key: cord-0842896-6cttilm8 authors: Hennighausen, Lothar; Lee, Hye Kyung title: Activation of the SARS-CoV-2 receptor Ace2 through JAK/STAT-dependent enhancers during pregnancy date: 2020-09-06 journal: Cell Rep DOI: 10.1016/j.celrep.2020.108199 sha: efd3fedf051e0fff2e5e9dc10be6678a73ff824f doc_id: 842896 cord_uid: 6cttilm8 ACE2 binds the coronavirus SARS-CoV-2 and facilitates its cellular entry. Interferons activate ACE2 expression in pneumocytes, suggesting a critical role of cytokines in SARS-CoV-2 target cells. Viral RNA was detected in breast milk in at least seven studies, raising the possibility that ACE2 is expressed in mammary tissue during lactation. Here we show that Ace2 expression in mouse mammary tissue is induced during pregnancy and lactation, which coincides with the activation of intronic enhancers. These enhancers are occupied by the prolactin-activated transcription factor STAT5 and additional regulatory factors, including Polymerase II. Deletion of Stat5a results in decommissioning of the enhancers and an 83% reduction of Ace2 mRNA. We also demonstrate that Ace2 expression increases during lactation in lung, but not in kidney and intestine. JAK/STAT components are present in a range of SARS-CoV-2 target cells opening the possibility that cytokines contribute to the viral load and extrapulmonary pathophysiology. Angiotensin-converting enzyme 2 (ACE2), the receptor for SARS-CoV (Imai et al., 2005) and SARS-CoV-2 (Hoffmann et al., 2020) , has been detected in a range of target cells, including absorptive enterocytes (Lamers et al., 2020) , colon organoids (Stanifer et al., 2020) , small intestine and colonocytes (Lee et al., 2020) , secretory goblet cells (Zhao, 2020 ), the olfactory system (Brann, 2020) and several epithelial cell types (Brann, 2020; J o u r n a l P r e -p r o o f Lukassen et al., 2020; Qi et al., 2020) . While SARS-CoV-2 infection of lung epithelium is a critical driver of disease, extrapulmonary manifestations of COVID-19 infection (Gupta et al., 2020) has been associated with direct viral damage of tissues that carry the ACE2 receptor, such as intestinal enterocytes and renal tissue (Monteil et al., 2020) . Deciphering the regulation of the ACE2 gene in SARS-CoV-2 target cells is a step forward in linking ACE2 levels with viral damage and COVID-19 pathology. This is relevant not only in the context of cytokine storms but also in patients with different physiological conditions, such as pregnancy and lactation. Although a body of work has focused on the pathology of COVID-19 during pregnancy (Khalil et al., 2020) , the extent to which hormones control ACE2 expression during pregnancy and lactation, and possibly the susceptibility of cells to SARS-CoV-2 infection, has not been investigated. One hallmark of pregnancy is the development of functional mammary tissue that produces large quantities of milk during lactation. Both mammary development and milk production are governed by the cytokine prolactin and the downstream Janus kinase (JAK)/STAT signaling pathway (Liu et al., 1997) . The Signal Transducer and Activator of Transcription (STAT) 5 is activated by prolactin and is essential for both mammary development and milk production. Its role in the activation of transcriptional enhancers during pregnancy is well established (Lee et al., 2018; Yamaji et al., 2013) . SARS-CoV-2 RNA has been detected in breast milk of infected individuals (Bastug et al., 2020; Buonsenso et al., 2020; Costa et al., 2020; Gross et al., 2020; Kirtsman et al., 2020; Tam et al., 2020; Wu et al., 2020) , suggesting the possibility of vertical transmission. However, the presence of ACE2 in mammary tissue and its regulation during pregnancy has not been investigated. Since interferons can activate ACE2 expression in pneumocytes (Ziegler et al., 2020) , the question arose whether other cytokines can regulate ACE2 in mammary cells through the JAK/STAT pathway. In addition, the widespread expression of JAK/STAT components and associated receptors, their overlapping activities and potential redundancy might have an impact on ACE2 expression in a range of epithelial cell types. Based on the presence of SARS-CoV-2 RNA in milk, we have now investigated the presence and regulation of Ace2 in mammary tissue throughout pregnancy and lactation in mice. Specifically, we have J o u r n a l P r e -p r o o f asked whether the hormonal milieu results in the activation of enhancer structures that induce Ace2 expression and the genetic role of the transcription factor STAT5 during pregnancy and lactation. Expression of the ACE2 gene in type II pneumocytes is activated by interferons (Ziegler et al., 2020) , opening the possibility that the cytokine storm in COVID-19 patients, and peptide hormones in general, might lead to increased levels of ACE2 in a range of putative SARS-CoV-2 target tissues. This in turn could result in elevated viral load and subsequent tissue damage. To explore the possibility that Ace2 gene expression is regulated by cytokines through the family of STAT transcription factors, we initially mined scRNA-seq data (Ziegler et al., 2020) . The abundant presence of interferon receptors (IFNAR) and its downstream mediators JAK1, JAK2, TYK2 as well as STATs 1, 3 and 5 supports a pivotal contribution of the JAK/STAT pathway in the activation of ACE2 by IFN-α/β and IFN-γ. STAT1 levels are increased sharply in cells treated with IFNs, further supporting the notion that an autoregulatory loop (Yuasa and Hijikata, 2016 ) is needed to activate interferon target genes. The presence of a wide range of cytokine receptors and JAK/STAT components suggests that ACE2 might be activated by a broad selection of cytokines, including prolactin, which controls mammary development and other physiological parameters during pregnancy and lactation. In a first step, we determined whether Ace2 expression in lung, kidney and intestine, well-established SARS-CoV-2 target tissues, is regulated during pregnancy and lactation in female mice ( Figure 1A ). While Ace2 levels in kidney and intestine were equivalent between non-parous and day ten lactating (L10) mice, a 2-fold increase was observed in lung tissue, which harbors the prolactin receptor and all necessary downstream signaling components. Following up on reports of SARS-CoV-2 RNA in milk, we explored Ace2 expression in mammary tissue. Ace2 mRNA was present in mammary tissue and an approximately 13-fold increase was observed during lactation ( Figure 1A ). Gene expression in mammary tissue during pregnancy and lactation is activated by prolactin through STAT5 (Liu et al., 1997) and increased J o u r n a l P r e -p r o o f expression of Stat5a during lactation is the result of an autoregulatory enhancer (Metser et al., 2016) . Next, we mined RNA-seq data from our lab and demonstrated increased Ace2 expression throughout pregnancy and lactation ( Figure 1B ) with a pattern similar to that of other prolactin-regulation genes (Lee et al., 2018; Yamaji et al., 2013) . In concordance, ACE2 protein levels increased during pregnancy and lactation ( Figure 1C ). In contrast, expression of the ACE2-associated serine protease Tmprss2 and the novel putative SARS-CoV-2 receptor neuropilin-1 (Nrp1) was not further induced, suggesting that they are not under overt control of the JAK/STAT pathway. Surfactant Protein D (SFTPD) is a secreted protein expressed in both lung tissue and mammary tissue, and its gene is induced during pregnancy and lactation ( Figure 1B ). Expression of Stat5a, a key driver of prolactin signaling in mammary tissue, increases during pregnancy and lactation ( Figure 1D ). The Ace2 expression pattern during pregnancy and lactation mirrored that of mammaryspecific prolactin-regulated genes, suggesting a key role of STAT5 in its regulation. To explore this further, we dug deeper and analyzed ChIP-seq profiles aimed at identifying mammary regulatory elements at L10 (Figure 2 ). ChIP-seq for H3K4me1 and H3K27ac suggested the presence of several enhancers in the extended locus ( Figure 2A ), with two putative intronic enhancers in the Ace2 gene ( Figure 2B ). STAT5 binding coincided with two GAS motifs, which constitute bona fide STAT binding sites. STAT5 binding was at sequences void of H3K27Ac marks, suggesting direct transcription factor binding to histone-free areas. In addition to STAT5, co-occupancy of several other transcription factors, including the glucocorticoid receptor (GR), nuclear factor 1 B (NFIB) and mediator complex subunit 1 (MED1), was observed ( Figure 2B ). Due to the absence of bona fide binding motifs for these factors, we propose that they bind through contacting STAT5, rather than binding independently to DNA. RNA polymerase II (Pol II) occupancy further supports the validity of this regulatory region. Of note, no STAT3 occupancy was observed, suggesting a predominance of STAT5. In order to directly link the activation of the intronic enhancer to increased Ace2 expression, we analyzed ChIP-seq data sets throughout pregnancy, lactation and J o u r n a l P r e -p r o o f involution ( Figure 3 ). While signs of enhancers were detected at day six of pregnancy (p6), the complete occupation with transcription factors and activation occurred only at L10 ( Figure 3A ). Upon cessation of lactation, the enhancers were decommissioned within 24 hours (I24), similar to that observed with other pregnancy-regulated genes (Willi et al., 2016) . Notably, coinciding with the loss of STAT5 occupancy at the enhancers, limited STAT3 binding was observed, similar to that of other mammary genes (Willi et al., 2016) . The Stat1 enhancer served as a control ( Figure 3B ). While the ChIP-seq experiments strongly suggest that Ace2 expression is under the control of STAT5, experimental genetics is required to proof a direct relationship. To directly address the contribution of STAT5, we analyzed mammary tissue from two lines of Stat5 mutant mice (Yamaji et al., 2013) . One line lacked the two Stat5a alleles (Stat5a -/-; Stat5b +/+ ) and the other lacked both Stat5 genes. Of note, in lactating mammary tissues STAT5a accounts to at least 75% of total STAT5 (Yamaji et al., 2013) . In the absence of STAT5a, Ace2 expression at day one of lactation (L1) was reduced by 83% ( Figure 4A) . A similar reduction was observed in the absence of both Stat5 genes, suggesting an insignificant contribution of STAT5b. ChIP-seq experiments validated the absence of STAT5a binding in mice lacking both Stat5a alleles ( Figure 4B ). Of note, also STAT5b binding was impaired in these mutant mice, suggesting that the presence of STAT5a is required for Stat5b binding or that a specific threshold of STAT5 is needed. The Cish gene served as a control for STAT5b binding in mutant mice ( Figure 4B ). Our study directly demonstrates that the Ace2 gene is expressed in mammary tissue and activated during pregnancy and lactation through intronic enhancers built on the transcription factor STAT5. Our findings built a framework needed to assess and understand the contributions of a wide range of cytokines faced under various physiological conditions in extrapulmonary manifestations of COVID-19 (Gupta et al., 2020) . The hormonal milieu associated with pregnancy and lactation is quite unique and J o u r n a l P r e -p r o o f high levels of prolactin, a cytokine that activates the pan JAK/STAT signaling pathway, control hundreds of target genes. While these target genes have been well characterized in mammary tissue (Yamaji et al., 2013) , less is known in other cell types. Since cytokine receptors and downstream JAK/STAT components are present in a wide range of cell types, it can be predicted that pregnancy hormones have a significant, possibly cell-preferential, impact on genetic programs. Our finding that Ace2 expression was increased in lung tissue during lactation is significant, as it might place women during pregnancy and lactation at a higher risk. A retrospective study would be warranted. While SARS-CoV-2 has been detected in breast milk in at least seven studies and our research has demonstrated that its receptor ACE2 is present in mammary tissue and highly induced during lactation, the impact of these findings on COVID-19 requires further investigations. Interrogating the mechanism of Ace2 regulation observed in mammary tissue during pregnancy through the use of cell lines will likely be futile since cell lines do not mimic the complexity of functional mammary tissue. The use of primary tissue and organoids from pregnant women cannot be pursued. Vertical transmission of SARS-CoV-2 through breast feeding (Lackey et al., 2020) would depend on the infectivity of the virus in milk and upon passing through the gastrointestinal tract. As human colonic organoids and gut enterocytes can be productively infected in vitro (Lamers et al., 2020; Stanifer et al., 2020) , the transmission of milk-borne SARS-CoV-2 to the infant needs further examination. Clearly, other viruses are vertically transmitted through milk (Reid et al., 1984) . Based on a pneumocyte study (Ziegler et al., 2020) and our results, we predict that cytokines regulate ACE2 levels in a range of SARS-CoV-2 target cells by drawing on JAK kinases and STAT transcription factors. While the impact on virus-induced nonpulmonary pathology remains to be determined, the effectiveness of JAK/STAT pathway inhibitors in mitigating ACE2 levels needs to be evaluated. Interfering with individual STATs result in the compensational recruitment of other STAT members to cytokine receptors (Cui et al., 2007) with all its transcriptional consequences (Hennighausen and Robinson, 2008) . In contrast, JAK inhibitors could prove effective. They are used to suppress cytokine storms induced by the pan JAK/STAT pathway and J o u r n a l P r e -p r o o f inflammation in COVID-19 patients treated with the JAK1/2 inhibitor ruxolitinib is reduced (La Rosee et al., 2020) . Similarly, Baricitinib, which inhibits the proinflammatory signal of several cytokines by suppressing JAK1/JAK2, has a beneficial impact in COVID-19 patients (Cantini et al., 2020) . Future investigations aimed at understanding the mechanism of ACE2 gene regulation in SARS-CoV-2 target tissues, such as kidney and intestine, need to address the entire range of cytokines and all components of pan JAK-STAT pathway. The presence of JAK/STAT components and their respective receptors are likely not sufficient to activate Ace2 expression, as shown in kidney and intestine of this study, and additional cell-specific transcription factors and receptors might be required. Candidates are mammary-enriched transcription factors NFIB, ELF5 and GR. Based on our study and previous data (Ziegler et al., 2020) it is likely that interferons and prolactin can activate Ace2 expression through STAT1 and STAT5. By including steroid hormones and males and females of different ages in future studies, insight into the sex differences seen in COVID-19 morbidity and mortality (Galbadage et al., 2020) Further information and requests for resources and reagents should be directed to and will be fulfilled by the Lead Contact Lothar Hennighausen (lotharh@nih.gov) . This study did not generate new unique reagents. RNA-seq data shown in Figure 1B LGP-17. Two-month-old C57BL/6 female mice (Charles River) were bred and the mammary gland tissue was harvested at days 6, 13 and 19 of pregnancy (p6, p13 and p18), at day 1 and 10 (L1 and L10) after parturition, and after 24 hours of involution (I24). For the I24 time point, pups were separated from lactating dams at day 10 of lactation and tissue was harvested after 24 hours. Quality filtering and alignment of the raw reads was done using Trimmomatic (Bolger et al., 2014) (version 0.36) and Bowtie (Langmead et al., 2009) mRNA-seq read quality control was done using Trimmomatic (Bolger et al., 2014) (version 0.36) and STAR RNA-seq (Dobin et al., 2013) (version STAR 2.5.4a) using 50bp paired-end mode was used to align the reads (mm10). HTSeq was to retrieve the raw counts and subsequently, R (https://www.R-project.org/), Bioconductor and DESeq2 (Love et al., 2014) were used. Additionally, the RUVSeq (Risso et al., 2014) package was applied to remove confounding factors. The data were pre-filtered keeping only those genes, which have at least ten reads in total. Genes were categorized as significantly differentially expressed with an adjusted p-value (pAdj) below 0.05 and a fold change > 2 for upregulated genes and a fold change of < -2 for down-regulated ones. The visualization was done using dplyr (https://CRAN.R-project.org/package=dplyr) and ggplot2 (Wickham, 2009 ). Proteins (100 µg) from mouse mammary tissues were extracted with lysis buffer (50 mM Tris-Cl pH 8.0, 150 mM NaCl, 0.5% Na-DOC, 1% NP-40, 0.1% SDS, 5 mM EDTA, 1 mM PMSF, and protease inhibitor cocktail), separated on a 4-12% NuPage gradient gel (Invitrogen) and transferred to a PVDF membrane (Invitrogen). Membranes were blocked for 1 h with 5% nonfat dry milk in PBS-T buffer (PBS containing 0.1% Tween 20) and incubated for 1.5 hr at 4°C with the primary antibody against ACE2 (Proteintech, 21115-1-AP) and GAPDH (Cell signaling, #5174). After washing, membranes were incubated for 1 h with HRP-conjugated secondary antibodies (Cell signaling). Labeled protein bands were detected using an enhanced chemiluminescence system (Thermo scientific) and Amersham Imager 600 (GE healthcare). Band density was analyzed using this imager. Mammary tissues were harvested from non-parous and day 10 of lactating female mice and homogenized using an electronic homogenizer. RNA was extracted using the PureLink RNA Mini Kit (Invitrogen) according to the manufacturer's instructions. cDNA was synthesized from total RNA using Superscript II (Invitrogen). Quantitative real-time PCR (qRT-PCR) was performed using TaqMan probes (Ace2, Mm01159006; Stat5a, Mm03053818; Stat5b, Mm00839889; Cish, Mm01230623, Thermo Fisher scientific; mouse Gapdh, 4352339E, Applied Biosystems) on the CFX384 Real-Time PCR Detection System (Bio-Rad) according to the manufacturer's instructions. PCR conditions were 95°C for 30s, 95°C for 15s, and 60°C for 30s for 40 cycles. All reactions were done in triplicate and normalized to the housekeeping gene Gapdh. Relative differences in PCR results were calculated using the comparative cycle threshold (C T ) method. For comparison of samples, data were presented as standard deviation in each group and were evaluated with a t-test and ANOVA multiple comparisons using PRISM J o u r n a l P r e -p r o o f GraphPad. Statistical significance was obtained by comparing the measures from wildtype or control group, and each mutant group. A value of *P < 0.05, **P < 0.001, ***P < 0.0001, ****P < 0.00001 was considered statistically significant. HTSeq--a Python framework to work with high-throughput sequencing data Virolactia in an Asymptomatic Mother with COVID-19 Trimmomatic: a flexible trimmer for Illumina sequence data Non-neuronal expression of SARS-CoV-2 entry genes in the olfactory system suggests mechanisms underlying COVID-19-associated anosmia Neonatal Late Onset Infection with Severe Acute Respiratory Syndrome Coronavirus 2 Beneficial impact of Baricitinib in COVID-19 moderate pneumonia; multicentre study Excretion of STAR: ultrafast universal RNA-seq aligner Systematic Review and Meta-Analysis of Sex-Specific COVID-19 Clinical Outcomes Detection of SARS-CoV-2 in human breastmilk Extrapulmonary manifestations of COVID-19 Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities Interpretation of cytokine signaling through the transcription factors STAT5A and STAT5B SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor Orchestrating high-throughput genomic analysis with Bioconductor Angiotensin-converting enzyme 2 protects from severe acute lung failure SARS-CoV-2 infection in pregnancy: A systematic review and meta-analysis of clinical features and pregnancy outcomes Probable congenital SARS-CoV-2 infection in a neonate born to a woman with active SARS-CoV-2 infection The Janus kinase 1/2 inhibitor ruxolitinib in COVID-19 with severe systemic hyperinflammation SARS-CoV-2 and human milk: What is the evidence? Matern Child Nutr SARS-CoV-2 productively infects human gut enterocytes Ultrafast and memoryefficient alignment of short DNA sequences to the human genome Progressing super-enhancer landscape during mammary differentiation controls tissue-specific gene regulation Relative Abundance of SARS-CoV-2 Entry Genes in the Enterocytes of the Lower Gastrointestinal Tract Stat5a is mandatory for adult mammary gland development and lactogenesis Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2 SARS-CoV-2 receptor ACE2 and TMPRSS2 are primarily expressed in bronchial transient secretory cells An autoregulatory enhancer controls mammaryspecific STAT5 functions Infections in Engineered Human Tissues Using Clinical-Grade Soluble Human ACE2 Single cell RNA sequencing of 13 human tissues identify cell types and receptors of human coronaviruses Transmission of louping-ill virus in goat milk Normalization of RNA-seq data using factor analysis of control genes or samples Critical Role of Type III Interferon in Controlling SARS-CoV-2 Infection in Human Intestinal Epithelial Cells Detectable severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in human breast milk of a mildly symptomatic patient with coronavirus disease 2019 (COVID-19) Integrative Genomics Viewer (IGV): high-performance genomics data visualization and exploration Ggplot2 : elegant graphics for data analysis Differential cytokine sensitivities of STAT5-dependent enhancers rely on Stat5 autoregulation Coronavirus disease 2019 among pregnant Chinese women: Case series data on the safety of vaginal birth and breastfeeding Single-cell RNA expression profiling of ACE2, the receptor of SARS-CoV-2 SARS-CoV-2 Receptor ACE2