key: cord-0942635-zhmgu35d authors: Ouyang, Yingshi; Bagalkot, Tarique; Fitzgerald, Wendy; Sadovsky, Elena; Chu, Tianjiao; Martínez-Marchal, Ana; Brieño-Enríquez, Miguel; Su, Emily J.; Margolis, Leonid; Sorkin, Alexander; Sadovsky, Yoel title: Term Human Placental Trophoblasts Express SARS-CoV-2 Entry Factors ACE2, TMPRSS2, and Furin date: 2021-04-14 journal: mSphere DOI: 10.1128/msphere.00250-21 sha: 3dc09e7629a041bc56430e542cf87112f52702ab doc_id: 942635 cord_uid: zhmgu35d The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has had a massive impact on human lives worldwide. While the airborne SARS-CoV-2 primarily affects the lungs, viremia is not uncommon. As placental trophoblasts are directly bathed in maternal blood, they are vulnerable to SARS-CoV-2. Intriguingly, the human fetus is largely spared from SARS-CoV-2 infection. We tested whether the human placenta expresses the main SARS-CoV-2 entry factors angiotensin-converting enzyme 2 (ACE2), transmembrane protease serine 2 (TMPRSS2), and furin and showed that ACE2 and TMPRSS2 are expressed in the trophoblast rather than in other placental villous cells. While furin is expressed in the main placental villous cell types, we surveyed, trophoblasts exhibit the highest expression. In line with the expression of these entry factors, we demonstrated that a SARS-CoV-2 pseudovirus could enter primary human trophoblasts. Mechanisms underlying placental defense against SARS-CoV-2 infection likely involve postentry processing, which may be germane for mitigating interventions against SARS-CoV-2. IMPORTANCE Pregnant women worldwide have been affected by COVID-19. As the virus is commonly spread to various organs via the bloodstream and because human placental trophoblasts are directly bathed in maternal blood, feto-placental infection by SARS-CoV-2 seems likely. However, despite the heightened risk to pregnant women, thus far the transmission risk of COVID-19 to the feto-placental unit seems extremely low. This has been recently attributed to a negligible expression of SARS-CoV-2 entry factors in the human placenta. We therefore sought to explore the expression of the entry factors ACE2 and TMPRSS2 in the different cell types of human placental villi. Using a combination of transcriptome sequencing (RNA-seq), real-time quantitative PCR (RT-qPCR), in situ hybridization, and immunofluorescence, we found that trophoblasts, but not the other main villous cell types, express ACE2 and TMPRSS2, with a broad expression of furin. Correspondingly, we also showed that primary human trophoblasts are permissive to entry of SARS-CoV-2 pseudovirus particles. on the presence of receptors and processing proteins that mediate trophoblast viral entry. Thus far, such data have been scant and relatively inconsistent. SARS-CoV-2 viral proteins, including S and N (nucleocapsid) proteins, were visualized in syncytiotrophoblasts, which are in direct contact with the maternal circulation (45) (46) (47) (48) . However, a recent analysis of single-cell transcriptome sequencing (RNA-seq) suggested negligible ACE2 and TMPRSS2 expression in the placenta (49) . In contrast, earlier studies showed that ACE2 is expressed in the human placenta, especially in syncytiotrophoblasts, the subjacent cytotrophoblasts, and endothelial cells of the umbilical cord (5, 45, (50) (51) (52) (53) (54) (55) (56) (57) (58) . The expression of placental ACE2 is increased in patients with preeclampsia, suggesting the functional role of ACE2 in countering hypertension at the human uteroplacental interface (5, 50) . Less information exists regarding ACE2 expression in the various villous cell types and the expression of TMPRSS2 and furin. We therefore sought to explore the expression of ACE2 and TMPRSS2 in the different cell types of placental villi. The expression of these processing proteins would suggest that trophoblasts are susceptible to SARS-CoV-2 infection and the ensuing transmission to the fetus. Using a combination of RNA-seq, real-time quantitative PCR (RT-qPCR), in situ hybridizationbased RNAscope, and immunofluorescence, we found that trophoblasts, but not the other main placental villous cell types, express ACE2 and TMPRSS2 with broad expression of furin. Correspondingly, we also showed that primary human trophoblasts derived from term placentas are permissive to entry of SARS-CoV-2 pseudovirus particles. The expression of ACE2, TMPRSS2, and furin mRNA in the human placenta and in villous cells. We first used RT-qPCR as a sensitive tool to quantify the expression of three viral entry factors, ACE2, TMPRSS2, and furin, in term human placenta and compared these data to the expression of these transcripts in other human organs, including the heart, small intestine, kidney, liver, lung, and testis. As shown in Fig. 1A , ACE2, TMPRSS2, and furin were clearly expressed in the human placenta, with average threshold cycle (C T ) values of 27.5, 29.9, and 22.7, respectively. Intriguingly, the expression level of ACE2 mRNA in the placenta was similar to its expression in the lung, the most common site for SARS-CoV-2 infection in humans. TMPRSS2 levels in most tissues were higher than the placenta, and the expression of furin was fairly comparable among the tissues (Fig. 1A) . To further determine the placental villous cell types that express mRNA of ACE2, TMPRSS2, and furin, we used RT-qPCR to measure these transcripts in the four main villous cell types: primary human trophoblasts (PHT cells), primary placental fibroblasts (PPF cells), primary human placental microvascular endothelial cells (HuPECs), and placental macrophage Hofbauer cells (HBCs). Notably, we routinely culture PHT cells up to 72 h, which has been rigorously tested to ensure their differentiation to the fused multinucleated syncytiotrophoblast phenotype. Interestingly, ACE2 was clearly expressed in PHT cells, whereas expression in PPF, HuPEC, and HBC cells was below the PCR detection threshold. Likewise, TMPRSS2 was also discernibly expressed in PHT cells, but not in the other three cell types. All four main placental cell types expressed furin, with PHT cells exhibiting the highest expression levels (Fig. 1B) . These results indicate that, among the main placental villous cell types, only trophoblasts express transcripts for the three key SARS-CoV-2 entry factors. Our data also indicate that furin is ubiquitously expressed in all major cell types of human placenta villi. We used bulk RNA-seq to validate the expression of ACE2, TMPRSS2, and furin in trophoblasts (Fig. 1C ). To profile their expression levels in differentiated syncytiotrophoblasts versus progenitor cytotrophoblasts, we cultured PHT cells in two conditions that are known to hinder the differentiation of cytotrophoblasts to syncytiotrophoblasts in vitro, namely, culture in the presence of 1.5% dimethyl sulfoxide (DMSO) or in hypoxia (,1% O 2 for 48 h) (59) (60) (61) (62) . We found that both conditions resulted in a moderate, yet statistically significant, decrease of ACE2 and furin expression in cytotrophoblasts compared to differentiated syncytiotrophoblasts. Intriguingly, the expression of TMPRSS2 mRNA was significantly increased with exposure to DMSO or hypoxia, compared to the standard condition, which promotes differentiation (Fig. 1C) . The expression of ACE2 and TMPRSS2 proteins in placental villi and in trophoblasts. Because of the lower and more selective expression pattern of ACE2 and TMPRSS2, we sought to spatially validate the expression of these two transcripts in trophoblasts. As shown in Fig. 2 , we performed in situ hybridization using the highly sensitive RNAscope technology. While there were no obvious punctae using the negative-control probe, when using the ACE2 or TMPRSS2 probes, we noted prominent punctae in trophoblasts, located at the outermost layer of the placental villi (Fig. 2 , arrows in insets). These results confirmed our findings using RT-qPCR and RNA-seq, with regard to trophoblastic expression of ACE2 and TMPRSS2. To further corroborate our mRNA expression findings, we examined the expression and location of ACE2 and TMPRSS2 proteins in the placental villi and PHT cells using immunofluorescence labeling. As shown in Fig. 3A and B, ACE2 and TMPRSS2 were significantly expressed in the outer, trophoblast layer at the periphery of the placental villi. Furthermore, we used Western blotting to confirm TMPRSS2 expression in cultured PHT cells (Fig. 3C) , providing further support to the expression of SARS-CoV-2 processing proteins in human villous trophoblasts. Whereas ACE2 proteins were enriched in the apical membrane of villous trophoblasts, we noted the presence of cytoplasmic punctae in the trophoblasts. This pattern of ACE2 expression prompted us to investigate whether the ACE2 protein undergoes endocytosis in trophoblasts. To address this possibility, we analyzed the colocalization of ACE2 with the canonical early endosome marker EEA.1 in both human villous cryosections and in cultured PHT cells. We found that ACE2-and EEA.1-demarcated endosomes were expressed in villous trophoblasts, with a lower-level expression in the villous core ( Fig. 4A to D) . Importantly, cytoplasmic ACE2 punctae partially colocalized with EEA.1 in both placental villi (Fig. 4D ) and in cultured PHT cells (Fig. 4E to G), suggesting that trophoblastic ACE2 is endocytosed into early endosomes. To assess this possibility, we incubated PHT cells with an Alexa Fluor 488-conjugated ACE2 antibody that recognizes the extracellular domain of ACE2, and we examined the localization of the ACE2-antibody complex in live PHT cells. As shown in Fig. 4H , ACE2 localized to the plasma membrane and intracellular vesicles (Fig. 4I , arrows), thus directly demonstrating endocytosis of ACE2 in trophoblasts. It should be noted that both immunofluorescence and live-cell imaging revealed considerable heterogeneity in the level of ACE2 expression, pattern of localization and endocytosis within the population of PHT cells, likely due to the diverse differentiation state of cells. Overall, our results indicate that the trophoblasts at the outmost layer of the human placental villi express SARS-CoV-2 entry factors, including ACE2, TMPRSS2, and furin, and that these cells are likely capable of ACE2 endocytosis. SARS-CoV-2 pseudovirus can enter PHT cells. In light of our findings that the three key SARS-CoV-2 entry factors, ACE2, TMPRSS2, and furin, are expressed in villous trophoblasts, we assessed the susceptibility of PHT cells to entry of replication-deficient lentiviral particles pseudotyped with SARS-CoV-2 viral proteins (pvSARS-CoV-2) membrane (M), nucleocapsid (N), and envelope (E), with (SMNE) or without (MNE) the spike (S) protein. We examined the amount of intracellular lentiviral p24 capsid proteins present in pvSARS-CoV-2 viruses as a measure of cellular viral entry. As expected, we detected robust expression of p24 in HEK293T cells overexpressing ACE2 following 2 days of infection with S-protein-expressing pvSARS-CoV-2, with some decline at day 5 ( Fig. 5A , inset), confirming the S-protein dependency of pseudovirus entry. Importantly, PHT cells were infected by pvSARS-CoV-2 SMNE at a significantly higher level than by pvSARS-CoV-2 MNE (Fig. 5A) . Overall, the degree of PHT cell infection was markedly lower than that using 293T cells. We confirmed the specificity of pvSARS-CoV-2 S1 entry into PHT cells by preincubating the cells with anti-ACE2 antibody, demonstrating that this significantly reduced viral entry, measured by intracellular p24. This reduction was not observed when we used preincubation with anti-DC-SIGN, which is irrelevant for SARS-CoV-2 entry (Fig. 5B ). As expected, PHT cells infected with pvSARS-CoV-2 S-or infected with inactivated HIV with conformationally intact gp160 displayed markedly lower level of viral entry. Overall, these results demonstrate the , derived from nine independent placentas, were resolved on SDS-PAGs and transferred to PVDF membranes that were blotted in three independent Western immunoblots using a mouse anti-TMPRSS2 antibody as detailed in Materials and Methods. Note that TMPRSS2 is expressed as a full-length zymogen protein (;70 kDa) and is processed by proteolytic cleavage (89, 90) , producing three activated mature forms with a smaller size (;37 kDa), derived from two isoforms. importance of ACE2 and spike protein for pseudovirus entry into PHT cells. These results are consistent with our expression data, suggesting that human trophoblasts are permissive to pvSARS-CoV-2 entry. Several lines of evidence support our conclusion that villous placental trophoblasts from term human pregnancies express ACE2, TMPRSS2, and furin. (i) We detected the transcripts for ACE2, TMPRSS2, and furin in term placental trophoblasts by RT-qPCR and RNA-seq. (ii) We used RNAscope to visualize intracellular ACE2 and TMPRSS2 mRNA punctae in trophoblasts of the term placental villi. (iii) We identified ACE2 and TMPRSS2 proteins in the term placental villi and in cultured term PHT cells. Whereas furin is expressed in all four cell types we examined, ACE2 and TMPRSS2 are predominantly expressed in the trophoblast layer. Functionally, we demonstrated that cultured PHT cells are permissive to the entry of pvSARS-CoV-2 that express the S protein. Together, our results indicate that the maternal blood-facing villous trophoblasts express the processing proteins that enable SARS-CoV-2 cell entry. The expression level of ACE2 varies during placental development. While it is expressed in trophoblasts across the various times of gestation surveyed (45, 54, 55, 63, 64) , placental ACE2 transcript levels decline from early pregnancy to term (56, 57) . The susceptibility of early gestation trophoblasts to SARS-CoV-2 infection remains unclear, and scant data exist regarding feto-placental transmission of SARS-CoV-2 in early pregnancy (65) . Interestingly, we found that PHT cell differentiation, which is enhanced as pregnancy progresses, is associated with higher ACE2 and furin expression, but not TMPRSS2 expression. This might have added to the variable level ACE2 protein expression in trophoblasts. The implications of these findings to feto-placental susceptibility to SARS-CoV-2 infection remain to be defined. TMPRSS2 is obligatory for cleavage of the viral S2 subunit, thus exposing the fusion peptide that facilitates SARS-CoV-2 cell entry and subsequent viral replication (7, 8) . Although others have noted that the expression of TMPRSS2 might be below the detection threshold (45, 49) , using several sensitive technologies, we were able to unequivocally document TMPRSS2 mRNA and protein expression in trophoblasts, but not in other villous cell types. Placental expression of furin endopeptidase has previously been demonstrated, where it was shown to be required for proteolytic activation of two fusogenic proteins, syncytin-1 and syncytin-2, which are essential for trophoblast fusion (66) (67) (68) (69) . Unlike the closely related coronavirus SARS-CoV, the S protein of SARS-CoV-2 has evolved to harbor a proteolytic cleavage site of furin, leading to the production of two subunits, S1 and S2, in SARS-CoV-2 permissive cells (70) . Importantly, furin-mediated processing of the S protein is critical for SARS-CoV-2induced cytopathic effects on host cells, cell-cell fusion-mediated viral spread, and SARS-CoV-2 production (8, 9, 71) . Following the initial interaction of SARS-CoV-2 with its receptor, ACE2, on the plasma membrane (6), SARS-CoV-2 usurps the endocytic pathway, culminating in release of its viral genome for viral propagation and assembly of progeny virions. Blocking endocytosis impairs SARS-CoV-2 replication (70, 72) , suggesting that proper spatiotemporal trafficking and sorting of SARS-CoV-2 within the endosomal network is required for viral replication in host cells. In line with this finding, we showed that ACE2 protein was colocalized with early endosomes, suggesting that ACE2 and its interacting SARS-CoV-2 are likely to enter endocytic pathways in human trophoblasts. Notwithstanding this observation, current epidemiological data suggest that the fetoplacental unit is largely resistant to SARS-CoV-2 infection. Based on these observations, it is likely that SARS-CoV-2 enters trophoblasts, albeit at a lower level than permissive lung epithelial cells. Thereby, we infer that SARS-CoV-2 replication in the human placenta may be limited by the failure to activate postentry pathways, such as endosomal escape or the lysosomal deacidification pathways (73) . In line with this intriguing possibility, a recent report demonstrated that interferon-induced transmembrane proteins (IFITM) family member IFITM3, an endosomal protein, restricts SARS-CoV-2 replication via its amphipathic helix domain, which is intimately connected with the endocytosis pathway (74) . Several studies have associated placental histological lesions with COVID-19 infection during pregnancy. While some of these histopathological patterns, such as intervillositis with macrophage or neutrophil infiltration (46) (47) (48) , are known to accompany other placental viral infections, other lesions are part of maternal or fetal malperfusion lesions (39, 46, 47, (75) (76) (77) (78) . Whether or not these lesions are caused by SARS-CoV-2 infection or by some other process that enables SARS-CoV-2 villous entry at the site of damage or are a mere coincidence remains to be established. Current data support the notion that transplacental fetal transmission of SARS-CoV-2 infection is a rare event (12) . Our data imply that placental resistance to SARS-CoV-2 infection cannot be assumed on the basis of deficient expression of the SARS-CoV-2 attachment and processing proteins ACE2 and TMPRSS2, as suggested by Pique-Regi et al. (49) , but rather is due to post-viral-entry events. Deeper insight into trophoblastic immune response (79) and endocytic pathways will be essential to deciphering the mechanisms underlying resistance to SARS-CoV-2 infection at the maternal-fetal interface. Cell culture and reagents. The institutional review board at the University of Pittsburgh approved all placental procurement, dispersal, and experimental protocols used in these studies. PHT cells were dispersed from term placentas, using a modification of previously published trypsin-DNase-dispase/ Percoll protocols (59, 80) and maintained up to 72 h in Dulbecco modified Eagle medium (DMEM; Corning, New York, NY) containing 10% bovine growth serum (BGS; HyClone, Logan, UT) and 1% penicillin-streptomycin (Sigma-Aldrich, St. Louis, MO) at 37°C in a 5% CO 2 air atmosphere. PPF cells were isolated by means of our routine standard placental cell isolation procedures and further purified using a mouse anti-CD9 antibody (catalog no. BS3022; Bioworld, St. Louis Park, MN) conjugated to magnetic beads. PPF cell purity was validated by flow cytometry using a mouse antivimentin antibody (clone V9) (catalog no. M0725; Dako, Carpinteria, CA). PPF cells were cultured in DMEM with 10% BGS and antibiotics. HBCs were isolated after our initial trypsin and DNase isolation of PHT cells, following the technique detailed by Tang et al. (81) . The purity of HBC cells was validated using the selective marker CD163, with negative signal for CD90, expressed by PPF and other nonmacrophage cell types (81) . HuPEC cells were isolated using a previously described protocol, with modifications (82) . The identity of HuPEC cells was confirmed via flow cytometry, with positive expression of CD31, endoglin (CD105), and melanoma cell adhesion molecule (CD146) and a lack of CD90 and protein tyrosine phosphatase receptor type C (CD45). RNA extraction, cDNA synthesis, and real-time quantitative PCR (RT-qPCR) or transcriptome sequencing (RNA-seq). For mRNA analysis, total RNA was extracted using TRI reagent (Thermo Fisher, Waltham, MA) according to the manufacturer's protocol. RNA samples were further purified with on-column RNase-free DNase (Qiagen, Germantown, MD). We purchased a FirstChoice Human Total RNA Survey Panel from Thermo Fisher (catalog no. AM6000), which consisted of total RNA from various healthy human tissues, including placenta, heart, intestine, kidney, liver, lung, and testis. Total RNA was reverse transcribed using a HiCapacity cDNA synthesis kit (Thermo Fisher) according to the manufacturer's protocol. Quantitative PCR was performed using SYBR Select in the ViiA 7 system (Thermo Fisher), and results were presented as threshold cycle (C T ) values, or calculated using the 2 2DCT method (83) and normalized to the expression of the housekeeping gene glyceraldehyde 3-phosphate dehydrogenase (GAPDH). Note that GAPDH was within 1 C T value across the human tissues and villous cells. All primer sequences were checked for specificity using BLAST (Basic Local Alignment Search Tool) and are presented in Table S1 in the supplemental material. Dissociation curves were run on all reactions to ensure amplification of a single product with the appropriate melting temperature. Control samples of H 2 O were included in each qPCR experiment. RNA libraries of primary human trophoblasts were constructed by different sequencing facilities and sequenced on an Illumina next-generation instrument. After trimming, the RNA sequencing libraries were aligned to human reference genome GRCh38, using the RNA-seq alignment tool STAR (84) . The number of reads per gene was calculated for each RNA-seq library, also using STAR. We then used the negative binomial test implemented in the R package DESeq2 (85) to identify ACE2, TMPRSS2, and furin genes that were differentially expressed either between the standard and hypoxia conditions or between the standard and 1.5% DMSO conditions (see also "Statistical analysis" below). In situ hybridization of ACE2 and TMPRSS2 using RNAscope technology. Fluorescence in situ hybridization was performed using the RNAscope Fluorescent Multiplex kit (catalog no. 323135; Advanced Cell Diagnostics, Newark, CA) according to the manufacturer's instructions with minor modifications. Briefly, targeting ACE2 (catalog no. 848151-C3) and TMPRSS2 (catalog no. 470341-C1) probes were purchased from Advanced Cell Diagnostics. Paraffin-embedded sections were deparaffinized twice with xylene, each for 5 min, followed by two 100% ethanol washes for 2 min each. After that, tissues were exposed to hydrogen peroxide for 10 min, heated in kit-provided target retrieval buffer for 15 min at 100°C, and then digested by the provided proteinase plus. After digestion, the probes were added and incubated at 40°C for 2 h. After the probes were rinsed, the signal was amplified using the preamplifier reagent, followed by the addition of Opal fluorophores. The reaction was then blocked, the slides were rinsed and mounted with 49,6-diamidino-2-phenylindole (DAPI)-Vectashield (Vector labs, Burlingame, CA). Sections were imaged with a Zeiss AxioImager M2 microscope with 40Â lens and processed using Zeiss Efficient Navigation microscope software (Carl Zeiss AG, Oberkochen, Germany). Immunofluorescence of ACE2 and TMPRSS2 in the human placenta. Cryosections of placental villi were washed and then processed in 1% hydrogen peroxide for 10 min to inhibit endogenous peroxidase. The tissues were then washed, permeabilized with 0.1% Triton X-100 for 1 h, and preincubated with a blocking buffer containing 10% healthy or normal donkey serum (NDS) (catalog no. D9663; Sigma-Aldrich), 3% bovine serum albumin (BSA) (catalog no. A2153; Sigma-Aldrich), 0.1% Triton X-100 (catalog no. X100-500ML; Sigma-Aldrich) in Tris-buffered saline for 1 h at room temperature (RT), followed by incubation with primary antibodies at 4°C in phosphate-buffered saline (PBS) containing 10% NDS, 0.1% Triton X-100, and 3% BSA for 48 h. The following primary antibodies were used: ACE2 (2 mg/ ml) (ab15348; ABCAM, Cambridge, MA), TMPRSS2 (1 mg/ml) (ab109131; ABCAM), and normal rabbit IgG (2 mg/ml) for comparison to ACE2, 1 mg/ml for comparison to TMPRSS2 (sc-2027; Santa Cruz Biotechnology, Dallas, TX). After three washes in PBS, the sections were incubated with the corresponding secondary antibodies for 1 h at RT. Nuclei were stained with Hoechst 33342 (catalog no. 62249; Thermo Fisher). All sections were mounted with ProLong Gold Antifade mounting medium (catalog no. P36930; Thermo Fisher). PHT cells were fixed with freshly prepared 4% paraformaldehyde for 15 min at RT and permeabilized with 0.1% Triton X-100 in PBS containing 0.2% BSA for 3 min at RT. Immunolabeling was then performed with mouse monoclonal antibody ACE2 (E-11; 2 mg/ml) (sc-390851; Santa Cruz Biotechnology) and rabbit polyclonal antibody to EEA.1 (2 mg/ml) (AB-2900; ABCAM) followed by the corresponding secondary antibodies. The nuclei were stained with Hoechst 33342, and cells were mounted with ProLong Gold as described above. To visualize ACE2 traffic in living cells, PHT cells were incubated with Alexa Fluor 488conjugated ACE2 antibody (4 mg/ml) (sc-390851; Santa Cruz Biotechnology) for 90 min at 37°C, and livecell imaging was performed at RT. To obtain high-resolution three-dimensional images of placenta slices and PHT cells, a z stack of confocal images was acquired using a spinning-disk confocal system based on a Zeiss Axio Observer Z1 inverted fluorescence microscope, equipped with a 63Â Plan Apo PH, 1.4-numerical-aperture (NA) objective, computer-controlled spherical aberration correction unit, Yokogawa CSU-W1, Vector photomanipulation module, Photometrics Evolve (used for imaging PHT cells), and Hamamatsu Orca-Flash4.0 complementary metal oxide semiconductor (CMOS) (used for imaging of placenta sections) cameras, environmental chamber, piezo stage controller and lasers (405, 445, 488, 515, 561, and 640 nm), all controlled by SlideBook 6 software (Intelligent Imaging Innovations, Denver, CO). Typically, 20 to 30 serial two-dimensional confocal images were recorded at 400-nm intervals. Colocalization of ACE2 with EEA.1 was determined by identifying clear overlapping structures that could be followed in multiple z planes. Western immunoblotting. Cells were lysed on ice for 30 min in lysis buffer (20 mM HEPES [pH 7.5], 150 mM NaCl, 1% Triton X-100) supplemented with protease inhibitor cocktail mini EDTA-free and PhoSTOP (Roche, Mannheim, Germany) and centrifuged at 16,000 Â g at 4°C for 10 min to remove cell debris. Lysate concentrations were determined with Pierce bicinchoninic acid (BCA) protein assay kit (Thermo Fisher) using a Versa Max microplate reader (Molecular Devices, San Jose, CA). Protein samples (60 mg) were resolved on a sodium dodecyl sulfate-polyacrylamide gel (SDS-PAG), and transferred onto 0.2-mm polyvinylidene difluoride (PVDF) membrane (Bio-Rad, Hercules, CA), using standard procedures. The PVDF membranes were immunoblotted with a mouse monoclonal anti-actin antibody (MAB1501; Sigma) (diluted 1:20,000) and a mouse anti-TMPRSS2 antibody (sc-515727; Santa Cruz Biotechnology) (diluted 1:500). The blots were processed for chemiluminescence using a WesternBright Sirius kit (catalog no. K-12043-D20; Advansta, San Jose, CA) and imaged with the ChemiDoc system (Bio-Rad). Pseudotyped virus entry of primary human trophoblasts. PHT cells and 293T-ACE2 (Integral Molecular, Philadelphia, PA) were seeded in 12-well plates at a density of 1.3 Â 10 6 cells/well for PHT (five donors) and 3 Â 10 5 cells/well for 293T-ACE2. Cells were infected with pvSARS-CoV-2 lentiviruses (100 ng/ml based on the p24 content) (Virongy, Manassas, VA) containing SMNE proteins (spike, membrane, nucleocapsid, and envelope), MNE proteins, S protein only, or no SARS-CoV-2 proteins, as well as aldrithiol-inactivated HIV-1 (86) as a negative control. For antibody inhibition experiments, PHT cells were preexposed to 20 mg/ml anti-ACE2 (catalog no. AF933; R&D Systems, Minneapolis, MN ) or anti-DC-SIGN (R&D catalog no. MAB161) for 30 min at 37°C before inoculation with the viruses (1, 7). Cells were harvested at day 2 and day 5 postinfection, and the cells were washed three times with 2 ml of PBS followed by 0.25% trypsin-EDTA for 10 min to release the cells from plastic and to remove any virus adhering to them. The cells were centrifuged at 400 Â g for 5 min to pellet cells, which was lysed with 1% Triton X-100 for 15 min at 37°C and centrifuged at 2,000 Â g for 5 min. The lysate supernatant was collected, and p24 was measured in the cell lysate using a cytometric bead assay (87) . Values were adjusted to normalize for volume differences (1 ml for each fraction). Statistical analysis. The main statistical analyses were performed with Prism software (GraphPad, San Diego, CA). Statistical significance for multiple comparison was calculated by one-way analysis of variance (ANOVA) and Tukey post hoc test or by two-tailed paired or unpaired t test, where appropriate. Significance was determined as P , 0.05. Values are presented as means 6 standard deviations (SD), derived from at least three independent experiments, as indicated in each figure legend. For statistical analysis of the RNA-seq-based expression of ACE2, TMPRSS2, and furin among the various experimental groups, the P values of differential expression tests were adjusted using Benjamini and Hochberg's method (88) to control the false discovery rate. In the plots, the expression of the genes was first normalized by the median of ratio method implemented in DESeq2 (85) and then log 2 transformed. Data availability. RNA-seq data have been deposited with the NCBI BioProject, accession numbers PRJNA674312, PRJNA674329, and PRJNA674366. Supplemental material is available online only. TABLE S1, PDF file, 0.02 MB. We thank Tiffany Coon for technical assistance, Lori Rideout for assistance during manuscript preparation, and Bruce Campbell for editing. Y. Sadovsky is a consultant at Illumina, Inc. The other authors report no conflicts. HCA Lung Biological Network. 2020. SARS-CoV-2 entry factors are highly expressed in nasal epithelial cells together with innate immune genes Cytokine storm in COVID-19-immunopathological mechanisms, clinical considerations, and therapeutic approaches: the REPROGRAM Consortium Position Paper Enhanced renal immunocytochemical expression of ANG-(1-7) and ACE2 during pregnancy Enhanced expression of Ang-(1-7) during pregnancy Distribution of angiotensin-(1-7) and ACE2 in human placentas of normal and pathological pregnancies A pneumonia outbreak associated with a new coronavirus of probable bat origin SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor TMPRSS2 and furin are both essential for proteolytic activation of SARS-CoV-2 in human airway cells A multibasic cleavage site in the spike protein of SARS-CoV-2 is essential for infection of human lung cells Microbial vertical transmission during human pregnancy Congenital viral infection: traversing the uterine-placental interface COVID-NET Surveillance Team. 2020. Characteristics and maternal and birth outcomes of hospitalized pregnant women with laboratory-confirmed COVID-19 2 COVID-NET, 13 states SARS-CoV-2 infection among hospitalized pregnant women: reasons for admission and pregnancy characteristics 2 eight U.S. health care centers Obesity and mortality among patients diagnosed with COVID-19: results from an integrated health care organization Coronavirus disease 2019 infection among asymptomatic and symptomatic pregnant women: two weeks of confirmed presentations to an affiliated pair of New York City hospitals Why are pregnant women susceptible to COVID-19? An immunological viewpoint Novel Influenza A (H1N1) Pregnancy Working Group. 2009. H1N1 2009 influenza virus infection during pregnancy in the USA Pandemic influenza and pregnant women Pregnancy and perinatal outcomes of women with severe acute respiratory syndrome Infants born to mothers with severe acute respiratory syndrome Maternal and perinatal outcomes in pregnant women infected by SARS-CoV-2: a meta-analysis A systematic review of 571 pregnancies affected by COVID-19 An analysis of 38 pregnant women with COVID-19, their newborn infants, and maternal-fetal transmission of SARS-CoV-2: maternal coronavirus infections and pregnancy outcomes Infections in pregnancy with COVID-19 and other respiratory RNA virus diseases are rarely, if ever, transmitted to the fetus: experiences with coronaviruses, HPIV, hMPV RSV, and influenza Transplacental transfer of influenza virus Detectable 2019-nCoV viral RNA in blood is a strong indicator for the further clinical severity Novel Coronavirus Outbreak Research Team. 2020. Epidemiologic features and clinical course of patients infected with SARS-CoV-2 in Singapore Clinical features of patients infected with 2019 novel coronavirus in Wuhan SARS-CoV-2 asymptomatic and symptomatic patients and risk for transfusion transmission Extrapulmonary manifestations of COVID-19 Neurological aspects of SARS-CoV-2 infection: mechanisms and manifestations A comprehensive review of manifestations of novel coronaviruses in the context of deadly COVID-19 global pandemic Multiorgan and renal tropism of SARS-CoV-2 Maternal and perinatal outcomes with COVID-19: a systematic review of 108 pregnancies Evidence for and against vertical transmission for severe acute respiratory syndrome coronavirus 2 Vertical transmission of coronavirus disease 19 (COVID-19) from infected pregnant mothers to neonates: a review Effect of coronavirus disease 2019 (COVID-19) on maternal, perinatal and neonatal outcome: systematic review Clinical characteristics and intrauterine vertical transmission potential of COVID-19 infection in nine pregnant women: a retrospective review of medical records SARS-CoV-2 infection of the placenta Visualization of SARS-CoV-2 virus invading the human placenta using electron microscopy Detection of SARS-COV-2 in placental and fetal membrane samples Possible vertical transmission of SARS-CoV-2 from an infected mother to her newborn Antibodies in infants born to mothers with COVID-19 pneumonia Can SARS-CoV-2 infection be acquired in utero? More definitive evidence is needed Consistent localization of SARS-CoV-2 spike glycoprotein and ACE2 over TMPRSS2 predominance in placental villi of 15 COVID-19 positive maternal-fetal dyads Vertical transmission of coronavirus disease 2019: severe acute respiratory syndrome coronavirus 2 RNA on the fetal side of the placenta in pregnancies with coronavirus disease 2019-positive mothers and neonates at birth Transplacental transmission of SARS-CoV-2 infection SARS-CoV2 vertical transmission with adverse effects on the newborn revealed through integrated immunohistochemical, electron microscopy and molecular analyses of placenta Does the human placenta express the canonical cell entry mediators for SARS-CoV-2? Elife Activation of local chorionic villi angiotensin II levels but not angiotensin (1-7) in preeclampsia SARS-CoV-2 can infect the placenta and is not associated with specific placental histopathology: a series of 19 placentas from COVID-19-positive mothers SARS-CoV-2 entry factors: ACE2 and TMPRSS2 are expressed in periimplantation embryos and the maternal-fetal interface Expression of severe acute respiratory syndrome coronavirus 2 cell entry genes, angiotensinconverting enzyme 2 and transmembrane protease serine 2, in the placenta across gestation and at the maternal-fetal interface in pregnancies complicated by preterm birth or preeclampsia The SARS-CoV-2 receptor ACE2 expression of maternal-fetal interface and fetal organs by single-cell transcriptome study Placental barrier against COVID-19 The expression and localization of the human placental prorenin/renin-angiotensin system throughout pregnancy: roles in trophoblast invasion and angiogenesis Is pregnancy an immunological contributor to severe or controlled COVID-19 disease? A single-cell RNA expression map of human coronavirus entry factors Hypoxia limits differentiation and up-regulates expression and activity of prostaglandin H synthase 2 in cultured trophoblast from term human placenta Differentiation of human trophoblast cells in vitro is inhibited by dimethylsulfoxide Trophoblast differentiation modulates the activity of caspases in primary cultures of term human trophoblasts The timing and duration of hypoxia determine gene expression patterns in cultured human trophoblasts Single-cell RNA expression profiling of ACE2 and TMPRSS2 in the human trophectoderm and placenta Single-Cell RNA-seq identifies cell subsets in human placenta that highly expresses factors driving pathogenesis of SARS-CoV-2 Persistence of SARS-CoV-2 in the first trimester placenta leading to vertical transmission and fetal demise from an asymptomatic mother Functional characterization of the placental fusogenic membrane protein syncytin Functional characterization of the human placental fusogenic membrane protein syncytin 2 Syncytin is a captive retroviral envelope protein involved in human placental morphogenesis Genomewide screening for fusogenic human endogenous retrovirus envelopes identifies syncytin 2, a gene conserved on primate evolution Characterization of spike glycoprotein of SARS-CoV-2 on virus entry and its immune crossreactivity with SARS-CoV Furin inhibitors block SARS-CoV-2 spike protein cleavage to suppress virus production and cytopathic effects Inhibition of PIKfyve kinase prevents infection by Zaire ebolavirus and SARS-CoV-2 Altan-Bonnet N. 2020. b-Coronaviruses use lysosomes for egress instead of the biosynthetic secretory pathway Opposing activities of IFITM proteins in SARS-CoV-2 infection Placental pathology in COVID-19 Placental pathology in COVID-19 positive mothers: preliminary findings Analysis of complement deposition and viral RNA in placentas of COVID-19 patients A structured review of placental morphology and histopathological lesions associated with SARS-CoV-2 infection Sinai Immunology Review Project. 2020. Immunology of COVID-19: current state of the science Purification, characterization, and in vitro differentiation of cytotrophoblasts from human term placentae Isolation of Hofbauer cells from human term placentas with high yield and purity Estrogen receptor-beta mediates cyclooxygenase-2 expression and vascular prostanoid levels in human placental villous endothelial cells Analysis of relative gene expression data using real-time quantitative PCR and the 2(-delta delta C(T)) method STAR: ultrafast universal RNA-seq aligner Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2 Inactivation of human immunodeficiency virus type 1 infectivity with preservation of conformational and functional integrity of virion surface proteins A highly sensitive and dynamic immunofluorescent cytometric bead assay for the detection of HIV-1 p24 Controlling the false discovery rate: a practical and powerful approach to multiple testing TMPRSS2 Isoform 1 activates respiratory viruses and is expressed in viral target cells Inhibition of influenza virus infection in human airway cell cultures by an antisense peptide-conjugated morpholino oligomer targeting the hemagglutininactivating protease TMPRSS2