific receptors of ACE2 on the host cell membrane (Li et al., 2003) and it is mainly expressed on type II alveolar epithelial cells, surface of epithelial cells in the oral, nasal mucosa and nasopharynx, with a weak expression indicating that the lungs are the primary target of SARS-CoV-2  (Hamming et al., 2004; Zou et al., 2020) and kidneys, heart and blood vessels, liver, pancreas and also regulates alterations in circulating lymphocytes and the immune system (Huang et al., 2020; Mehta et al., 2020; Xu et al., 2021). Increase in the release of inflammatory cytokines and chemokines in the fluid followed by SARS CoV2 infection leading to acute respiratory distress and multiple organ failure (Deshmukh et al., 2021). 
       
In wild animals, mainly canids had seroprevalence against CCoVs (1.7%), felids to FCoVs (2%) and various bovids to BCoVs (range, 6.6-13.3%) {Evermann et al., 1980; Foreyt and Evermann, 1985; Evermann et al., 1988; Tsunemitsu et al., 1995}.
       
In this review paper, describe the various microscopical lesions of COVID 19 infection and compare the microscopic pathology of coronavirus infection in animals.
 
Microscopic pathology on the respiratory system
 
Human
 
Microscopically lung revealed interalveolar haemorrhages, diffuse alveolar damage, desquamation of alveolar epithelial cells, amphophilc granular cytoplasm, type II pneumocyte hyperplasia,  and hyaline membrane formation (Xu et al., 2020; Cai et al., 2020; Carsana et al., 2020; Fox et al., 2020, Magro et al., 2020; Bradley et al., 2020) interalveolar neutrophilic infiltration, pulmonary congestion, edema (Magro et al., 2020; Bradley et al., 2020), plug formation, microthrombi formation, fibrinoid necrosis of the blood vessels, infiltration of mononuclear cells predominantly lymphocytes (Cai et al., 2020; Kuang et al., 2020; Luo et al., 2020; Xu et al., 2020; Menter et al., 2020; Pernazza et al., 2020; Yao et al., 2020; Zeng et al.,2020) and monocytes (Xu et al., 2020; Pernazza et al., 2020; Konopka et al., 2020; Tian et al., 2020a; Tian et al.,2020b; Yao et al., 2020), presence of syncytial giant cells and interstitial fibrosis (Copin et al., 2020).
       
The microscopical lesions in the infection increase with other comorbid conditions hypertension, chronic kidney disease, obstructive sleep apnea and metabolic diseases like diabetes and obesity (Zhu et al., 2020; Tian et al., 2020a) characterized by intra-alveolar hemorrhages, plug formation, hyaline membrane, type II pneumocyte hyperplasia, fibrinoid necrosis of the small vasculature and abundant intra-alveolar neutrophil infiltration and it was suggestive of bacterial infection leading to  bronchopneumonia (Huang et al., 2020).
 
Animals
 
The microscopical lesion in lungs of felines revealed phlebitis is observed in small to medium sized veins is mediated by virus infected monocytes/macrophage with few T-cells and neutrophils causes progression into granuloma (Kipar et al., 2005). Metalloproteinase-9 enzyme expression was increased by activated monocytes leads to endothelial barrier dysfunction and subsequent extravasation of monocytes causing perivascular and vascular damage (Kipar et al., 2001; Drechsler et al., 2011; Tekes et al., 2016).
       
Histological lesions caused by canine corona virus in lungs of dogs showed fibrinopurulent infiltrate extending from alveoli into bronchioles and bronchi, macrophages with erythrophagocytosis in the alveoli, perivascular serous-fibrinous edema and mural fibrinoid vascular necrosis, with infiltration of monocytes and neutrophils (Evermann et al., 2005; Zappulli et al., 2008).
       
BCoVs infection caused the respiratory lesions of petechiae with mucopurulent material in the trachea and bronchointerstitial pneumonia with intra-bronchiolar syncytial cells (Ellis, 2019), type II pneumocytes hyperplasia (Park et al., 2007).
       
In ferret, the microscopical lesions in lung such as severe lymphoplasmacytic perivasculitis and vasculitis, more type II pneumocytes, macrophages and neutrophils in the alveolar septa and alveolar lumen and a mild peribronchitis (Shi et al., 2020).
       
In wild cats (Felis silvestris), lungs revealed acute inflammation of alveoli, marked intra-alveolar hemorrhage, infiltration of macrophages were seen in alveolar and bronchiolar lumen and in addition to that marginated mononuclear cells were also seen in the pulmonary veins and arterioles (Watt et al., 1993).
       
MHV strain (MHV-S) in mouse produces the following histological lesion such as mild olfactory mucosal necrosis, neuronal necrosis of olfactory bulbs and interstitial pneumonia (Barthold and Smith, 1983; Leibowitz et al., 2010). In the lung revealed hyaline membrane and fibrin deposition, infiltration with lymphocytes, macrophages and neutrophils (Leibowitz et al., 2010).
       
Parker’s Rat Corona Virus (PRC) caused respiratory tract lesions of necrotizing rhinitis, laryngitis, tracheitis, bronchitis, hyperplasia and infiltration of lymphocytes, plasma cells and macrophages (Liang et al., 1995; Bihun and Percy, 1995; Miura et al., 2007; Percy and Barthold, 2016). Interstitial pneumonia characterized by leukocyte infiltration in the alveolar wall and type I pneumocytes hyperplasia was recorded (Funk et al., 2009; Miura et al., 2021). Type II pneumocytes can be a target cell of sialodacryoadenitis virus (SDAV) in respiratory infections and spontaneous animal model to study HCoVs in Rat coronavirus infection (Funk et al., 2009).
       
Experimental infection with SARS-CoV in guinea pigs showed the lesions of alveolar walls thickening due to hyperplasia of type II pneumocyte, fibrin exudate within the interstitium, hyaline membrane formation and scattered fibroblast proliferation in the lungs (Liang et al., 2005).
       
In birds, Infectious bronchitis (IB) is caused by Avian Corona Virus (AvCoVs) (Catroxo et al., 2011) belongs to the Gammacoronaviruses, subgenus Igacovirus (Zhuang et al., 2020). The microscopic lesions of IBV are loss of cilia, sloughing of nasal mucosal epithelial cells, epithelial cell hyperplasia, mucosal gland hypertrophy with heterophilic and lymphocytic infiltration. Pneumonia with hemorrhages, oedema, desquamation, fibrinous exudate with infiltration of lymphocytes and heterophils. Wild birds carry gamma-coronaviruses asymptomatically and it could act as a genetic reservoir for future emerging pathogenic CoVs (Chu et al., 2011). In this regard, surveillance of wild birds is a very important.
 
Microscopic pathology on the cardiovascular system
 
Human
 
Heart revealed mild pericardial oedema (Liu et al., 2020), serosanguinous pericardial effusion (Fox et al., 2020), mild myocardial edema (Yao et al., 2020; Tian et al., 2020b), interstitial fibrosis (Tian et al., 2020b), interstitial mononuclear cells  (Xu et al., 2020; Fox et al., 2020; Yao et al., 2020; Bradley et al., 2020; Tavazzi et al., 2020) and lymphocytic infiltration ( Bradley et al., 2020; Liu et al., 2020; Wichmann et al., 2020). Endothelial cell inflammation was also observed (Varga et al., 2020).
 
Animals
 
Vascular necrosis and necrotizing myocarditis were observed in the heart of mountain lion (Stephenson et al., 2013).
 
Microscopic pathology of blood vessels
 
Human
 
The presence of viral inclusions along with inflammatory cells and apoptotic bodies in the endothelial cells (Xiao et al., 2020). Oedema in the alveolar capillaries and small vessels with the presence of fibrin thrombi, neutrophils and CD61+ megakaryocytes (Fox et al., 2020).
 
Animals
 
Ferret systemic coronavirus showed granulomas around the vessels and includes adventitia of vessels.
       
In wild animals, cheetahs showed histological lesions in blood vessels such as systemic vasculitis and perivasculitis and cheetah virus compared to other FCoV strains did not s show cell fusion in the form of syncytia in in vitro studies (Evermann et al., 1988).
       
In wild cats (Felis silvestris), the following histological lesions of marked thickening of the tunica adventitia, with infiltration of macrophages, lymphocytes and neutrophils in the medium-sized arteries and veins (Watt et al., 1993).
 
Microscopic pathology on the liver and biliary system
 
Human
 
The liver revealed mild steatosis, patchy hepatic necrosis, Kupffer cell hyperplasia, sinusoidal dilatation (Xu et al., 2020; Yao et al., 2020; Tian et al., 2020b; Yao et al., 2020; Bradley et al., 2020; Varga et al., 2020; Schweitzer et al., 2020), lymphocytic infiltration (Xu et al., 2020; Tian et al., 2020a; Bradley et al., 2020) and endotheilitis (Varga et al., 2020).
 
Animals
 
Histological lesions in dogs revealed hepatocellular necrosis (Zappulli et al., 2008).
       
In wild cats (Felis silvestris), the corona virus infection produces the marked thickening of serosal surface by fibrin deposition with diffuse leucocytic infiltration in the liver. Sequential stages of hepatic necrosis associated with syncytial formation were also observed (Watt et al., 1993).
       
In mouse hepatitis virus (MHV- Murine CoV) produces multifocal to coalesce acute necrosis with syncytia in the parenchymal and endothelial cells of liver. In addition to that syncytia was not seen in immunocompetent mice (Percy and Barthold, 2016). Microthrombi in sinusoids (Belouzard et al., 2012).
       
In avian IBV infection caused congestion of the central vein and hepatic sinusoids, hepatocellular degeneration, necrosis, thickening and hyalinization of the portal vein wall and infiltration of lymphocytes and heterophils (Xia et al., 2018; Ghany and Elseddawy, 2019; Jackwood and De Wit, 2020).
 
Microscopic pathology on the kidney
 
Human
 
The SARS COV-2 infected kidney revealed segmental glomerulosclerosis (Menter et al., 2020; Diao et al., 2020), podocyte vacuolation, plasma accumulation in bowman’s space, congestion, arteriosclerosis (Menter et al., 2020; Su et al., 2020; Kissling et al., 2020), proximal tubular epithelial cell degeneration (Bradley et al., 2020),  necrosis, fibrin and hyaline thrombi (Yao et al., 2020; Liu et al., 2020) and interstitial lymphocytic infiltration (Yao et al., 2020; Su et al., 2020).ACE2 is to be upregulated in the COVID 19 patients and tubules showing positive to the immunostaining with SARS-CoV-2 nucleoprotein antibody (Yao et al., 2020).
 
Animals
 
Histological lesions in dogs revealed tubular epithelial cell necrosis (Zappulli et al., 2008).
       
Kidney of IBV infected birds revealed interstitial nephritis with hemorrhages, tubular degeneration and necrosis, edema of Bowman’s capsule and dilated renal tubules with urate crystals, cast and lymphocytic and heterophilic infiltration (Xia et al., 2018; Ghany and Elseddawy, 2019; Jackwood and De Wit, 2020).
 
Microscopic pathology of oesophagus, stomach and intestine
 
Human
 
The oesophagus showed lymphocytic infiltration. Degeneration and necrosis of gastric mucosa, congestion and edema, mononuclear dell infiltration (lymphocytes, monocytes and plasma cells) in the stomach and segmental dilatation, lymphoplasmacytic infiltration in the lamina propria of the intestine (Yao et al., 2020; Tian et al., 2020b; Liu et al., 2020; Xiao et al., 2020). Specific epithelial damage and endotheilitis were also recorded in the gastrointestinal tract.
 
Animals
 
CCoVs infection in dogs revealed chronic lymphoplasmacytic inflammation with mild fibrosis, crypts necrosis and villi necrosis (Zappulli et al., 2008; Evermann et al., 2005).
       
BCoVs infections in cattle produced the lesions of sloughed villi, villous atrophy, villous fusion, increased crypt depth, crypt micro-abscesses, crypt hyperplasia and hemorrhages in the lamina propria with mononuclear cell infiltration (Park et al., 2007; Singh et al., 2020).
       
In equine corona virus infection in equines revealed diffuse necrotizing enteritis (Shi et al., 2020). Hemorrhages, attenuation of villi, loss of mucosal epithelium, crypt micro-abscesses, fibrin and pseudomembrane deposition, often associated with secondary bacterial colonization, infiltration of histiocytes, lymphocytes, neutrophils and eosinophils in the lamina propria and the submucosa along with fibrin thrombi mainly on post capillary submucosal venules (microthrombosis). In addition to that Crypt enterocytes showed single 1.5 to 3 µm diameter, irregularly round intracytoplasmic eosinophilic inclusion bodies within clear vacuoles up to 4 µm diameter and lymphocytolysis of Peyer’s patches of ileum (Giannitti et al., 2015).
       
In Ferret enteric CoV caused the lesions of hyperemic mucosa, atrophy, fusion and blunting and vacuolar degeneration and necrosis of the apical epithelium diffuse lymphocytic enteritis, with villus. The antigen was not localized in the large intestine, lymph nodes, spleen, esophagus, stomach and parotid salivary glands (Wise et al., 2006). Systemic corona virus in ferret showed granuloma in various organs (Murray et al., 2010; Autieri et al., 2015; Lescano et al., 2015; Doria-Torra et al., 2016).
       
In swine corona virus infection in pigs revealed the lesions of villous atrophy and necrosis (Madson et al., 2014), attenuation, swelling, flattening, karyomegaly and cytoplasmic vacuolation of superficial enterocytes (Park and Shin, 2014; Madson et al., 2014; Wang et al., 2016) crypt hyperplasia and syncytia formation (Stevenson et al., 2013) in the intestine.
       
Vasculitis and fibrinoid nesrosis was seen in the small, large intestine and mesenteric lymph node of Mountain lion affected with corona virus infection (Stephenson et al., 2013).
       
In the mouse, the lesions caused by enterotropic MHV strains viz. severe necrotizing enterocolitis, haemorrhages, attenuation of villi, enterocytes and endothelial cells of mesenteric vessels had syncytia, leukocytic infiltration, occasionally hemorrhages and presence of eosinophilic intracytoplasmic inclusions (Percy and Barthold, 2016; Compton et al., 2004).
       
Guinea pigs affected with CoVs revealed the following lesions of blunting, fusion of villi, necrosis and syncytia in the enterocytes of distal ileum (Jaax et al., 1990).
       
In rabbits infected with Rabbit Enteric Corona Virus (RECoV) revealed blunting of villi, vacuolation and necrosis of enterocytes, mucosal edema, neutrophilic and mononuclear cell infiltration. Lymphoid cell necrosis in the lymphoid follicles and increased mitotic figures and more cells in intestinal crypts (Descoteaux and Lussier, 1990; Cavanagh, 2005).
       
In Turkey CoVs produced the lesions in the intestine viz. villi atrophy, multifocal marked pseudostratification of the epithelium, lymphocytic infiltration, lymphoid aggregates in the lamina propria and submucosa, heterophilic and plasmacytic infiltration (Gomaa et al., 2009; Wickramasinghe et al., 2014).
 
Microscopic pathology of brain
 
Human
 
The brain revealed hypoxic ischemia, hyperemia, oedema (Menter et al., 2020) and neuronal degeneration (Solomon et al., 2020; Moriguchi et al., 2020; Mahammedi et al., 2020). Subarachnoid hemorrhages were observed in one case (Bradley et al., 2020). The possible route of entry of the virus into the brain by hematogenous route by breaching blood–brain barrier or retrograde neuronal spread involving olfactory nerves, (Zubair et al., 2020). The possible route of entry of SARS-CoV-1 and MERS-CoV in mice models is olfactory nerve (Agrawal et al., 2015; Solomon et al., 2020).
 
Animals
 
In ECoVs infection in the brain showed Alzheimer Type II astrocytes in the cerebral cortex with hyperammonemia (Giannitti et al., 2015; Fielding et al., 2015). In wild cats, the brain showed dilated perivascular spaces in the white matter (Mwase et al., 2015). IBV affected birds brain revealed neuronal cell degeneration, satellitosis and neuronophagia and perivascular lymphocytic cuffing (Xia et al., 2018; Ghany and Elseddawy, 2019; Jackwood and De Wit, 2020).
 
Microscopic pathology of the spleen
 
Human
 
Spleen revealed atrophy, congestion, hemorrhages, infarction, lymphoid cell depletion and necrosis (Xu et al., 2020; Yao et al., 2020; Wichmann et al., 2020; Chen et al., 2020).
 
Animals
 
Feline corona virus causes blood vascular wall necrosis and sporadic smooth muscle hyperplasia are mainly due to perivascular macrophages (Drechsler et al., 2011) and monocytes rarely infiltrate the wall and usually adhere to endothelial cells (Kipar et al., 2005). Macrophages repaved by B-cells and plasma cells further progresses into the granuloma (Kipar and Meli, 2014). Lymphoid cells mainly T- and B-cells depletion in the Lymphoid tissues and in the splenic red pulp, increase in a number of macrophages were seen (Kipar et al., 2001).
       
CCoVs produced multifocal hyperemia and diffuse severe lymphoid depletion in the spleen, it was also observed in thymus and lymph nodes (Zappulli et al., 2008).
 
IBV affected spleen showed thickening and hyalinization of central arterioles with endotheliosis and lymphoid cell depletion (Xia et al., 2018; Ghany and Elseddawy, 2019; Jackwood and De Wit, 2020).
 
Microscopic pathology of skin
 
Human
 
Dense perivascular lymphoplasmacytic infiltration was seen surrounding the swollen blood vessels with extravasation of red blood cells and intraluminal thrombi (Gianotti et al., 2020; Kolivras et al., 2020). COVID-19 positive patients showed parakeratosis, acanthosis, dyskeratotic keratinocytes, necrotic keratinocytes, acantholytic clefts along lymphocytes satellitisms (Gianotti et al., 2020). The virus reaches the cutaneous tissue through blood vessels. Endothelial cells are expressing more ACE2 and they can easily bind to spike protein and leads to viral invasion into the skin tissue followed by producing pathogenesis. Vasculitis is observed in the viral invasion by the inflammatory cell infiltration.  The immune response begins to activation of Langerhans cells causing cascade reaction (Sungnak et al., 2020).
 
Microscopic pathology of testes and prostate
 
Human
 
Extensive germ cell destruction, basement membrane thickening, peritubular fibrosis, congestion and infiltration of leucocytes (Xu et al., 2006) and prostate showed thrombi (Wichmann et al., 2020). Association of coronavirus family and orchitis in humans has been found (Xu et al., 2006). ACE2 receptor is present in seminiferous tubules, Leydig cells, Sertoli cells and spermatogonia (Li et al., 2020). Histiocytic and lymphocytic infiltration in the testicular tissue (Shen et al., 2020).
 
Microscopic pathology of the adrenal gland
 
Human
 
The adrenal gland revealed systemic angiopathy (Menter et al., 2020).
 
Microscopic pathology of eye
 
Human
 
SARS-COV-2 affected patients revealed conjunctivitis, hyperemia in the conjunctiva and chemosis (Wu et al., 2020).
 
Animals
 
African lions (Panthera leo) revealed the histological lesion of bilateral panuveitis with retinal detachment. Lymphoplasmacytic infiltration with scattered macrophages were seen diffusely or in the small vein area.  The pigmented layer of the iris is lost and melanophages were seen scattered in the thickened iris and retina. Anterior chamber revealed eosinophilic proteinaceous exudate (Mwase et al., 2015).
 
Microscopic pathology of the placenta
 
Human
 
The placenta from covid 19 affected patient revealed inflammatory infiltrates in the placenta and funisitis (Baud et al., 2020). SARS Cov 2 affected placenta showed epithelial damage and infiltration of inflammatory cells (Polak et al., 2020).
 
Discussion
 
Various animal and human diseases caused by corona viruses indicate the widespread prevalence in the ecosystem due to its change in shape, formation of new variant strains leads to new emergence of coronavirus in the world and it is now the recent outbreak is also the evidence of the nature of the coronavirus.  Corona virus affects most of all animals, birds, human beings and this might be caused the control of this disease is very critical and difficult.
       
CoVs is having positive-strand RNA viruses is having the ability to acquire genetic diversity by having some typical features of infidelity of the RNA-dependent RNA polymerase, the high frequency of homologous and heterologous RNA recombination and the large genomes (Gouilh et al., 2011). This kind of genetic variability of this CoVs confirms its high potential of evolution that sometimes allowing them to overcome species barriers and host specificity (Baric et al., 1995; Baric et al., 1997; Thackray and Holmes, 2004).
       
The genomic diversity of CoVs produces their variation in species adaptation related to receptor binding ability, tissue tropism, causing localized to systemic diseases affecting different organs. SARS-CoV uses ACE2 as a receptor and SARS-COV is mainly infects ciliated bronchial epithelial cells and type II pneumocytes, whereas MERS-CoV uses dipeptidyl peptidase 4 (DPP4) and infects non-ciliated bronchial epithelial cells and type II pneumocytes (Cui et al., 2019).
              
The microscopical lesions in the animals and human cases are more or less similar mainly produces vascular thrombosis, fibrinous exudation, syncytia formation, lymphoid organ depletion and intestinal epithelial cell damage. Based on the lesions in animals and human beings the animals could be used as a model, important ring in epidemiological chain to be studied and monitored (Zappulli et al., 2020). The SARS Cov2 infection-causing fatality mainly by DAD, coagulopathy and hemodynamic derangements.
The SARS CoV-2 infection-causing fatality mainly by DAD, coagulopathy and hemodynamic derangements. Pathogenesis, epidemiological, clinicopathological findings are essential to find out the target organ and treat the patient accordingly. The SARS CoV-2 virus is mainly affecting the respiratory system followed by the immune, cardiovascular, kidneys, gastrointestinal tract, testes and nervous system and is more. The findings are more severe in comorbid patients and elderly patients. The microscopical lesions in animals more or less similar to the human cases. In this regard, the animals could be used as an experimental animal model to study SARS CoV-2.
None.

  1. Agrawal, A.S., Garron, T., Tao, X. et al. (2015).  Generation of a transgenic mouse model of middle East respiratory syndrome coronavirus infection and disease. Journal of Virology. 89: 3659-70.

  2. Alagaili, A.N., Briese, T., Mishra, N., Kapoor, V., Sameroff, S.C., Burbelo, P.D. et al. (2014). Middle East respiratory syndrome coronavirus infection in dromedary camels in Saudi Arabia. M Bio. 5: e884-e814. doi: 10.1128/mBio. 00884-14.

  3. Autieri, C.R., Miller, C.L., Scott, K.E., Kilgore, A., Papscoe, V.A., Garner, M.M., Haupt, J.L., Bakthavatchalu, V., Muthupalani,  S., Fox, J.G. (2015). Systemic coronaviral disease in 5 ferrets. Comparative Medicine. 65: 508-516.

  4. Baric, R.S., Fu, K., Chen, W., Yount, B. (1995).  High Recombination and Mutation Rates in Mouse Hepatitis Virus Suggest that Coronaviruses may be Potentially Important Emerging  Viruses. In: Corona and Related Viruses. Advances in Experimental Medicine and Biology; [Talbot, P., Levy, G., (Eds)]; Springer: Boston, MA, USA, pp. 571-576. 

  5. Baric, R.S., Yount, B., Hensley, L., Peel, S.A., Chen, W. (1997).  Episodic evolution mediates interspecies transfer of a murine coronavirus. Journal of Virology. 71: 1946-1955. 

  6. Barthold, S.W. and Smith, A.L. (1983). Mouse hepatitis virus S in weanling Swiss mice following intranasal inoculation. Laboratory Animal Science. 33: 355-360.

  7. Baud, D., Greub, G., Favre, G., Gengler, C., Jaton, K., Dubruc, E. et al. (2020). Second-trimester miscarriage in a pregnant woman with SARSCoV-2 infection. Journal of American Medical Association. 323: 2198-220.

  8. Belouzard, S., Millet, J.K., Licitra, B.N., Whittaker, G.R. (2012). Mechanisms of coronavirus cell entry mediated by the viral spike protein. Viruses. 4: 1011-1033.

  9. Bihun, C.G. and Percy, D.H. (1995). Morphologic changes in the nasal cavity associated with sialodacryoadenitis virus infection in the Wistar rat. Veterinary Pathology. 32: 1-10.

  10. Bradley, B.T., Maioli, H., Johnston, R., Chaudhry, I., Fink, S.L., Xu, H. et al. (2020). Histopathology and ultrastructural findings of fatal COVID-19 infections. Med Rxiv. 2020.04.17.20058545.

  11. Cai, Y., Hao, Z., Gao, Y., Ping, W., Wang, Q., Peng, S. et al. (2020). COVID-19 in the perioperative period of lung resection: A brief report from a single thoracic surgery department in Wuhan, China. Journal of Thoracic Oncology. 15: 1065-72.

  12. Carsana, L., Sonzogni, A., Nasr, A., Rossi, R., Pellegrinelli, A., Zerbi, P. et al. (2020). Pulmonary post-mortem findings in a large series of COVID19 cases from Northern Italy. Med Rxiv. 2020.04.19.200542.

  13. Catroxo, M.H.B., Martins, A.M.C.R.P.F., Petrella, S., Curi, N.A., Melo, N. (2011). Research of viral agent in free-living pigeon feces (Columba livia) in the City of São Paulo, SP, Brazil, for transmission electron microscopy. International  Journal of Morphology. 29: 628-635.

  14. Cavanagh, D. (2005). Coronaviruses in poultry and other birds. Avian Pathology. 34: 439-448.

  15. Chen, Y., Feng, Z., Diao, B., Wang, R., Wang, G., Wang, C. et al. (2020). The novel severe acute respiratory syndrome Coronavirus 2 (SARSCoV-2) directly decimates human spleens and lymph nodes. Med Rxiv. 2020.03.27.20045427.

  16. Chu, D.K.W., Leung, C.Y.H., Gilbert, M., Joyner, P.H., Ng, E.M., Tse, T.M., Guan, Y., Peiris, J.S.M., Poon, L.L.M. (2011). Avian coronavirus in wild aquatic birds. Journal of Virology.  85: 12815-12820.

  17. Compton, S.R., Ball-Goodrich, L.J., Paturzo, F.X., Macy, J.D. (2004).  Transmission of enterotropic mouse hepatitis virus from immunocompetent and immunodeficient mice. Comparative  Medicine. 54: 29-35.

  18. Copin, M.C., Parmentier, E., Duburcq, T., Poissy, J., Mathieu, D. (2020). Time to consider histologic pattern of lung injury to treat critically ill patients with COVID-19 infection. Intensive Care Medicine. 10.1007/s00134-020-06057-8.

  19. Cui, J., Li, F., Shi, Z.L. (2019). Origin and evolution of pathogenic coronaviruses. Nature Reviews Microbiology. 17: 181-192.

  20. Descoteaux, J.P. and Lussier, G. (1990). Experimental infection of young rabbits with a rabbit enteric coronavirus. Canadian Journal of Veterinary Research.  54: 473-476. 

  21. Deshmukh, V., Motwani, R., Kumar, A., Kumari, C., Raza, K. (2021). Histopathological observations in COVID-19: A systematic  review. Journal of clinical Pathology. 74: 76-83.

  22. Diao, B., Wang, C., Wang, R., Feng, Z., Tan, Y., Wang, H. et al. (2020). Human kidney is a target for novel severe acute respiratory syndrome Coronavirus 2 (SARS-CoV-2) infection. Med Rxiv. 2020.03.04.20031120.

  23. Dina, R., Eldin, H.S., Taeimah, M., Khattab, R., Salem, R. (2020). The COVID-19 Cytokine Storm; What We Know So Far. Frontiers in Immunology. https://doi.org/10.3389/fimmu. 2020.01446.

  24. Doria-Torra, G., Vidaña, B., Ramis, A., Amarilla, S.P., Martínez, J. (2016). Coronavirus infection in ferrets: Antigen distribution  and inflammatory response. Veterinary Pathology. 53: 1180-1186. 

  25. Drechsler, Y., Alcaraz, A., Bossong, F.J., Collisson, E.W., Diniz, P.P.V.P. (2011). Feline Coronavirus in multicat environments.  Veterinary Clinics of North America: Small Animal Practice.    41: 1133-1169.

  26. Ellis, J. (2019). What is the evidence that bovine coronavirus is a biologically significant respiratory pathogen in cattle? Canadian Veterinary Journal. 60: 147-152.

  27. Eurosurveillance Editorial Team. (2020). Note from the editors: World Health Organization declares novel coronavirus (2019-nCoV) sixth public health emergency of international  concern. Eurosurveillance. 25: 200131e. doi: 10.2807/ 1560-7917.ES.2020.25.5.200131e.

  28. Evermann, J.F., Foreyt, W., Maag-Miller, L., Leathers, C.W., McKeirnan,  A.J., LeaMaster, B. (1980). Acute hemorrhagic enteritis associated with canine coronavirus and parvovirus infections in a captive coyote population. Journal of the American Medical Association. 177: 784-786. 

  29. Evermann, J.F., Heeney, J.L., Roelke, M.E., McKeirnan, A.J., O’Brien, S.J. (1988). Biological and pathological consequences  of feline infectious peritonitis virus infection in the cheetah.  Archives of Virology. 102: 155-171.

  30. Evermann, J.F., Abbott, J.R., Han, S. (2005).Canine coronavirus- associated puppy mortality without evidence of concurrent  canine parvovirus infection. The Journal of Veterinary Diagnostic Investigation. 17: 610-614. 

  31. Fehr, A.R. and Perlman, S. (2015). Coronaviruses: An overview of their replication and pathogenesis. In: Coronaviruses: Methods and Protocols.  p. 1-23. doi: 10.1007/978-1- 4939-2438-7_1.

  32. Fielding, C.L., Higgins, J.K., Higgins, J.C., Mcintosh, S., Scott, E., Giannitti, F., Mete, A., Pusterla, N. (2015). Disease associated with equine coronavirus infection and high case fatality rate. Journal of Veterinary Internal Medicine. 29: 307-310.

  33. Foreyt, W.J. and   Evermann, J.F. (1985). Serologic survey of canine coronavirus in wild coyotes in the western United States, 1972-1982. Journal of Wildlife Diseases. 21: 428-430. 

  34. Fox, S.E., Akmatbekov, A., Harbert, J.L., Li, G., Brown, J.Q., Heide, S.V. (2020). Pulmonary and cardiac pathology in Covid- 19: The first autopsy series from New Orleans. Med Rxiv. 2020.04.06.20050575.

  35. Funk, C.J., Manzer, R., Miura, T.A., Groshong, S.D., Ito, Y., Travanty, E.A., Leete, J., Holmes, K.V., Mason, R.J. (2009). Rat respiratory coronavirus infection: Replication in airway and alveolar epithelial cells and the innate immune response. Journal of General Virology. 90: 2956-2964.

  36. Ghany, H.M.A. and Elseddawy, N.M. (2019). Diagnostic studies of infectious bronchitis disease in broilers using pathological and molecular investigations in Kaliobeya Governorate, egypt. Advances in Environmental Biology. 13: 1-6.

  37. Giannitti, F., Diab, S., Mete, A., Stanton, J.B., Fielding, L., Crossley, B., Sverlow, K., Fish, S., Mapes, S., Scott, L. et al. (2015). Necrotizing enteritis and hyperammonemic encephalopathy  associated with equine coronavirus infection in equids. Veterinary Pathology. 52: 1148-1156.

  38. Gianotti, R., Veraldi, S., Recalcati, S., Cusini, M., Ghislanzoni, M., Boggio, F. et al. (2020) Cutaneous clinico-pathological findings in three COVID-19-positive patients observed in the metropolitan area of milan, Italy. Acta Dermatolo- Venereologica. 100(8): adv00124. doi: 10.2340/000 15555-3490.

  39. Gomaa, M.H., Yoo, D., Ojkic, D., Barta, J.R. (2009). Infection with a pathogenic turkey coronavirus isolate negatively affects growth performance and intestinal morphology of young turkey poults in Canada. Avian Pathology. 38: 279-286.

  40. Gouilh, M.A., Puechmaille, S.J., Gonzalez, J.P., Teeling, E., Kittayapong,  P., Manuguerra, J.C. (2011). SARS-Coronavirus ancestor’s  foot-prints in South-East Asian bat colonies and the refuge theory. Infection Genetics and Evolution. 11: 1690-1702.

  41. Hamming, I., Timens, W., Bulthuis, M., Lely, A., Navis, G., van Goor, H. (2004). Tissue distribution of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding SARS pathogenesis. Journal of Pathology. 203: 631-7.

  42. Huang, C., Wang, Y., Li, X. et al. (2020). Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 395: 497-506.

  43. Jaax, G.P., Jaax, N.K., Petrali, J.P., Corcoran, K.D., Vogel, A.P. (1990). Coronavirus-like virions associated with a wasting syndrome in guinea pigs. Laboratory Animal Science. 40: 375-378.

  44. Jackwood, M. and De Wit, S. (2020). Infectious Bronchitis. In: Diseases of Poultry; [Swayne, D., (Ed)]; John Wiley and Sons: Hoboken, NJ, USA, pp. 167-188.

  45. Jiang, G. and Korteweg, C. (2007). Pathology and pathogenesis of severe acute respiratory syndrome. The American Journal  of Pathology. 170(4): 1136-1147. 

  46. Kipar, A. and Meli, M.L. (2014). Feline infectious peritonitis: Still an enigma? Veterinary Pathology. 51: 505-526. 

  47. Kipar, A., Köhler, K., Leukert, W., Reinacher, M.A. (2001). Comparison  of lymphatic tissues from cats with spontaneous feline infectious peritonitis (FIP), cats with FIP virus infection but no FIP and cats with no infection. The Journal of  Comparative Pathology. 125: 182-191.

  48. Kipar, A., May, H., Menger, S., Weber, M., Leukert, W., Reinacher, M. (2005). Morphologic features and development of granulomatous vasculitis in feline infectious peritonitis. Veterinary Pathology. 42: 321-330. 

  49. Kissling, S., Rotman, S., Gerber, C., Halfon, M., Lamoth, F., Comte, D. et al. (2020). Collapsing glomerulopathy in a COVID- 19 patient. Kidney International. 10.1016/j.kint.2020.04.006.

  50. Kolivras, A., Dehavay, F., Delplace, D., Feoli, F., Meiers, I, Milone, L. et al.(2020). Coronavirus (COVID-19) infection-induced chilblains: A case report with histopathological findings. JAAD Case Reports. 6: 489-92.

  51. Konopka, K.E., Wilson, A., Myersm, J.L. (2020). Postmortem lung findings in an asthmatic with Coronavirus disease 2019 (COVID-19). Chest. 10.1016/j.chest.2020.04.032.

  52. Kuang, D., Xu, S.P., Hu, Y., Liu, C., Duan, Y.Q., Wang, G.P. (2020). The pathological changes and related studies of novel coronavirus infected surgical specimen. Zhonghuabing Li Xue Za Zhi. 49: E008.

  53. Lam, T.T., Shum, M.H., Zhu, H.C., Tong, Y.G., Ni, X.B., Liao, Y.S. et al. (2020). Identifying SARS-CoV-2 related coronaviruses  in Malayan pangolins. Nature. 583: 282-285. doi: 10.1038/ s41586-020-2169-0.

  54. Lechien, J.R., Chiesa-Estomba, C.M., De Siati, D.R., Horoi, M., Bon, S.D.L., Rodriguez, A. et al. (2020). Olfactory and gustatory dysfunctions as a clinical presentation of mild- to-moderate forms of the coronavirus disease (COVID- 19): A multicenter European study. European Archives of Oto-rhino-laryngology. 277: 2251-2261. doi: 10.1007/ s00405-020-05965-1.

  55. Leibowitz, J.L., Srinivasa, R., Williamson, S.T., Chua, M.M., Liu, M., Wu, S., Kang, H., Ma, X.-Z., Zhang, J., Shalev, I. et al. (2010). Genetic Determinants of mouse hepatitis virus strain 1 pneumovirulence. Journal of Virology. 84: 9278- 9291.

  56. Lescano, J., Quevedo, M., Gonzales-Viera, O., Luna, L., Keel, M.K., Gregori, F. (2015). First case of systemic coronavirus infection in a domestic ferret (Mustela putorius furo) in peru. Transboundary and Emerging Diseases. 62: 581-585.

  57. Li, M.Y., Li, L., Zhang, Y. et al. (2020).  Expression of the SARS- CoV-2 cell receptor gene ACE2 in a wide variety of human tissues. Infectious Diseases of Poverty. 9: 45. doi: 10.1186/s40249-020-00662-x.

  58. Li, W., Moore, M., Vasilieva, N., Sui, J., Wong, S., Berne, M, (2003).  Angiotensin converting enzyme 2 is a functional receptor for the SARS coronavirus. Nature. 426: 450-4.

  59. Liang, L., He, C., Lei, M., Li, S., Hao, Y., Zhu, H., Duan, Q. (2005). Pathology of guinea pigs experimentally infected with a novel reovirus and coronavirus isolated from SARS patients. DNA Cell Biology. 24: 485-490.

  60. Liang, S.C., Schoeb, T.R. Davis, J.K., Simecka, J.W., Cassell, G.H., Lindsey, J.R. (1995). Comparative severity of respiratory lesions of sialodacryoadenitis virus and Sendai virus infections in LEW and F344 rats. Veterinary Pathology. 32: 661-667. 

  61. Lin, L., Jiang, X., Zhang, Z., Huang, S., Zhang, Z., Fang, Z. et al. (2020). Gastrointestinal symptoms of 95 cases with SARS-CoV-2 infection. Gut. 69: 997-1001. doi: 10.1136/ gutjnl-2020-321013.

  62. Liu, Q., Wang, R.S., Qu, G.Q., Wang, Y.Y., Liu, P., Zhu, Y.Z. (2020).  Gross examination report of a COVID-19 death autopsy. Fa Yi Xue Za Zhi. 36: 21-3.

  63.  Luo, W., Yu, H., Gou, J., Li, X., Sun, Y., Li, J. et al. (2020). Clinical pathology of critical patient with novel coronavirus pneumonia  (COVID-19). Preprints. 2020020407.

  64. Madson, D.M., Magstadt, D.R., Arruda, P.H.E., Hoang, H., Sun, D., Bower, L.P., Bhandari, M., Burrough, E.R., Gauger, P.C., Pillatzki, A.E. et al. (2014). Pathogenesis of porcine epidemic diarrhea virus isolate (US/Iowa/18984/2013) in 3-week-old weaned pigs. Veterinary Microbiology. 174: 60-68.  

  65. Magro, C., Mulvey, J.J., Berlin, D., Nuovo, G., Salvatore, S., Harp, J. et al. (2020). Complement associated microvascular injury and thrombosis in the pathogenesis of severe COVID-19 infection: a report of five cases. Translational Research. S1931-5244:30070-0.

  66. Mahammedi, A., Saba L., Vagal, A. et al. (2020). Imaging in neurological  disease of hospitalized COVID-19 patients: An Italian multicenter retrospective observational study. Radiology. 201933:201933.

  67. Mehta, P., McAuley, D.F., Brown, M. et al. (2020). COVID-19: Consider cytokine storm syndromes and immunosuppression.  Lancet. 395: 1033-4.

  68. Menter, T., Haslbauer, J.D., Nienhold, R., Savic, S., Hopfer, H., Deigendesch, N. (2020). Posyt-mortem examination of COVID19 patients reveals diffuse alveolar damage with severe capillary congestion and variegated findings of lungs and other organs suggesting vascular dysfunction. Histopathology. 10.1111/his.14134.

  69. Miura, T.A., Wang, J., Holmes, K.V., Mason, R.J. (2007). Rat coronaviruses infect rat alveolar type I epithelial cells and induce expression of CXC chemokines. Virology. 369: 288-298. 

  70. Miura, F., Kitajima, M., Omori, R. (2021). Duration of SARS-CoV-2 viral shedding in faeces as a parameter for wastewater- based epidemiology: Re-analysis of patient data using a shedding dynamics model. Science of the Total Environment.  https://doi.org/10.1016/j.scitotenv.2020.144549.

  71. Moriguchi, T., Harii, N., Goto, J. et al. (2020). A first case of meningitis/ encephalitis associated with SARS-Coronavirus-2. International Journal of Infectious Diseases. 94: 55-8.

  72. Murray, J.,Kiupel, M., Maes, R.K. (2010). Ferret coronavirus-associated diseases. Veterinary Clinics: Exotic Animal Practice. 13: 543-560.

  73. Mwase, M., Shimada, K., Mumba, C., Yabe, J., Squarre, D., Madarame,  H. (2015).  Positive immunolabelling for feline infectious peritonitis in an african lion (Pantheraleo) with bilateral panuveitis. Journal of Comparative Pathology. 152: 265- 268.

  74. Park, J. and Shin, H. (2014). Porcine epidemic diarrhea virus infects and replicates in porcine alveolar macrophages. Virus Research. 191: 143-152.

  75. Park, S.J., Kim, G.Y., Choy, H.E., Hong, Y.J., Saif, L.J., Jeong, J.H., Park, S.I., Kim, H.H., Kim, S.K., Shin, S.S. et al. (2007). Dual enteric and respiratory tropisms of winter dysentery bovine coronavirus in calves. Archives of Virology.  152: 1885-1900.

  76. Percy, D.H. and Barthold, S.W. (2016). Pathology of Laboratory Rodents and Rabbits, 4th ed.; [Barthold, S.W., Griffey, S.M., Percy, D.H. (Eds)]; John Wiley and Sons, Inc. Chichester, UK, ISBN 9781118924051.

  77. Pernazza, A., Mancini, M., Rullo, E., Bassi, M., De Giacomo, T., Rocca, C.D. et al. (2020).  Early histologic findings of pulmonary SARSCoV-2 infection detected in a surgical specimen. Virchow’s Archive. 1-6.

  78. Polak, S.B., Gool, I.C.V., Cohen, D., Von der Thüsen, J.H.J., Paasse, J.V. (2020). A systematic review of pathological findings in COVID-19: A pathophysiological timeline and possible mechanisms of disease progression. Modern Pathology. 33: 2128-2138 https://doi.org/10.1038/ s41379-020-0603-3.

  79. Schweitzer, W., Ruder, T., Baumeister, R., Bolliger, S., Thali, M., Meixner, E. et al. (2020). Implications for forensic death investigations from first Swiss case of non-hospital treatment with COVID-19. Forensic Imaging. 21: 200378.

  80. Shen, Q., Xiao, X., Aierken, A. et al. (2020). The ACE2 expression in Sertoli cells and germ cells may cause male reproductive  disorder after SARS-CoV-2 infection. Journal of Cellular and Molecular Medicine. 24(16): 9472-9477. doi: 10.1111/ jcmm.15541.

  81. Shi, J., Wen, Z., Zhong, G., Yang, H., Wang, C., Huang, B., Liu, R., He, X., Shuai, L., Sun, Z. et al. (2020). Susceptibility of ferrets, cats, dogs and other domesticated animals to SARS-coronavirus 2. Science. 368: 1016-1020.

  82. Singh, S., Singh, R., Singh, K.P., Singh, V., Malik, Y.P.S., Kamdi, B., Singh, R., Kashyap, G. (2020). Immunohistochemical and molecular detection of natural cases of bovine rotavirus and coronavirus infection causing enteritis in dairy calves. Microbial Pathogenesis. 138: 103814.

  83. Solomon, I.H., Normandin, E., Bhattacharyya, S., Mukerji, S.S., Keller, K., Ali, A.S. et al. (2020). Neuropathological features of Covid-19. New England Journal of Medicine. 383(10): 989-92. 

  84. Stephenson, N., Swift, P., Moeller, R.B., Worth, S.J., Foley, J. (2013). Feline infectious peritonitis in a mountain lion (Puma concolor), California, USA. Journal of Wil life Diseases. 49: 408-412.

  85. Stevenson, G.W., Hoang, H., Schwartz, K.J., Burrough, E.R., Sun, D., Madson, D., Cooper, V.L., Pillatzki, A., Gauger, P., Schmitt, B.J. (2013). Emergence of porcine epidemic diarrhea virus in the United States: Clinical signs, lesions and viral genomic sequences. Journal of Veterinary Diagnostic Investigation. 25: 649-654.

  86. Su, H., Yang, M., Wan, C. et al. (2020). Renal histopathological analysis of 26 postmortem findings of patients with COVID-19 in China. Kidney International. 98: 219-27.

  87. Sungnak, W., Huang, N., Bécavin, C. et al. (2020). SARS-CoV-2 entry factors are highly expressed in nasal epithelial cells together with innate immune genes. Nature Medicine. 26: 681-7.

  88. Tavazzi, G., Pellegrini, C., Maurelli, M., Belliato, M., Sciutti, F., Bottazzi,  A. et al. (2020). Myocardial localization of coronavirus in COVID19 cardiogenic shock. European Journal of Heart Failure. 22: 911-5.

  89. Tekes, G. and Thiel, H.J. (2016).  Feline Coronaviruses: Pathogenesis  of Feline Infectious Peritonitis, 1st ed.; Elsevier Inc. Amsterdam,  The Netherlands, Volume 96.

  90. Thackray, L.B. and Holmes, K.V. (2004). Amino acid substitutions and an insertion in the spike glycoprotein extend the host range of the murine coronavirus MHV-A59. Virology. 324: 510-524.

  91. Tian, S., Hu, W., Niu, L., Liu, H., Xu, H., Xiao, S.Y. (2020a).  Pulmonary  pathology of early-phase 2019 novel Coronavirus (COVID-19)  pneumonia in two patients with lung cancer. Journal of Thoracic Oncology. 15: 700-4.

  92. Tian, S., Xiong, Y., Liu, H., Niu, L., Guo, J., Liao, M. et al. (2020b). Pathological study of the 2019 novel coronavirus disease (COVID-19) through postmortem core biopsies. Modern Pathology. 10.1038/s41379- 020-0536-x.

  93. Tsunemitsu, H., El-Kanawati, Z.R., Smith, D.R., Reed, H.H., Saif, L.J. (1995). Isolation of coronaviruses antigenically indistinguishable from bovine coronavirus from wild ruminants with diarrhea. Journal of Clinical Microbiology. 33: 3264-3269.

  94. Varga, Z., Flammer, A.J., Steiger, P., Haberecker, M. andermatt, R., Zinkernagel, A.S. et al. (2020).  Endothelial cell infection and endotheliitis in COVID-19. Lancet. 395: 1417-8.

  95. Wang, D., Hu, B., Hu, C., Zhu, F., Liu, X., Zhang, J. et al. (2020). Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus infected pneumonia in Wuhan, China. Journal of the American Medical Association. 323: 1061-1069. DOI: 10.1001/ jama.2020.1585.

  96. Wang, L., Hayes, J., Sarver, C., Byrum, B., Zhang, Y. (2016). Porcine  deltacoronavirus: Histological lesions and genetic characterization.  Archives of Virology. 161: 171-175.

  97. Wang, W., Xu, Y., Gao, R., Lu, R., Han, K., Wu,G. et al. (2020).  Detection of SARS-CoV-2 in different types of clinical specimens. Journal of American Medical Association. 323: 1843-4.

  98. Watt, N.J., MacIntyre, N.J., McOrist, S. (1993). An extended outbreak of infectious peritonitis in a closed colony of european wildcats (Felissilvestris). Journal of Comparative  Pathology. 108: 73-79.

  99. Wichmann, D., Sperhake, J-P., Lütgehetmann, M., Steurer, S., Edler, C., Heinemann, A. et al. (2020). Autopsy findings and venous thromboembolism in patients With COVID- 19: A prospective cohort study. Annals of Internal Medicine.  M20-2003.

  100. Wickramasinghe, I.N.A., van Beurden, S.J., Weerts, E.A.W.S., Verheije, M.H. (2014). The avian coronavirus spike protein.  Virus Research. 194: 37-48.

  101. Wise, A.G., Kiupel, M., Maes, R.K. (2006). Molecular characterization  of a novel coronavirus associated with epizootic catarrhal enteritis (ECE) in ferrets. Virology. 349: 164-174.

  102. Wu, F., Zhao, S., Yu, B., Chen, Y. M., Wang, W., Song, Z. G. Yi, Hu., Zhao-Wu, Tao., Jun-Hua, T., Yuan-Yuan, P.  et al. (2020). A new coronavirus associated with human respiratory disease in China. Nature. 579: 265-269. doi: 10.1038/s41586-020-2008-3. 

  103. Xia, J., He, X., Du, L.J., Liu, Y.Y., You, G.J., Li, S.Y., Liu, P., Cao, S.J., Han, X.F., Huang, Y. (2018). Preparation and protective efficacy of a chicken embryo kidney cell- attenuation GI-19/QX-like avian infectious bronchitis virus vaccine. Vaccine. 36: 4087-4094.

  104. Xia, J., He, X., Du, L.J., Liu, Y.Y., You, G.J., Li, S.Y., Liu, P., Cao, S.J., Han, X.F., Huang, Y. (2018). Preparation and protective  efficacy of a chicken embryo kidney cell-attenuation GI- 19/QX-like avian infectious bronchitis virus vaccine. Vaccine. 36: 4087-4094.

  105. Xiao, F., Tang, M., Zheng, X. et al. (2020). Evidence for gastrointestinal  infection of SARS-CoV-2. Gastroenterology.158: 1831-3.

  106. Xu, S.P., Kuang, D., Hu, Y., Liu, C., Duan, Y.Q., Wang, G.P. (2020).  Detection of 2019-nCoV in the pathological paraffin embedded tissue. Zhonghuabing Li Xuezazhi. 49: E004.

  107. Xu, X., Yu, C., Qu, J., Zhang, L., Jiang, S., Huang, D. et al. (2020). Imaging and clinical features of patients with 2019 novel coronavirus SARS-CoV-2. European Journal of Nuclear Medicine and Molecular Imaging. 47: 1275-1280. doi: 10.1007/s00259-020-04735-9.

  108. Xu, Z., Shi, L., Wang, Y. et al.  (2020). Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respiratory Medicine. 8: 420-2.

  109. Xu, X., Zheng, X.,   Li, S., Lama, N.Z.,   Wang, Y., Chu, D.K.W., Poon, L.L.M., Tun, H.T., Peiris, M., Deng, Y., Leung, G.M., Zhang, T. (2021). The first case study of wastewater- based epidemiology of COVID-19 in Hong Kong. Science of the Total Environment. https://doi.org/10.1016/j.scitotenv. 2021.148000.

  110. Yao, X.H., He, Z.C., Li, T.Y., Zhang, H.R., Wang, Y., Mou, H. et al.  (2020). Pathological evidence for residual SARS-CoV-2 in pulmonary tissues of a ready-for-discharge patient. Cell Research. 30: 541-3.

  111. Yao, X.H., Li, T.Y., He, Z.C. et al. (2020). A pathological report of three COVID-19 cases by minimal invasive autopsies. Zhonghua Bing Li Xue Za Zhi. 49: 411-7.

  112. Yao, X.H., Li, T.Y., He, Z.C., Ping, Y.F., Liu, H.W., Yu, S.C. et al. (2020). A pathological report of three COVID-19 cases by minimally invasive autopsies. Zhonghua Bing Li Xue Za Zhi.49: E009.

  113. Zappulli, V., Caliari, D., Cavicchioli, L.., Tinelli, A., Castagnaro, M. (2008). Systemic fatal type II coronavirus infection in a dog: Pathological findings and immunohistochemistry. Research in Veterinary Science. 84: 278-282. 

  114. Zappulli, V., Ferro, S., Bonsembiante, F., Brocca, G., Calore, A., Cavicchioli, C., Centelleghe, C., Corazzola, G., De Vreese,  S. et al. (2020). Pathology of coronavirus infections: A review of lesions in animals in the one-health perspective. Animals. 377: 1-41. doi: 10.3390/ani10122377.

  115. Zeng, Z., Xu, L., Xie, X.Y., Yan, H.L., Xie, B.J., Xu, W.Z. et al. (2020).  Pulmonary pathology of early phase COVID-19 pneumonia in a patient with a benign lung lesion. Histopathology. 10.1111/his.14138.

  116. Zhang, C., Shi, L., Wang, F.S. (2020). Liver injury in COVID-19: Management and challenges. Lancet Gastroenterology and Hepatology. 5: 428-430. doi: 10.1016/s2468-1253 (20)30057-1.

  117. Zhang, T., Wu, Q., Zhang, Z. (2020). Probable pangolin origin of SARS CoV-2 associated with the COVID-19 outbreak. Current Biology. 30: 1346-1351.e2. doi: 10.1016/j.cub. 2020.03.022.

  118. Zhou, P., Yang, X. L., Wang, X. G., Hu, B., Zhang, L., Zhang, W. et al.  (2020). A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature. 579: 270-273. doi: 10.1038/s41586-020-2012-7.

  119. Zhu, N., Zhang, D., Wang, W., Li, X., Yang, B., Song, J. et al. (2020). A novel coronavirus from patients with pneumonia in China, 2019. New England Journal of Medicine. 382: 727-733. doi: 10.1056/NEJMoa2001017.

  120. Zhuang, Q., Liu, S., Zhang, X., Jiang, W., Wang, K., Wang, S., Peng, C., Hou, G., Li, J., Yu, X. et al. (2020). Surveillance and taxonomic analysis of the coronavirus dominant in pigeons in China. Transboundary and Emerging Diseases.  67: 1981-1990.

  121. Zou, X., Chen, K., Zou, J., Han, P., Hao, J., Han, Z. (2020). The single-cell RNA-seq data analysis on the receptor ACE2 expression reveals the potential risk of different human organs vulnerable to Wuhan 2019-nCoV infection. Frontiers  in Medicine. 14: 185-92.

  122. Zubair, A.S., McAlpine, L.S., Gardin, T. et al. (2020).  Neuropathogenesis  and neurologic manifestations of the coronaviruses in the age of coronavirus disease 2019: A review. Journal of American Medical Association Neurology. doi: 10.1001/ jamaneurol.2020.2065.

Editorial Board

View all (0)