Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2): a Systemic Infection

REVIEW

Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2): a Systemic Infection

Aleksandra Synowiec,a  Artur Szczepan ski,a,b  Emilia Barreto-Duran,a  Laurensius Kevin Lie,a  Krzysztof Pyrca aVirogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland

bMicrobiology Department, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland

SUMMARY …………………………………………………………………………1 INTRODUCTION ……………………………………………………………………1 HOSTFACTORSDETERMININGCELLTROPISM ………………………………………2 THERESPIRATORYTRACT ………………………………………………………….3 THEGASTROINTESTINALTRACT ……………………………………………………8 THECARDIOVASCULARSYSTEM …………………………………………………..10

Vascular Events ………………………………………………………………….11

The Heart ……………………………………………………………………….12 THEIMMUNESYSTEM …………………………………………………………….13 THEKIDNEY ………………………………………………………………………13 THELIVER ………………………………………………………………………..14 THE PANCREAS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .14 THENEUROLOGICALSYSTEM ……………………………………………………..15

The Eye …………………………………………………………………………16 REPRODUCTIVESYSTEM ………………………………………………………….16 CONCLUSIONSANDKEYTAKEAWAYMESSAGES …………………………………..18 ACKNOWLEDGMENTS …………………………………………………………….18 REFERENCES ……………………………………………………………………..18 AUTHORBIOS …………………………………………………………………….31

SUMMARY To date, seven identified coronaviruses (CoVs) have been found to infect humans; of these, three highly pathogenic variants have emerged in the 21st century. The newest member of this group, severe acute respiratory syndrome co- ronavirus 2 (SARS-CoV-2), was first detected at the end of 2019 in Hubei province, China. Since then, this novel coronavirus has spread worldwide, causing a pan- demic; the respiratory disease caused by the virus is called coronavirus disease 2019 (COVID-19). The clinical presentation ranges from asymptomatic to mild respi- ratory tract infections and influenza-like illness to severe disease with accompany- ing lung injury, multiorgan failure, and death. Although the lungs are believed to be the site at which SARS-CoV-2 replicates, infected patients often report other symptoms, suggesting the involvement of the gastrointestinal tract, heart, cardio- vascular system, kidneys, and other organs; therefore, the following question arises: is COVID-19 a respiratory or systemic disease? This review aims to summarize exist- ing data on the replication of SARS-CoV-2 in different tissues in both patients and ex vivo models.

KEYWORDS COVID-19, SARS-CoV-2, coronavirus, disease, infection, organoids, organs, systemic

INTRODUCTION

Coronaviruses (CoVs), enveloped, nonsegmented, positive-sense single-stranded RNA (ssRNA) viruses that belong to the Coronaviridae family, can infect both humans and animals. To date, seven CoVs have been reported to infect humans, of which four (human CoV-NL63 [HCoV-NL63] [1], HCoV-OC43 [2, 3], HCoV-229E [2, 3], and

Citation Synowiec A, Szczepan ski A, Barreto- Duran E, Lie LK, Pyrc K. 2021. Severe acute

 

April 2021 Volume 34 Issue 2 e00133-20 Clinical Microbiology Reviews cmr.asm.org 1

respiratory syndrome coronavirus 2 (SARS- CoV-2): a systemic infection. Clin Microbiol Rev 34:e00133-20. https://doi.org/10.1128/CMR .00133-20.
Copyright © 2021 American Society for Microbiology. All Rights Reserved.
Address correspondence to Krzysztof Pyrc, k.a.pyrc@uj.edu.pl.
Published 13 January 2021

Downloaded from http://cmr.asm.org/ on January 21, 2021 by guest

Synowiec et al.

Clinical Microbiology Reviews

HCoV-HKU1 [4]) circulate worldwide and cause mild, seasonal respiratory tract disease. Importantly, three of seven CoVs emerged in the 21st century and are associated with severe acute respiratory tract infections. Severe acute respiratory syndrome CoV (SARS- CoV) emerged in late 2002 in Guangdong province, China, and spread rapidly to other countries and continents, accounting for ;8,000 confirmed cases and a fatality rate of 9.6% (5, 6). SARS-CoV is a betacoronavirus that originated in horseshoe bats and subse- quently leaked into the population of wild animals, including palm civets, in China; the virus adapted and ultimately was transmitted to humans by direct animal–human con- tact (7). Even though human-to-human transmission of the virus was efficient, the epi- demic burned out in May 2004 due to the seasonal nature of the virus and imposed health care measures; since then, no case of SARS-CoV has been reported. Middle East respiratory syndrome CoV (MERS-CoV) emerged 10 years later and caused outbreaks in Saudi Arabia and South Korea (8, 9). Similar to SARS-CoV, MERS-CoV originated in bats, but dromedary camels were identified as an intermediate host (10). It is still not clear how the virus was transmitted between these animals, and one may speculate that another intermediate host may have been involved (11). While human-to-human trans- mission of MERS-CoV accounts for almost half of cases, it is limited to households or nosocomial outbreaks, and close and prolonged contact is required (12). Despite that, MERS has accounted for ;2400 cases in the last 8 years, with an unsettling fatality rate of 34% (13). These two highly pathogenic coronaviruses caught the attention of researchers and triggered the number of studies on the potential of zoonotic coronavi- ruses to cause pandemics in humans. The discovery of a large pool of SARS-like corona- viruses in bats in Yunnan, China (14), led to the conclusion that we may encounter the SARS virus again. Indeed, 2019 brought us such a novel zoonotic coronavirus, which appears to be a close relative of the 2002 SARS-CoV. Severe acute respiratory syndrome CoV 2 (SARS-CoV-2) emerged in Hubei province, China (15). The virus, initially named “2019-nCoV,” belongs to the SARS-like virus cluster (15, 16) and shares 86% homology on the nucleotide level with the first detected SARS-CoV (17). The disease caused by the virus was named coronavirus disease 2019 (COVID-19). The clinical picture ranges from asymptomatic, through mild respiratory tract infections and influenza-like illness (mainly fever, cough, and fatigue), to severe disease with accompanying lung injury, multiorgan failure, and death (18, 19). Unsurprisingly, the lungs are the main gate of infection; however, SARS-CoV-2 RNA was detected in the kidneys, liver, heart, brain, and blood samples at autopsy (20). This is in agreement with reports showing that COVID-19 patients frequently exhibit other symptoms, suggesting multiorgan involve- ment and a rare but severe complication of SARS-CoV-2 replication, which is a multisys- tem inflammatory syndrome (MIS) in children (MIS-C) and adults (MIS-A) (21–30). This review aims to summarize and pull together existing data about the replication of SARS-CoV-2 in different tissues.

HOST FACTORS DETERMINING CELL TROPISM

Virus entry into a cell is a complex process that requires both viral and cellular fac- tors. The first steps are interaction with an adhesion receptor, binding to the entry re- ceptor, cell internalization/fusion, and transport to the site of replication (cytoplasm or nucleus). Coronavirus particles comprise at least four structural proteins: spike (S), en- velope (E), membrane (M), and nucleocapsid (N). Schematic SARS-CoV-2 structure and protein localization are presented in Fig. 1.

The S protein is responsible for receptor binding and determines the host range and cell tropism (31). This large protein comprises a short C-terminal tail located inside the virion, a transmembrane domain, a rod-like S2 domain responsible for the fusion process, and a large globular S1 domain, within which the receptor-binding domain is located. In advance of interaction with the entry receptor, the virus binds to adhesion receptors; this concentrates the virus on the cell surface. Next, the virus binds to the entry receptor, which initiates a fusion of the viral and cellular membranes. Finally, the

April 2021 Volume 34 Issue 2 e00133-20

cmr.asm.org 2

Downloaded from http://cmr.asm.org/ on January 21, 2021 by guest

SARS-CoV-2: a Systemic Infection

Clinical Microbiology Reviews

FIG 1 Schematic structure of the SARS-CoV-2 virion.

viral nucleoprotein enters the cytoplasm. The adhesion and entry receptors used by human coronaviruses (32–42) are presented in Fig. 2.

The in vitro and ex vivo models that are permissive to infection by SARS-CoV-2 are listed in Tables 1 and 2.

The internalization site depends on the availability of the proteases required to trig- ger a transformation of the S protein into the fusogenic state. In vitro models show that human coronaviruses use an endocytic entry pathway in which gradual acidifica- tion of the microenvironment activates endosomal cathepsin B (catB) and cathepsin L (catL), which effectively prime the S protein and initiate entry (43, 44). However, recent studies showed that human coronaviruses bypass this process and use serine pro- teases (transmembrane protease serine 2 [TMPRSS2], kallikrein 13) present on the cell surface (Fig. 2). In such cases, the fusion occurs on the cell surface and endocytosis is not required (45–50). Interestingly, the concentration of cathepsins in the endosomal compartments of primary cells lining the respiratory tract is too low for virus activation. Endocytosis does not allow virus fusion in vivo.

Focusing on SARS-CoV-2, Sungnak et al. (48) evaluated the expression of angioten- sin (Ang)-converting enzyme 2 (ACE2), which is an entry receptor for this virus (40), and of TMPRSS2 (a spike-priming protease) in different cell types. For their study, they used single-cell RNA sequencing (scRNA-seq) data sets from healthy donors generated by the Human Cell Atlas consortium. The authors focused mainly on evaluating the expression of ACE2 in epithelial cell types within the lung and airways. They found that even though the level of ACE2 expression was in general low, it was expressed by numerous epithelial cell types (e.g., alveolar type II [AT2], bronchial secretory, ciliated, and basal), with higher expression levels detected on nasal goblet and ciliated cells (48). Interestingly, although the lungs are considered to be the SARS-CoV-2 target organ, only ;2% of cells in this tissue are ACE2 positive, whereas ACE2-positive cells are found extensively in the small intestine, gallbladder, kidneys, testes, thyroid, adi- pose tissue, heart muscle, vagina, breast, ovary, and pancreas (51, 52). To give some examples, high ACE2 expression was found in ileal epithelial cells (about 30% of cells were found to be ACE2 positive). High expression of this protein was also found in myocardial cells and kidney proximal tubule cells (7.5% and 4% positive, respectively) (52). The widespread tissue distribution of the ACE2 protein explains the multiorgan dysfunction reported in patients. Moreover, it draws attention to the fact that COVID- 19 may be a systemic disease.

THE RESPIRATORY TRACT

The novel human coronavirus mainly affects the respiratory system, causing a respi- ratory disease characterized by cough (mostly dry), dyspnea, fatigue, and, in severe cases, pneumonia or respiratory failure (corroborated by radiographic bilateral ground-

April 2021 Volume 34 Issue 2 e00133-20

cmr.asm.org 3

Downloaded from http://cmr.asm.org/ on January 21, 2021 by guest

Synowiec et al. Clinical Microbiology Reviews

FIG 2 The entry of human coronaviruses into the host cell. Coronaviruses first interact with an adhesion molecule (e.g., heparan sulfate proteoglycans [HSPGs] for HCoV-NL63 [32], SARS-CoV [33], and [possibly] SARS-CoV-2 [409]; N-acetyl-9-O-acetylneuraminic acid

April 2021 Volume 34 Issue 2 e00133-20

cmr.asm.org 4

(Continued on next page)

Downloaded from http://cmr.asm.org/ on January 21, 2021 by guest

SARS-CoV-2: a Systemic Infection

Clinical Microbiology Reviews

glass opacity) (53–55). Damage to the airway tract and lungs was evident during bi- opsy and autopsy studies (53–55). Diffuse alveolar damage (DAD) and airway inflam- mation have been reported both in humans and in nonhuman primates (53, 56–63). The leading cause of mortality for SARS-CoV-2 is respiratory failure from acute respira- tory distress syndrome (ARDS) (64). ARDS can be related to airway remodeling caused by pulmonary fibrosis and systemic inflammation (65, 66). The exact molecular mecha- nism of airway remodeling during the COVID-19 remains unknown and is associated with both viral replication in the tissue and dysregulation of natural pathways such as cytokine production or oxidative stress. Finally, the identification of viral cellular targets may shed some light on potential therapeutic and preventive strategies that may be used in COVID-19 patients with ARDS in the future.

While it is known that the respiratory tract is an entry point for SARS-CoV-2, it is vital to identify the cells that are the primary targets of the infection. First, in vitro analyses carried out by Hoffmann et al. demonstrated that SARS-CoV-2 pseudoviruses entered human cell lines derived from the airways, including Calu3, A549, BEAS-2B, and H1299 cells (49), with Calu3 cells being the most permissive (49). While efficient SARS-CoV-2 replication in the Calu3 cell line was also demonstrated by others (54, 67–71), A549 cells were not found to be permissive unless they overexpressed ACE2 (54, 70, 72–75).

Data mining allowed the identification of cell types that may be permissive to infec- tion in vivo (48, 52, 73, 76–78). The cells present in the human respiratory tract are shown in Fig. 3. In general, lung and bronchial tissues show low expression of ACE2 (73, 79); alveolar type II cells (AT2) show higher expression of ACE2 and TMPRSS2 (48, 49, 52, 77, 80–82). Hikmet et al. reported expression of ACE2 in more than 150 cell types from different tissues (immunohistochemical analysis) (73), but in that study, the level of expression of ACE2 in the respiratory system was limited. Aguiar et al. showed similar results using microarrays and scRNA-seq data set analysis (79). Sungnak et al. reported high expression of both ACE2 and TMPRSS2 in nasal goblet and ciliated cells (48). They corroborated these results by performing an independent scRNA-seq study of nasal brushings and studies using an in vivo nasal human airway epithelium (HAE) model. In accordance with those results, Lukassen et al. evaluated healthy human lung tissues (biopsy specimens) and bronchial HAE air-liquid interphase (ALI) cultures (HBEC); they reported that “transient secretory cells” showed expression of ACE2 and TMPRSS2 (81). These cells were reported to be cells transiting from a club or goblet phenotype to a differentiated ciliated phenotype (81). Tindle et al. demonstrated the expression of ACE2 in club cells using immunofluorescence staining of human lung sections from infected and noninfected patients (66). Zhang et al. analyzed airway epi- thelia using bulk RNA sequencing, scRNA-seq, and microarrays. They found that ACE2 is expressed in basal, club, goblet, and ciliated cells of the small airway, large airway, and trachea (83). Valyaeva et al. proposed that levels of expression of ACE2 and other SARS-CoV-2 entry factors might be underestimated when using 39 scRNA-seq data sets rather than full-length scRNA-seq data. They showed that ACE2 levels in basal cells were almost 10 times higher when evaluated using full-length scRNA-seq data, which is in accordance with results of ex vivo lung experiments showing basal cell infection (425).

Different approaches have been used to identify the cells that constitute the real targets for the virus; studies have examined primary human airway cells, tissue explants, and tissue cultures (49, 80, 84–87). Zhang et al. and Tindle et al. reported high expression of the viral N protein in alveolar epithelial cells within immunostained

FIG 2 Legend (Continued)
[Neu5Ac] for HCoV-HKU1 and HCoV-OC43 [34]; or carcinoembryonic antigen-related cell adhesion molecule 5 [CEACAM5] for MERS- CoV [35]). Next, the virus interacts with the entry receptor (aminopeptidase N [APN] for HCoV-229E [36]; dipeptidyl peptidase 4 [DPP4] for MERS-CoV [37]; 9-O-acetylated sialic acid for HCoV-OC43 [39]; or angiotensin-converting enzyme 2 [ACE2] for HCoV-NL63, SARS-CoV, and SARS-CoV-2 [40]). Recently, neuropilin 1 (NRP1) was reported to enhance the SARS-COV-2 entry (41, 42). To enter the cell, the S protein requires proteolytic priming, which may occur on the cell surface (TMPRSS2, TMPRSS4, kallikrein 13) or after endosomal entry (cathepsin B [catB] and cathepsin L [catL]) (43–50, 410–414).

April 2021 Volume 34 Issue 2 e00133-20 cmr.asm.org 5

Downloaded from http://cmr.asm.org/ on January 21, 2021 by guest

Synowiec et al.

Clinical Microbiology Reviews

TABLE 1 Cell lines that support the replication of SARS-CoV-2

CPEa Additional information (reference[s])
1/2 Robust replication, no cell death detected, also

susceptible to SARS-CoV, one group reports

visible cytopathic effect (131)
1/2 Robust replication, no cell death detected, also

susceptible to SARS-CoV, some groups report

visible cytopathic effect (71, 415)
2 Robust replication, highly permissive (higher virus

titer than a parental line), no cell death detected 2 Robust replication

1 Robust replication. Also susceptible to SARS-CoV 2 Robust (80) or modest (416) replication; also

susceptible to SARS-CoV
2 Robust (80) or modest (416) replication; also

susceptible to SARS-CoV 2 Modest replication

1 Cessation of beating after 72 h of infection

2 Alpha, beta, and delta cells; alpha and beta cells were permissive for VSV-based SARS-CoV-2

pseudoviruses
2 The entry of pseudoparticles harboring spike protein

2 The entry of pseudoparticles harboring spike protein

1 Robust replication, cell rounding, detachment, degeneration, and syncytium formation; also

susceptible to SARS-CoV
1 Robust replication, cell rounding, detachment,

degeneration, and syncytium formation; also

susceptible to SARS-CoV
2 Robust replication; also susceptible to SARS-CoV

2 Also susceptible to SARS-CoV
2 Also susceptible to SARS-CoV
1/2 Robust replication; also susceptible to SARS-CoV 2 Modest replication

lung tissue biopsy specimens from a SARS-CoV-2-infected patient, suggesting that these cells may be effectively infected (53, 66). Hui et al. used ex vivo cultures of human bronchus and lung to show that AT1 cells, ciliated cells, club cells, and goblet cells, but not basal cells, are susceptible to SARS-CoV-2 infection (85). They also showed that the level of SARS-CoV-2 replication was higher than that of SARS-CoV in ex vivo bronchial cultures. Zhou et al. also demonstrated higher infectivity and replication of SARS-CoV- 2 than SARS-CoV in the airway organoids and confirmed the observation using subge- nomic mRNA analysis, transmission electron microscopy (TEM), and immunofluores- cence staining (88). Likewise, Chu et al. demonstrated replication and cell tropism of SARS-CoV-2 and SARS-CoV using ex vivo lung explants (80). The authors used plaque assay, quantitative reverse transcription-PCR (RT-qPCR), and confocal microscopy to show that SARS-CoV-2 infected and replicated more efficiently in human lung tissues than SARS-CoV. These findings are in agreement with results of studies performed with the Calu3 cell line (80, 85). The human airway epithelium (HAE) cultures are ALI models, which are used commonly to study human respiratory tract diseases due to their re- semblance to in vivo airway tissue (89–91). The ALI methodology promotes epithelial cell differentiation into different cell types (e.g., basal, ciliated, club, and goblet cells); besides, it allows the production of mucus and beating cilia, thereby providing a more reliable model of virus infection and cell tropism than traditional cell culture models (92–96). The first study to use HAE as a model for SARS-CoV-2 was presented by

Cell line

Caco-2

Calu3

C2BBe1 (Caco-2 subclone)

T84 CL14 Huh7

293T

U251 hiPSC-MC

hPSC

BEAS-2B H1299 Vero E6

FRhK4

LLC-MK2

Origin Species

Colorectal adenocarcinoma Human

Lung adenocarcinoma Human

Colorectal adenocarcinoma Human

Colorectal adenocarcinoma Human Colorectal adenocarcinoma Human Hepatocellular carcinoma Human

Embryonic kidney epithelia Human

Glioblastoma Human Induced pluripotent stem Human

cell-derived

cardiomyocytes
hPSC-derived pancreatic Human

endocrine cells

Nontumorigenic bronchial Human epithelium

Non-small-cell lung Human carcinoma

Kidney African monkey

Kidney Rhesus monkey

Kidney Rhesus monkey

Reference

54, 80, 130, 131

67–71, 80, 130, 331, 415–419

130

128 131 80, 416

80

80 242

309

49

49, 415

69, 70, 80, 416, 418, 420

80

80

80
80
80, 130 130

Kidney Cat Kidney Rabbit Kidney Pig Intestine Pig

CRFK
RK-13
PK-15
IPEC-J2
aCPE, cytopathic effect. 1, positive; 2, negative; 1/2, ambiguous result.

green

April 2021 Volume 34 Issue 2 e00133-20

cmr.asm.org 6

Downloaded from http://cmr.asm.org/ on January 21, 2021 by guest

SARS-CoV-2: a Systemic Infection

Clinical Microbiology Reviews

TABLE 2 Ex vivo models used to study SARS-CoV-2 infection

Model

Human airway epithelium (HAE) cultures, ALI cultures

Primary human airway epithelial cells Primary cell-derived lung organoids hPSC-derived lung and macrophage

coculture system
hPSC-derived lung organoids Human lung organoids with mixed

proximodistal epithelia
Human embryonic stem cell (hESC)-

derived organoid 3D alveolar organoids Lung-on-chip

hESC-derived SEAM eye organoids

Human intestinal organoids (HIOs) hPSC-derived colon organoids

(hPSC-COs)
Human gastric organoids (HGOs)

Human tonsil organoids
Human blood vessel organoids Human kidney organoids
Human liver ductal organoids Human bronchial organoids (HBOs)

Human brain organoids

hPSC-derived choroid plexus organoids

Bat intestinal organoids

Additional information

Also susceptible to SARS-CoV; the virus infects primarily ciliated cells; cessation of cilium beating

Also susceptible to SARS-CoV
Also susceptible to SARS-CoV; SARS-CoV-2 infection of ciliated and basal cells M2 and M1 macrophages have inhibitory effects on SARS-CoV-2 infection

Mainly composed of AT2 and AT1 cells
Composed of both proximal and distal airway epithelia

Differentiated human airway organoids from hESC

Distal lung epithelial cells with or without lung fibroblasts
Cultures are composed of human airway epithelial and endothelial cells;

macrophages were also present in some experiments
Organoids are composed of four distinct zones of ocular tissues, including retinal

pigment epithelium (RPE), neural retina, ciliary body, lens, and cornea; highly

active SARS-CoV-2 replication in the corneal limbus
The virus replicates in enterocytes, cytopathic effect; also susceptible to SARS-CoV hPSC-derived organoids, composed of enterocytes, goblet cells, transit-amplifying

(TA) cells, enteroendocrine (EE) cells, and LGR51 or BMI11 stem cells; viral RNA

was detected in all five cell populations
Organoids derived from human fetal and pediatric tissue; standard and reversed-

polarity organoids included; robust viral replication in pediatric-derived

organoids but not fetal ones
Obtained from tonsil tissues, secretion of the progeny viral particles
iPSC-derived organoids, infectious viral progeny production
iPSC-derived organoids, infectious viral progeny production
Robust replication in cholangiocytes
Generated from commercially available cryopreserved human bronchial epithelial

cells
iPSC-derived organoids; SARS-CoV-2 enters into neuronal cells and targets cortical

region, but replication is probably abortive; neuronal cell death Simulated the blood-cerebrospinal fluid barrier; productive SARS-CoV-2

replication was observed, with SARS-CoV-2 preferentially infecting the choroid

plexus epithelium Progressive cytopathic effect

References

15, 67–69, 81, 98, 102–105, 421, 422

49 88 62

108 66

107

100, 107, 110–112 423, 424

347

123, 126, 136 108

137

246

98

98

298

84

317, 318 318, 335

126

Milewska et al. (97). The quantitative results indicated that the virus infects ciliated cells and is released on the apical side of the culture, not the basolateral side; this means that viral infection is effective in the airway lumen (97). Subsequent reports by others confirmed these observations (15, 98–101). Zhu et al. reported that ciliated, club, and goblet cells were infected in their HAE model and that the cytopathic effect (CPE) was observed (101). Ravindra et al. showed that the virus primarily infects ciliated cells and that during infection other cells (basal and club) can become infected (97, 102). They used scRNA-seq to show that goblet cells, neuroendocrine cells, tuft cells, and mono- cytes are rarely infected (102). TEM revealed that infection of human airway epithelial models of nasal and bronchial origin induced remodeling of the cellular ultrastructure of the ciliated, goblet, and (to a lesser extent) basal cells (102, 103). Following the results obtained reported by Ravindra et al., Mulay et al. used immunostaining to dem- onstrate that SARS-CoV-2 predominantly infected ciliated cells and a small portion of goblet cells in their HAE model (100). The HAE model has also been efficiently used by different research groups to evaluate different SARS-CoV-2 inhibitors (67–69, 72, 100, 104–106), suggesting that it is also a suitable model for this approach. Pei et al. showed that human embryonic stem cell (hESC)-derived organoids reflected the natural micro- environment. In this model, more than 90% of ciliated cells, less than 10% of club cells, and no basal or goblet cells were infected with SARS-CoV-2 (107). Tindle et al. devel- oped an adult stem cell-derived human lung organoid model composed of both proxi- mal and distal airway epithelia. They showed that the proximal airway epithelium is

April 2021 Volume 34 Issue 2 e00133-20

cmr.asm.org 7

Downloaded from http://cmr.asm.org/ on January 21, 2021 by guest

Synowiec et al.

Clinical Microbiology Reviews

FIG 3 Cell types and their localization within the human respiratory tract.

more permissive to SARS-CoV-2 than the distal alveolar tissue (66). Han et al. demon- strated SARS-CoV-2 pseudovirus entry and SARS-CoV-2 infection in a human pluripo- tent stem cell (hPSC)-derived lung organoid model composed mainly of AT2 cells, AT1 cells, stroma cells, neuroendocrine cells, and airway epithelial cells (108). Similarly, Huang et al. proved the infection of iAT2 (AT2 cells derived from induced pluripotent stem cell [iPSC]) organoids in ALI culture (109) and Youk et al. the infection of the alve- olar stem cell-derived organoids (110). The results obtained by others (100, 107, 111, 112) are consistent with these observations.

THE GASTROINTESTINAL TRACT

Although coronaviral infections in humans are associated mainly with respiratory tract disease, accompanying symptoms in the gastrointestinal (GI) tract have been reported (113–119). According to one study, during a SARS-CoV outbreak in March 2003 in Hong Kong, 19.6% of infected patients developed nausea, diarrhea, and/or vomiting (113). Another study reported that 38% of patients experienced diarrhea during their illness (114). Interestingly, some patients (5.8%) with fever and diarrhea did not develop a respiratory disease (114). Consequently, viral replication in the small and large intestine of patients with SARS-CoV was confirmed (114). Infection by the second highly pathogenic coronavirus, MERS-CoV, was also associated with GI symptoms. Descriptive studies from 2012 to 2013 reported that a quarter of MERS-positive patients had accompanying GI symptoms, including diarrhea and vomiting (119). Importantly, not only highly pathogenic coronaviruses but also sea- sonal human coronaviruses are associated with GI infections. As an example, 33% of HCoV-NL63-positive patients and 57% of HCoV-OC43-positive patients in France developed digestive problems such as abdominal pain, diarrhea, and vomiting (116, 118). These data clearly show that the fecal-oral route of coronavirus transmis- sion is an important research area that needs further investigation during the COVID-19 pandemic.

After the emergence of SARS-CoV-2, it was observed that COVID-19 patients often suffered from GI tract disease symptoms (120, 121) and that up to 53% of patients

April 2021 Volume 34 Issue 2 e00133-20

cmr.asm.org 8

Downloaded from http://cmr.asm.org/ on January 21, 2021 by guest

SARS-CoV-2: a Systemic Infection

Clinical Microbiology Reviews

FIG 4 Cell types and their localization in the human intestine.

infected with SARS-CoV-2 tested positive for viral RNA in stool specimens (117, 122, 123). Moreover, viral RNA can be detected in fecal samples for up to 5 weeks after re- spiratory samples become virus negative. In contrast, in some patients, an occurrence of GI tract symptoms does not correlate with the detection of viral RNA in fecal sam- ples (124). Some may speculate that such symptoms may be related to alterations in the gut microbiota and/or dysbiosis during COVID-19 (125). These findings make it uncertain whether SARS-CoV-2 replicates in the GI tract. Immunostaining of viral pro- teins in gastrointestinal tissue samples collected from affected patients shed some light on this by providing evidence for viral replication within these tissues, suggesting that the fecal-oral route is indeed a relevant transmission route (117). Moreover, some groups have reported successful isolation of infectious virus from stool samples (126, 127).

Efforts to model GI infection in vitro led to identification of four colon carcinoma cell lines (human intestinal epithelial cells [IECs]) that are permissive to SARS-CoV-2 infection: Caco-2 (49, 128) (also susceptible to the SARS-CoV infection) (129); C2BBe1, the Caco-2 brush border-expressing subclone (130); CL14 (131); and T84 (128). However, most niche-mimicking models and models of the GI tract are based on the use of human intestinal organoids (HIOs), which are currently the most advanced tool available. HIOs are differentiated, nontransformed, and physiologically active cultures, containing multiple intestinal epithelial cell types such as enterocytes, goblet cells, tuft cells, enteroendocrine cells (EECs), and Paneth cells (132). Cell types present in intes- tines are shown in Fig. 4. Importantly, a recent study showed that HIOs allow replica- tion of MERS-CoV (133), along with other viruses that could not be cultured using the standard cell lines (134, 135). HIOs, which can be grown in three-dimensional (3D) or 2D monolayers, support replication of SARS-CoV-2 and SARS-CoV in the ileum, duode- num, and colon-derived organoids (126, 128, 136). Importantly, the intestines are not the only affected part of the digestive system; viral nucleocapsid protein was visualized in gastric tissue derived from COVID-19 patients (117). Unsurprisingly, human gastric organoids (HGOs) derived from pediatric patients supported SARS-CoV-2 replication (137). Of note, human organoids are not the only organoids permissive to novel coro- navirus; bat intestinal organoids also support SARS-CoV-2 infection, which is in agree- ment with the virus origin predictions (126, 138).

Generally, ACE2 is an entry receptor for the virus, and TMPRSS2 is the spike priming protease. Intriguingly, the level of ACE2 expression in intestinal tissues is much higher

April 2021 Volume 34 Issue 2 e00133-20

cmr.asm.org 9

Downloaded from http://cmr.asm.org/ on January 21, 2021 by guest

Synowiec et al.

Clinical Microbiology Reviews

than that seen in the lungs (139). To be more precise, ACE2 is abundantly expressed in stomach epithelial cells and in enterocytes from the small intestine, including the duo- denum, jejunum, and ileum, and it is poorly expressed in colonocytes (140). Unsurprisingly, human colonoids are affected to a lesser extent than organoids deriv- ing from the small intestine (128, 136). Consequently, SARS-CoV and SARS-CoV-2 infect only enterocytes and not goblet cells, EECs, tuft cells, or Paneth cells (123, 136). Mature enterocytes express higher ACE2 levels than immature ones, but the levels of replica- tion are comparable. This may indicate that a low level of ACE2 expression is sufficient for the virus to enter the cell (123, 136) or that there is an additional restriction factor present in mature enterocytes. What is interesting is that ACE2 expression increases during gastric (141) and colorectal (142) cancer development. Increased expression of ACE2 is also observed in patients with inflammatory bowel disease (IBD) (143, 144). Although ACE2 is not the only factor required during the infection, one might think that cancer or/and IBD patients might experience more-severe gastrointestinal symp- toms. Nevertheless, it is still an understudied research area that needs to be addressed. Human intestinal enteroid monolayer models confirmed that SARS-CoV-2 efficiently infects and replicates in the enterocytes and that the virus is released from the apical side (123). Except for ACE2, there are additional “players” during virus entry, and in intestines, the spike protein, similarly to other organs, is primed by TMPRSS2 (49) and possibly also by TMPRSS4 (123). As in the case of the respiratory tract, the role of cathe- psins in in vivo and ex vivo activity seems to be limited.

Nevertheless, one can imagine that bowel inflammation can lead to the “leaky gut” syndrome. This may result in systemic distribution of the virus and infection of other organs, for example, the lungs or heart. No reports have shown that the infectious virus can be found in blood, but viral RNA was found in 15% of plasma samples from COVID-19 patients in one study (139). Further, the systemic distribution of the virus confirms that SARS-CoV-2 may be spread either by blood or by blood cells. A similar study was carried out for MERS-CoV, when humanized dipeptidyl peptidase 4 (DPP4) mice were intragastrically administered with the virus; in addition to GI disease, ani- mals developed lung and brain infections (133). If the situation is similar in COVID-19 patients, the results may support clinical reports suggesting that gastrointestinal tract disease precedes respiratory tract symptoms (145). While infectious viral progeny are produced by gut organoids (136) and infectious SARS-CoV-2 can be isolated from stool samples (126, 127), the importance of the fecal-oral transmission route for SARS-CoV-2 remains unclear. Although the GI tract seems to be a replication site, it is worth men- tioning that in order to employ this route, the virus needs to cross the GI tract and remain infectious. This is questionable, as the recombinant SARS-CoV-2 mNeonGreen reporter virus was previously shown to be susceptible to inactivation by human gastric fluids (123). A similar phenomenon was reported for MERS-CoV, wherein the virus appeared to be tolerant of gastric and intestinal fluids produced during the fed state but not during fasting (133). Taking the data altogether, it remains unclear whether the GI tract can serve as the primary site of infection. Further investigations and devel- opment of appropriate animal models are needed.

THE CARDIOVASCULAR SYSTEM

The cardiovascular system was also thought to be a target for SARS-CoV-2 infection. Cardiovascular sequelae have been reported for other highly pathogenic human coro- naviruses. In SARS-CoV patients, these are usually mild and self-limiting (146), but MERS-CoV is associated with acute myocarditis and heart failure (147). It is well recog- nized that patients with preexisting cardiovascular diseases are more likely to suffer COVID-19 complications and to require admission to an intensive care unit (ICU) (148–154). Furthermore, myocardial injury and heart failure are considered to be sequelae of COVID-19 (51, 152, 153, 155). Nevertheless, one may say that cardiovascu- lar clinical manifestations may be solely the result of thrombosis.

April 2021 Volume 34 Issue 2 e00133-20

cmr.asm.org 10

Downloaded from http://cmr.asm.org/ on January 21, 2021 by guest

SARS-CoV-2: a Systemic Infection

Clinical Microbiology Reviews

Vascular Events

FIG 5 Cell types and their localization in the cardiovascular system.

Endothelial cells are another cell population in the lungs but also in the cardiovascular system; importantly, they express ACE2 receptors and TMPRSS2 protease, as well as some other molecules that may mediate infection (e.g., CD147) (140, 156–160). The presence of SARS-CoV-2 virions was confirmed within endothelial cells; moreover, endotheliitis and ele- vated levels of circulating endothelial cells were observed (156, 157, 161–163). Cell types present in the cardiovascular system are shown in Fig. 5. The infection results in the pro- duction of virulent progeny viruses, which was confirmed using human capillary organoids (98).

Interestingly, severe illness is rare in children (164); however, several Kawasaki-like disease cases have been reported, first in Bergamo province in Italy and in England and later in other regions (22–24, 26, 165–175). Kawasaki disease is an acute pediatric vasculitis of unknown origin and is associated with coronary artery aneurysms. It is believed to be an aberrant response of the immune system and it was previously thought to be triggered by human coronaviruses (26, 164, 166). Diagnosed children are generally older than is usual for Kawasaki syndrome and present with more-severe disease; some require circulatory and respiratory assistance, with coronary artery aneurysms appearing to be frequent complications. Based on these cases, a definition of MIS-C, also called pediatric multisystem inflammatory syndrome (PMIS/PIMS), was formulated (21–23, 26, 27, 164, 166–168, 176). Similar symptoms were later observed in adolescents and adults, leading to the recognition of multisystem inflammatory syn- drome in adults (MIS-A). In contrast to other severe cases of COVID-19, patients with MIS-C or MIS-A have minimal respiratory symptoms and often test negative in PCR tests for SARS-CoV-2, suggesting that the symptoms constitute pathological sequelae of the infection (25, 177–184).

The renin-angiotensin system (RAS) is believed to play a central role in the pathoge- nesis of COVID-19, and medications that modulate the RAS pathway have been pro- posed as potential therapeutics (185). Under physiological conditions, a decrease in

April 2021 Volume 34 Issue 2 e00133-20

cmr.asm.org 11

Downloaded from http://cmr.asm.org/ on January 21, 2021 by guest

Synowiec et al.

Clinical Microbiology Reviews

renal blood flow stimulates the secretion of renin and generation of angiotensin I (AngI). The angiotensin-converting enzyme (ACE) then converts AngI to angiotensin II (AngII), which mediates effects such as vasoconstriction; sodium and fluid retention in a kidney; fibrosis; inflammation; and vascular permeability. It also leads to accelerated thrombosis by activating the coagulation cascade and flux of neutrophils and macro- phages to the affected tissues. In contrast, ACE2 generates angiotensin fragments (Ang1 to Ang9 and Ang1 to Ang7) which have vasodilatory, anti-inflammatory, antipro- liferative, antifibrotic, and cardioprotective properties (186–190). SARS-CoV-2 infection facilitates loss of the ACE2 catalytic effect, downregulates its expression, and promotes shedding from the cell surface, leading to accumulation of AngII and, through this, to endothelial dysfunction, inflammation, and thrombosis (187, 188, 191–193). While ACE inhibitors (ACEIs) and receptor blockers (ARBs) might be beneficial, the advisability of their usage is debatable (185, 194–197).

Furthermore, coagulopathy and resulting thromboembolic events were observed in COVID-19 patients. Importantly, these conditions were recognized as a cause of death in up to one-third of cases (158, 198–203). In consequence, the International Society on Thrombosis and Hemostasis recommends prophylactic doses of low-molecular- weight heparin (LMWH) for all patients who require hospital admission (202–204), which results in significantly lower mortality (205, 206). Interestingly, this result is a consequence not only of anticoagulative activity of LMWH but also of its anti-inflam- matory activity and LMWH-mediated inhibition of viral adhesion to the cells (205–209). The exact mechanism underlying coagulopathy is unknown; however, recent reports suggest a role of RAS axis dysregulation, inflammation and complement activation, for- mation of neutrophil extracellular traps (NETs), prolonged immobilization of patients, and activation of endothelial cells and platelets (161, 210–218). Endothelial cells are in constant contact with blood and endothelial glycocalyx, providing anticoagulant prop- erties and preventing platelet activation and aggregation. Endothelial damage may easily alter this situation and contribute to the development of disseminated intravas- cular coagulation. Additionally, while formation of NETs is part of the body’s defense against pathogens, dysregulation of this process during COVID-19 may also result in endothelial damage and blood vessel occlusion. Consequently, SARS-CoV-2 may con- tribute to the hypercoagulation observed in patients and multiorgan failure in more- severe cases (158, 159, 161, 199, 216, 217, 219–226). Among the other SARS-CoV-2 manifestations most likely related to endothelial damage are chilblain-like skin lesions, also known as “COVID toes.” While, based on PCR data, evidence of infection is not consistently found, viral particles and proteins were previously observed in endothelial cells from skin biopsy specimens (227–229).

The Heart

There are several hypotheses about the mechanism of underlying cardiac injury during the course of COVID-19; these include direct injury mediated by SARS-CoV-2 virus invasion, pulmonary infection, induced severe cases of hypoxia resulting in damage to myocardial cells, cardiotoxicity of antiviral drugs, and indirect damage mediated by excessive inflam- matory responses. Such indirect damage is especially relevant in patients with preexisting conditions, as inflammation may be associated with rupture of the coronary atheroscler- otic plaques. Furthermore, endothelial cell damage and loss of the cardioprotection pro- vided by Ang1 to Ang7 may also lead to myocardial injury (150, 155, 230–238). Several reports document elevated levels of serum troponin, creatinine kinase, and lactate dehy- drogenase in individuals with COVID-19 (51, 150–152, 155, 230, 239–241). A higher con- centration of troponins, reflecting cardiac injury, is present in 5% to 27.8% of hospitalized patients and is associated with significantly worse prognosis and increased risk of mortality (151, 152, 155, 230, 242, 243). High expression of ACE2 in the heart suggests that direct injury is possible (152, 231, 244); indeed, pericytes are thought to be the target cardiac cells for SARS-CoV-2 due to high ACE2 expression (220, 240). Viral particles have been detected in cardiac tissue (157, 245), and viral replication was shown in human induced

April 2021 Volume 34 Issue 2 e00133-20

cmr.asm.org 12

Downloaded from http://cmr.asm.org/ on January 21, 2021 by guest

SARS-CoV-2: a Systemic Infection

Clinical Microbiology Reviews

pluripotent stem cell (iPSC)-derived cardiomyocytes which led to visible cytopathic effects and a decrease in contractility (242).

THE IMMUNE SYSTEM

At the moment, not much data concerning the effects of SARS-CoV-2 on the immune system are available. Palatine tonsils are among the first lines of defense, and SARS-CoV-2 was reported to infect and replicate in 3D tonsil organoids, reflecting the in vivo tonsil epithelium (246). Further, other organs responsible for the immune responses were investigated, and cell degeneration or necrosis was also observed in the spleen (220, 247, 248). Additionally, Diao et al. (249) showed that lymphocytopenia is common among COVID-19 patients, and that finding was confirmed by other stud- ies. It was suggested that components of the immune system might be infected by SARS-CoV-2 and that poor prognoses might be related to loss of specific T-cell subsets (250–254). It was also demonstrated that the virus infects alveolar macrophages (255), as well as ACE2-positive and CD68-positive macrophages, and induces interleukin-6 (IL-6) secretion, which is in some cases associated with a fatal outcome (139, 220, 256–261). A similar effect was observed for SARS-CoV and MERS-CoV, and while most laboratories report poor, incomplete, or abortive replication, these viruses seem to prime macrophages and dendritic cells to release proinflammatory cytokines, leading to systemic hyperinflammation (“cytokine storm”) (252, 262–267). What is more, SARS- CoV-2 was frequently detected in monocytes and B cells and, to a lesser extent, in T cells of COVID-19 patients. The permissiveness of these cells was further confirmed using peripheral blood mononuclear cells (PBMCs) from healthy donors (254, 260). The permissiveness of T-lymphocytes is noteworthy, considering the low level of ACE2 expression; however, there is a need for further study to confirm this phenomenon, as it remains debatable (254, 268). These results are similar to those reported for MERS- CoV, which infects T cells and induces their apoptosis; surprisingly, T cells are resistant to infection by SARS-CoV (269). The entry of SARS-CoV-2 into lymphocytes is unex- pected because MERS-CoV infection correlates with surface levels of DPP4 (269); how- ever, ACE2 expression in T cells is almost nonexistent (268). An alternative route of entry might be a CD147 receptor-dependent route, as this molecule is expressed widely by T lymphocytes or DPP4 as the interaction between Spike S1 domain and DPP4 was predicted. However, those data were not validated experimentally and should be interpreted with caution (270–277). While the complement system repre- sents the first response of the immune system to infection, there is growing evidence that virus-induced activation of this system plays a role in COVID-19 pathogenesis. There are still many unknowns, but postmortem analysis of COVID-19 patients with ARDS revealed deposits of complement components, including membrane attack com- plex (C5b-9), C3, C4, and mannose-binding lectin (MBL)-associated serine protease 2 (MASP2) (278–280). Results of animal studies showed that C3- and C4-deficient mice exhibited lower levels of respiratory dysfunction and body weight loss than wild-type mice. Further, C3 activation was already noted in the lungs 1 day after the infection (280–282). Interestingly, a humanized anti-C5 antibody (eculizumab) was shown to improve patients’ parameters (283, 284).

THE KIDNEY

Acute renal injury was first considered to be an extrapulmonary clinical presenta- tion of SARS-CoV-2 infection (285, 286). Renal involvement was first suggested in reports describing the isolation of infectious viral particles from patients’ urine (287, 288). Chu et al. demonstrated that SARS-CoV-2 replicates in multiple kidney cell lines (54). Among these, the virus productively replicates in CRFK (feline), PK-15 (porcine), RK-13 (rabbit), and LLCMK2 (monkey) cells (54). They also observed SARS-CoV-2 replica- tion in 293T human embryonic kidney cells (54). However, they observed CPE forma- tion only in nonhuman primate kidney cell lines Vero E6 and FRhK-4, where infected cells visibly rounded together and detached from the monolayer (54). Another recent

April 2021 Volume 34 Issue 2 e00133-20

cmr.asm.org 13

Downloaded from http://cmr.asm.org/ on January 21, 2021 by guest

Synowiec et al.

Clinical Microbiology Reviews

study by Monteil et al. demonstrated robust SARS-CoV-2 replication in a human kidney organoid model (98). Several RNA-seq studies identified multiple cell types in the kid- ney that showed extensive ACE2 expression. These included podocytes, glomerular pa- rietal epithelial cells, basal epithelial cells, and tubular epithelial cells (52, 77, 98). Heightened expression of TMPRSS2 and cathepsin L (two suspected facilitators of SARS-CoV-2 infection) was reported in multiple cell types in the kidney (20). Indeed, postmortem electron microscopic analyses of kidney tissues revealed the presence of viral particles in proximal tubules accompanied by abnormal formations of the double- membraned vesicles (289–291). Further immunohistochemical analyses by Diao et al. revealed the presence of macrophage and CD81 T-lymphocyte infiltrates, as well as significant deposition of C5b-9 complement components (290), which is indicative of cytokine release syndrome (292). Further studies are required to establish the pathol- ogy, understand the interplay between host immunity and the infected kidney tissue, and understand the intercellular dissemination of SARS-CoV-2 in this organ.

THE LIVER

Liver injury has been reported in some patients with severe SARS-CoV-2; the avail- able data show that 2% to 11% of COVID-19 patients had liver comorbidities (293). This suggests that this organ is a potential secondary infection site for SARS-CoV-2 (18, 294). Importantly, liver impairment has been previously reported in patients infected with SARS-CoV or MERS-CoV (295, 296). Indicatively, significant elevation of serum ala- nine aminotransferase (ALT), aspartate aminotransferase (AST), and gamma-glutamyl transferase (GGT) levels has been reported in patients with severe SARS-CoV-2 cases (257, 293, 297), as well as abnormal bilirubin levels (18).

Recently, replication of SARS-CoV-2 in the human hepatocellular carcinoma cell line Huh7 was reported (54). Moreover, two separate studies on the RNA-sequence libraries of human tissues identified the cholangiocyte as a potential target for SARS-CoV-2 infection due to high levels of ACE2 expression (52, 77). This was confirmed by Zhao and colleagues using a human liver ductal organoid model in which they observed ro- bust SARS-CoV-2 replication (298). Dysregulated expression of tight junction protein claudin-1 and two bile acid transporters (apical sodium-dependent bile acid trans- porter [ASBT] and cystic fibrosis transmembrane conductance regulator [CFTR]) was also observed, indicating defective tight junction formation and bile transport in chol- angiocytes due to the SARS-CoV-2 infection (298). It remains unclear whether liver injury in severe cases of SARS-CoV-2 is due to viral infection or excessive immune responses. Analysis of cholangiocyte intercellular interaction networks indicates possi- ble interactions between these cells and Kupffer cells via an interaction between CD74 and macrophage migration inhibitory factor (MIF) (77), which triggers a proinflamma- tory response in various organs (299–301). Another point of contention lies in how pre- existing liver conditions increase the risk of severe SARS-CoV-2 infection; this is because ACE2 expression is upregulated significantly in a cirrhotic liver (302, 303). Conversely, Biquard et al. examined patients with metabolic-associated fatty liver dis- ease and reported no significant change in expression levels of ACE2 or TMPRSS2 in the liver (304). Enhanced infection models are therefore needed to evaluate the activity of resident inflammatory cells in the liver during SARS-CoV-2 infection, along with the relationship between changes in expression of SARS-CoV-2 receptors and lipid metab- olism in the liver.

THE PANCREAS

The pancreas is also a potential target for SARS-CoV-2. Pancreatitis was reported in ferrets infected with a feline coronavirus (305, 306). In the case of SARS-CoV-2, clinical reports have described acute hyperglycemia and transient diabetes in COVID-19 patients without a history of type 2 diabetes, which may indicate pancreatic injury (258). Of note, Liu et al. observed increased levels of amylase and lipase in the sera of patients with severe SARS-CoV-2, and some of those patients also presented focal

April 2021 Volume 34 Issue 2 e00133-20

cmr.asm.org 14

Downloaded from http://cmr.asm.org/ on January 21, 2021 by guest

SARS-CoV-2: a Systemic Infection

Clinical Microbiology Reviews

pancreatic enlargement and dilatation of the pancreatic duct under computed tomog- raphy scanning (307). Furthermore, ACE2 is highly expressed by both pancreatic islets and exocrine glands (307, 308). These observations suggest that SARS-CoV-2 may tran- siently infect the pancreatic islets and disrupt glucose metabolism (258). Indeed, Yang et al. demonstrated the permissiveness of human pancreatic alpha and beta cells to SARS-CoV-2, using induced hPSC-derived pancreatic islets and vesicular stomatitis virus (VSV)-based SARS-CoV-2 pseudoviruses (309). Further studies are required to deter- mine the clinical relevance of these observations and possibly also to assess the impact of the infection on patients’ metabolism.

THE NEUROLOGICAL SYSTEM

The involvement of human coronaviruses in a neurological disease was suggested a long time ago. For example, an immunocompromised child with OC43 coronavirus developed fatal progressive encephalitis (310). The neurotropic potential of OC43 and 229E coronaviruses was demonstrated through experimental infection of several microglial, oligodendrocytic, and astrocytic cell lines (311–313). Neurological symp- toms, including headache, confusion, and impaired consciousness, have also been reported in some patients with COVID-19 (314–316). Modest SARS-CoV-2 replication was observed in U251 human glioblastoma cells, which may indicate the neurotropic potential of this virus (54). Very recently, some groups utilized a human brain organoid model to study the pathophysiology of SARS-CoV-2 (317, 318). Although they observed inefficient SARS-CoV-2 replication in this model, they showed that SARS-CoV-2 targets the soma of cortical neurons and is associated with Tau missortment in the axons and soma (317). They also observed colocalization of SARS-CoV-2 particles with Tau phos- phorylated at threonine-231, which is associated with neuronal apoptosis and is indica- tive of the early stage of neurodegeneration (317, 319, 320).

Different routes of coronavirus neuroinvasion have been proposed. Intranasal inoc- ulation of transgenic mice with SARS-CoV expressing human ACE2 results in neuronal dissemination into the brain through the olfactory bulb (321, 322). In the human brain, ACE2 is expressed predominantly in neurons, astrocytes, and oligodendrocytes of the middle temporal gyrus and posterior singular cortex, as well as by endothelial and arte- rial smooth muscle cells (140, 323, 324). Unlike in mice, ACE2 and TMPRSS2 are not expressed in the human olfactory sensory and bulb neurons (325). However, they are expressed in the supporting cells, olfactory basal cells, and perivascular cells (325). These observations not only indicate the possibility of intranasal entry of SARS-CoV-2 into a human brain but could also explain the onset of hyposmia and hypogeusia reported at the early stage of SARS-CoV-2 infection (156, 326). It is worth remembering that the observed neurological symptoms in SARS-CoV-2 patients may also be associ- ated with improper blood coagulation (327–329), resulting in thrombosis of blood ves- sels and ischemic tissue damage. This is indicated by reports describing patients with severe SARS-CoV-2 cases who suffer seizures and impaired consciousness, which are accompanied by ischemic stroke (330, 331). Alternatively, SARS-CoV has also been detected in circulating monocytes (332) and has been shown to induce activation of microglia (321, 333). Furthermore, both monocytic and lymphocytic infiltrates were observed in the brain tissue of a deceased SARS-CoV patient, indicating possible neu- roinflammation during SARS-CoV infection (334). It remains unclear if SARS-CoV-2 can similarly manipulate host innate immune responses to induce inflammatory damage to the blood-brain barrier in order to disseminate into the central nervous system. However, using choroid plexus organoid models, Pellegrini et al. and Fadi et al. demon- strated that SARS-CoV-2 can disrupt the blood-cerebrospinal fluid barrier. They found that SARS-CoV-2 preferentially infected mature choroid plexus epithelium, which abun- dantly expressed ACE2. This resulted in the disruption of tight junction integrity and subsequent leakage of cerebrospinal fluid (318, 335). Nevertheless, further studies using neuronal tissue and blood-brain barrier models are required to investigate SARS- CoV-2 dissemination and pathology in the neurological system.

April 2021 Volume 34 Issue 2 e00133-20

cmr.asm.org 15

Downloaded from http://cmr.asm.org/ on January 21, 2021 by guest

Synowiec et al.

Clinical Microbiology Reviews

The Eye

Eyes were suggested to be potential entry points for SARS-CoV-2 and secondary infection sites. Clinical signs of SARS-CoV-2 infection in the eyes ranged from mild (e.g., chemosis, epiphora, and conjunctival hyperemia) to visual impairment, ophthalmopa- resis, and retinitis (336–338). In multiple cases, viral RNA was detected in ocular dis- charges of SARS-CoV-2 patients both with and without conjunctivitis. The onset of con- junctivitis in some cases precluded the respiratory symptoms (339, 340), and it is hypothesized that SARS-CoV-2 may be transferred from the eyes to the respiratory sys- tem through the nasolacrimal duct connecting the eyes and the nasal cavity (341). Conversely, an onset of ophthalmic clinical signs had also been reported at later stages of COVID-19 (342). Among the components of the human ocular system, expression of SARS-CoV-2 receptor ACE2 had been observed in the conjunctival epithelium, retina, and aqueous humor (343–346). More recently, Makovoz et al. used eye organoids rep- resenting hESC-derived self-formed ectodermal autonomous multizone of ocular cells (SEAM) to study SARS-CoV-2 ocular infection (347). This study identified distinct sub- sets of ACE2-expressing corneal cells, furin-expressing corneal cells, and a presumptive subset of TMPRSS2-expressing corneal cells by bulk RNA sequencing (347). Subsequent infection of eye organoids revealed low levels of SARS-CoV-2 replication in a central cornea and efficient replication in the corneal limbus—the site of corneal and conjunc- tival stem cells (347, 348). Moreover, type I and III interferon responses appeared to be suppressed during SARS-CoV-2 infection of eye organoids, but the NF-κB-mediated inflammatory response was upregulated (347). The replication trend of SARS-CoV-2 observed in the eye organoid was similar to what was observed in intestinal organoids by Lamers et al. (136), highlighting the preference of SARS-CoV-2 for actively proliferat- ing cells. Taking the data together, further studies are required to understand the role of the ocular tissues on SARS-CoV-2 spread.

REPRODUCTIVE SYSTEM

Among the organs affected during COVID-19, reproductive organs have been reported rarely (349, 350). Only a limited number of studies on this topic have been carried out. Bioinformatic analyses and data mining suggest that the testes show a high level of expression of the ACE2 protein (82, 85, 351–354), with the spermatogonia, seminiferous ducts (Sertoli cells), and Leydig cells showing the highest levels (353, 355–362). While the majority of publications postulate that the testes express ACE2, infection of the male reproductive organs by SARS-CoV-2 is not obvious (363, 364). Bian et al. reported the presence of SARS-CoV-2 in testes tissue of deceased COVID-19 patients. This was demonstrated using PCR, immunohistochemistry, and TEM (63). A similar study was carried out by Yang et al., but in this case, 11 of 12 samples tested negative for SARS-COV-2 (365). Li and colleagues evaluated the presence of SARS-CoV- 2 in semen samples from 23 COVID-19 patients in the acute or recovery stage and found 6 of 38 samples positive (366). Song et al. reported that SARS-CoV-2 was not present in semen samples obtained from 12 patients during the recovery phase or in a testicular biopsy specimen from a patient who died during the acute phase (367). In agreement with this, Pan et al. showed that SARS-CoV-2 was not detected in the semen of 34 adult Chinese males recovering from COVID-19 (368), Guo et al. showed that SARS-CoV-2 was not detected in 23 samples collected from patients in the acute and recovery infection phases (369), and Nora et al. did not detect SARS-CoV-2 in 18 semen samples from recovered patients or in two samples from patients with active COVID-19 infection (370). Besides, the virus was not detected in prostatic secretions from 23 COVID-19 patients (371). It is worth noting that Ma et al. and Xu et al. analyzed sex-related hormones levels in 119 and 39 men infected with SARS-CoV-2, respectively. Ma et al. reported some alterations in the hormone levels, whereas Xu et al. did not observe such changes (372, 373).

Except for some transcriptomic studies that evaluated the susceptibility to infection of the female reproductive system (85, 352, 353, 361, 374), data on this subject are

April 2021 Volume 34 Issue 2 e00133-20

cmr.asm.org 16

Downloaded from http://cmr.asm.org/ on January 21, 2021 by guest

SARS-CoV-2: a Systemic Infection

Clinical Microbiology Reviews

FIG 6 Organs affected by COVID-19. The solid and dotted lines indicate direct and indirect viral replication, respectively.

limited (19, 375). Jing et al. reported ubiquitous expression of ACE2 in the ovary, uterus, vagina, and placenta (376). Goad et al., using single-cell sequencing of uterus, myometrium, ovary, fallopian tube, and breast epithelium, found that none of these tissues had high expression of ACE2 and none of them showed coexpression with TMPRSS2 (377). Qiu et al. tested vaginal fluid from 10 women with severe COVID-19 disease, but all the samples were negative for the virus (378). Similar results were obtained in other studies that evaluated vaginal fluid samples and breast milk samples from pregnant patients (379–382). Studies of pregnant women with COVID-19 showed that placenta, amniotic fluid, and/or cord blood analysis results were also negative for SARS-CoV-2 (160, 382–387). However, Fenizia et al. analyzed the presence of the viral RNA in nasopharyngeal swabs from the mothers and the newborns; vaginal swabs; maternal and umbilical cord plasma, placenta, and umbilical cord biopsy specimens; amniotic fluids; and milk. SARS-CoV-2 RNA was found in one blood sample from an umbilical cord, two placenta samples, one vaginal mucosa sample, and one milk sam- ple (388). Additionally, three studies identified an infection in the placenta by qPCR, histological examination, and electron microscopy (389–392). It is difficult at this stage to ultimately determine the long-term effect of the infection in pregnant women for the women and their newborns (393–399). Some studies have shown the absence of vertical transmission or complication in the pregnancy or neonates (383, 386, 387, 395, 400, 401), and there are other studies that have reported vertical transmission of the vi- rus (388, 402–404).

Taking into account all of the cited studies, it is evident that the subject should be further evaluated to determine the effect of SARS-CoV-2 on male and female reproduc- tive systems. There is no evidence of sexual transmission of SARS-CoV-2, but the conse- quences regarding male fertility as well as female fertility and perinatal outcomes are not evident at the moment. Nevertheless, it should be a topic of further study and dis- cussion (396, 405–408).

April 2021 Volume 34 Issue 2 e00133-20

cmr.asm.org 17

Downloaded from http://cmr.asm.org/ on January 21, 2021 by guest

Synowiec et al.

Clinical Microbiology Reviews

CONCLUSIONS AND KEY TAKEAWAY MESSAGES

SARS-CoV-2 is a recently emerged virus that has caused a pandemic that has para- lyzed the world. Our understanding of the threat is still limited, and aside from the mortality rate, the long-term consequences of the infection must be discussed widely, particularly when different epidemic management strategies are considered. While the main COVID-19 outcome involves lungs, other organs are also reported to be affected (Fig. 6). During the COVID-19 pandemic, we have witnessed an incredible boost in the research on coronaviruses. In our opinion, some of the most important work encompasses the employment of human organoids, which are three-dimensional, miniaturized, and sim- plified versions of natural organs. The organoids may be used to mirror in vivo tissue orga- nization and complexity, and the relevance of these models has been proven well, as the results obtained using organoids were in several cases confirmed in the clinic. Importantly, the possible sites of infection impact the person-to-person transmission that shapes the pandemic. Some of the observations, however, still require confirmation in vivo, but even the slight possibility of permanent damage to neural or reproductive tissue, cardiac tissue, or blood vessels in children needs to be verified; this is because adoption of the herd im- munity concept may result in a permanent detrimental effect on society that extends beyond that of the pandemic itself.

ACKNOWLEDGMENTS

This work was supported by the funds provided by the Ministry of Science and Higher Education for research on SARS-CoV-2 (K.P.), by grants from the National Science Center (grants UMO-2017/27/B/NZ6/02488 to K.P.), and by EU-Horizon2020 ITN OrganoVir grant 812673.

We declare no conflict of interest. The funders had no role in the preparation of the manuscript.

REFERENCES

1. van der Hoek L, Pyrc K, Jebbink MF, Vermeulen-Oost W, Berkhout RJ, Wolthers KC, Wertheim-van Dillen PM, Kaandorp J, Spaargaren J, Berkhout B. 2004. Identification of a new human coronavirus. Nat Med 10:368–373. https://doi.org/10.1038/nm1024.

2. Tyrrell DA, Bynoe ML. 1965. Cultivation of a novel type of common-cold virus in organ cultures. Br Med J 1:1467–1470. https://doi.org/10.1136/ bmj.1.5448.1467.

3. Hamre D, Procknow JJ. 1966. A new virus isolated from the human respi- ratory tract. Proc Soc Exp Biol Med 121:190–193. https://doi.org/10.3181/ 00379727-121-30734.

4. Woo PC, Lau SK, Chu CM, Chan KH, Tsoi HW, Huang Y, Wong BH, Poon RW, Cai JJ, Luk WK, Poon LL, Wong SS, Guan Y, Peiris JS, Yuen KY. 2005. Characterization and complete genome sequence of a novel coronavi- rus, coronavirus HKU1, from patients with pneumonia. J Virol 79:884–895. https://doi.org/10.1128/JVI.79.2.884-895.2005.

5. Ksiazek TG, Erdman D, Goldsmith CS, Zaki SR, Peret T, Emery S, Tong S, Urbani C, Comer JA, Lim W, Rollin PE, Dowell SF, Ling AE, Humphrey CD, Shieh WJ, Guarner J, Paddock CD, Rota P, Fields B, DeRisi J, Yang JY, Cox N, Hughes JM, LeDuc JW, Bellini WJ, Anderson LJ, Group SW, SARS Work- ing Group. 2003. A novel coronavirus associated with severe acute respi- ratory syndrome. N Engl J Med 348:1953–1966. https://doi.org/10.1056/ NEJMoa030781.

6. Drosten C, Gunther S, Preiser W, van der Werf S, Brodt HR, Becker S, Rabenau H, Panning M, Kolesnikova L, Fouchier RA, Berger A, Burguiere AM, Cinatl J, Eickmann M, Escriou N, Grywna K, Kramme S, Manuguerra JC, Muller S, Rickerts V, Sturmer M, Vieth S, Klenk HD, Osterhaus AD, Schmitz H, Doerr HW. 2003. Identification of a novel coronavirus in patients with severe acute respiratory syndrome. N Engl J Med 348:1967–1976. https://doi.org/ 10.1056/NEJMoa030747.

7. Li W, Shi Z, Yu M, Ren W, Smith C, Epstein JH, Wang H, Crameri G, Hu Z, Zhang H, Zhang J, McEachern J, Field H, Daszak P, Eaton BT, Zhang S, Wang LF. 2005. Bats are natural reservoirs of SARS-like coronaviruses. Sci- ence 310:676–679. https://doi.org/10.1126/science.1118391.

8. Zaki AM, van Boheemen S, Bestebroer TM, Osterhaus AD, Fouchier RA. 2012. Isolation of a novel coronavirus from a man with pneumonia in

April 2021 Volume 34 Issue 2 e00133-20

cmr.asm.org 18

Saudi Arabia. N Engl J Med 367:1814–1820. https://doi.org/10.1056/

NEJMoa1211721.
9. de Groot RJ, Baker SC, Baric RS, Brown CS, Drosten C, Enjuanes L, Fouchier

RA, Galiano M, Gorbalenya AE, Memish ZA, Perlman S, Poon LL, Snijder EJ, Stephens GM, Woo PC, Zaki AM, Zambon M, Ziebuhr J. 2013. Middle East respiratory syndrome coronavirus (MERS-CoV): announcement of the Co- ronavirus Study Group. J Virol 87:7790–7792. https://doi.org/10.1128/JVI .01244-13.

10. Alagaili AN, Briese T, Mishra N, Kapoor V, Sameroff SC, Burbelo PD, de Wit E, Munster VJ, Hensley LE, Zalmout IS, Kapoor A, Epstein JH, Karesh WB, Daszak P, Mohammed OB, Lipkin WI. 2014. Middle East respiratory syndrome coronavirus infection in dromedary camels in Saudi Arabia. mBio 5:e00884-14. https://doi.org/10.1128/mBio.00884-14.

11. Omrani AS, Al-Tawfiq JA, Memish ZA. 2015. Middle East respiratory syn- drome coronavirus (MERS-CoV): animal to human interaction. Pathog Glob Health 109:354–362. https://doi.org/10.1080/20477724.2015.1122852.

12. Hui DS, Azhar EI, Kim YJ, Memish ZA, Oh MD, Zumla A. 2018. Middle East respiratory syndrome coronavirus: risk factors and determinants of pri- mary, household, and nosocomial transmission. Lancet Infect Dis 18: e217–e227. https://doi.org/10.1016/S1473-3099(18)30127-0.

13. WHO. 2019. MERS monthly summary, November 2019. https://www.who .int/emergencies/mers-cov/en/. Accessed 5 March 2020.

14. Hu B, Zeng LP, Yang XL, Ge XY, Zhang W, Li B, Xie JZ, Shen XR, Zhang YZ, Wang N, Luo DS, Zheng XS, Wang MN, Daszak P, Wang LF, Cui J, Shi ZL. 2017. Discovery of a rich gene pool of bat SARS-related coronaviruses provides new insights into the origin of SARS coronavirus. PLoS Pathog 13:e1006698. https://doi.org/10.1371/journal.ppat.1006698.

15. Zhu N, Zhang D, Wang W, Li X, Yang B, Song J, Zhao X, Huang B, Shi W, Lu R, Niu P, Zhan F, Ma X, Wang D, Xu W, Wu G, Gao GF, Tan W, China Novel Coronavirus Investigating and Research Team. 2020. A novel coro- navirus from patients with pneumonia in China, 2019. N Engl J Med 382:727–733. https://doi.org/10.1056/NEJMoa2001017.

16. Dhama K, Khan S, Tiwari R, Sircar S, Bhat S, Malik YS, Singh KP, Chaicumpa W, Bonilla-Aldana DK, Rodriguez-Morales AJ. 2020. Coronavirus disease 2019–COVID-19. Clin Microbiol Rev 33:e00028-20. https://doi.org/10.1128/ CMR.00028-20.

Downloaded from http://cmr.asm.org/ on January 21, 2021 by guest

SARS-CoV-2: a Systemic Infection

Clinical Microbiology Reviews

17. Chan JF, Kok KH, Zhu Z, Chu H, To KK, Yuan S, Yuen KY. 2020. Genomic characterization of the 2019 novel human-pathogenic coronavirus isolated from a patient with atypical pneumonia after visiting Wuhan. Emerg Microbes Infect 9:221–236. https://doi.org/10.1080/22221751 .2020.1719902.

18. GuanWJ,NiZY,HuY,LiangWH,OuCQ,HeJX,LiuL,ShanH,LeiCL,Hui DSC, Du B, Li LJ, Zeng G, Yuen KY, Chen RC, Tang CL, Wang T, Chen PY, Xiang J, Li SY, Wang JL, Liang ZJ, Peng YX, Wei L, Liu Y, Hu YH, Peng P, Wang JM, Liu JY, Chen Z, Li G, Zheng ZJ, Qiu SQ, Luo J, Ye CJ, Zhu SY, Zhong NS, China Medical Treatment Expert Group for Covid-19. 2020. Clinical characteristics of coronavirus disease 2019 in China. N Engl J Med 382:1708–1720. https://doi.org/10.1056/NEJMoa2002032.

19. Fu L, Wang B, Yuan T, Chen X, Ao Y, Fitzpatrick T, Li P, Zhou Y, Lin YF, DuanQ,LuoG,FanS,LuY,FengA,ZhanY,LiangB,CaiW,ZhangL,Du X, Li L, Shu Y, Zou H. 2020. Clinical characteristics of coronavirus disease 2019 (COVID-19) in China: a systematic review and meta-analysis. J Infect 80:656–665. https://doi.org/10.1016/j.jinf.2020.03.041.

20. Puelles VG, Lutgehetmann M, Lindenmeyer MT, Sperhake JP, Wong MN, Allweiss L, Chilla S, Heinemann A, Wanner N, Liu S, Braun F, Lu S, Pfefferle S, Schroder AS, Edler C, Gross O, Glatzel M, Wichmann D, Wiech T, Kluge S, Pueschel K, Aepfelbacher M, Huber TB. 2020. Multiorgan and renal tropism of SARS-CoV-2. N Engl J Med 383:590–592. https://doi.org/ 10.1056/NEJMc2011400.

21. Abrams JY, Godfred-Cato SE, Oster ME, Chow EJ, Koumans EH, Bryant B, Leung JW, Belay ED. 2020. Multisystem inflammatory syndrome in chil- dren (MIS-C) associated with SARS-CoV-2: a systematic review. J Pediatr 226:45–54.e1. https://doi.org/10.1016/j.jpeds.2020.08.003.

22. Belhadjer Z, Meot M, Bajolle F, Khraiche D, Legendre A, Abakka S, Auriau J, Grimaud M, Oualha M, Beghetti M, Wacker J, Ovaert C, Hascoet S, Selegny M, Malekzadeh-Milani S, Maltret A, Bosser G, Giroux N, Bonnemains L, Bordet J, Di Filippo S, Mauran P, Falcon-Eicher S, Thambo JB, Lefort B, Moceri P, Houyel L, Renolleau S, Bonnet D. 2020. Acute heart failure in multisystem inflammatory syndrome in children (MIS-C) in the context of global SARS-CoV-2 pandemic. Circulation 142:429–436. https://doi.org/10 .1161/CIRCULATIONAHA.120.048360.

23. Chiotos K, Bassiri H, Behrens EM, Blatz AM, Chang J, Diorio C, Fitzgerald JC, Topjian A, John ARO. 2020. Multisystem inflammatory syndrome in children during the COVID-19 pandemic: a case series. J Pediatric Infect Dis Soc 9:393–398. https://doi.org/10.1093/jpids/piaa069.

24. Dufort EM, Koumans EH, Chow EJ, Rosenthal EM, Muse A, Rowlands J, Barranco MA, Maxted AM, Rosenberg ES, Easton D, Udo T, Kumar J, Pulver W, Smith L, Hutton B, Blog D, Zucker H, New York State and Cen- ters for Disease Control and Prevention Multisystem Inflammatory Syn- drome in Children Investigation Team. 2020. Multisystem inflammatory syndrome in children in New York State. N Engl J Med 383:347–358. https://doi.org/10.1056/NEJMoa2021756.

25. Morris SB, Schwartz NG, Patel P, Abbo L, Beauchamps L, Balan S, Lee EH, Paneth-Pollak R, Geevarughese A, Lash MK, Dorsinville MS, Ballen V, Eiras DP, Newton-Cheh C, Smith E, Robinson S, Stogsdill P, Lim S, Fox SE, Richardson G, Hand J, Oliver NT, Kofman A, Bryant B, Ende Z, Datta D, Belay E, Godfred-Cato S. 2020. Case series of multisystem inflammatory syndrome in adults associated with SARS-CoV-2 infection – United King- dom and United States, March-August 2020. MMWR Morb Mortal Wkly Rep 69:1450–1456. https://doi.org/10.15585/mmwr.mm6940e1.

26. Rauf A, Vijayan A, John ST, Krishnan R, Latheef A. 2020. Multisystem inflam- matory syndrome with features of atypical Kawasaki disease during COVID-19 pandemic. Indian J Pediatr 87:745–747. https://doi.org/10 .1007/s12098-020-03357-1.

27. Singh-Grewal D, Lucas R, Macartney K, Cheng AC, Wood N, Ostring G, Britton P, Crawford N, Burgner D. 2020. Update on the COVID-19-associ- ated inflammatory syndrome in children and adolescents; paediatric inflammatory multisystem syndrome-temporally associated with SARS- CoV-2. J Paediatr Child Health 56:1173–1177. https://doi.org/10.1111/ jpc.15049.

28. Valverde I, Singh Y, Sanchez-de-Toledo J, Theocharis P, Chikermane A, Di Filippo S, Kucinska B, Mannarino S, Tamariz-Martel A, Gutierrez-Larraya F, Soda G, Vandekerckhove K, Gonzalez Barlatey F, McMahon CJ, Marcora SA, Pace Napoleone C, Duong P, Tuo G, Deri A, Nepali G, Ilina M, Ciliberti P, Miller O. 9 November 2020, posting date. Acute cardiovascular manifesta- tions in 286 children with multisystem inflammatory syndrome associated with COVID-19 infection in Europe. Circulation https://doi.org/10.1161/ circulationaha.120.050065.

29. Alsaied T, Tremoulet AH, Burns JC, Saidi A, Dionne A, Lang SM, Newburger JW, de Ferranti S, Friedman KG. 9 November 2020, posting date. Review of

cardiac involvement in multisystem inflammatory syndrome in children.

Circulation https://doi.org/10.1161/CIRCULATIONAHA.120.049836.
30. Tabaac S, Kothari P, Cassidy-Smith T. 5 November 2020, posting date. Multisystem inflammatory syndrome in children. J Emerg Med https://

doi.org/10.1016/j.jemermed.2020.10.009.
31. Masters PS. 2006. The molecular biology of coronaviruses. Adv Virus Res

66:193–292. https://doi.org/10.1016/S0065-3527(06)66005-3.
32. Milewska A, Zarebski M, Nowak P, Stozek K, Potempa J, Pyrc K. 2014. Human coronavirus NL63 utilizes heparan sulfate proteoglycans for attachment to

target cells. J Virol 88:13221–13230. https://doi.org/10.1128/JVI.02078-14. 33. Lang J, Yang N, Deng J, Liu K, Yang P, Zhang G, Jiang C. 2011. Inhibition of SARS pseudovirus cell entry by lactoferrin binding to heparan sulfate proteoglycans. PLoS One 6:e23710. https://doi.org/10.1371/journal.pone

.0023710.
34. Vlasak R, Luytjes W, Spaan W, Palese P. 1988. Human and bovine corona-

viruses recognize sialic acid-containing receptors similar to those of influenza C viruses. Proc Natl Acad Sci U S A 85:4526–4529. https://doi .org/10.1073/pnas.85.12.4526.

35. Chan CM, Chu H, Wang Y, Wong BH, Zhao X, Zhou J, Yang D, Leung SP, Chan JF, Yeung ML, Yan J, Lu G, Gao GF, Yuen KY. 2016. Carcinoembry- onic antigen-related cell adhesion molecule 5 is an important surface attachment factor that facilitates entry of Middle East respiratory syn- drome coronavirus. J Virol 90:9114–9127. https://doi.org/10.1128/JVI .01133-16.

36. Yeager CL, Ashmun RA, Williams RK, Cardellichio CB, Shapiro LH, Look AT, Holmes KV. 1992. Human aminopeptidase N is a receptor for human coro- navirus 229E. Nature 357:420–422. https://doi.org/10.1038/357420a0.

37. Raj VS, Mou H, Smits SL, Dekkers DH, Muller MA, Dijkman R, Muth D, Demmers JA, Zaki A, Fouchier RA, Thiel V, Drosten C, Rottier PJ, Osterhaus AD, Bosch BJ, Haagmans BL. 2013. Dipeptidyl peptidase 4 is a functional receptor for the emerging human coronavirus-EMC. Nature 495:251–254. https://doi.org/10.1038/nature12005.

38. Collins AR. 1993. HLA class I antigen serves as a receptor for human co- ronavirus OC43. Immunol Invest 22:95–103. https://doi.org/10.3109/ 08820139309063393.

39. Hulswit RJG, Lang Y, Bakkers MJG, Li W, Li Z, Schouten A, Ophorst B, van Kuppeveld FJM, Boons GJ, Bosch BJ, Huizinga EG, de Groot RJ. 2019. Human coronaviruses OC43 and HKU1 bind to 9-O-acetylated sialic acids via a conserved receptor-binding site in spike protein domain A. Proc Natl Acad Sci U S A 116:2681–2690. https://doi.org/10.1073/pnas.1809667116.

40. Hofmann H, Pyrc K, van der Hoek L, Geier M, Berkhout B, Pohlmann S. 2005. Human coronavirus NL63 employs the severe acute respiratory syndrome coronavirus receptor for cellular entry. Proc Natl Acad Sci U S A 102:7988–7993. https://doi.org/10.1073/pnas.0409465102.

41. Cantuti-Castelvetri L, Ojha R, Pedro LD, Djannatian M, Franz J, Kuivanen S, van der Meer F, Kallio K, Kaya T, Anastasina M, Smura T, Levanov L, Szirovicza L, Tobi A, Kallio-Kokko H, Osterlund P, Joensuu M, Meunier FA, Butcher SJ, Winkler MS, Mollenhauer B, Helenius A, Gokce O, Teesalu T, Hepojoki J, Vapalahti O, Stadelmann C, Balistreri G, Simons M. 2020. Neu- ropilin-1 facilitates SARS-CoV-2 cell entry and infectivity. Science 370:856–860. https://doi.org/10.1126/science.abd2985.

42. Daly JL, Simonetti B, Klein K, Chen KE, Williamson MK, Anton-Plagaro C, Shoemark DK, Simon-Gracia L, Bauer M, Hollandi R, Greber UF, Horvath P, Sessions RB, Helenius A, Hiscox JA, Teesalu T, Matthews DA, Davidson AD, Collins BM, Cullen PJ, Yamauchi Y. 2020. Neuropilin-1 is a host factor for SARS-CoV-2 infection. Science 370:861–865. https://doi.org/10.1126/ science.abd3072.

43. Simmons G, Gosalia DN, Rennekamp AJ, Reeves JD, Diamond SL, Bates P. 2005. Inhibitors of cathepsin L prevent severe acute respiratory syndrome coronavirus entry. Proc Natl Acad Sci U S A 102:11876–11881. https://doi .org/10.1073/pnas.0505577102.

44. Kawase M, Shirato K, van der Hoek L, Taguchi F, Matsuyama S. 2012. Si- multaneous treatment of human bronchial epithelial cells with serine and cysteine protease inhibitors prevents severe acute respiratory syn- drome coronavirus entry. J Virol 86:6537–6545. https://doi.org/10.1128/ JVI.00094-12.

45. Matsuyama S, Nagata N, Shirato K, Kawase M, Takeda M, Taguchi F. 2010. Efficient activation of the severe acute respiratory syndrome coronavirus spike protein by the transmembrane protease TMPRSS2. J Virol 84:12658–12664. https://doi.org/10.1128/JVI.01542-10.

46. Glowacka I, Bertram S, Muller MA, Allen P, Soilleux E, Pfefferle S, Steffen I, Tsegaye TS, He Y, Gnirss K, Niemeyer D, Schneider H, Drosten C, Pohlmann S. 2011. Evidence that TMPRSS2 activates the severe acute respiratory syn- drome coronavirus spike protein for membrane fusion and reduces viral

April 2021 Volume 34 Issue 2 e00133-20

cmr.asm.org 19

Downloaded from http://cmr.asm.org/ on January 21, 2021 by guest

Synowiec et al.

Clinical Microbiology Reviews

control by the humoral immune response. J Virol 85:4122–4134. https://

doi.org/10.1128/JVI.02232-10.

47. Shulla A, Heald-Sargent T, Subramanya G, Zhao J, Perlman S, Gallagher T.
2011. A transmembrane serine protease is linked to the severe acute re- spiratory syndrome coronavirus receptor and activates virus entry. J Virol 85:873–882. https://doi.org/10.1128/JVI.02062-10.

48. Sungnak W, Huang N, Becavin C, Berg M, Queen R, Litvinukova M, Talavera-Lopez C, Maatz H, Reichart D, Sampaziotis F, Worlock KB, Yoshida M, Barnes JL, HCA Lung Biological Network. 2020. SARS-CoV-2 entry factors are highly expressed in nasal epithelial cells together with innate immune genes. Nat Med 26:681–687. https://doi.org/10.1038/ s41591-020-0868-6.

49. Hoffmann M, Kleine-Weber H, Schroeder S, Kruger N, Herrler T, Erichsen S, Schiergens TS, Herrler G, Wu NH, Nitsche A, Muller MA, Drosten C, Pohlmann S. 2020. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell 181:271–280. e8. https://doi.org/10.1016/j.cell.2020.02.052.

50. Milewska A, Falkowski K, Kalinska M, Bielecka E, Naskalska A, Mak P, Lesner A, Ochman M, Urlik M, Potempa J, Kantyka T, Pyrc K. 2020. Kallik- rein 13: a new player in coronaviral infections. bioRxiv https://doi.org/10 .1101/2020.03.01.971499.

51. ZhouF,YuT,DuR,FanG,LiuY,LiuZ,XiangJ,WangY,SongB,GuX, GuanL,WeiY,LiH,WuX,XuJ,TuS,ZhangY,ChenH,CaoB.2020.Clini- cal course and risk factors for mortality of adult inpatients with COVID- 19 in Wuhan, China: a retrospective cohort study. Lancet 395:1054–1062. https://doi.org/10.1016/S0140-6736(20)30566-3.

52. Zou X, Chen K, Zou J, Han P, Hao J, Han Z. 2020. Single-cell RNA-seq data analysis on the receptor ACE2 expression reveals the potential risk of dif- ferent human organs vulnerable to 2019-nCoV infection. Front Med 14:185–192. https://doi.org/10.1007/s11684-020-0754-0.

53. ZhangH,ZhouP,WeiY,YueH,WangY,HuM,ZhangS,CaoT,YangC,Li M,GuoG,ChenX,ChenY,LeiM,LiuH,ZhaoJ,PengP,WangC-Y,DuR. 2020. Histopathologic changes and SARS–CoV-2 immunostaining in the lung of a patient with COVID-19. Ann Intern Med 173:185–192. https:// doi.org/10.7326/m20-0533.

54. Chu H, Chan JF-W, Yuen TT-T, Shuai H, Yuan S, Wang Y, Hu B, Yip CC-Y, Tsang JO-L, Huang X, Chai Y, Yang D, Hou Y, Chik KK-H, Zhang X, Fung AY-F, Tsoi H-W, Cai J-P, Chan W-M, Ip JD, Chu AW-H, Zhou J, Lung DC, Kok K-H, To KK-W, Tsang OT-Y, Chan K-H, Yuen K-Y. 2020. Comparative tropism, replication kinetics, and cell damage profiling of SARS-CoV-2 and SARS-CoV with implications for clinical manifestations, transmissibil- ity, and laboratory studies of COVID-19: an observational study. Lancet Microbe 1:e14–e23. https://doi.org/10.1016/S2666-5247(20)30004-5.

55. Iwasawa T, Sato M, Yamaya T, Sato Y, Uchida Y, Kitamura H, Hagiwara E, Komatsu S, Utsunomiya D, Ogura T. 2020. Ultra-high-resolution com- puted tomography can demonstrate alveolar collapse in novel coronavi- rus (COVID-19) pneumonia. Jpn J Radiol 38:394–398. https://doi.org/10 .1007/s11604-020-00956-y.

56. Xu Z, Shi L, Wang Y, Zhang J, Huang L, Zhang C, Liu S, Zhao P, Liu H, Zhu L, Tai Y, Bai C, Gao T, Song J, Xia P, Dong J, Zhao J, Wang F-S. 2020. Patho- logical findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir Med 8:420–422. https://doi.org/10.1016/S2213 -2600(20)30076-X.

57. Barton LM, Duval EJ, Stroberg E, Ghosh S, Mukhopadhyay S. 2020. COVID-19 autopsies, Oklahoma, USA. Am J Clin Pathol 153:725–733. https://doi.org/10.1093/ajcp/aqaa062.

58. Tian S, Hu W, Niu L, Liu H, Xu H, Xiao SY. 2020. Pulmonary pathology of early-phase 2019 novel coronavirus (COVID-19) pneumonia in two patients with lung cancer. J Thorac Oncol 15:700–704. https://doi.org/10 .1016/j.jtho.2020.02.010.

59. Tian S, Xiong Y, Liu H, Niu L, Guo J, Liao M, Xiao SY. 2020. Pathological study of the 2019 novel coronavirus disease (COVID-19) through post- mortem core biopsies. Mod Pathol 33:1007–1014. https://doi.org/10 .1038/s41379-020-0536-x.

60. Rockx B, Kuiken T, Herfst S, Bestebroer T, Lamers MM, Oude Munnink BB, de Meulder D, van Amerongen G, van den Brand J, Okba NMA, Schipper D, van Run P, Leijten L, Sikkema R, Verschoor E, Verstrepen B, Bogers W, Langermans J, Drosten C, Fentener van Vlissingen M, Fouchier R, de Swart R, Koopmans M, Haagmans BL. 2020. Comparative pathogenesis of COVID-19, MERS, and SARS in a nonhuman primate model. Science 368:1012–1015. https://doi.org/10.1126/science.abb7314.

61. Schaefer I-M, Padera RF, Solomon IH, Kanjilal S, Hammer MM, Hornick JL, Sholl LM. 2020. In situ detection of SARS-CoV-2 in lungs and airways of

April 2021 Volume 34 Issue 2 e00133-20

patients with COVID-19. Mod Pathol 33:2104–2114. https://doi.org/10

.1038/s41379-020-0595-z.
62. Duan F, Guo L, Yang L, Han Y, Thakur A, Nilsson-Payant BE, Wang P,

Zhang Z, Ma CY, Zhou X, Han T, Zhang T, Wang X, Xu D, Duan X, Xiang J, Tse HF, Liao C, Luo W, Huang FP, Chen YW, Evans T, Schwartz RE, tenOever B, Ho DD, Chen S, Lian Q, Chen HJ. 20 August 2020, posting date. Modeling COVID-19 with human pluripotent stem cell-derived cells reveals synergistic effects of anti-inflammatory macrophages with ACE2 inhibition against SARS-CoV-2. Res Sq https://doi.org/10.21203/rs.3.rs -62758/v1.

63. Bian X-W, Yao X-H, Ping Y-F, Yu S, Shi Y, Luo T, He Z-C, Tang R, Chen C, Fu W-J, Zhang H, Zhang H-R, Xiang D-F, Li Q-R, Huang X, Li T, Zhao P, Wang C,FeiX,CaiJ,ZhaoL,ZhangH,LiuZ,LiuL,WangG,NieX,ZhouY,RenL, LiuQ,WangY,AoQ,WangX,DuanY,LiJ,XiongJ,XuS,ZhangJ,Huang S,YangM,HuangB,LiX,PengL,XiP,HuaX,SuH,WangchengS,YuC, Wu H, Li H, Ren Y, The COVID-19 Pathology Team, et al. 2020. Autopsy of COVID-19 patients in China. National Science Rev 7:1414–1418. https:// doi.org/10.1093/nsr/nwaa123.

64. Ruan Q, Yang K, Wang W, Jiang L, Song J. 2020. Clinical predictors of mortality due to COVID-19 based on an analysis of data of 150 patients from Wuhan. Intensive Care Med 46:846–848. https://doi.org/10.1007/ s00134-020-05991-x.

65. Chen G, Wu D, Guo W, Cao Y, Huang D, Wang H, Wang T, Zhang X, Chen H,YuH,ZhangX,ZhangM,WuS,SongJ,ChenT,HanM,LiS,LuoX, Zhao J, Ning Q. 2020. Clinical and immunological features of severe and moderate coronavirus disease 2019. J Clin Invest 130:2620–2629. https:// doi.org/10.1172/JCI137244.

66. Tindle C, Fuller M, Fonseca A, Taheri S, Ibeawuchi S-R, Beutler N, Claire A, Castillo V, Hernandez M, Russo H, Duran J, Crotty Alexander LE, Tipps A, Lin G, Thistlethwaite PA, Chattopadhyay R, Rogers TF, Sahoo D, Ghosh P, Das S. 2020. Adult stem cell-derived complete lung organoid models emulate lung disease in COVID-19. bioRxiv https://doi.org/10.1101/2020 .10.17.344002.

67. Pruijssers AJ, George AS, Schäfer A, Leist SR, Gralinksi LE, Dinnon KH, Yount BL, Agostini ML, Stevens LJ, Chappell JD, Lu X, Hughes TM, Gully K, Martinez DR, Brown AJ, Graham RL, Perry JK, Du Pont V, Pitts J, Ma B, Babusis D, Murakami E, Feng JY, Bilello JP, Porter DP, Cihlar T, Baric RS, Denison MR, Sheahan TP. 2020. Remdesivir potently inhibits SARS-CoV-2 in human lung cells and chimeric SARS-CoV expressing the SARS-CoV-2 RNA polymerase in mice. bioRxiv https://doi.org/10.1101/2020.04.27 .064279.

68. Sheahan TP, Sims AC. 2020. An orally bioavailable broad-spectrum anti- viral inhibits SARS-CoV-2 in human airway epithelial cell cultures and multiple coronaviruses in mice. Sci Transl Med 12:eabb5883. https://doi .org/10.1126/scitranslmed.abb5883.

69. Salgado-Benvindo C, Thaler M, Tas A, Ogando NS, Bredenbeek PJ, Ninaber DK, Wang Y, Hiemstra PS, Snijder EJ, van Hemert MJ. 2020. Sura- min inhibits SARS-CoV-2 infection in cell culture by interfering with early steps of the replication cycle. Antimicrob Agents Chemother 64:e00900- 20. https://doi.org/10.1128/AAC.00900-20.

70. Jureka AS, Silvas JA, Basler CF. 2020. Propagation, inactivation, and safety testing of SARS-CoV-2. Viruses 12:622. https://doi.org/10.3390/ v12060622.

71. Hsin F, Chao T-L, Chan Y-R, Kao H-C, Liu W-D, Wang J-T, Pang Y-H, Lin C- H, Tsai Y-M, Lin J-Y, Chang S-Y, Liu HM. 2020. Distinct inductions of and responses to type I and type III interferons promote infections in two SARS-CoV-2 isolates. bioRxiv https://doi.org/10.1101/2020.04.30.071357.

72. Milewska A, Chi Y, Szczepanski A, Barreto-Duran E, Liu K, Liu D, Guo X, Ge Y, Li J, Cui L, Ochman M, Urlik M, Rodziewicz-Motowidlo S, Zhu F, Szczubialka K, Nowakowska M, Pyrc K. 20 November 2020, posting date. HTCC as a highly effective polymeric inhibitor of SARS-CoV-2 and MERS- CoV. J Virol https://doi.org/10.1128/JVI.01622-20.

73. Hikmet F, Méar L, Uhlén M, Lindskog C. 2020. The protein expression profile of ACE2 in human tissues. 16:e9610. https://doi.org/10.15252/ msb.20209610.

74. Cagno V. 2020. SARS-CoV-2 cellular tropism. Lancet Microbe 1:e2–e3. https://doi.org/10.1016/S2666-5247(20)30008-2.

75. Blanco-Melo D, Nilsson-Payant BE, Liu W-C, Uhl S, Hoagland D, Møller R, Jordan TX, Oishi K, Panis M, Sachs D, Wang TT, Schwartz RE, Lim JK, Albrecht RA, tenOever BR. 2020. Imbalanced host response to SARS-CoV- 2 drives development of COVID-19. Cell 181:1036–1045.e9. https://doi .org/10.1016/j.cell.2020.04.026.

76. Zhao Y, Zhao Z, Wang Y, Zhou Y, Ma Y, Zuo W. 2020. Single-cell RNA cmr.asm.org 20

Downloaded from http://cmr.asm.org/ on January 21, 2021 by guest

SARS-CoV-2: a Systemic Infection

Clinical Microbiology Reviews

expression profiling of ACE2, the putative receptor of Wuhan 2019-nCov.

bioRxiv https://www.biorxiv.org/content/10.1101/2020.01.26.919985v2.

77. Qi F, Qian S, Zhang S, Zhang Z. 2020. Single cell RNA sequencing of 13 human tissues identify cell types and receptors of human coronaviruses. Biochem Biophys Res Commun 526:135–140. https://doi.org/10.1016/j
.bbrc.2020.03.044.

78. Li H, Liu SM, Yu XH, Tang SL, Tang CK. 2020. Coronavirus disease 2019
(COVID-19): current status and future perspectives. Int J Antimicrob
Agents 55:105951. https://doi.org/10.1016/j.ijantimicag.2020.105951.

79. Aguiar JA, Tremblay BJ-M, Mansfield MJ, Woody O, Lobb B, Banerjee A, Chandiramohan A, Tiessen N, Cao Q, Dvorkin-Gheva A, Revill S, Miller MS, Carlsten C, Organ L, Joseph C, John A, Hanson P, Austin R, McManus BM, Jenkins G, Mossman K, Ask K, Doxey AC, Hirota JA. 2020. Gene expression and in situ protein profiling of candidate SARS-CoV-2 recep- tors in human airway epithelial cells and lung tissue. Eur Respir J
526:2001123. https://doi.org/10.1183/13993003.01123-2020.

80. Chu H, Chan JF, Wang Y, Yuen TT, Chai Y, Hou Y, Shuai H, Yang D, Hu B, Huang X, Zhang X, Cai JP, Zhou J, Yuan S, Kok KH, To KK, Chan IH, Zhang AJ, Sit KY, Au WK, Yuen KY. 2020. Comparative replication and immune activation profiles of SARS-CoV-2 and SARS-CoV in human lungs: an ex vivo study with implications for the pathogenesis of COVID-19. Clin
Infect Dis 71:1400–1409. https://doi.org/10.1093/cid/ciaa410.

81. Lukassen S, Chua RL, Trefzer T, Kahn NC, Schneider MA, Muley T, Winter H, Meister M, Veith C, Boots AW, Hennig BP, Kreuter M, Conrad C, Eils R. 2020. SARS-CoV-2 receptor ACE2 and TMPRSS2 are primarily expressed in bronchial transient secretory cells. EMBO J 39:e105114. https://doi
.org/10.15252/embj.20105114.

82. Zhou L, Niu Z, Jiang X, Zhang Z, Zheng Y, Wang Z, Zhu Y, Gao L, Wang X,
Sun Q. 2020. Systemic analysis of tissue cells potentially vulnerable to SARS-CoV-2 infection by the protein-proofed single-cell RNA profiling of ACE2, TMPRSS2 and Furin proteases. iScience 23:101744. https://doi.org/ 10.1016/j.isci.2020.101744.

83. Zhang H, Rostami MR, Leopold PL, Mezey JG, O’Beirne SL, Strulovici- Barel Y, Crystal RG. 2020. Expression of the SARS-CoV-2 ACE2 receptor in the human airway epithelium. Am J Respir Crit Care Med 202:219–229. https://doi.org/10.1164/rccm.202003-0541OC.

84. Suzuki T, Itoh Y, Sakai Y, Saito A, Okuzaki D, Motooka D, Minami S, Kobayashi T, Yamamoto T, Okamoto T, Takayama K. 2020. Generation of human bronchial organoids for SARS-CoV-2 research. bioRxiv https://doi .org/10.1101/2020.05.25.115600.

85. Hui KPY, Cheung M-C, Perera RAPM, Ng K-C, Bui CHT, Ho JCW, Ng MMT, Kuok DIT, Shih KC, Tsao S-W, Poon LLM, Peiris M, Nicholls JM, Chan MCW. 2020. Tropism, replication competence, and innate immune responses of the coronavirus SARS-CoV-2 in human respiratory tract and conjunc- tiva: an analysis in ex-vivo and in-vitro cultures. Lancet Respir Med 8:687–695. https://doi.org/10.1016/S2213-2600(20)30193-4.

86. Chua RL, Lukassen S, Trump S, Hennig BP, Wendisch D, Pott F, Debnath O, Thürmann L, Kurth F, Völker MT. 2020. COVID-19 severity correlates with airway epithelium–immune cell interactions identified by single- cell analysis. Nat Biotechnol 38:970–979. https://doi.org/10.1038/s41587 -020-0602-4.

87. Chua RL, Lukassen S, Trump S, Hennig BP, Wendisch D, Pott F, Debnath O, Thürmann L, Kurth F, Kazmierski J, Timmermann B, Twardziok S, Schneider S, Machleidt F, Müller-Redetzky H, Krannich A, Schmidt S, Balzer F, Liebig J, Loske J, Eils J, Ishaque N, von Kalle C, Hocke A, Witzenrath M, Goffinet C, Drosten C, Laudi S, Lehmann I, Conrad C, Sander L-E, Eils R. 2020. Cross-talk between the airway epithelium and activated immune cells defines sever- ity in COVID-19. medRxiv https://doi.org/10.1101/2020.04.29.20084327.

88. Zhou J, Chiu MC, Cun L, Liu X, Zhao X, Wang D, Wei Y, Chu H, Cai J-P, Yip CC-Y. 9 September 2020, posting date. Human airway organoids model SARS-CoV-2 high infectiousness and evasion of interferon response. Res Sq https://doi.org/10.21203/rs.3.rs-67556/v1.

89. Milewska A, Ciejka J, Kaminski K, Karewicz A, Bielska D, Zeglen S, Karolak W, Nowakowska M, Potempa J, Bosch BJ, Pyrc K, Szczubialka K. 2013. Novel polymeric inhibitors of HCoV-NL63. Antiviral Res 97:112–121. https://doi.org/10.1016/j.antiviral.2012.11.006.

90. Pyrc K, Sims AC, Dijkman R, Jebbink M, Long C, Deming D, Donaldson E, Vabret A, Baric R, van der Hoek L, Pickles R. 2010. Culturing the uncultur- able: human coronavirus HKU1 infects, replicates, and produces progeny virions in human ciliated airway epithelial cell cultures. J Virol 84:11255–- 11263. https://doi.org/10.1128/JVI.00947-10.

91. Huang X, Dong W, Milewska A, Golda A, Qi Y, Zhu QK, Marasco WA, Baric RS, Sims AC, Pyrc K, Li W, Sui J. 2015. Human coronavirus HKU1 spike pro- tein uses O-acetylated sialic acid as an attachment receptor determinant

and employs hemagglutinin-esterase protein as a receptor-destroying

enzyme. J Virol 89:7202–7213. https://doi.org/10.1128/JVI.00854-15.
92. Banach BS, Orenstein JM, Fox LM, Randell SH, Rowley AH, Baker SC. 2009. Human airway epithelial cell culture to identify new respiratory viruses: coronavirus NL63 as a model. J Virol Methods 156:19–26. https://doi.org/

10.1016/j.jviromet.2008.10.022.
93. Milewska A, Nowak P, Owczarek K, Szczepanski A, Zarebski M, Hoang A,

Berniak K, Wojarski J, Zeglen S, Baster Z, Rajfur Z, Pyrc K. 2017. Entry of human coronavirus NL63 into the cell. J Virol 92:e01933-17. https://doi .org/10.1128/JVI.01933-17.

94. Pyrc K, Stoz_ek K, Wojcik K, Gawron K, Zeglen S, Karolak W, Wojarski J, Ochman M, Hubalewska-Mazgaj M, Bochenek G, Sanak M, Zembala M, Szczeklik A, Potempa J. 2012. Use of sensitive, broad-spectrum molecu- lar assays and human airway epithelium cultures for detection of respi- ratory pathogens. PLoS One 7:e32582. https://doi.org/10.1371/journal .pone.0032582.

95. Farsani SM, Deijs M, Dijkman R, Molenkamp R, Jeeninga RE, Ieven M, Goossens H, van der Hoek L. 2015. Culturing of respiratory viruses in well-differentiated pseudostratified human airway epithelium as a tool to detect unknown viruses. Influenza Other Respir Viruses 9:51–57. https://doi.org/10.1111/irv.12297.

96. Owczarek K, Szczepanski A, Milewska A, Baster Z, Rajfur Z, Sarna M, Pyrc K. 2018. Early events during human coronavirus OC43 entry to the cell. Sci Rep 8:7124. https://doi.org/10.1038/s41598-018-25640-0.

97. Milewska A, Kula-Pacurar A, Wadas J, Suder A, Szczepanski A, Dabrowska A, Owczarek K, Ochman M, Stacel T, Rajfur Z, Labaj P, Branicki W, Pyrc K. 2020. Replication of SARS-CoV-2 in human respiratory epithelium. bio- Rxiv https://doi.org/10.1101/2020.03.20.999029.

98. Monteil V, Kwon H, Prado P, Hagelkruys A, Wimmer RA, Stahl M, Leopoldi A, Garreta E, Hurtado Del Pozo C, Prosper F, Romero JP, Wirnsberger G, Zhang H, Slutsky AS, Conder R, Montserrat N, Mirazimi A, Penninger JM. 2020. Inhibition of SARS-CoV-2 infections in engineered human tissues using clinical-grade soluble human ACE2. Cell 181:905–913.e7. https:// doi.org/10.1016/j.cell.2020.04.004.

99. Vanderheiden A, Ralfs P, Chirkova T, Upadhyay AA, Zimmerman MG, Bedoya S, Aoued H, Tharp GM, Pellegrini KL, Manfredi C, Sorscher E, Mainou B, Lobby JL, Kohlmeier JE, Lowen AC, Shi P-Y, Menachery VD, Anderson LJ, Grakoui A, Bosinger SE, Suthar MS. 2020. Type I and type III interferons restrict SARS-CoV-2 infection of human airway epithelial cul- tures. J Virol 94:e00985-20. https://doi.org/10.1128/JVI.00985-20.

100. Mulay A, Konda B, Garcia G, Yao C, Beil S, Sen C, Purkayastha A, Kolls JK, Pociask DA, Pessina P, Sainz de Aja J, Garcia-de-Alba C, Kim CF, Gomperts B, Arumugaswami V, Stripp BR. 2020. SARS-CoV-2 infection of primary human lung epithelium for COVID-19 modeling and drug discovery. bio- Rxiv https://doi.org/10.1101/2020.06.29.174623.

101. Zhu N, Wang W, Liu Z, Liang C, Wang W, Ye F, Huang B, Zhao L, Wang H, Zhou W, Deng Y, Mao L, Su C, Qiang G, Jiang T, Zhao J, Wu G, Song J, Tan W. 2020. Morphogenesis and cytopathic effect of SARS-CoV-2 infec- tion in human airway epithelial cells. Nat Commun 11:3910. https://doi .org/10.1038/s41467-020-17796-z.

102. Ravindra NG, Alfajaro MM, Gasque V, Wei J, Filler RB, Huston NC, Wan H, Szigeti-Buck K, Wang B, Montgomery RR, Eisenbarth SC, Williams A, Pyle AM, Iwasaki A, Horvath TL, Foxman EF, van Dijk D, Wilen C. 2020. Single- cell longitudinal analysis of SARS-CoV-2 infection in human bronchial epithelial cells. bioRxiv https://doi.org/10.1101/2020.05.06.081695.

103. Pizzorno A, Padey B, Julien T, Trouillet-Assant S, Traversier A, Errazuriz- Cerda E, Fouret J, Dubois J, Gaymard A, Lescure F-X, Dulière V, Brun P, Constant S, Poissy J, Lina B, Yazdanpanah Y, Terrier O, Rosa-Calatrava M. 2020. Characterization and treatment of SARS-CoV-2 in nasal and bron- chial human airway epithelia. bioRxiv https://doi.org/10.1101/2020.03 .31.017889.

104. Dinnon KH, Leist SR, Schäfer A, Edwards CE, Martinez DR, Montgomery SA, West A, Yount BL, Hou YJ, Adams LE, Gully KL, Brown AJ, Huang E, Bryant MD, Choong IC, Glenn JS, Gralinski LE, Sheahan TP, Baric RS. 2020. A mouse-adapted model of SARS-CoV-2 to test COVID-19 countermeas- ures. Nature 586:560–566. https://doi.org/10.1038/s41586-020-2708-8.

105. Terrier O, Dilly S, Pizzorno A, Henri J, Berenbaum F, Lina B, Fève B, Adnet F, Sabbah M, Rosa-Calatrava M, Maréchal V, Schwok AS. 2020. Broad- spectrum antiviral activity of naproxen: from Influenza A to SARS-CoV-2 Coronavirus. bioRxiv https://doi.org/10.1101/2020.04.30.069922.

106. Mykytyn AZ, Breugem TI, Riesebosch S, Schipper D, van den Doel PB, Rottier RJ, Lamers MM, Haagmans BL. 2020. The SARS-CoV-2 multibasic cleavage site facilitates early serine protease-mediated entry into

April 2021 Volume 34 Issue 2 e00133-20

cmr.asm.org 21

Downloaded from http://cmr.asm.org/ on January 21, 2021 by guest

Synowiec et al.

Clinical Microbiology Reviews

organoid-derived human airway cells. bioRxiv https://doi.org/10.1101/

2020.09.07.286120.

107. PeiR,FengJ,ZhangY,SunH,LiL,YangX,HeJ,XiaoS,XiongJ,LinY,
Wen K, Zhou H, Chen J, Rong Z, Chen X. 2020. Human embryonic stem cell-derived lung organoids: a model for SARS-CoV-2 infection and drug test. bioRxiv https://doi.org/10.1101/2020.08.10.244350.

108. Han Y, Duan X, Yang L, Nilsson-Payant BE, Wang P, Duan F, Tang X, Yaron TM, Zhang T, Uhl S, Bram Y, Richardson C, Zhu J, Zhao Z, Redmond D, Houghton S, Nguyen DT, Xu D, Wang X, Jessurun J, Borczuk A, Huang Y, Johnson JL, Liu Y, Xiang J, Wang H, Cantley LC, tenOever BR, Ho DD, Pan FC, Evans T, Chen HJ, Schwartz RE, Chen S. 28 October 2020, posting date. Identification of SARS-CoV-2 inhibitors using lung and co- lonic organoids. Nature https://doi.org/10.1038/s41586-020-2901-9.

109. Huang J, Hume AJ, Abo KM, Werder RB, Villacorta-Martin C, Alysandratos K-D, Beermann ML, Simone-Roach C, Olejnik J, Suder EL, Bullitt E, Hinds A, Sharma A, Bosmann M, Wang R, Hawkins F, Burks EJ, Saeed M, Wilson AA, Mühlberger E, Kotton DN. 2020. SARS-CoV-2 infection of pluripotent stem cell-derived human lung alveolar type 2 cells elicits a rapid epithe- lial-intrinsic inflammatory response. bioRxiv https://doi.org/10.1101/2020 .06.30.175695.

110. Youk J, Kim T, Evans KV, Jeong Y-I, Hur Y, Hong SP, Kim JH, Yi K, Kim SY, Na KJ, Bleazard T, Kim HM, Fellows M, Mahbubani KT, Saeb-Parsy K, Kim SY, Kim YT, Koh GY, Choi B-S, Ju YS, Lee J-H. 21 October 2020, posting date. Three-dimensional human alveolar stem cell culture models reveal infection response to SARS-CoV-2. Cell Stem Cell https://doi.org/10 .1016/j.stem.2020.10.004.

111. Salahudeen AA, Choi SS, Rustagi A, Zhu J, de la OSM, Flynn RA, Margalef- Català M, Santos AJM, Ju J, Batish A, van Unen V, Usui T, Zheng GXY, Edwards CE, Wagar LE, Luca V, Anchang B, Nagendran M, Nguyen K, Hart DJ, Terry JM, Belgrader P, Ziraldo SB, Mikkelsen TS, Harbury PB, Glenn JS, Garcia KC, Davis MM, Baric RS, Sabatti C, Amieva MR, Blish CA, Desai TJ, Kuo CJ. 2020. Progenitor identification and SARS-CoV-2 infec- tion in long-term human distal lung organoid cultures. bioRxiv https:// doi.org/10.1101/2020.07.27.212076.

112. Youk J, Kim T, Evans KV, Jeong Y-I, Hur Y, Hong SP, Kim JH, Yi K, Kim SY, Na KJ, Bleazard T, Kim HM, Ivory N, Mahbubani KT, Saeb-Parsy K, Kim YT, Koh GY, Choi B-S, Ju YS, Lee J-H. 2020. Robust three-dimensional expan- sion of human adult alveolar stem cells and SARS-CoV-2 infection. bio- Rxiv https://doi.org/10.1101/2020.07.10.194498.

113. Lee N, Hui D, Wu A, Chan P, Cameron P, Joynt GM, Ahuja A, Yung MY, Leung CB, To KF, Lui SF, Szeto CC, Chung S, Sung JJ. 2003. A major out- break of severe acute respiratory syndrome in Hong Kong. N Engl J Med 348:1986–1994. https://doi.org/10.1056/NEJMoa030685.

114. Leung WK, To KF, Chan PK, Chan HL, Wu AK, Lee N, Yuen KY, Sung JJ. 2003. Enteric involvement of severe acute respiratory syndrome-associ- ated coronavirus infection. Gastroenterology 125:1011–1017. https://doi .org/10.1016/s0016-5085(03)01215-0.

115. Booth CM, Matukas LM, Tomlinson GA, Rachlis AR, Rose DB, Dwosh HA, Walmsley SL, Mazzulli T, Avendano M, Derkach P, Ephtimios IE, Kitai I, Mederski BD, Shadowitz SB, Gold WL, Hawryluck LA, Rea E, Chenkin JS, Cescon DW, Poutanen SM, Detsky AS. 2003. Clinical features and short- term outcomes of 144 patients with SARS in the greater Toronto area. JAMA 289:2801–2809. https://doi.org/10.1001/jama.289.21.JOC30885.

116. Vabret A, Mourez T, Dina J, van der Hoek L, Gouarin S, Petitjean J, Brouard J, Freymuth F. 2005. Human coronavirus NL63. Emerg Infect Dis 11:1225–1229. https://doi.org/10.3201/eid1108.050110.

117. Xiao F, Tang M, Zheng X, Liu Y, Li X, Shan H. 2020. Evidence for gastroin- testinal infection of SARS-CoV-2. Gastroenterology 158:1831–1833.e3. https://doi.org/10.1053/j.gastro.2020.02.055.

118. Vabret A, Mourez T, Gouarin S, Petitjean J, Freymuth F. 2003. An out- break of coronavirus OC43 respiratory infection in Normandy, France. Clin Infect Dis 36:985–989. https://doi.org/10.1086/374222.

119. Assiri A, Al-Tawfiq JA, Al-Rabeeah AA, Al-Rabiah FA, Al-Hajjar S, Al-Barrak A, Flemban H, Al-Nassir WN, Balkhy HH, Al-Hakeem RF, Makhdoom HQ, Zumla AI, Memish ZA. 2013. Epidemiological, demographic, and clinical characteristics of 47 cases of Middle East respiratory syndrome coronavi- rus disease from Saudi Arabia: a descriptive study. Lancet Infect Dis 13:752–761. https://doi.org/10.1016/S1473-3099(13)70204-4.

120. Song Y, Liu P, Shi XL, Chu YL, Zhang J, Xia J, Gao XZ, Qu T, Wang MY. 2020. SARS-CoV-2 induced diarrhoea as onset symptom in patient with COVID- 19. Gut 69:1143–1144. https://doi.org/10.1136/gutjnl-2020-320891.

121. Cheung KS, Hung IF, Chan PP, Lung KC, Tso E, Liu R, Ng YY, Chu MY, Chung TW, Tam AR, Yip CC, Leung KH, Yim-Fong Fung A, Zhang RR, Lin Y, Cheng HM, Zhang AJ, To KK, Chan KH, Yuen KY, Leung WK. 2020.

Gastrointestinal manifestations of SARS-CoV-2 infection and virus load in fecal samples from the Hong Kong Cohort and systematic review and meta-analysis. Gastroenterology 159:81–95. https://doi.org/10.1053/j.gastro .2020.03.065.

122. Holshue ML, DeBolt C, Lindquist S, Lofy KH, Wiesman J, Bruce H, Spitters C, Ericson K, Wilkerson S, Tural A, Diaz G, Cohn A, Fox L, Patel A, Gerber SI, Kim L, Tong S, Lu X, Lindstrom S, Pallansch MA, Weldon WC, Biggs HM, Uyeki TM, Pillai SK, Washington State 2019-nCoV Case Investigation Team. 2020. First case of 2019 novel coronavirus in the United States. N Engl J Med 382:929–936. https://doi.org/10.1056/NEJMoa2001191.

123. Zang R, Gomez Castro MF, McCune BT, Zeng Q, Rothlauf PW, Sonnek NM, Liu Z, Brulois KF, Wang X, Greenberg HB, Diamond MS, Ciorba MA, Whelan SPJ, Ding S. 2020. TMPRSS2 and TMPRSS4 promote SARS-CoV-2 infection of human small intestinal enterocytes. Sci Immunol 5: eabc3582. https://doi.org/10.1126/sciimmunol.abc3582.

124. WuY,GuoC,TangL,HongZ,ZhouJ,DongX,YinH,XiaoQ,TangY,QuX, Kuang L, Fang X, Mishra N, Lu J, Shan H, Jiang G, Huang X. 2020. Prolonged presence of SARS-CoV-2 viral RNA in faecal samples. Lancet Gastroenterol Hepatol 5:434–435. https://doi.org/10.1016/S2468-1253(20)30083-2.

125. Zuo T, Zhang F, Lui GCY, Yeoh YK, Li AYL, Zhan H, Wan Y, Chung A, Cheung CP, Chen N, Lai CKC, Chen Z, Tso EYK, Fung KSC, Chan V, Ling L, Joynt G, Hui DSC, Chan FKL, Chan PKS, Ng SC. 2020. Alterations in gut microbiota of patients with COVID-19 during time of hospitalization. Gastroenterology 159:944–955.e8. https://doi.org/10.1053/j.gastro.2020 .05.048.

126. Zhou J, Li C, Liu X, Chiu MC, Zhao X, Wang D, Wei Y, Lee A, Zhang AJ, Chu H, Cai JP, Yip CC, Chan IH, Wong KK, Tsang OT, Chan KH, Chan JF, To KK, Chen H, Yuen KY. 2020. Infection of bat and human intestinal orga- noids by SARS-CoV-2. Nat Med 26:1077–1083. https://doi.org/10.1038/ s41591-020-0912-6.

127. Zhang Y, Chen C, Zhu S, Shu C, Wang D, Song J, Song Y, Zhen W, Feng Z, Wu G, Xu J, Xu W, National Health Commission Key Laboratory for Medi- cal Virology, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China. 2020. Isolation of 2019-nCoV from a stool specimen of a laboratory-confirmed case of the coronavirus disease 2019 (COVID-19). China CDC Wkly 2:123–124. https://doi.org/10.46234/ccdcw2020.033.

128. Stanifer ML, Kee C, Cortese M, Zumaran CM, Triana S, Mukenhirn M, Kraeusslich HG, Alexandrov T, Bartenschlager R, Boulant S. 2020. Critical role of type III interferon in controlling SARS-CoV-2 infection in human intestinal epithelial cells. Cell Rep 32:107863. https://doi.org/10.1016/j .celrep.2020.107863.

129. Cinatl J, Jr, Hoever G, Morgenstern B, Preiser W, Vogel JU, Hofmann WK, Bauer G, Michaelis M, Rabenau HF, Doerr HW. 2004. Infection of cultured intestinal epithelial cells with severe acute respiratory syndrome corona- virus. Cell Mol Life Sci 61:2100–2112. https://doi.org/10.1007/s00018 -004-4222-9.

130. Lee S, Yoon GY, Myoung J, Kim SJ, Ahn DG. 2020. Robust and persistent SARS-CoV-2 infection in the human intestinal brush border expressing cells. Emerg Microbes Infect 9:2169–2179. https://doi.org/10.1080/ 22221751.2020.1827985.

131. Bojkova D, McGreig JE, McLaughlin K-M, Masterson SG, Widera M, Krähling V, Ciesek S, Wass MN, Michaelis M, Cinatl J. 2020. SARS-CoV-2 and SARS-CoV differ in their cell tropism and drug sensitivity profiles. bioRxiv https://doi.org/10.1101/2020.04.03.024257.

132. Sato T, Stange DE, Ferrante M, Vries RG, Van Es JH, Van den Brink S, Van Houdt WJ, Pronk A, Van Gorp J, Siersema PD, Clevers H. 2011. Long-term expansion of epithelial organoids from human colon, adenoma, adeno- carcinoma, and Barrett’s epithelium. Gastroenterology 141:1762–1772. https://doi.org/10.1053/j.gastro.2011.07.050.

133. Zhou J, Li C, Zhao G, Chu H, Wang D, Yan HH, Poon VK, Wen L, Wong BH, Zhao X, Chiu MC, Yang D, Wang Y, Au-Yeung RKH, Chan IH, Sun S, Chan JF, To KK, Memish ZA, Corman VM, Drosten C, Hung IF, Zhou Y, Leung SY, Yuen KY. 2017. Human intestinal tract serves as an alternative infec- tion route for Middle East respiratory syndrome coronavirus. Sci Adv 3: eaao4966. https://doi.org/10.1126/sciadv.aao4966.

134. Ettayebi K, Crawford SE, Murakami K, Broughman JR, Karandikar U, Tenge VR, Neill FH, Blutt SE, Zeng XL, Qu L, Kou B, Opekun AR, Burrin D, Graham DY, Ramani S, Atmar RL, Estes MK. 2016. Replication of human noroviruses in stem cell-derived human enteroids. Science 353:1387–1393. https://doi.org/10.1126/science.aaf5211.

135. Zou WY, Blutt SE, Crawford SE, Ettayebi K, Zeng XL, Saxena K, Ramani S, Karandikar UC, Zachos NC, Estes MK. 2019. Human intestinal enteroids:

April 2021 Volume 34 Issue 2 e00133-20

cmr.asm.org 22

Downloaded from http://cmr.asm.org/ on January 21, 2021 by guest

SARS-CoV-2: a Systemic Infection

Clinical Microbiology Reviews

new models to study gastrointestinal virus infections. Methods Mol Biol

1576:229–247. https://doi.org/10.1007/7651_2017_1.

136. Lamers MM, Beumer J, van der Vaart J, Knoops K, Puschhof J, Breugem TI, Ravelli RBG, van Schayck JP, Mykytyn AZ, Duimel HQ, van Donselaar E, Riesebosch S, Kuijpers HJH, Schipper D, van de Wetering WJ, de Graaf M, Koopmans M, Cuppen E, Peters PJ, Haagmans BL, Clevers H. 2020. SARS-CoV-2 productively infects human gut enterocytes. Science
369:50–54. https://doi.org/10.1126/science.abc1669.

137. Giobbe GG, Bonfante F, Zambaiti E, Gagliano O, Jones BC, Luni C, Laterza
C, Perin S, Stuart HT, Pagliari M, Bortolami A, Mazzetto E, Manfredi A, Colantuono C, Di Filippo L, Pellegata A, Li VSW, Eaton S, Thapar N, Cacchiarelli D, Elvassore N, De Coppi P. 2020. SARS-CoV-2 infection and replication in human fetal and pediatric gastric organoids. bioRxiv https://doi.org/10.1101/2020.06.24.167049.

138. Andersen KG, Rambaut A, Lipkin WI, Holmes EC, Garry RF. 2020. The proximal origin of SARS-CoV-2. Nat Med 26:450–452. https://doi.org/10 .1038/s41591-020-0820-9.

139. HuangC,WangY,LiX,RenL,ZhaoJ,HuY,ZhangL,FanG,XuJ,GuX, ChengZ,YuT,XiaJ,WeiY,WuW,XieX,YinW,LiH,LiuM,XiaoY,GaoH, Guo L, Xie J, Wang G, Jiang R, Gao Z, Jin Q, Wang J, Cao B. 2020. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 395:497–506. https://doi.org/10.1016/S0140-6736(20)30183-5.

140. Hamming I, Timens W, Bulthuis M, Lely A, Navis G, van Goor H. 2004. Tis- sue distribution of ACE2 protein, the functional receptor for SARS coro- navirus. A first step in understanding SARS pathogenesis. J Pathol 203:631–637. https://doi.org/10.1002/path.1570.

141. XuJ,ChuM,ZhongF,TanX,TangG,MaiJ,LaiN,GuanC,LiangY,Liao G. 2020. Digestive symptoms of COVID-19 and expression of ACE2 in di- gestive tract organs. Cell Death Discov 6:76. https://doi.org/10.1038/ s41420-020-00307-w.

142. ChenH,XuanB,YanY,ZhuX,ShenC,ZhaoG,JiL,XuD,XiongH,YuT, Li X, Liu Q, Chen Y, Cui Y, Hong J, Fang J-Y. 2020. Profiling ACE2 expres- sion in colon tissue of healthy adults and colorectal cancer patients by single-cell transcriptome analysis. medRxiv https://doi.org/10.1101/2020 .02.15.20023457.

143. Garg M, Royce SG, Tikellis C, Shallue C, Batu D, Velkoska E, Burrell LM, Patel SK, Beswick L, Jackson A, Britto K, Lukies M, Sluka P, Wardan H, Hirokawa Y, Tan CW, Faux M, Burgess AW, Hosking P, Monagle S, Thomas M, Gibson PR, Lubel J. 2020. Imbalance of the renin-angiotensin system may contribute to inflammation and fibrosis in IBD: a novel therapeutic target? Gut 69:841–851. https://doi.org/10.1136/gutjnl-2019-318512.

144. AnP,JiM,RenH,SuJ,DingNS,KangJ,YinA,ZhouQ,ShenL,ZhaoL, JiangX,XiaoY,TanW,LvX,LiJ,LiuS,ZhouJ,ChenH,XuY,LiuJ,Chen M, Cao J, Zhou Z, Shen L, Tan S, Yu H, Dong W, Ding Y. 2020. Prevention of COVID-19 in patients with inflammatory bowel disease in Wuhan. Lancet Gastroenterol Hepatol 5:525–527. https://doi.org/10.1016/S2468 -1253(20)30121-7.

145. PanL,MuM,YangP,SunY,WangR,YanJ,LiP,HuB,WangJ,HuC,JinY, Niu X, Ping R, Du Y, Li T, Xu G, Hu Q, Tu L. 2020. Clinical characteristics of COVID-19 patients with digestive symptoms in Hubei, China: a descrip- tive, cross-sectional, multicenter study. Am J Gastroenterol 115:766–773. https://doi.org/10.14309/ajg.0000000000000620.

146. Yu CM, Wong RS, Wu EB, Kong SL, Wong J, Yip GW, Soo YO, Chiu ML, Chan YS, Hui D, Lee N, Wu A, Leung CB, Sung JJ. 2006. Cardiovascular complications of severe acute respiratory syndrome. Postgrad Med J 82:140–144. https://doi.org/10.1136/pgmj.2005.037515.

147. Alhogbani T. 2016. Acute myocarditis associated with novel Middle East respiratory syndrome coronavirus. Ann Saudi Med 36:78–80. https://doi .org/10.5144/0256-4947.2016.78.

148. Emami A, Javanmardi F, Pirbonyeh N, Akbari A. 2020. Prevalence of underlying diseases in hospitalized patients with COVID-19: a systematic review and meta-analysis. Arch Acad Emerg Med 8:e35.

149. ZhangJ,LuS,WangX,JiaX,LiJ,LeiH,LiuZ,LiaoF,JiM,LvX,KangJ,Tian S, Ma J, Wu D, Gong Y, Xu Y, Dong W. 2020. Do underlying cardiovascular diseases have any impact on hospitalised patients with COVID-19? Heart 106:1148–1153. https://doi.org/10.1136/heartjnl-2020-316909.

150. Kang Y, Chen T, Mui D, Ferrari V, Jagasia D, Scherrer-Crosbie M, Chen Y, Han Y. 2020. Cardiovascular manifestations and treatment considera- tions in Covid-19. Heart 106:1132–1141. https://doi.org/10.1136/heartjnl -2020-317056.

151. Slawinski G, Lewicka E. 2020. What should a cardiologist know about coronavirus disease 2019? Kardiol Pol 78:278–283. https://doi.org/10 .33963/KP.15302.

152. Cheng P, Zhu H, Witteles RM, Wu JC, Quertermous T, Wu SM, Rhee JW.

April 2021 Volume 34 Issue 2 e00133-20

2020. Cardiovascular risks in patients with COVID-19: potential mecha- nisms and areas of uncertainty. Curr Cardiol Rep 22:34. https://doi.org/ 10.1007/s11886-020-01293-2.

153. Karbalai Saleh S, Oraii A, Soleimani A, Hadadi A, Shajari Z, Montazeri M, Moradi H, Talebpour M, Sadat Naseri A, Balali P, Akhbari M, Ashraf H. 2020. The association between cardiac injury and outcomes in hospital- ized patients with COVID-19. Intern Emerg Med 15:1415–1424. https:// doi.org/10.1007/s11739-020-02466-1.

154. Sisti N, Valente S, Mandoli GE, Santoro C, Sciaccaluga C, Franchi F, Cameli P, Mondillo S, Cameli M. 30 July 2020, posting date. COVID-19 in patients with heart failure: the new and the old epidemic. Postgrad Med J https://doi.org/10.1136/postgradmedj-2020-138080.

155. Zhu H, Rhee JW, Cheng P, Waliany S, Chang A, Witteles RM, Maecker H, Davis MM, Nguyen PK, Wu SM. 2020. Cardiovascular complications in patients with COVID-19: consequences of viral toxicities and host immune response. Curr Cardiol Rep 22:32. https://doi.org/10.1007/ s11886-020-01292-3.

156. Paniz-Mondolfi A, Bryce C, Grimes Z, Gordon RE, Reidy J, Lednicky J, Sordillo EM, Fowkes M. 2020. Central nervous system involvement by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). J Med Virol 92:699–702. https://doi.org/10.1002/jmv.25915.

157. Varga Z, Flammer AJ, Steiger P, Haberecker M, Andermatt R, Zinkernagel AS, Mehra MR, Schuepbach RA, Ruschitzka F, Moch H. 2020. Endothelial cell infection and endotheliitis in COVID-19. Lancet 395:1417–1418. https://doi.org/10.1016/S0140-6736(20)30937-5.

158. Hess DC, Eldahshan W, Rutkowski E. 2020. COVID-19-related stroke. Transl Stroke Res 11:322–325. https://doi.org/10.1007/s12975-020-00818-9.
159. Sardu C, Gambardella J, Morelli MB, Wang X, Marfella R, Santulli G. 2020.

Is COVID-19 an endothelial disease? Clinical and basic evidence. Pre-

Prints https://www.preprints.org/manuscript/202004.0204/v1.
160. GuoJ,WeiX,LiQ,LiL,YangZ,ShiY,QinY,ZhangX,WangX,ZhiX, Meng D. 2020. Single-cell RNA analysis on ACE2 expression provides insights into SARS-CoV-2 potential entry into the bloodstream and heart

injury. J Cell Physiol 235:9884–9894. https://doi.org/10.1002/jcp.29802. 161. Jung F, Kruger-Genge A, Franke RP, Hufert F, Kupper JH. 2020. COVID-19 and the endothelium. Clin Hemorheol Microcirc 75:7–11. https://doi

.org/10.3233/CH-209007.
162. Guervilly C, Burtey S, Sabatier F, Cauchois R, Lano G, Abdili E, Daviet F,

Arnaud L, Brunet P, Hraiech S, Jourde-Chiche N, Koubi M, Lacroix R, Pietri L, Berda Y, Robert T, Degioanni C, Velier M, Papazian L, Kaplanski G, Dignat-George F. 2020. Circulating endothelial cells as a marker of endo- thelial injury in severe COVID-19. J Infect Dis 222:1789–1793. https://doi .org/10.1093/infdis/jiaa528.

163. Nizzoli ME, Merati G, Tenore A, Picone C, Consensi E, Perotti L, Ferretti VV, Sambo M, Di Sabatino A, Iotti GA, Arcaini L, Bruno R, Belliato M. 2020. Circu- lating endothelial cells in COVID-19. Am J Hematol 95:E187–E188. https:// doi.org/10.1002/ajh.25881.

164. Viner RM, Whittaker E. 2020. Kawasaki-like disease: emerging complica- tion during the COVID-19 pandemic. Lancet 395:1741–1743. https://doi .org/10.1016/S0140-6736(20)31129-6.

165. Riphagen S, Gomez X, Gonzalez-Martinez C, Wilkinson N, Theocharis P. 2020. Hyperinflammatory shock in children during COVID-19 pandemic. Lancet 395:1607–1608. https://doi.org/10.1016/S0140-6736(20)31094-1.

166. Verdoni L, Mazza A, Gervasoni A, Martelli L, Ruggeri M, Ciuffreda M, Bonanomi E, D’Antiga L. 2020. An outbreak of severe Kawasaki-like dis- ease at the Italian epicentre of the SARS-CoV-2 epidemic: an observatio- nal cohort study. Lancet 395:1771–1778. https://doi.org/10.1016/S0140 -6736(20)31103-X.

167. Jones VG, Mills M, Suarez D, Hogan CA, Yeh D, Bradley Segal J, Nguyen EL, Barsh GR, Maskatia S, Mathew R. 2020. COVID-19 and Kawasaki dis- ease: novel virus and novel case. Hosp Pediatr 10:537–540. https://doi .org/10.1542/hpeds.2020-0123.

168. Licciardi F, Pruccoli G, Denina M, Parodi E, Taglietto M, Rosati S, Montin D. 2020. SARS-CoV-2-induced Kawasaki-like hyperinflammatory syn- drome: a novel COVID phenotype in children. Pediatrics 146:e20201711. https://doi.org/10.1542/peds.2020-1711.

169. Dhanalakshmi K, Venkataraman A, Balasubramanian S, Madhusudan M, Amperayani S, Putilibai S, Sadasivam K, Ramachandran B, Ramanan AV. 2020. Epidemiological and clinical profile of pediatric inflammatory mul- tisystem syndrome – temporally associated with SARS-CoV-2 (PIMS-TS) in Indian children. Indian Pediatr 57:1010–1014. https://doi.org/10.1007/ s13312-020-2025-1.

170. Heidemann SM, Tilford B, Bauerfeld C, Martin A, Garcia RU, Yagiela L, Sarnaik AP. 2020. Three cases of pediatric multisystem inflammatory

cmr.asm.org 23

Downloaded from http://cmr.asm.org/ on January 21, 2021 by guest

Synowiec et al.

Clinical Microbiology Reviews

syndrome associated with COVID-19 due to SARS-CoV-2. Am J Case Rep

21:e925779. https://doi.org/10.12659/AJCR.925779.

171. Godfred-Cato S, Bryant B, Leung J, Oster ME, Conklin L, Abrams J, Roguski
K, Wallace B, Prezzato E, Koumans EH, Lee EH, Geevarughese A, Lash MK, Reilly KH, Pulver WP, Thomas D, Feder KA, Hsu KK, Plipat N, Richardson G, Reid H, Lim S, Schmitz A, Pierce T, Hrapcak S, Datta D, Morris SB, Clarke K, Belay E, California MIS-C Response Team. 2020. COVID-19-associated mul- tisystem inflammatory syndrome in children – United States, March-July 2020. MMWR Morb Mortal Wkly Rep 69:1074–1080. https://doi.org/10 .15585/mmwr.mm6932e2.

172. Belot A, Antona D, Renolleau S, Javouhey E, Hentgen V, Angoulvant F, Delacourt C, Iriart X, Ovaert C, Bader-Meunier B, Kone-Paut I, Levy-Bruhl D. 2020. SARS-CoV-2-related paediatric inflammatory multisystem syn- drome, an epidemiological study, France, 1 March to 17 May 2020. Euro Surveill 25:2001010. https://doi.org/10.2807/1560-7917.ES.2020.25.22 .2001010.

173. Khan KS, Ullah I. 2 August 2020, posting date. SARS-CoV-2 causes Kawa- saki-like disease in children: cases reported in Pakistan. J Med Virol https://doi.org/10.1002/jmv.26340.

174. Moraleda C, Serna-Pascual M, Soriano-Arandes A, Simo S, Epalza C, Santos M, Grasa C, Rodriguez M, Soto B, Gallego N, Ruiz Y, Urretavizcaya- Martinez M, Pareja M, Sanz-Santaeufemia FJ, Fumado V, Lanaspa M, Jordan I, Prieto L, Belda S, Toral-Vazquez B, Rincon E, Gil-Villanueva N, Mendez-Echevarria A, Castillo-Serrano A, Riviere JG, Soler-Palacin P, Rojo P, Tagarro A. 25 July 2020, posting date. Multi-inflammatory syndrome in children related to SARS-CoV-2 in Spain. Clin Infect Dis https://doi .org/10.1093/cid/ciaa1042.

175. Loke YH, Berul CI, Harahsheh AS. 2020. Multisystem inflammatory syn- drome in children: is there a linkage to Kawasaki disease? Trends Cardio- vasc Med 30:389–396. https://doi.org/10.1016/j.tcm.2020.07.004.

176. Davies P, Evans C, Kanthimathinathan HK, Lillie J, Brierley J, Waters G, Johnson M, Griffiths B, Du Pre P, Mohammad Z, Deep A, Playfor S, Singh D, Inwald D, Jardine M, Ross O, Shetty N, Worrall M, Sinha R, Koul A, Whittaker E, Vyas H, Scholefield BR, Ramnarayan P. 2020. Intensive care admissions of children with paediatric inflammatory multisystem syndrome temporally associated with SARS-CoV-2 (PIMS-TS) in the UK: a multicentre observatio- nal study. Lancet Child Adolesc Health 4:669–677. https://doi.org/10.1016/ S2352-4642(20)30215-7.

177. DeBiasi RL, Song X, Delaney M, Bell M, Smith K, Pershad J, Ansusinha E, Hahn A, Hamdy R, Harik N, Hanisch B, Jantausch B, Koay A, Steinhorn R, Newman K, Wessel D. 2020. Severe coronavirus disease-2019 in children and young adults in the Washington, DC, metropolitan region. J Pediatr 223:199–203.e1. https://doi.org/10.1016/j.jpeds.2020.05.007.

178. Lee PY, Day-Lewis M, Henderson LA, Friedman KG, Lo J, Roberts JE, Lo MS, Platt CD, Chou J, Hoyt KJ, Baker AL, Banzon TM, Chang MH, Cohen E, de Ferranti SD, Dionne A, Habiballah S, Halyabar O, Hausmann JS, Hazen MM, Janssen E, Meidan E, Nelson RW, Nguyen AA, Sundel RP, Dedeoglu F, Nigrovic PA, Newburger JW, Son MBF. 2020. Distinct clinical and immunological features of SARS-CoV-2-induced multisystem inflamma- tory syndrome in children. J Clin Invest 130:5942–5950. https://doi.org/ 10.1172/JCI141113.

179. Jiang L, Tang K, Levin M, Irfan O, Morris SK, Wilson K, Klein JD, Bhutta ZA. 2020. COVID-19 and multisystem inflammatory syndrome in children and adolescents. Lancet Infect Dis 20:e276–e288. https://doi.org/10 .1016/S1473-3099(20)30651-4.

180. Chowdhary A, Joy E, Plein S, Abdel-Rahman SE. 4 September 2020, post- ing date. Multisystem inflammatory syndrome in an adult with SARS- CoV-2 infection. Eur Heart J Cardiovasc Imaging https://doi.org/10.1093/ ehjci/jeaa232.

181. Ng KF, Kothari T, Bandi S, Bird PW, Goyal K, Zoha M, Rai V, Tang JW. 2020. COVID-19 multisystem inflammatory syndrome in three teenagers with confirmed SARS-CoV-2 infection. J Med Virol 92:2880–2886. https:// doi.org/10.1002/jmv.26206.

182. Toubiana J, Poirault C, Corsia A, Bajolle F, Fourgeaud J, Angoulvant F, Debray A, Basmaci R, Salvador E, Biscardi S, Frange P, Chalumeau M, Casanova JL, Cohen JF, Allali S. 2020. Kawasaki-like multisystem inflam- matory syndrome in children during the covid-19 pandemic in Paris, France: prospective observational study. BMJ 369:m2094. https://doi .org/10.1136/bmj.m2094.

183. Burgi Vieira C, Ferreira AT, Botelho Cardoso F, Pelicano Paulos J, Germano N. 2020. Kawasaki-like syndrome as an emerging complication of SARS-CoV-2 infection in young adults. Eur J Case Rep Intern Med 7:001886. https://doi.org/10.12890/2020_001886.

184. Kofman AD, Sizemore EK, Detelich JF, Albrecht B, Piantadosi AL. 2020. A

April 2021 Volume 34 Issue 2 e00133-20

young adult with COVID-19 and multisystem inflammatory syndrome in children (MIS-C)-like illness: a case report. BMC Infect Dis 20:716. https:// doi.org/10.1186/s12879-020-05439-z.

185. Sturrock BR, Milne KM, Chevassut TJ. 2020. The renin-angiotensin system – a therapeutic target in COVID-19? Clin Med (Lond) 20:e72–e75. https:// doi.org/10.7861/clinmed.2020-0146.

186. Amirfakhryan H. 2020. Kawasaki-like disease in children with COVID-19: a hypothesis. Med Hypotheses 143:110117. https://doi.org/10.1016/j .mehy.2020.110117.

187. Thakkar AN, Tea I, Al-Mallah MH. 2020. Cardiovascular implications of COVID-19 infections. Methodist Debakey Cardiovasc J 16:146–154. https://doi.org/10.14797/mdcj-16-2-146.

188. Cohen JB, Hanff TC, Bress AP, South AM. 2020. Relationship between ACE2 and other components of the renin-angiotensin system. Curr Hypertens Rep 22:44. https://doi.org/10.1007/s11906-020-01048-y.

189. Samavati L, Uhal BD. 2020. ACE2, much more than just a receptor for SARS-COV-2. Front Cell Infect Microbiol 10:317. https://doi.org/10.3389/ fcimb.2020.00317.

190. Gheblawi M, Wang K, Viveiros A, Nguyen Q, Zhong JC, Turner AJ, Raizada MK, Grant MB, Oudit GY. 2020. Angiotensin converting enzyme 2: SARS- CoV-2 receptor and regulator of the renin-angiotensin system. Circ Res 126:1456–1474. https://doi.org/10.1161/CIRCRESAHA.120.317015.

191. Cano F, Gajardo M, Freundlich M. 2020. Renin angiotensin axis, angioten- sin converting enzyme 2 and coronavirus. Rev Chil Pediatr 91:330–338. (In Spanish.) https://doi.org/10.32641/rchped.vi91i3.2548.

192. Janardhan V, Janardhan V, Kalousek V. 2020. COVID-19 as a blood clot- ting disorder masquerading as a respiratory illness: a cerebrovascular perspective and therapeutic implications for stroke thrombectomy. J Neuroimaging 30:555–561. https://doi.org/10.1111/jon.12770.

193. Gavriatopoulou M, Korompoki E, Fotiou D, Ntanasis-Stathopoulos I, Psaltopoulou T, Kastritis E, Terpos E, Dimopoulos MA. 2020. Organ-spe- cific manifestations of COVID-19 infection. Clin Exp Med 20:493–506. https://doi.org/10.1007/s10238-020-00648-x.

194. Bian J, Zhao R, Zhai S, Li Z. 2020. Letter to the editor: anti-RAS drugs and SARS-CoV-2 infection. Acta Pharm Sin B 10:1251–1252. https://doi.org/ 10.1016/j.apsb.2020.04.013.

195. Kai H, Kai M. 2020. Interactions of coronaviruses with ACE2, angiotensin II, and RAS inhibitors-lessons from available evidence and insights into COVID-19. Hypertens Res 43:648–654. https://doi.org/10.1038/s41440 -020-0455-8.

196. MengJ,XiaoG,ZhangJ,HeX,OuM,BiJ,YangR,DiW,WangZ,LiZ,GaoH, Liu L, Zhang G. 2020. Renin-angiotensin system inhibitors improve the clini- cal outcomes of COVID-19 patients with hypertension. Emerg Microbes Infect 9:757–760. https://doi.org/10.1080/22221751.2020.1746200.

197. Vaduganathan M, Vardeny O, Michel T, McMurray JJV, Pfeffer MA, Solomon SD. 2020. Renin-angiotensin-aldosterone system inhibitors in patients with Covid-19. N Engl J Med 382:1653–1659. https://doi.org/10 .1056/NEJMsr2005760.

198. Beun R, Kusadasi N, Sikma M, Westerink J, Huisman A. 2020. Throm- boembolic events and apparent heparin resistance in patients infected with SARS-CoV-2. Int J Lab Hematol 42:19–20. https://doi.org/10.1111/ ijlh.13230.

199. Wichmann D, Sperhake JP, Lutgehetmann M, Steurer S, Edler C, Heinemann A, Heinrich F, Mushumba H, Kniep I, Schroder AS, Burdelski C, de Heer G, Nierhaus A, Frings D, Pfefferle S, Becker H, Bredereke-Wiedling H, de Weerth A, Paschen HR, Sheikhzadeh-Eggers S, Stang A, Schmiedel S, Bokemeyer C, Addo MM, Aepfelbacher M, Puschel K, Kluge S. 2020. Autopsy findings and venous thromboembolism in patients with COVID-19: a prospective cohort study. Ann Intern Med 173:268–277. https://doi.org/10.7326/M20-2003.

200. Manjunath M, Miranda J, Fraenkel L, Johansen PM, Phinney B, Valli- Harwood G, Callahan C, Alsmaan H, Oelberg D. 2020. Acute pulmonary embolism in critically ill patients with COVID-19. medRxiv https://doi .org/10.1101/2020.05.22.20110270.

201. Tang N, Li D, Wang X, Sun Z. 2020. Abnormal coagulation parameters are associated with poor prognosis in patients with novel coronavirus pneumonia. J Thromb Haemost 18:844–847. https://doi.org/10.1111/jth .14768.

202. Llitjos JF, Leclerc M, Chochois C, Monsallier JM, Ramakers M, Auvray M, Merouani K. 2020. High incidence of venous thromboembolic events in anti- coagulated severe COVID-19 patients. J Thromb Haemost 18:1743–1746. https://doi.org/10.1111/jth.14869.

203. Lodigiani C, Iapichino G, Carenzo L, Cecconi M, Ferrazzi P, Sebastian T, Kucher N, Studt JD, Sacco C, Alexia B, Sandri MT, Barco S, Humanitas C- TF, Humanitas COVID-19 Task Force. 2020. Venous and arterial

cmr.asm.org 24

Downloaded from http://cmr.asm.org/ on January 21, 2021 by guest

SARS-CoV-2: a Systemic Infection

Clinical Microbiology Reviews

thromboembolic complications in COVID-19 patients admitted to an academic hospital in Milan, Italy. Thromb Res 191:9–14. https://doi.org/ 10.1016/j.thromres.2020.04.024.

204. Thachil J, Tang N, Gando S, Falanga A, Cattaneo M, Levi M, Clark C, Iba T. 2020. ISTH interim guidance on recognition and management of coagul- opathy in COVID-19. J Thromb Haemost 18:1023–1026. https://doi.org/ 10.1111/jth.14810.

205. Tang N, Bai H, Chen X, Gong J, Li D, Sun Z. 2020. Anticoagulant treatment is associated with decreased mortality in severe coronavirus disease 2019 patients with coagulopathy. J Thromb Haemost 18:1094–1099. https://doi .org/10.1111/jth.14817.

206. Paranjpe I, Fuster V, Lala A, Russak A, Glicksberg BS, Levin MA, Charney AW, Narula J, Fayad ZA, Bagiella E, Zhao S, Nadkarni GN. 2020. Associa- tion of treatment dose anticoagulation with in-hospital survival among hospitalized patients with COVID-19. J Am Coll Cardiol 76:122–124. https://doi.org/10.1016/j.jacc.2020.05.001.

207. Rico-Mesa JS, Rosas D, Ahmadian-Tehrani A, White A, Anderson AS, Chilton R. 2020. The role of anticoagulation in COVID-19-induced hyper- coagulability. Curr Cardiol Rep 22:53. https://doi.org/10.1007/s11886 -020-01328-8.

208. Bandyopadhyay D, Akhtar T, Hajra A, Gupta M, Das A, Chakraborty S, Pal I, Patel N, Amgai B, Ghosh RK, Fonarow GC, Lavie CJ, Naidu SS. 2020. COVID-19 pandemic: cardiovascular complications and future implica- tions. Am J Cardiovasc Drugs 20:311–324. https://doi.org/10.1007/s40256 -020-00420-2.

209. Costanzo L, Palumbo FP, Ardita G, Antignani PL, Arosio E, Failla G, Italian Society for Vascular Investigation and the Italian Society of Vascular Medicine. 2020. Coagulopathy, thromboembolic complications, and the use of heparin in COVID-19 pneumonia. J Vasc Surg Venous Lymphat Disord 8:711–716. https://doi.org/10.1016/j.jvsv.2020.05.018.

210. Griffin DO, Jensen A, Khan M, Chin J, Chin K, Parnell R, Awwad C, Patel D. 2020. Arterial thromboembolic complications in COVID-19 in low risk patients despite prophylaxis. Br J Haematol 190:e11–e13. https://doi .org/10.1111/bjh.16792.

211. Thachil J, Agarwal S. 2020. Understanding the COVID-19 coagulop- athy spectrum. Anaesthesia 75:1432–1436. https://doi.org/10.1111/ anae.15141.

212. Viecca M, Radovanovic D, Forleo GB, Santus P. 2020. Enhanced platelet inhibition treatment improves hypoxemia in patients with severe Covid- 19 and hypercoagulability. A case control, proof of concept study. Phar- macol Res 158:104950. https://doi.org/10.1016/j.phrs.2020.104950.

213. Franchini M, Marano G, Cruciani M, Mengoli C, Pati I, Masiello F, Veropalumbo E, Pupella S, Vaglio S, Liumbruno GM. 2020. COVID-19- associated coagulopathy. Diagnosis (Berl) 7:357–363. https://doi.org/10 .1515/dx-2020-0078.

214. Lo MW, Kemper C, Woodruff TM. 2020. COVID-19: complement, coagula- tion, and collateral damage. J Immunol 205:1488–1495. https://doi.org/ 10.4049/jimmunol.2000644.

215. O’Sullivan JM, Gonagle DM, Ward SE, Preston RJS, O’Donnell JS. 2020. Endothelial cells orchestrate COVID-19 coagulopathy. Lancet Haematol 7:e553–e555. https://doi.org/10.1016/S2352-3026(20)30215-5.

216. Marchetti M. 2020. COVID-19-driven endothelial damage: complement, HIF-1, and ABL2 are potential pathways of damage and targets for cure. Ann Hematol 99:1701–1707. https://doi.org/10.1007/s00277-020-04138-8.

217. Pons S, Fodil S, Azoulay E, Zafrani L. 2020. The vascular endothelium: the cornerstone of organ dysfunction in severe SARS-CoV-2 infection. Crit Care 24:353. https://doi.org/10.1186/s13054-020-03062-7.

218. Singhania N, Bansal S, Nimmatoori DP, Ejaz AA, McCullough PA, Singhania G. 2020. Current overview on hypercoagulability in COVID-19. Am J Cardio- vasc Drugs 20:393–403. https://doi.org/10.1007/s40256-020-00431-z.

219. Walborn A, Rondina M, Mosier M, Fareed J, Hoppensteadt D. 2019. Endo- thelial dysfunction is associated with mortality and severity of coagulop- athy in patients with sepsis and disseminated intravascular coagulation. Clin Appl Thromb Hemost 25:1076029619852163. https://doi.org/10 .1177/1076029619852163.

220. Guzik TJ, Mohiddin SA, Dimarco A, Patel V, Savvatis K, Marelli-Berg FM, Madhur MS, Tomaszewski M, Maffia P, D’Acquisto F, Nicklin SA, Marian AJ, Nosalski R, Murray EC, Guzik B, Berry C, Touyz RM, Kreutz R, Wang DW, Bhella D, Sagliocco O, Crea F, Thomson EC, McInnes IB. 2020. COVID-19 and the cardiovascular system: implications for risk assess- ment, diagnosis, and treatment options. Cardiovasc Res 116:1666–1687. https://doi.org/10.1093/cvr/cvaa106.

221. Cure E, Cure MC. 2020. COVID-19 may predispose to thrombosis by

April 2021 Volume 34 Issue 2 e00133-20

affecting both vascular endothelium and platelets. Clin Appl Thromb

Hemost 26:1076029620933945. https://doi.org/10.1177/1076029620933945. 222. Becker RC. 2020. COVID-19-associated vasculitis and vasculopathy. J Thromb Thrombolysis 50:499–511. https://doi.org/10.1007/s11239-020

-02230-4.
223. Cyranoski D. 2020. Why children avoid the worst coronavirus complica-

tions might lie in their arteries. Nature 582:324–325. https://doi.org/10

.1038/d41586-020-01692-z.
224. Okada H, Yoshida S, Hara A, Ogura S, Tomita H. 2020. Vascular endothe-

lial injury exacerbates coronavirus disease 2019: the role of endothelial glycocalyx protection. Microcirculation 582:e12654. https://doi.org/10 .1111/micc.12654:e12654.

225. Leppkes M, Knopf J, Naschberger E, Lindemann A, Singh J, Herrmann I, Sturzl M, Staats L, Mahajan A, Schauer C, Kremer AN, Volkl S, Amann K, Evert K, Falkeis C, Wehrfritz A, Rieker RJ, Hartmann A, Kremer AE, Neurath MF, Munoz LE, Schett G, Herrmann M. 2020. Vascular occlusion by neutrophil extracellular traps in COVID-19. EBioMedicine 58:102925. https://doi.org/10.1016/j.ebiom.2020.102925.

226. Gupta A, Madhavan MV, Sehgal K, Nair N, Mahajan S, Sehrawat TS, Bikdeli B, Ahluwalia N, Ausiello JC, Wan EY, Freedberg DE, Kirtane AJ, Parikh SA, Maurer MS, Nordvig AS, Accili D, Bathon JM, Mohan S, Bauer KA, Leon MB, Krumholz HM, Uriel N, Mehra MR, Elkind MSV, Stone GW, Schwartz A, Ho DD, Bilezikian JP, Landry DW. 2020. Extrapulmonary manifestations of COVID-19. Nat Med 26:1017–1032. https://doi.org/10 .1038/s41591-020-0968-3.

227. Colmenero I, Santonja C, Alonso-Riano M, Noguera-Morel L, Hernandez- Martin A, Andina D, Wiesner T, Rodriguez-Peralto JL, Requena L, Torrelo A. 2020. SARS-CoV-2 endothelial infection causes COVID-19 chilblains: histopathological, immunohistochemical and ultrastructural study of seven paediatric cases. Br J Dermatol 183:729–737. https://doi.org/10 .1111/bjd.19327.

228. Santonja C, Heras F, Nunez L, Requena L. 2020. COVID-19 chilblain-like lesion: immunohistochemical demonstration of SARS-CoV-2 spike pro- tein in blood vessel endothelium and sweat gland epithelium in a poly- merase chain reaction-negative patient. Br J Dermatol 183:778–780. https://doi.org/10.1111/bjd.19338.

229. Massey PR, Jones KM. 2020. Going viral: a brief history of chilblain-like skin lesions (“COVID toes”) amidst the COVID-19 pandemic. Semin Oncol 47:330–334. https://doi.org/10.1053/j.seminoncol.2020.05.012.

230. Tan W, Aboulhosn J. 2020. The cardiovascular burden of coronavirus dis- ease 2019 (COVID-19) with a focus on congenital heart disease. Int J Car- diol 309:70–77. https://doi.org/10.1016/j.ijcard.2020.03.063.

231. Zheng YY, Ma YT, Zhang JY, Xie X. 2020. COVID-19 and the cardiovascu- lar system. Nat Rev Cardiol 17:259–260. https://doi.org/10.1038/s41569 -020-0360-5.

232. Wei S, Zhang L, Cui H, Jiang S. 2020. Progress in treatment of myocardial injury in patients with 2019-nCoV: a Chinese experience. Heart Surg Fo- rum 23:E426–E429. https://doi.org/10.1532/hsf.2959.

233. Ka B, Chaudhuri D. 2020. A review of acute myocardial injury in coronavi- rus disease 2019. Cureus 12:e8426. https://doi.org/10.7759/cureus.8426. 234. Turshudzhyan A. 2020. Severe acute respiratory syndrome coronavirus 2

(SARS-CoV-2)-induced cardiovascular syndrome: etiology, outcomes, and management. Cureus 12:e8543. https://doi.org/10.7759/cureus .8543.

235. Figueiredo Neto JA, Marcondes-Braga FG, Moura LZ, Figueiredo A, Figueiredo V, Mourilhe-Rocha R, Mesquita ET. 2020. Coronavirus disease 2019 and the myocardium. Arq Bras Cardiol 114:1051–1057. https://doi .org/10.36660/abc.20200373.

236. Montone RA, Iannaccone G, Meucci MC, Gurgoglione F, Niccoli G. 2020. Myocardial and microvascular injury due to coronavirus disease 2019. Eur Cardiol 15:e52. https://doi.org/10.15420/ecr.2020.22.

237. Wu L, O’Kane AM, Peng H, Bi Y, Motriuk-Smith D, Ren J. 2020. SARS-CoV- 2 and cardiovascular complications: from molecular mechanisms to pharmaceutical management. Biochem Pharmacol 178:114114. https:// doi.org/10.1016/j.bcp.2020.114114.

238. Gonzalez-Jaramillo N, Low N, Franco OH. 2020. The double burden of disease of COVID-19 in cardiovascular patients: overlapping conditions could lead to overlapping treatments. Eur J Epidemiol 35:335–337. https://doi.org/10.1007/s10654-020-00628-1.

239. Kazory A, Ronco C, McCullough PA. 16 April 2020, posting date. SARS- CoV-2 (COVID-19) and intravascular volume management strategies in the critically ill. Proc (Bayl Univ Med Cent) https://doi.org/10.1080/ 08998280.2020.1754700.

240. Chen L, Li X, Chen M, Feng Y, Xiong C. 2020. The ACE2 expression in cmr.asm.org 25

Downloaded from http://cmr.asm.org/ on January 21, 2021 by guest

Synowiec et al.

Clinical Microbiology Reviews

human heart indicates new potential mechanism of heart injury among patients infected with SARS-CoV-2. Cardiovasc Res 116:1097–1100. https://doi.org/10.1093/cvr/cvaa078.

241. Mardani R, Ahmadi Vasmehjani A, Zali F, Gholami A, Mousavi Nasab SD, Kaghazian H, Kaviani M, Ahmadi N. 2020. Laboratory parameters in detection of COVID-19 patients with positive RT-PCR; a diagnostic accu- racy study. Arch Acad Emerg Med 8:e43.

242. Sharma A, Garcia G, Jr, Wang Y, Plummer JT, Morizono K, Arumugaswami V, Svendsen CN. 2020. Human iPSC-derived cardiomyocytes are susceptible to SARS-CoV-2 infection. Cell Rep Med 1:100052. https://doi.org/10.1016/j .xcrm.2020.100052.

243. Gupta AK, Jneid H, Addison D, Ardehali H, Boehme AK, Borgaonkar S, Boulestreau R, Clerkin K, Delarche N, DeVon HA, Grumbach IM, Gutierrez J, Jones DA, Kapil V, Maniero C, Mentias A, Miller PS, Ng SM, Parekh JD, Sanchez RH, Sawicki KT, Te Riele ASJM, Remme CA, London B. 2020. Cur- rent perspectives on Coronavirus 2019 (COVID-19) and cardiovascular disease: a white paper by the JAHA editors. JAHA 9:e017013. https://doi .org/10.1161/JAHA.120.017013.

244. Zhou L, Niu Z, Jiang X, Zhang Z, Zheng Y, Wang Z, Zhu Y, Gao L, Wang X, Sun Q. 2020. Systemic analysis of tissue cells potentially vulnerable to SARS-CoV-2 infection by the protein-proofed single-cell RNA profiling of ACE2, TMPRSS2 and Furin proteases. bioRxiv https://doi.org/10.1101/ 2020.04.06.028522.

245. Tavazzi G, Pellegrini C, Maurelli M, Belliato M, Sciutti F, Bottazzi A, Sepe PA, Resasco T, Camporotondo R, Bruno R, Baldanti F, Paolucci S, Pelenghi S, Iotti GA, Mojoli F, Arbustini E. 2020. Myocardial localization of coronavirus in COVID-19 cardiogenic shock. Eur J Heart Fail 22:911–915. https://doi.org/10.1002/ejhf.1828.

246. Kim HK, Kim H, Lee MK, Choi WH, Jang Y, Shin JS, Park J-Y, Hyun S-I, Kim KH, Han HW, Kim M, Lim YC, Yoo J. 2020. Generation of tonsil organoids as an ex vivo model for SARS-CoV-2 infection. bioRxiv https://doi.org/10 .1101/2020.08.06.239574.

247. Yao XH, Li TY, He ZC, Ping YF, Liu HW, Yu SC, Mou HM, Wang LH, Zhang HR,FuWJ,LuoT,LiuF,ChenC,XiaoHL,GuoHT,LinS,XiangDF,ShiY,Li QR, Huang X, Cui Y, Li XZ, Tang W, Pan PF, Huang XQ, Ding YQ, Bian XW. 2020. A pathological report of three COVID-19 cases by minimally inva- sive autopsies. Zhonghua Bing Li Xue Za Zhi 49:411–417. (In Chinese.) https://doi.org/10.3760/cma.j.cn112151-20200312-00193.

248. Santos Leite Pessoa M, Franco Costa Lima C, Farias Pimentel AC, Godeiro Costa JC, Bezerra Holanda JL. 2020. Multisystemic infarctions in COVID- 19: focus on the spleen. Eur J Case Rep Intern Med 7:001747. https://doi .org/10.12890/2020_001747.

249. DiaoB,WangC,TanY,ChenX,LiuY,NingL,ChenL,LiM,LiuY,WangG, Yuan Z, Feng Z, Zhang Y, Wu Y, Chen Y. 2020. Reduction and functional exhaustion of T cells in patients with coronavirus disease 2019 (COVID- 19). Front Immunol 11:827. https://doi.org/10.3389/fimmu.2020.00827.

250. Bouadma L, Wiedemann A, Patrier J, Surénaud M, Wicky PH, Foucat E, Diehl JL, Hejblum BP, Sinnah F, de Montmollin E, Lacabaratz C, Thiébaut R, Timsit JF, Lévy Y. 2020. Immune alterations in a patient with SARS- CoV-2-related acute respiratory distress syndrome. J Clin Immunol 40:1082–1092. https://doi.org/10.1007/s10875-020-00839-x.

251. Lombardi A, Trombetta E, Cattaneo A, Castelli V, Palomba E, Tirone M, Mangioni D, Lamorte G, Manunta M, Prati D, Ceriotti F, Gualtierotti R, Costantino G, Aliberti S, Scaravilli V, Grasselli G, Gori A, Porretti L, Bandera A. 2020. Early phases of COVID-19 are characterized by a reduc- tion of lymphocyte populations and the presence of atypical mono- cytes. medRxiv https://doi.org/10.1101/2020.05.01.20087080.

252. Yao Z, Zheng Z, Wu K, Junhua Z. 2020. Immune environment modula- tion in pneumonia patients caused by coronavirus: SARS-CoV, MERS- CoV and SARS-CoV-2. Aging (Albany NY) 12:7639–7651. https://doi.org/ 10.18632/aging.103101.

253. Wang Y, Wang Y, Chen Y, Qin Q. 2020. Unique epidemiological and clinical features of the emerging 2019 novel coronavirus pneumonia (COVID-19) implicate special control measures. J Med Virol 92:568–576. https://doi .org/10.1002/jmv.25748.

254. Pontelli MC, Castro IA, Martins RB, Veras FP, Serra LL, Nascimento DC, Cardoso RS, Rosales R, Lima TM, Souza JP, Caetité DB, de Lima MHF, Kawahisa JT, Giannini MC, Bonjorno LP, Lopes MIF, Batah SS, Siyuan L, Assad RL, Almeida SCL, Oliveira FR, Benatti MN, Pontes LLF, Santana RC, Vilar FC, Martins MA, Cunha TM, Calado RT, Alves-Filho JC, Zamboni DS, Fabro A, Louzada-Junior P, Oliveira RDR, Cunha FQ, Arruda E. 2020. Infection of human lymphomononuclear cells by SARS-CoV-2. bioRxiv https://doi.org/10.1101/2020.07.28.225912.

255. Song X, Hu W, Yu H, Zhao L, Zhao Y, Zhao Y. 2020. High expression of

April 2021 Volume 34 Issue 2 e00133-20

angiotensin-converting enzyme-2 (ACE2) on tissue macrophages that may be targeted by virus SARS-CoV-2 in COVID-19 patients. bioRxiv https://doi.org/10.1101/2020.07.18.210120.

256. Chen Y, Feng Z, Diao B, Wang R, Wang G, Wang C, Tan Y, Liu L, Wang C, Liu Y, Liu Y, Yuan Z, Ren L, Wu Y. 2020. The novel severe acute respira- tory syndrome coronavirus 2 (SARS-CoV-2) directly decimates human spleens and lymph nodes. medRxiv https://doi.org/10.1101/2020.03.27 .20045427.

257. WangD,HuB,HuC,ZhuF,LiuX,ZhangJ,WangB,XiangH,ChengZ, Xiong Y, Zhao Y, Li Y, Wang X, Peng Z. 2020. Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus-infected pneu- monia in Wuhan, China. JAMA 323:1061–1069. https://doi.org/10.1001/ jama.2020.1585.

258. ChenN,ZhouM,DongX,QuJ,GongF,HanY,QiuY,WangJ,LiuY,Wei Y, Xia J, Yu T, Zhang X, Zhang L. 2020. Epidemiological and clinical char- acteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study. Lancet 395:507–513. https://doi.org/10.1016/ S0140-6736(20)30211-7.

259. ZhouY,HanT,ChenJ,HouC,HuaL,HeS,GuoY,ZhangS,WangY,Yuan J,ZhaoC,ZhangJ,JiaQ,ZuoX,LiJ,WangL,CaoQ,JiaE.21April2020, posting date. Clinical and autoimmune characteristics of severe and crit- ical cases of COVID-19. Clin Transl Sci https://doi.org/10.1111/cts.12805.

260. Merad M, Martin JC. 2020. Pathological inflammation in patients with COVID-19: a key role for monocytes and macrophages. Nat Rev Immunol 20:355–362. https://doi.org/10.1038/s41577-020-0331-4.

261. Chen X, Zhao B, Qu Y, Chen Y, Xiong J, Feng Y, Men D, Huang Q, Liu Y, Yang B, Ding J, Li F. 2020. Detectable serum SARS-CoV-2 viral load (RNAaemia) is closely correlated with drastically elevated interleukin 6 (IL-6) level in critically ill COVID-19 patients. Clin Infect Dis 71:1937–1942. https:// doi.org/10.1093/cid/ciaa449.

262. Tynell J, Westenius V, Ronkko E, Munster VJ, Melen K, Osterlund P, Julkunen I. 2016. Middle East respiratory syndrome coronavirus shows poor replication but significant induction of antiviral responses in human monocyte-derived macrophages and dendritic cells. J Gen Virol 97:344–355. https://doi.org/10.1099/jgv.0.000351.

263. Chu H, Zhou J, Wong BH, Li C, Cheng ZS, Lin X, Poon VK, Sun T, Lau CC, Chan JF, To KK, Chan KH, Lu L, Zheng BJ, Yuen KY. 2014. Productive repli- cation of Middle East respiratory syndrome coronavirus in monocyte- derived dendritic cells modulates innate immune response. Virology 454–455:197–205. https://doi.org/10.1016/j.virol.2014.02.018.

264. Zhou J, Chu H, Li C, Wong BH, Cheng ZS, Poon VK, Sun T, Lau CC, Wong KK, Chan JY, Chan JF, To KK, Chan KH, Zheng BJ, Yuen KY. 2014. Active replication of Middle East respiratory syndrome coronavirus and aber- rant induction of inflammatory cytokines and chemokines in human macrophages: implications for pathogenesis. J Infect Dis 209:1331–1342. https://doi.org/10.1093/infdis/jit504.

265. Lau YL, Peiris JS, Law HK. 2012. Role of dendritic cells in SARS coronavi- rus infection. Hong Kong Med J 18(Suppl 3):28–30.

266. Yen YT, Liao F, Hsiao CH, Kao CL, Chen YC, Wu-Hsieh BA. 2006. Modeling the early events of severe acute respiratory syndrome coronavirus infec- tion in vitro. J Virol 80:2684–2693. https://doi.org/10.1128/JVI.80.6.2684 -2693.2006.

267. Tseng CT, Perrone LA, Zhu H, Makino S, Peters CJ. 2005. Severe acute re- spiratory syndrome and the innate immune responses: modulation of effec- tor cell function without productive infection. J Immunol 174:7977–7985. https://doi.org/10.4049/jimmunol.174.12.7977.

268. Burgueno JF, Reich A, Hazime H, Quintero MA, Fernandez I, Fritsch J, Santander AM, Brito N, Damas OM, Deshpande A, Kerman DH, Zhang L, Gao Z, Ban Y, Wang L, Pignac-Kobinger J, Abreu MT. 2020. Expression of SARS-CoV-2 entry molecules ACE2 and TMPRSS2 in the gut of patients with IBD. Inflamm Bowel Dis 26:797–808. https://doi.org/10.1093/ibd/ izaa085.

269. Chu H, Zhou J, Wong BH, Li C, Chan JF, Cheng ZS, Yang D, Wang D, Lee AC, Li C, Yeung ML, Cai JP, Chan IH, Ho WK, To KK, Zheng BJ, Yao Y, Qin C, Yuen KY. 2016. Middle East respiratory syndrome coronavirus effi- ciently infects human primary T lymphocytes and activates the extrinsic and intrinsic apoptosis pathways. J Infect Dis 213:904–914. https://doi .org/10.1093/infdis/jiv380.

270. Koch C, Staffler G, Hüttinger R, Hilgert I, Prager E, Cerný J, Steinlein P, Majdic O, Horejší V, Stockinger H. 1999. T cell activation-associated epi- topes of CD147 in regulation of the T cell response, and their definition by antibody affinity and antigen density. Int Immunol 11:777–786. https://doi.org/10.1093/intimm/11.5.777.

271. Ulrich H, Pillat MM. 2020. CD147 as a target for COVID-19 treatment: cmr.asm.org 26

Downloaded from http://cmr.asm.org/ on January 21, 2021 by guest

SARS-CoV-2: a Systemic Infection

Clinical Microbiology Reviews

suggested effects of azithromycin and stem cell engagement. Stem Cell

Rev Rep 16:434–440. https://doi.org/10.1007/s12015-020-09976-7.

272. Luo P, Liu Y, Qiu L, Liu X, Liu D, Li J. 2020. Tocilizumab treatment in COVID-19: a single center experience. J Med Virol 92:814–818. https://
doi.org/10.1002/jmv.25801.

273. Griffin DO, Jensen A, Khan M, Chin J, Chin K, Saad J, Parnell R, Awwad C,
Patel D. 2020. Pulmonary embolism and increased levels of d-dimer in patients with coronavirus disease. Emerg Infect Dis 26:1941–1943. https:// doi.org/10.3201/eid2608.201477.

274. Alattar R, Ibrahim TBH, Shaar SH, Abdalla S, Shukri K, Daghfal JN, Khatib MY, Aboukamar M, Abukhattab M, Alsoub HA, Almaslamani MA, Omrani AS. 2020. Tocilizumab for the treatment of severe coronavirus disease 2019. J Med Virol 92:2042–2049. https://doi.org/10.1002/jmv.25964.

275. Mazzoni A, Salvati L, Maggi L, Capone M, Vanni A, Spinicci M, Mencarini J, Caporale R, Peruzzi B, Antonelli A, Trotta M, Zammarchi L, Ciani L, Gori L, Lazzeri C, Matucci A, Vultaggio A, Rossi O, Almerigogna F, Parronchi P, Fontanari P, Lavorini F, Peris A, Rossolini GM, Bartoloni A, Romagnani S, Liotta F, Annunziato F, Cosmi L. 2020. Impaired immune cell cytotoxicity in severe COVID-19 is IL-6 dependent. J Clin Invest 130:4694–4703. https://doi.org/10.1172/JCI138554.

276. Vankadari N, Wilce JA. 2020. Emerging WuHan (COVID-19) coronavirus: glycan shield and structure prediction of spike glycoprotein and its interaction with human CD26. Emerg Microbes Infect 9:601–604. https://doi.org/10.1080/22221751.2020.1739565.

277. Prajapat M, Sarma P, Shekhar N, Prakash A, Avti P, Bhattacharyya A, Kaur H, Kumar S, Bansal S, Sharma AR, Medhi B. 2020. Update on the target structures of SARS-CoV-2: a systematic review. Indian J Pharmacol 52:142–149. https://doi.org/10.4103/ijp.IJP_338_20.

278. Polycarpou A, Howard M, Farrar CA, Greenlaw R, Fanelli G, Wallis R, Klavinskis LS, Sacks S. 2020. Rationale for targeting complement in COVID-19. EMBO Mol Med 12:e12642. https://doi.org/10.15252/emmm .202012642.

279. Sokolowska M, Lukasik ZM, Agache I, Akdis CA, Akdis D, Akdis M, Barcik W, Brough HA, Eiwegger T, Eljaszewicz A, Eyerich S, Feleszko W, Gomez- Casado C, Hoffmann-Sommergruber K, Janda J, Jimenez-Saiz R, Jutel M, Knol EF, Kortekaas Krohn I, Kothari A, Makowska J, Moniuszko M, Morita H, O’Mahony L, Nadeau K, Ozdemir C, Pali-Scholl I, Palomares O, Papaleo F, Prunicki M, Schmidt-Weber CB, Sediva A, Schwarze J, Shamji MH, Tramper-Stranders GA, van de Veen W, Untersmayr E. 2020. Immunol- ogy of COVID-19: mechanisms, clinical outcome, diagnostics, and per- spectives–a report of the European Academy of Allergy and Clinical Immunology (EAACI). Allergy 75:2445–2476. https://doi.org/10.1111/ all.14462.

280. Noris M, Benigni A, Remuzzi G. 2020. The case of complement activation in COVID-19 multiorgan impact. Kidney Int 98:314–322. https://doi.org/ 10.1016/j.kint.2020.05.013.

281. Java A, Apicelli AJ, Liszewski MK, Coler-Reilly A, Atkinson JP, Kim AH, Kulkarni HS. 2020. The complement system in COVID-19: friend and foe? JCI Insight 5:e140711. https://doi.org/10.1172/jci.insight.140711.

282. Bosmann M. 2020. Complement activation during critical illness: current findings and an outlook in the era of COVID-19. Am J Respir Crit Care Med 202:163–165. https://doi.org/10.1164/rccm.202005-1926ED.

283. Diurno F, Numis FG, Porta G, Cirillo F, Maddaluno S, Ragozzino A, De Negri P, Di Gennaro C, Pagano A, Allegorico E, Bressy L, Bosso G, Ferrara A, Serra C, Montisci A, D’Amico M, Schiano Lo Morello S, Di Costanzo G, Tucci AG, Marchetti P, Di Vincenzo U, Sorrentino I, Casciotta A, Fusco M, Buonerba C, Berretta M, Ceccarelli M, Nunnari G, Diessa Y, Cicala S, Facchini G. 2020. Eculizumab treatment in patients with COVID-19: pre- liminary results from real life ASL Napoli 2 Nord experience. Eur Rev Med Pharmacol Sci 24:4040–4047. https://doi.org/10.26355/eurrev_202004 _20875.

284. Giudice V, Pagliano P, Vatrella A, Masullo A, Poto S, Polverino BM, Gammaldi R, Maglio A, Sellitto C, Vitale C, Serio B, Cuffa B, Borrelli A, Vecchione C, Filippelli A, Selleri C. 2020. Combination of ruxolitinib and eculizumab for treatment of severe SARS-CoV-2-related acute respiratory distress syndrome: a controlled study. Front Pharmacol 11:857. https://doi .org/10.3389/fphar.2020.00857.

285. Fanelli V, Fiorentino M, Cantaluppi V, Gesualdo L, Stallone G, Ronco C, Castellano G. 2020. Acute kidney injury in SARS-CoV-2 infected patients. Crit Care 24:155. https://doi.org/10.1186/s13054-020-02872-z.

286. Naicker S, Yang C-W, Hwang S-J, Liu B-C, Chen J-H, Jha V. 2020. The novel coronavirus 2019 epidemic and kidneys. Kidney Int 97:824–828. https://doi.org/10.1016/j.kint.2020.03.001.

287. Sun J, Zhu A, Li H, Zheng K, Zhuang Z, Chen Z, Shi Y, Zhang Z, Chen S-b,

April 2021 Volume 34 Issue 2 e00133-20

LiuX,DaiJ,LiX,HuangS,HuangX,LuoL,WenL,ZhuoJ,LiY,WangY, Zhang L, Zhang Y, Li F, Feng L, Chen X, Zhong N, Yang Z, Huang J, Zhao J, Li Y-m. 2020. Isolation of infectious SARS-CoV-2 from urine of a COVID- 19 patient. Emerg Microbes Infecti 9:991–993. https://doi.org/10.1080/ 22221751.2020.1760144.

288. PengL,LiuJ,XuW,LuoQ,ChenD,LeiZ,HuangZ,LiX,DengK,LinB, Gao Z. 2020. SARS-CoV-2 can be detected in urine, blood, anal swabs, and oropharyngeal swabs specimens. J Med Virol 92:1676–1680. https:// doi.org/10.1002/jmv.25936.

289. Farkash EA, Wilson AM, Jentzen JM. 2020. Ultrastructural evidence for direct renal infection with SARS-CoV-2. J Am Soc Nephrol 31:1683–1687. https://doi.org/10.1681/ASN.2020040432.

290. DiaoB,WangC,WangR,FengZ,TanY,WangH,WangC,LiuL,LiuY,Liu Y, Wang G, Yuan Z, Ren L, Wu Y, Chen Y. 2020. Human kidney is a target for novel severe acute respiratory syndrome coronavirus (SARS-CoV-2) infection. medRxiv https://doi.org/10.1101/2020.03.04.20031120.

291. Yin W, Zhang PL. 2020. Infectious pathways of SARS-CoV-2 in renal tis- sue. J Nephropathol 9:e37. https://doi.org/10.34172/jnp.2020.37.

292. Moore JB, June CH. 2020. Cytokine release syndrome in severe COVID- 19. Science 368:473–474. https://doi.org/10.1126/science.abb8925.
293. Zhang C, Shi L, Wang F-S. 2020. Liver injury in COVID-19: management

and challenges. Lancet Gastroenterol Hepatol 5:428–430. https://doi

.org/10.1016/S2468-1253(20)30057-1.
294. Xu L, Liu J, Lu M, Yang D, Zheng X. 2020. Liver injury during highly path-

ogenic human coronavirus infections. Liver Int 40:998–1004. https://doi

.org/10.1111/liv.14435.
295. Alsaad KO, Hajeer AH, Al Balwi M, Al Moaiqel M, Al Oudah N, Al Ajlan A,

AlJohani S, Alsolamy S, Gmati GE, Balkhy H, Al-Jahdali HH, Baharoon SA, Arabi YM. 2018. Histopathology of Middle East respiratory syndrome coronovirus (MERS-CoV) infection – clinicopathological and ultrastructural study. Histopathology 72:516–524. https://doi.org/10.1111/his.13379.

296. Chau TN, Lee KC, Yao H, Tsang TY, Chow TC, Yeung YC, Choi KW, Tso YK, Lau T, Lai ST, Lai CL. 2004. SARS-associated viral hepatitis caused by a novel coronavirus: report of three cases. Hepatology 39:302–310. https://doi.org/ 10.1002/hep.20111.

297. Weber S, Mayerle J, Irlbeck M, Gerbes AL. 2020. Severe liver failure dur- ing SARS-CoV-2 infection. Gut 69:1365–1367. https://doi.org/10.1136/ gutjnl-2020-321350.

298. ZhaoB,NiC,GaoR,WangY,YangL,WeiJ,LvT,LiangJ,ZhangQ,XuW, Xie Y, Wang X, Yuan Z, Liang J, Zhang R, Lin X. 2020. Recapitulation of SARS-CoV-2 infection and cholangiocyte damage with human liver organoids. Protein Cell 11:771–775. https://doi.org/10.1007/s13238-020 -00718-6.

299. Heinrichs D, Knauel M, Offermanns C, Berres M-L, Nellen A, Leng L, Schmitz P, Bucala R, Trautwein C, Weber C, Bernhagen J, Wasmuth HE. 2011. Macrophage migration inhibitory factor (MIF) exerts antifibrotic effects in experimental liver fibrosis via CD74. Proc Natl Acad Sci U S A 108:17444–17449. https://doi.org/10.1073/pnas.1107023108.

300. Li JH, Tang Y, Lv J, Wang XH, Yang H, Tang PMK, Huang XR, He ZJ, Zhou ZJ, Huang QY, Klug J, Meinhardt A, Fingerle-Rowson G, Xu AP, Zheng ZH, Lan HY. 2019. Macrophage migration inhibitory factor promotes renal injury induced by ischemic reperfusion. J Cell Mol Med 23:3867–3877. https://doi.org/10.1111/jcmm.14234.

301. Takahashi K, Koga K, Linge HM, Zhang Y, Lin X, Metz CN, Al-Abed Y, Ojamaa K, Miller EJ. 2009. Macrophage CD74 contributes to MIF-induced pulmonary inflammation. Respir Res 10:33. https://doi.org/10.1186/1465 -9921-10-33.

302. Paizis G, Tikellis C, Cooper ME, Schembri JM, Lew RA, Smith AI, Shaw T, Warner FJ, Zuilli A, Burrell LM, Angus PW. 2005. Chronic liver injury in rats and humans upregulates the novel enzyme angiotensin converting enzyme 2. Gut 54:1790–1796. https://doi.org/10.1136/gut.2004.062398.

303. Casey S, Schierwagen R, Mak KY, Klein S, Uschner F, Jansen C, Praktiknjo M, Meyer C, Thomas D, Herath C, Jones R, Trebicka J, Angus P. 2019. Acti- vation of the alternate renin-angiotensin system correlates with the clin- ical status in human cirrhosis and corrects post liver transplantation. J Clin Med 8:419. https://doi.org/10.3390/jcm8040419.

304. Biquard L, Valla D, Rautou P-E. 2020. No evidence for an increased liver uptake of SARS-CoV-2 in metabolic associated fatty liver disease. J Hepa- tol 73:717–718. https://doi.org/10.1016/j.jhep.2020.04.035.

305. Wills SE, Beaufrère HH, Brisson BA, Fraser RS, Smith DA. 2018. Pancreati- tis and systemic coronavirus infection in a ferret (Mustela putorius furo). Comp Med 68:208–211. https://doi.org/10.30802/AALAS-CM-17-000109.

306. Garner MM, Ramsell K, Morera N, Juan-Sallés C, Jiménez J, Ardiaca M, Montesinos A, Teifke JP, Löhr CV, Evermann JF, Baszler TV, Nordhausen

cmr.asm.org 27

Downloaded from http://cmr.asm.org/ on January 21, 2021 by guest

Synowiec et al.

Clinical Microbiology Reviews

RW, Wise AG, Maes RK, Kiupel M. 2008. Clinicopathologic features of a systemic coronavirus-associated disease resembling feline infectious peri- tonitis in the domestic ferret (Mustela putorius). Vet Pathol 45:236–246. https://doi.org/10.1354/vp.45-2-236.

307. Liu F, Long X, Zhang B, Zhang W, Chen X, Zhang Z. 2020. ACE2 expres- sion in pancreas may cause pancreatic damage after SARS-CoV-2 infec- tion. Clin Gastroenterol Hepatol 18:2128–2130.e2. https://doi.org/10 .1016/j.cgh.2020.04.040.

308. Yang J-K, Lin S-S, Ji X-J, Guo L-M. 2010. Binding of SARS coronavirus to its receptor damages islets and causes acute diabetes. Acta Diabetol 47:193–199. https://doi.org/10.1007/s00592-009-0109-4.

309. Yang L, Han Y, Nilsson-Payant BE, Gupta V, Wang P, Duan X, Tang X, Zhu J, Zhao Z, Jaffré F, Zhang T, Kim TW, Harschnitz O, Redmond D, Houghton S, Liu C, Naji A, Ciceri G, Guttikonda S, Bram Y, Nguyen D-HT, Cioffi M, Chandar V, Hoagland DA, Huang Y, Xiang J, Wang H, Lyden D, Borczuk A, Chen HJ, Studer L, Pan FC, Ho DD, tenOever BR, Evans T, Schwartz RE, Chen S. 2020. A human pluripotent stem cell-based plat- form to study SARS-CoV-2 tropism and model virus infection in human cells and organoids. Cell Stem Cell 27:125–136.e7. https://doi.org/10 .1016/j.stem.2020.06.015.

310. Nilsson A, Edner N, Albert J, Ternhag A. 2020. Fatal encephalitis associ- ated with coronavirus OC43 in an immunocompromised child. Infect Dis 52:419–422. https://doi.org/10.1080/23744235.2020.1729403.

311. Arbour N, Ekandé S, Côté G, Lachance C, Chagnon F, Tardieu M, Cashman NR, Talbot PJ. 1999. Persistent infection of human oligoden- drocytic and neuroglial cell lines by human coronavirus 229E. J Virol 73:3326–3337. https://doi.org/10.1128/JVI.73.4.3326-3337.1999.

312. Arbour N, Côté G, Lachance C, Tardieu M, Cashman NR, Talbot PJ. 1999. Acute and persistent infection of human neural cell lines by human co- ronavirus OC43. J Virol 73:3338–3350. https://doi.org/10.1128/JVI.73.4 .3338-3350.1999.

313. Arbour N, Day R, Newcombe J, Talbot PJ. 2000. Neuroinvasion by human respiratory coronaviruses. J Virol 74:8913–8921. https://doi.org/10.1128/ jvi.74.19.8913-8921.2000.

314. Moriguchi T, Harii N, Goto J, Harada D, Sugawara H, Takamino J, Ueno M, Sakata H, Kondo K, Myose N, Nakao A, Takeda M, Haro H, Inoue O, Suzuki-Inoue K, Kubokawa K, Ogihara S, Sasaki T, Kinouchi H, Kojin H, Ito M, Onishi H, Shimizu T, Sasaki Y, Enomoto N, Ishihara H, Furuya S, Yamamoto T, Shimada S. 2020. A first case of meningitis/encephalitis associated with SARS-Coronavirus-2. Int J Infect Dis 94:55–58. https://doi .org/10.1016/j.ijid.2020.03.062.

315. Mao L, Jin H, Wang M, Hu Y, Chen S, He Q, Chang J, Hong C, Zhou Y, Wang D, Miao X, Li Y, Hu B. 2020. Neurologic manifestations of hospital- ized patients with coronavirus disease 2019 in Wuhan, China. JAMA Neurol 77:683. https://doi.org/10.1001/jamaneurol.2020.1127.

316. Helms J, Kremer S, Merdji H, Clere-Jehl R, Schenck M, Kummerlen C, Collange O, Boulay C, Fafi-Kremer S, Ohana M, Anheim M, Meziani F. 2020. Neurologic features in severe SARS-CoV-2 infection. N Engl J Med 382:2268–2270. https://doi.org/10.1056/NEJMc2008597.

317. Ramani A, Müller L, Ostermann PN, Gabriel E, Abida-Islam P, Müller- Schiffmann A, Mariappan A, Goureau O, Gruell H, Walker A, Andrée M, Hauka S, Houwaart T, Dilthey A, Wohlgemuth K, Omran H, Klein F, Wieczorek D, Adams O, Timm J, Korth C, Schaal H, Gopalakrishnan J. 2020. SARS-CoV-2 targets neurons of 3D human brain organoids. EMBO J 39:e106230. https://doi.org/10.15252/embj.2020106230.

318. Jacob F, Pather SR, Huang WK, Zhang F, Wong SZH, Zhou H, Cubitt B, Fan W, Chen CZ, Xu M, Pradhan M, Zhang DY, Zheng W, Bang AG, Song H, Carlos de la Torre J, Ming GL. 2020. Human pluripotent stem cell- derived neural cells and brain organoids reveal SARS-CoV-2 neurotro- pism predominates in choroid plexus epithelium. Cell Stem Cell https:// doi.org/10.1016/j.stem.2020.09.016.

319. Alonso AD, Di Clerico J, Li B, Corbo CP, Alaniz ME, Grundke-Iqbal I, Iqbal K. 2010. Phosphorylation of Tau at Thr212, Thr231, and Ser262 combined causes neurodegeneration. J Biol Chem 285:30851–30860. https://doi.org/10.1074/jbc.M110.110957.

320. Buerger K, Otto M, Teipel SJ, Zinkowski R, Blennow K, DeBernardis J, Kerkman D, Schröder J, Schönknecht P, Cepek L, McCulloch C, Möller H- J, Wiltfang J, Kretzschmar H, Hampel H. 2006. Dissociation between CSF total tau and tau protein phosphorylated at threonine 231 in Creutz- feldt-Jakob disease. Neurobiol Aging 27:10–15. https://doi.org/10.1016/j .neurobiolaging.2004.12.003.

321. Netland J, Meyerholz DK, Moore S, Cassell M, Perlman S. 2008. Severe acute respiratory syndrome coronavirus infection causes neuronal death

in the absence of encephalitis in mice transgenic for human ACE2. J Virol

82:7264–7275. https://doi.org/10.1128/JVI.00737-08.
322. McCray PB, Pewe L, Wohlford-Lenane C, Hickey M, Manzel L, Shi L,

Netland J, Jia HP, Halabi C, Sigmund CD, Meyerholz DK, Kirby P, Look DC, Perlman S. 2007. Lethal infection of K18-hACE2 mice infected with severe acute respiratory syndrome coronavirus. J Virol 81:813–821. https://doi .org/10.1128/JVI.02012-06.

323. Chen R, Wang K, Yu J, Chen Z, Wen C, Xu Z. 2020. The spatial and cell- type distribution of SARS-CoV-2 receptor ACE2 in human and mouse brain. bioRxiv https://doi.org/10.1101/2020.04.07.030650.

324. Yashavantha Rao HC, Jayabaskaran C. 2020. The emergence of a novel coronavirus (SARS-CoV-2) disease and their neuroinvasive propensity may affect in COVID-19 patients. J Med Virol 92:786–790. https://doi .org/10.1002/jmv.25918.

325. Brann DH, Tsukahara T, Weinreb C, Lipovsek M, Van den Berge K, Gong B, Chance R, Macaulay IC, Chou H-J, Fletcher RB, Das D, Street K, de Bezieux HR, Choi Y-G, Risso D, Dudoit S, Purdom E, Mill J, Hachem RA, Matsunami H, Logan DW, Goldstein BJ, Grubb MS, Ngai J, Datta SR. 2020. Non-neuronal expression of SARS-CoV-2 entry genes in the olfactory system suggests mechanisms underlying COVID-19-associated anosmia. SciAdv 6:eabc5801. https://doi.org/10.1126/sciadv.abc5801.

326. Finsterer J, Stollberger C. 2020. Causes of hypogeusia/hyposmia in SARS-CoV2 infected patients. J Med Virol 92:1793–1794. https://doi .org/10.1002/jmv.25903.

327. Giannis D, Ziogas IA, Gianni P. 2020. Coagulation disorders in coronavi- rus infected patients: COVID-19, SARS-CoV-1, MERS-CoV and lessons from the past. J Clin Virol 127:104362. https://doi.org/10.1016/j.jcv.2020 .104362.

328. LuR,ZhaoX,LiJ,NiuP,YangB,WuH,WangW,SongH,HuangB,ZhuN, Bi Y, Ma X, Zhan F, Wang L, Hu T, Zhou H, Hu Z, Zhou W, Zhao L, Chen J, Meng Y, Wang J, Lin Y, Yuan J, Xie Z, Ma J, Liu WJ, Wang D, Xu W, Holmes EC, Gao GF, Wu G, Chen W, Shi W, Tan W. 2020. Genomic charac- terisation and epidemiology of 2019 novel coronavirus: implications for virus origins and receptor binding. Lancet 395:565–574. https://doi.org/ 10.1016/S0140-6736(20)30251-8.

329. Tan CW, Low JGH, Wong WH, Chua YY, Goh SL, Ng HJ. 2020. Critically ill COVID-19 infected patients exhibit increased clot waveform analysis pa- rameters consistent with hypercoagulability. Am J Hematol 95:E156–E158. https://doi.org/10.1002/ajh.25822.

330. Avula A, Nalleballe K, Narula N, Sapozhnikov S, Dandu V, Toom S, Glaser A, Elsayegh D. 2020. COVID-19 presenting as stroke. Brain Behav Immun 87:115–119. https://doi.org/10.1016/j.bbi.2020.04.077.

331. Sun J, Ye F, Wu A, Yang R, Pan M, Sheng J, Zhu W, Mao L, Wang M, Huang B, Tan W, Jiang T. 2020. Comparative transcriptome analysis reveals the intensive early-stage responses of host cells to SARS-CoV-2 infection. bioRxiv https://doi.org/10.1101/2020.04.30.071274.

332. Yilla M, Harcourt BH, Hickman CJ, McGrew M, Tamin A, Goldsmith CS, Bellini WJ, Anderson LJ. 2005. SARS-coronavirus replication in human peripheral monocytes/macrophages. Virus Res 107:93–101. https://doi .org/10.1016/j.virusres.2004.09.004.

333. Desforges M, Le Coupanec A, Dubeau P, Bourgouin A, Lajoie L, Dubé M, Talbot PJ. 2019. Human coronaviruses and other respiratory viruses: underestimated opportunistic pathogens of the central nervous system? Viruses 12:14. https://doi.org/10.3390/v12010014.

334. XuJ,ZhongS,LiuJ,LiL,LiY,WuX,LiZ,DengP,ZhangJ,ZhongN,Ding Y, Jiang Y. 2005. Detection of severe acute respiratory syndrome corona- virus in the brain: potential role of the chemokine mig in pathogenesis. Clin Infect Dis 41:1089–1096. https://doi.org/10.1086/444461.

335. Pellegrini L, Albecka A, Mallery DL, Kellner MJ, Paul D, Carter AP, James LC, Lancaster MA. 13 October 2020, posting date. SARS-CoV-2 infects the brain choroid plexus and disrupts the blood-CSF barrier in human brain organoids. Cell Stem Cell https://doi.org/10.1016/j.stem.2020.10 .001.

336. Douglas KAA, Douglas VP, Moschos MM. 2020. Ocular manifestations of COVID-19 (SARS-CoV-2): a critical review of current literature. In Vivo 34:1619–1628. https://doi.org/10.21873/invivo.11952.

337. Dinkin M, Gao V, Kahan J, Bobker S, Simonetto M, Wechsler P, Harpe J, Greer C, Mints G, Salama G, Tsiouris A, Leifer D. 2020. COVID-19 present- ing with ophthalmoparesis from cranial nerve palsy. Neurology 95:221–223. https://doi.org/10.1212/WNL.0000000000009700.

338. Chen L, Liu M, Zhang Z, Qiao K, Huang T, Chen M, Xin N, Huang Z, Liu L, Zhang G, Wang J. 2020. Ocular manifestations of a hospitalised patient with confirmed 2019 novel coronavirus disease. Br J Ophthalmol 104:748–751. https://doi.org/10.1136/bjophthalmol-2020-316304.

April 2021 Volume 34 Issue 2 e00133-20

cmr.asm.org 28

Downloaded from http://cmr.asm.org/ on January 21, 2021 by guest

SARS-CoV-2: a Systemic Infection

Clinical Microbiology Reviews

339. ZhangX,ChenX,ChenL,DengC,ZouX,LiuW,YuH,ChenB,SunX. 2020. The evidence of SARS-CoV-2 infection on ocular surface. Ocul Surf 18:360–362. https://doi.org/10.1016/j.jtos.2020.03.010.

340. Sun C-B, Wang Y-Y, Liu G-H, Liu Z. 2020. Role of the eye in transmitting human coronavirus: what we know and what we do not know. Front Public Health 8:155–155. https://doi.org/10.3389/fpubh.2020.00155.

341. Durán CSC, Mayorga GDC. 15 July 2020, posting date. The eye: “an organ that must not be forgotten in coronavirus disease 2019 (COVID-2019) pandemic.” J Optom https://doi.org/10.1016/j.optom.2020.07.002.

342. Cheema M, Aghazadeh H, Nazarali S, Ting A, Hodges J, McFarlane A, Kanji JN, Zelyas N, Damji KF, Solarte C. 2020. Keratoconjunctivitis as the initial medical presentation of the novel coronavirus disease 2019 (COVID-19). Can J Ophthalmol 55:e125–e129. https://doi.org/10.1016/j .jcjo.2020.03.003.

343. Holappa M, Vapaatalo H, Vaajanen A. 2017. Many faces of renin-angio- tensin system – focus on eye. Open Ophthalmol J 11:122–142. https:// doi.org/10.2174/1874364101711010122.

344. Panoutsopoulos AA. 2020. Conjunctivitis as a sentinel of SARS-CoV-2 infection: a need of revision for mild symptoms. Sn Compr Clin Med 2:859–864. https://doi.org/10.1007/s42399-020-00360-7.

345. Senanayake P, Drazba J, Shadrach K, Milsted A, Rungger-Brandle E, Nishiyama K, Miura S-I, Karnik S, Sears JE, Hollyfield JG. 2007. Angioten- sin II and its receptor subtypes in the human retina. Invest Ophthalmol Vis Sci 48:3301–3311. https://doi.org/10.1167/iovs.06-1024.

346. Holappa M, Valjakka J, Vaajanen A. 2015. Angiotensin(1–7) and ACE2, “the hot spots” of renin-angiotensin system, detected in the human aqueous humor. Open Ophthalmol J 9:28–32. https://doi.org/10.2174/ 1874364101509010028.

347. Makovoz B, Moeller R, Zebitz Eriksen A, tenOever BR, Blenkinsop TA. 15 July 2020, posting date. SARS-CoV-2 infection of ocular cells from human adult donor eyes and hESC-derived eye organoids. SSRN https:// doi.org/10.2139/ssrn.3650574.

348. Nakatsu MN, Ding Z, Ng MY, Truong TT, Yu F, Deng SX. 2011. Wnt/b-cat- enin signaling regulates proliferation of human cornea epithelial stem/ progenitor cells. Invest Ophthalmol Vis Sci 52:4734–4741. https://doi .org/10.1167/iovs.10-6486.

349. Kim J, Thomsen T, Sell N, Goldsmith AJ. 2020. Abdominal and testicular pain: an atypical presentation of COVID-19. Am J Emerg Med 38:1542. e1–1542.e3. https://doi.org/10.1016/j.ajem.2020.03.052.

350. La Marca A, Busani S, Donno V, Guaraldi G, Ligabue G, Girardis M. 2020. Testicular pain as an unusual presentation of COVID-19: a brief review of SARS-CoV-2 and the testis. Reprod Biomed Online 41:903–906. https:// doi.org/10.1016/j.rbmo.2020.07.017.

351. Suryawanshi H, Morozov P, Muthukumar T, tenOever BR, Yamaji M, Williams Z, Tuschl T. 2020. Cell-type-specific expression of renin-an- giotensin-system components in the human body and its relevance to SARS-CoV-2 infection. bioRxiv https://doi.org/10.1101/2020.04.11 .034603.

352. Shastri A, Wheat J, Agrawal S, Chaterjee N, Pradhan K, Goldfinger M, Kornblum N, Steidl U, Verma A, Shastri J. 2020. Delayed clearance of SARS-CoV2 in male compared to female patients: high ACE2 expression in testes suggests possible existence of gender-specific viral reservoirs. medRxiv https://doi.org/10.1101/2020.04.16.20060566.

353. ZhangJ,WuY,WangR,LuK,TuM,GuoH,XieW,QinZ,LiS,ZhuP, Wang X. 2020. Bioinformatic analysis reveals that the reproductive sys- tem is potentially at risk from SARS-CoV-2. PrePrints https://www .preprints.org/manuscript/202002.0307/v1.

354. Younis JS, Abassi Z, Skorecki K. 2020. Is there an impact of the COVID-19 pandemic on male fertility? The ACE2 connection. Am J Physiol Endocri- nol Metab 318:E878–E880. https://doi.org/10.1152/ajpendo.00183.2020.

355. Fan C, Li K, Ding Y, Lu WL, Wang J. 2020. ACE2 expression in kidney and testis may cause kidney and testis damage after 2019-nCoV infection. medRxiv https://doi.org/10.1101/2020.02.12.20022418.

356. Wang Z, Xu X. 2020. scRNA-seq profiling of human testes reveals the presence of the ACE2 receptor, a target for SARS-CoV-2 infection in sper- matogonia, Leydig and Sertoli cells. Cells 9:920. https://doi.org/10.3390/ cells9040920.

357. Shen Q, Xiao X, Aierken A, Liao M, Hua J. 2020. The ACE2 expression in Sertoli cells and germ cells may cause male reproductive disorder after SARS-CoV-2 infection. J Cell Mol Med 24:9472–9477. https://doi.org/10 .1111/jcmm.15541.

358. Liu X, Chen Y, Tang W, Zhang L, Chen W, Yan Z, Yuan P, Yang M, Kong S, Yan L, Qiao J. 2020. Single-cell transcriptome analysis of the novel coro- navirus (SARS-CoV-2) associated gene ACE2 expression in normal and

April 2021 Volume 34 Issue 2 e00133-20

non-obstructive azoospermia (NOA) human male testes. Sci China Life

Sci 63:1006–1015. https://doi.org/10.1007/s11427-020-1705-0.
359. RenX,WeiX,LiG,RenS,ChenX,ZhangT,ZhangX,LuZ,YouZ,WangS, Qin C, Song N, Wang Z. 2020. Multiple expression assessments of ACE2 and TMPRSS2 SARS-CoV-2 entry molecules in the urinary tract and their associations with clinical manifestations of COVID-19. bioRxiv https://doi

.org/10.1101/2020.05.08.083618.
360. Reis AB, Araújo FC, Pereira VM, Dos Reis AM, Santos RA, Reis FM. 2010.

Angiotensin (1–7) and its receptor Mas are expressed in the human tes- tis: implications for male infertility. J Mol Histol 41:75–80. https://doi .org/10.1007/s10735-010-9264-8.

361. Zupin L, Pascolo L, Zito G, Ricci G, Crovella S. 2020. SARS-CoV-2 and the next generations: which impact on reproductive tissues? J Assist Reprod Genet 37:2399–2403. https://doi.org/10.1007/s10815-020-01917-0.

362. Vishvkarma R, Rajender S. 2020. Could SARS-CoV-2 affect male fertility? Andrologia 52:e13712. https://doi.org/10.1111/and.13712.

363. Paoli D, Pallotti F, Colangelo S, Basilico F, Mazzuti L, Turriziani O, Antonelli G, Lenzi A, Lombardo F. 2020. Study of SARS-CoV-2 in semen and urine samples of a volunteer with positive naso-pharyngeal swab. J Endocrinol Invest 43:1819–1822. https://doi.org/10.1007/s40618-020 -01261-1.

364. Paoli D, Pallotti F, Turriziani O, Mazzuti L, Antonelli G, Lenzi A, Lombardo F. 26 May 2020, posting date. SARS-CoV-2 presence in seminal fluid: myth or reality? Andrology https://doi.org/10.1111/andr.12825.

365. Yang M, Chen S, Huang B, Zhong J-M, Su H, Chen Y-J, Cao Q, Ma L, He J, Li X-F, Li X, Zhou J-J, Fan J, Luo D-J, Chang X-N, Arkun K, Zhou M, Nie X. 2020. Pathological findings in the testes of COVID-19 patients: clinical implications. Eur Urol Focus 6:1124–1129. https://doi.org/10.1016/j.euf .2020.05.009.

366. Li D, Jin M, Bao P, Zhao W, Zhang S. 2020. Clinical characteristics and results of semen tests among men with coronavirus disease 2019. JAMA Netw Open 3:e208292. https://doi.org/10.1001/jamanetworkopen.2020 .8292.

367. SongC,WangY,LiW,HuB,ChenG,XiaP,WangW,LiC,HuZ,YangX, Yao B, Liu Y. 2020. Detection of 2019 novel coronavirus in semen and testicular biopsy specimen of COVID-19 patients. medRxiv https://doi .org/10.1101/2020.03.31.20042333.

368. Pan F, Xiao X, Guo J, Song Y, Li H, Patel DP, Spivak AM, Alukal JP, Zhang X, Xiong C, Li PS, Hotaling JM. 2020. No evidence of SARS-CoV-2 in semen of males recovering from COVID-19. Fertil Steril 113:1135–1139. https://doi.org/10.1016/j.fertnstert.2020.04.024.

369. GuoL,ZhaoS,LiW,WangY,LiL,JiangS,RenW,YuanQ,ZhangF,Kong F, Lei J, Yuan M. 29 June 2020, posting date. Absence of SARS-CoV-2 in semen of a COVID-19 patient cohort. Andrology https://doi.org/10 .1111/andr.12848.

370. Nora H, Philippos E, Marcel A, Cornelius D, Dunja B-B, Ortwin A, Jan- Steffen K, Petra BA. 2020. Assessment of SARS-CoV-2 in human semen – a cohort study. Fertil Steril 114:233–238. https://doi.org/10.1016/j.fertnstert .2020.05.028.

371. QuanW,ZhengQ,TianJ,ChenJ,LiuZ,ChenX,WuT,JiZ,TangJ,ChuH, Xu H, Zhao Y. 2020. No SARS-CoV-2 in expressed prostatic secretion of patients with coronavirus disease 2019: a descriptive multicentre study in China. medRxiv https://doi.org/10.1101/2020.03.26.20044198.

372. Xu H, Wang Z, Feng C, Yu W, Chen Y, Zeng X, Liu C. 5 November 2020, posting date. Effects of SARS-CoV-2 infection on male sex-related hor- mones in recovering patients. Andrology https://doi.org/10.1111/andr .12942.

373. MaL,XieW,LiD,ShiL,YeG,MaoY,XiongY,SunH,ZhengF,ChenZ, Qin J, Lyu J, Zhang Y, Zhang M. 4 July 2020, posting date. Evaluation of sex-related hormones and semen characteristics in reproductive-aged male COVID-19 patients. J Med Virol https://doi.org/10.1002/jmv.26259.

374. Henarejos-Castillo I, Sebastian-Leon P, Devesa-Peiro A, Pellicer A, Diaz- Gimeno P. 2020. SARS-CoV-2 infection risk assessment in the endome- trium: viral infection-related gene expression across the menstrual cycle. Fertil Steril 114:223–232. https://doi.org/10.1016/j.fertnstert.2020.06.026.

375. Stanley KE, Thomas E, Leaver M, Wells D. 2020. Coronavirus disease (COVID-19) and fertility: viral host entry protein expression in male and female reproductive tissues. Fertil Steril 114:33–43. https://doi.org/10 .1016/j.fertnstert.2020.05.001.

376. Jing Y, Run-Qian L, Hao-Ran W, Hao-Ran C, Ya-Bin L, Yang G, Fei C. 2020. Potential influence of COVID-19/ACE2 on the female reproductive sys- tem. Mol Hum Reprod 26:367–373. https://doi.org/10.1093/molehr/ gaaa030.

377. Goad J, Rudolph J, Rajkovic A. 2020. Female reproductive tract has low cmr.asm.org 29

Downloaded from http://cmr.asm.org/ on January 21, 2021 by guest

Synowiec et al.

Clinical Microbiology Reviews

concentration of SARS-CoV2 receptors. bioRxiv https://doi.org/10.1101/

2020.06.20.163097.

378. QiuL,LiuX,XiaoM,XieJ,CaoW,LiuZ,MorseA,XieY,LiT,ZhuL.2April
2020, posting date. SARS-CoV-2 is not detectable in the vaginal fluid of women with severe COVID-19 infection. Clin Infect Dis https://doi.org/ 10.1093/cid/ciaa375.

379. Juan J, Gil MM, Rong Z, Zhang Y, Yang H, Poon LC. 2020. Effect of coro- navirus disease 2019 (COVID-19) on maternal, perinatal and neonatal outcome: systematic review. Ultrasound Obstet Gynecol 56:15–27. https://doi.org/10.1002/uog.22088.

380. Cui P, Chen Z, Wang T, Dai J, Zhang J, Ding T, Jiang J, Liu J, Zhang C, Shan W, Wang S, Rong Y, Chang J, Miao X, Ma X, Wang S. 2020. Clinical features and sexual transmission potential of SARS-CoV-2 infected female patients: a descriptive study in Wuhan, China. medRxiv https:// doi.org/10.1101/2020.02.26.20028225.

381. Dong L, Tian J, He S, Zhu C, Wang J, Liu C, Yang J. 2020. Possible vertical transmission of SARS-CoV-2 from an infected mother to her newborn. JAMA 323:1846–1848. https://doi.org/10.1001/jama.2020.4621.

382. Lamouroux A, Attie-Bitach T, Martinovic J, Leruez-Ville M, Ville Y. 2020. Evidence for and against vertical transmission for SARS-CoV-2 (COVID-19). Am J Obstet Gynecol 223:91.e1–91.e4. https://doi.org/10 .1016/j.ajog.2020.04.039.

383. ChenH,GuoJ,WangC,LuoF,YuX,ZhangW,LiJ,ZhaoD,XuD,Gong Q, Liao J, Yang H, Hou W, Zhang Y. 2020. Clinical characteristics and intrauterine vertical transmission potential of COVID-19 infection in nine pregnant women: a retrospective review of medical records. Lancet 395:809–815. https://doi.org/10.1016/S0140-6736(20)30360-3.

384. Rodrigues C, Baia I, Domingues R, Barros H. 2020. Pregnancy and breast- feeding during COVID-19 pandemic: a systematic review of published pregnancy cases. medRxiv https://doi.org/10.1101/2020.04.25.20079509.

385. FanC,LeiD,FangC,LiC,WangM,LiuY,BaoY,SunY,HuangJ,GuoY, Yu Y, Wang S. 17 March 2020, posting date. Perinatal transmission of COVID-19-associated SARS-CoV-2: should we worry? Clin Infect Dis https://doi.org/10.1093/cid/ciaa226.

386. Chen S, Huang B, Luo DJ, Li X, Yang F, Zhao Y, Nie X, Huang BX. 2020. Pregnancy with new coronavirus infection: clinical characteristics and placental pathological analysis of three cases. Zhonghua Bing Li Xue Za Zhi 49:418–423. (In Chinese.) https://doi.org/10.3760/cma.j.cn112151 -20200225-00138.

387. Karimi-Zarchi M, Neamatzadeh H, Dastgheib SA, Abbasi H, Mirjalili SR, Behforouz A, Ferdosian F, Bahrami R. 2020. Vertical transmission of co- ronavirus disease 19 (COVID-19) from infected pregnant mothers to neonates: a review. Fetal Pediatr Pathol 39:246–250. https://doi.org/10 .1080/15513815.2020.1747120.

388. Fenizia C, Biasin M, Cetin I, Vergani P, Mileto D, Spinillo A, Gismondo MR, Perotti F, Callegari C, Mancon A, Cammarata S, Beretta I, Nebuloni M, Trabattoni D, Clerici M, Savasi V. 2020. Analysis of SARS-CoV-2 vertical transmission during pregnancy. Nat Commun 11:5128. https://doi.org/ 10.1038/s41467-020-18933-4.

389. Penfield CA, Brubaker SG, Limaye MA, Lighter J, Ratner AJ, Thomas KM, Meyer JA, Roman AS. 2020. Detection of severe acute respiratory syn- drome coronavirus 2 in placental and fetal membrane samples. Am J Obstet Gynecol MFM 2:100133. https://doi.org/10.1016/j.ajogmf.2020 .100133.

390. Hosier H, Farhadian SF, Morotti RA, Deshmukh U, Lu-Culligan A, Campbell KH, Yasumoto Y, Vogels CBF, Casanovas-Massana A, Vijayakumar P, Geng B, Odio CD, Fournier J, Brito AF, Fauver JR, Liu F, Alpert T, Tal R, Szigeti- Buck K, Perincheri S, Larsen C, Gariepy AM, Aguilar G, Fardelmann KL, Harigopal M, Taylor HS, Pettker CM, Wyllie AL, Cruz CD, Ring AM, Grubaugh ND, Ko AI, Horvath TL, Iwasaki A, Reddy UM, Lipkind HS. 2020. SARS–CoV-2 infection of the placenta. J Clin Invest 130:4947–4953. https://doi.org/10.1172/JCI139569.

391. Algarroba GN, Hanna NN, Rekawek P, Vahanian SA, Khullar P, Palaia T, Peltier MR, Chavez MR, Vintzileos AM. 2020. Confirmatory evidence of the visualization of severe acute respiratory syndrome coronavirus 2 invading the human placenta using electron microscopy. Am J Obstet Gynecol 223:953–954. https://doi.org/10.1016/j.ajog.2020.08.106.

392. Algarroba GN, Rekawek P, Vahanian SA, Khullar P, Palaia T, Peltier MR, Chavez MR, Vintzileos AM. 2020. Visualization of severe acute respiratory syndrome coronavirus 2 invading the human placenta using electron microscopy. Am J Obstet Gynecol 223:275–278. https://doi.org/10.1016/ j.ajog.2020.05.023.

393. Phoswa WN, Khaliq OP. 2020. Is pregnancy a risk factor of COVID-19? Eur

April 2021 Volume 34 Issue 2 e00133-20

J Obstet Gynecol Reprod Biol 252:605–609. https://doi.org/10.1016/j

.ejogrb.2020.06.058.
394. Hanna N, Hanna M, Sharma S. 5 August 2020, posting date. Is pregnancy

an immunological contributor to severe or controlled COVID-19 disease?

Am J Reprod Immunol https://doi.org/10.1111/aji.13317.
395. Dashraath P, Wong JLJ, Lim MXK, Lim LM, Li S, Biswas A, Choolani M, Mattar C, Su LL. 2020. Coronavirus disease 2019 (COVID-19) pandemic and pregnancy. Am J Obstet Gynecol 222:521–531. https://doi.org/10

.1016/j.ajog.2020.03.021.
396. Mehan A, Venkatesh A, Girish M. 2020. COVID-19 in pregnancy: risk of

adverse neonatal outcomes. J Med Virol 92:2295–2297. https://doi.org/

10.1002/jmv.25959.
397. Li G, Li W, Song B, Wu H, Tang D, Wang C, He X, Cao Y. 2020. SARS-CoV-2

and the reproductive system: assessment of risk and considerations for infection control in reproductive departments. Syst Biol Reprod Med 66:343–346. https://doi.org/10.1080/19396368.2020.1817627.

398. Singh B, Gornet M, Sims H, Kisanga E, Knight Z, Segars J. 2020. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its effect on gametogenesis and early pregnancy. Am J Reprod Immunol 84:e13351. https://doi.org/10.1111/aji.13351.

399. Khalil A, Kalafat E, Benlioglu C, O’Brien P, Morris E, Draycott T, Thangaratinam S, Le Doare K, Heath P, Ladhani S, von Dadelszen P, Magee LA. 2020. SARS- CoV-2 infection in pregnancy: a systematic review and meta-analysis of clini- cal features and pregnancy outcomes. EClinicalMedicine 25:100446. https:// doi.org/10.1016/j.eclinm.2020.100446.

400. LeiD,WangC,LiC,FangC,YangW,ChenB,WeiM,XuX,YangH,Wang S, Fan C. 2020. Clinical characteristics of COVID-19 in pregnancy: analysis of nine cases. Chin J Perinat Med 23. https://doi.org/10.3760/cma.j .cn113903-20200216-00117.

401. Peng Z, Wang J, Mo Y, Duan W, Xiang G, Yi M, Bao L, Shi Y. 2020. Unlikely SARS-CoV-2 vertical transmission from mother to child: a case report. J Infect Public Health 13:818–820. https://doi.org/10.1016/j.jiph.2020.04 .004.

402. Deniz M, Tezer H. 21 July 2020, posting date. Vertical transmission of SARS CoV-2: a systematic review. Matern Fetal Neonatal Med https://doi .org/10.1080/14767058.2020.1793322.

403. Vivanti AJ, Vauloup-Fellous C, Prevot S, Zupan V, Suffee C, Do Cao J, Benachi A, De Luca D. 2020. Transplacental transmission of SARS-CoV-2 infection. Nat Commun 11:3572. https://doi.org/10.1038/s41467-020 -17436-6.

404. Zeng H, Xu C, Fan J, Tang Y, Deng Q, Zhang W, Long X. 2020. Antibodies in infants born to mothers with COVID-19 pneumonia. JAMA 323:1848–1849. https://doi.org/10.1001/jama.2020.4861.

405. Xu J, Qi L, Chi X, Yang J, Wei X, Gong E, Peh S, Gu J. 2006. Orchitis: a com- plication of severe acute respiratory syndrome (SARS). Biol Reprod 74:410–416. https://doi.org/10.1095/biolreprod.105.044776.

406. DingY,HeL,ZhangQ,HuangZ,CheX,HouJ,WangH,ShenH,QiuL,Li Z, Geng J, Cai J, Han H, Li X, Kang W, Weng D, Liang P, Jiang S. 2004. Organ distribution of severe acute respiratory syndrome (SARS) associ- ated coronavirus (SARS-CoV) in SARS patients: implications for pathoge- nesis and virus transmission pathways. J Pathol 203:622–630. https://doi .org/10.1002/path.1560.

407. Dutta S, Sengupta P. 10 July 2020, posting date. SARS-CoV-2 and male infertility: possible multifaceted pathology. Reprod Sci https://doi.org/ 10.1007/s43032-020-00261-z.

408. Hallak J, Teixeira TA, Bernardes FS, Carneiro F, Duarte SAS, Pariz JR, Esteves SC, Kallas E, Saldiva PHN. 1 September 2020, posting date. SARS- CoV-2 and its relationship with the genitourinary tract: implications for male reproductive health in the context of COVID-19 pandemic. Androl- ogy https://doi.org/10.1111/andr.12896.

409. Liu L, Chopra P, Li X, Wolfert MA, Tompkins SM, Boons G-J. 2020. SARS- CoV-2 spike protein binds heparan sulfate in a length- and sequence-de- pendent manner. bioRxiv https://doi.org/10.1101/2020.05.10.087288.

410. Qian Z, Dominguez SR, Holmes KV. 2013. Role of the spike glycoprotein of human Middle East respiratory syndrome coronavirus (MERS-CoV) in virus entry and syncytia formation. PLoS One 8:e76469. https://doi.org/ 10.1371/journal.pone.0076469.

411. Shirato K, Kanou K, Kawase M, Matsuyama S. 2017. Clinical isolates of human coronavirus 229E bypass the endosome for cell entry. J Virol 91: e01387-16. https://doi.org/10.1128/JVI.01387-16.

412. Shirato K, Kawase M, Matsuyama S. 2018. Wild-type human coronavi- ruses prefer cell-surface TMPRSS2 to endosomal cathepsins for cell entry. Virology 517:9–15. https://doi.org/10.1016/j.virol.2017.11.012.

413. Shen LW, Mao HJ, Wu YL, Tanaka Y, Zhang W. 2017. TMPRSS2: a cmr.asm.org 30

Downloaded from http://cmr.asm.org/ on January 21, 2021 by guest

SARS-CoV-2: a Systemic Infection

Clinical Microbiology Reviews

potential target for treatment of influenza virus and coronavirus infec-

tions. Biochimie 142:1–10. https://doi.org/10.1016/j.biochi.2017.07.016.

414. Shang J, Wan Y, Luo C, Ye G, Geng Q, Auerbach A, Li F. 2020. Cell entry mechanisms of SARS-CoV-2. Proc Natl Acad Sci U S A 117:11727–11734.
https://doi.org/10.1073/pnas.2003138117.

415. Emanuel W, Kirstin M, Vedran F, Asija D, Theresa GL, Roberto A, Filippos
K, David K, Salah A, Christopher B, Anja R, Ivano L, Andranik I, Tommaso M, Simone DG, Patrick PJ, Alexander MM, Daniela N, Matthias S, Altuna A, Nikolaus R, Christian D, Markus L. 2020. Bulk and single-cell gene expression profiling of SARS-CoV-2 infected human cell lines identifies molecular targets for therapeutic intervention. bioRxiv https://doi.org/ 10.1101/2020.05.05.079194.

416. Harcourt J, Tamin A, Lu X, Kamili S, Sakthivel SK, Wang L, Murray J, Queen K, Lynch B, Whitaker B, Tao Y, Paden CR, Zhang J, Li Y, Uehara A, Wang H, Goldsmith C, Bullock HA, Gautam R, Schindewolf C, Lokugamage KG, Scharton D, Plante JA, Mirchandani D, Widen SG, Narayanan K, Makino S, Ksiazek TG, Plante KS, Weaver SC, Lindstrom S, Tong S, Menachery VD, Thornburg NJ. 2020. Isolation and characterization of SARS-CoV-2 from the first US COVID-19 patient. bioRxiv https://doi.org/10.1101/2020.03.02 .972935.

417. Pruijssers AJ, George AS, Schäfer A, Leist SR, Gralinksi LE, Dinnon KH, Yount BL, Agostini ML, Stevens LJ, Chappell JD, Lu X, Hughes TM, Gully K, Martinez DR, Brown AJ, Graham RL, Perry JK, Du Pont V, Pitts J, Ma B, Babusis D, Murakami E, Feng JY, Bilello JP, Porter DP, Cihlar T, Baric RS, Denison MR, Sheahan TP. 2020. Remdesivir inhibits SARS-CoV-2 in human lung cells and chimeric SARS-CoV expressing the SARS-CoV-2 RNA polymerase in mice. Cell Rep 32:107940. https://doi.org/10.1016/j .celrep.2020.107940.

418. Ko M, Jeon S, Ryu W-S, Kim S. 2020. Comparative analysis of antiviral effi- cacy of FDA-approved drugs against SARS-CoV-2 in human lung cells: nafamostat is the most potent antiviral drug candidate. bioRxiv https:// doi.org/10.1101/2020.05.12.090035.

419. Yamamoto M, Kiso M, Sakai-Tagawa Y, Iwatsuki-Horimoto K, Imai M,

Aleksandra Synowiec is a Ph.D. Student at
Malopolska Centre of Biotechnology,
Jagiellonian University, Krakow, Poland. She
completed her master’s degree in molecular
biotechnology at the Faculty of Biochemistry,
Biophysics and Biotechnology, Jagiellonian
University, Krakow, Poland, in 2019. Her area
of interest is molecular biology of viral entry
as well as antiviral research, with a focus on
human coronaviruses and human norovirus.
She has been awarded several awards and
scholarships such as L’Oréal-UNESCO For Women in Science Award or Ministry of Science and Higher Education Scholarship for scientific achievements.

Takeda M, Kinoshita N, Ohmagari N, Gohda J, Semba K, Matsuda Z, Kawaguchi Y, Kawaoka Y, Inoue J-i. 2020. The anticoagulant nafamostat potently inhibits SARS-CoV-2 infection in vitro: an existing drug with multiple possible therapeutic effects. bioRxiv https://doi.org/10.1101/ 2020.04.22.054981.

420. Ogando NS, Dalebout TJ, Zevenhoven-Dobbe JC, Limpens RWAL, van der Meer Y, Caly L, Druce J, de Vries JJC, Kikkert M, Bárcena M, Sidorov I, Snijder EJ. 2020. SARS-coronavirus-2 replication in Vero E6 cells: replication kinetics, rapid adaptation and cytopathology. J Gen Virol 101:925–940. https://doi.org/10.1099/jgv.0.001453.

421. Milewska A, Kula-Pacurar A, Wadas J, Suder A, Szczepanski A, Dabrowska A, Owczarek K, Marcello A, Ochman M, Stacel T, Rajfur Z, Sanak M, Labaj P, Branicki W, Pyrc K. 2020. Replication of severe acute respiratory syn- drome coronavirus 2 in human respiratory epithelium. J Virol 94:e00957- 20. https://doi.org/10.1128/JVI.00957-20.

422. Milewska A, Chi Y, Szczepanski A, Barreto-Duran E, Liu K, Liu D, Guo X, Ge Y, Li J, Cui L, Ochman M, Urlik M, Rodziewicz-Motowidlo S, Zhu F, Szczubialka K, Nowakowska M, Pyrc K. 2020. HTCC as a highly effective polymeric inhibitor of SARS-CoV-2 and MERS-CoV. bioRxiv https://doi .org/10.1101/2020.03.29.014183.

423. Thacker VV, Sharma K, Dhar N, Mancini G-F, Sordet-Dessimoz J, Mckinney JD. 2020. Rapid endothelialitis and vascular inflammation characterise SARS-CoV-2 infection in a human lung-on-chip model. bioRxiv https://doi .org/10.1101/2020.08.10.243220.

424. Si L, Bai H, Rodas M, Cao W, Oh CY, Jiang A, Nurani A, Zhu DY, Goyal G, Gilpin SE, Prantil-Baun R, Ingber DE. 2020. Human organs-on-chips as tools for repurposing approved drugs as potential influenza and COVID19 therapeutics in viral pandemics. bioRxiv https://doi.org/10 .1101/2020.04.13.039917.

425. Valyaeva AA, Zharikova AA, Kasianov AS, Vassetzky YS, Sheval EV. 2020. Expression of SARS-CoV-2 entry factors in lung epithelial stem cells and its potential implications for COVID-19. Sci Rep 10:17772. https://doi .org/10.1038/s41598-020-74598-5.

Artur Szczepan ski is a Ph.D. Student at
Faculty of Biochemistry, Biophysics and
Biotechnology, Jagiellonian University,
Krakow, Poland. He is finishing his thesis on the
early stages of Canine Respiratory Coronavirus
entry to the cell. He completed his master’s
degree in biotechnology at the Faculty of
Biochemistry, Biophysics and Biotechnology,
Jagiellonian University, Krakow, Poland, in 2013.
His area of interest includes molecular virology
of viral entry with a focus on human and animal
coronaviruses, antiviral research, and advanced methods of imaging.

  

April 2021 Volume 34 Issue 2 e00133-20

cmr.asm.org 31

Downloaded from http://cmr.asm.org/ on January 21, 2021 by guest

Synowiec et al.

Clinical Microbiology Reviews

Emilia Barreto-Duran studied Biology at Los
Andes University, Bogota, Colombia, and did
her master’s in Cell Biology at the Pontificia
Universidad Javeriana, also in Bogota. Her
master’s thesis was aimed at developing an
organotypic spheroid model for the study of
the human bone marrow microenvironment.
She is currently a Marie Skłodowska-Curie
Fellow within the OrganoVIR Training Net-
work (MSCA-ITN) at the Jagiellonian Univer-
sity in Krakow, Poland. Emilia is part of Professor’sKrzysztofPyr cresearchgroup,ViroGenticslaboratory,inThe Malopolska Centre of Biotechnology. The objective of her Ph.D. research project is the development of a complex coculture model that resembles the human airway epithelium microenvironment for the study of coronavi- rus pathogenesis.

Laurensius Kevin Lie is a Ph.D. student at
Malopolska Center of Biotechnology, Jagiel-
lonian University, Krakow, Poland. He is an
early-stage researcher enrolled in the Marie
Skłodowska-Curie Action – Integrated Train-
ing Network program OrganoVIR, which
focuses on the study of viruses using orga-
noid technology. He completed his master’s
degree in biomedical science at the Catholic
University of Leuven, Belgium, in 2015. He
had previously worked as a research and
teaching staff in the Laboratory of Hygienic Sciences of Kobe Pharmaceutical University in Japan in 2016. His area of interest is in molecu- lar virology of virus dissemination, with a particular focus on understanding cross-tissue and cross-species dissemination of corona-viruses using organoids.

Krzysztof Pyrc, Ph.D. is a full professor and a
leader of the Virogenetics team at Malopolska
Centre of Biotechnology, Jagiellonian Univer-
sity, Krakow, Poland. Ph.D. in 2007 at the
University of Amsterdam, the Netherlands,
habilitation in 2013 at the University of Lodz,
Poland, and title of professor by the president
of Poland in 2019. Laureate of several national
and international awards and research grants.
Official governmental advisor during the
COVID-19 pandemic, author of almost 100 sci-
entific publications in journals as Nature Medicine, Science Translational Medicine, PNAS, ACS Applied Materials & Interfaces, Journal of Virology, and others. Scientific interests: virology with particular focus on coronavi- ruses and flaviviruses.

 

April 2021 Volume 34 Issue 2 e00133-20

cmr.asm.org 32

Downloaded from http://cmr.asm.org/ on January 21, 2021 by guest

Leave a Reply

Fill in your details below or click an icon to log in:

WordPress.com Logo

You are commenting using your WordPress.com account. Log Out /  Change )

Twitter picture

You are commenting using your Twitter account. Log Out /  Change )

Facebook photo

You are commenting using your Facebook account. Log Out /  Change )

Connecting to %s

%d bloggers like this: