key: cord-0714743-kjx36r71 authors: Garcia-Flores, Valeria; Romero, Roberto; Xu, Yi; Theis, Kevin; Arenas-Hernandez, Marcia; Miller, Derek; Peyvandipour, Azam; Galaz, Jose; Levenson, Dustyn; Bhatti, Gaurav; Gershater, Meyer; Pusod, Errile; Kracht, David; Florova, Violetta; Leng, Yaozhu; Tao, Li; Faucett, Megan; Para, Robert; Hsu, Chaur-Dong; Zhang, Gary; Tarca, Adi L.; Pique-Regi, Roger; Gomez-Lopez, Nardhy title: Maternal-Fetal Immune Responses in Pregnant Women Infected with SARS-CoV-2 date: 2021-03-31 journal: Res Sq DOI: 10.21203/rs.3.rs-362886/v1 sha: 5f50a139cf002aeef83ed78fd3e21683ef03ddb4 doc_id: 714743 cord_uid: kjx36r71 Pregnant women are a high-risk population for severe/critical COVID-19 and mortality. However, the maternal-fetal immune responses initiated by SARS-CoV-2 infection, and whether this virus is detectable in the placenta, are still under investigation. Herein, we report that SARS-CoV-2 infection during pregnancy primarily induced specific maternal inflammatory responses in the circulation and at the maternal-fetal interface, the latter being governed by T cells and macrophages. SARS-CoV-2 infection during pregnancy was also associated with a cytokine response in the fetal circulation (i.e. umbilical cord blood) without compromising the cellular immune repertoire. Moreover, SARS-CoV-2 infection neither altered fetal cellular immune responses in the placenta nor induced elevated cord blood levels of IgM. Importantly, SARS-CoV-2 was not detected in the placental tissues, nor was the sterility of the placenta compromised by maternal viral infection. This study provides insight into the maternal-fetal immune responses triggered by SARS-CoV-2 and further emphasizes the rarity of placental infection. To date, over 65,000 pregnant women in the United States have been infected with 54 SARS-CoV-2 1 , the virus responsible for the coronavirus disease 2019 . During 55 pregnancy, SARS-CoV-2 infection can lead to variable outcomes, which range from 56 experiencing no symptoms to developing severe/critical disease 2,3 . Most pregnant women with 57 SARS-CoV-2 infection are asymptomatic or only experience mild symptoms 4,5 . Regardless, in 58 the first six months of the COVID-19 pandemic, it was documented that pregnant women with 59 SARS-CoV-2 were at an increased risk for hospitalization, mechanical ventilation, intensive care 60 unit admission, and preterm birth 2,3,6-8 , but rates of maternal mortality were reported to be similar 61 between pregnant and non-pregnant women 6 . More recently, it has been clearly shown that 62 pregnant women are at a high risk for severe/critical disease and mortality as well as preterm 63 birth 9-12 . Therefore, investigating host immune responses in pregnant women infected with 64 SARS-CoV-2, even if they are asymptomatic, is timely. 8 VEGF (1.56-log2 fold change), IL-5 (1.23-log2 fold change), and IL-8 (0.99-log2 fold change) 145 than those born to control mothers (Fig. 1C, Supplementary Fig. 2, Supplementary Table 2 ). 146 Such inflammatory changes in the neonates were not solely driven by the severe COVID-19 147 case. Based on an unsupervised analysis, the primary source of variability in the maternal and between SARS-CoV-2 cases and controls. In general, fetal cell types were minimally altered by 195 the presence of SARS-CoV-2 infection in the mother (Fig. 4D&E) . 196 The effects of SARS-CoV-2 on gene expression in maternal T cells from the CAM and 197 PVBP were compared to those from peripheral T cells from hospitalized COVID-19 patients 40 , 198 which we will refer to as the reference database hereafter. Maternal T-cell gene expression with SARS-CoV-2 41 ) but not in the placental tissues from women with SARS-CoV-2 infection 238 ( Supplementary Fig. 6B&C ). 239 Subsequently, we investigated the presence of viral RNA in the CAM, basal plate (BP), 240 and placental villi (PV) using RT-qPCR for the N1 and N2 viral genes ( Supplementary Fig. 7A ). SARS-CoV-2 N1 and N2 proteins were not detected in any of the placental samples from women 242 with SARS-CoV-2 infection or healthy controls ( Supplementary Fig. 7B ). Yet, in the spike-in 243 positive control, N1 and N2 RNA was detected in the CAM, BP, and PV. A sensitivity assay 244 revealed that 10 is the minimum confident copy number of viral particles detectable in the 245 placental villi using RT-qPCR ( Supplementary Fig. 7C ). Next, we determined whether the spike and nucleocapsid proteins were detected in the 247 placental tissues of women with SARS-CoV-2 infection using immunohistochemistry (Fig. 6A ). Several histological slides from the CAM, BP, and PV were included in our evaluation, 249 including negative and spike-in positive controls (Supplementary Table 4 were also included. None of the placentas from women with SARS-CoV-2 infection or controls 260 had detectable levels of N1 and N2 RNA viral genes; yet, the spike-in positive controls were 261 detected (Fig. 6F ). Collectively, these data show that SARS-CoV-2 is not detected in the placental tissues, 263 including the chorioamniotic membranes, of women infected with SARS-CoV-2. profiles of the placental tissues, we did not find evidence that the same is true for maternal 282 SARS-CoV-2 infection. This study provides evidence that, in a largely asymptomatic population, SARS-CoV-2 285 infection in pregnancy is primarily associated with maternal inflammatory responses in the 286 circulation and at the maternal-fetal interface. First, we showed that pregnant women with IgM cannot cross the placenta due to its large molecular weight, and thus the detection of this 298 immunoglobulin in the cord blood represents an acute fetal response in the clinical setting 47,48 . Therefore, the absence of detectable IgM in the cord blood suggests that vertical transmission in 300 utero of SARS-CoV-2 was unlikely to occur in our study population. In the current study, we report that pregnant women mount a mild systemic inflammatory 302 response to SARS-CoV-2, which is consistent with observations in asymptomatic non-pregnant 303 individuals with SARS-CoV-2 infection 49 . Interestingly, we found that neonates born to SARS- de-multiplexing. The fastq files were then aligned using kallisto 122 , and bustools 123 summarized 563 the cell/gene transcript counts in a matrix for each sample using the "lamanno" workflow for 564 scRNAseq. Each library was then processed using DIEM 124 to eliminate debris and empty 565 droplets. In parallel, "cellranger counts" was also used to align the scRNAseq reads using the 566 STAR 125 aligner to produce the bam files necessary for demultiplexing the individual of origin, 567 28 based on the genotype information using souporcell 126 and demuxlet 127 . We removed any 568 droplet/GEM barcode that was assigned to doublet or ambiguous cells in demuxlet or souporcell, 569 and only those cells that could be assigned a pregnancy case and maternal/fetal origin were kept. All count data matrices were then normalized and combined using the "NormalizeData," the FDR method 135 . The functions "enrichPathway", "enrichKEGG", and "gseGO" from 616 "clusterProfiler" were used to perform the ORA and GSEA analyses separately for each list of 617 genes obtained as differentially expressed for each cell type, placental compartment, and 618 maternal/fetal origin. Only results that were significant after correction were reported with q < 619 0.05 being considered statistically significant. Reactions were incubated at 25°C for 2 min followed by 50°C for 15 mins. Initial denaturation 657 was set for 2 min at 95°C followed by 45 amplification cycles at 95°C for 3 sec and 55°C for 30 were both calculated from the control group. The Z-scores were visualized as a heat map and 820 compared between SARS-CoV-2 (+) and control groups using two-sample t-tests. P-values were 821 adjusted for multiple comparisons using the false discovery rate method to obtain q-values. A q-822 value < 0.1 was considered statistically significant. The principal components (PC) of the flow 823 cytometry data were also determined, and PC1, PC2, and PC3 were plotted on a 3D scatter plot. Longitudinal analyses reveal immunological misfiring in severe COVID-927 19 Immunological and inflammatory profiles in mild and severe cases of 929 COVID-19 Longitudinal Multi-omics Analyses Identify Responses of 931 Severe COVID-19 Is Marked by a Dysregulated Myeloid 934 Impaired type I interferon activity and inflammatory responses in severe 936 COVID-19 patients What is the placenta? Placenta: the forgotten organ Single cell transcriptional signatures of the human placenta in term 942 and preterm parturition Imbalance of Regulatory and Cytotoxic SARS-CoV-2-Reactive 944 CD4(+) T Cells in COVID-19 Host-Viral Infection Maps Reveal Signatures of Severe Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) 949 Antibodies at Delivery in Women, Partners, and Newborns Impact of SARS-CoV-2 Infection on Pregnancy Outcomes: A 952 Population-Based Study An IgG-transporting Fc receptor 954 expressed in the syncytiotrophoblast of human placenta Isolation from human placenta of the IgG transporter, FcRn, and 957 localization to the syncytiotrophoblast: implications for maternal-fetal antibody transport Compromised SARS-CoV-2-specific placental antibody transfer IL-6: 993 Relevance for immunopathology of SARS-CoV-2 Interleukin-6 in pregnancy and 996 gestational disorders The role of interleukin-6 in monitoring severe case of coronavirus disease 998 2019 Serum interleukin-6 is an indicator for severity in 901 patients with 1000 -2 infection: a cohort study Lymphopenia during 1003 the COVID-19 infection: What it shows and what can be learned T-Cell Subset Counts in Peripheral Blood Can Be Used as Discriminatory 1006 Biomarkers for Diagnosis and Severity Prediction of Coronavirus Disease High levels of SARS-CoV-2-specific T cells with restricted functionality 1009 in severe courses of COVID-19 T cell responses in patients with COVID-19 Complex Immune Dysregulation in COVID-19 Patients with Severe Respiratory Failure Histologic and Immunohistochemical Evaluation of 65 Placentas from 1104 Women with Polymerase Chain Reaction-proven Severe Acute Respiratory Syndrome 1105 Coronavirus 2 (SARS-CoV-2) Infection Clinical characteristics and intrauterine vertical transmission potential of 1108 COVID-19 infection in nine pregnant women: a retrospective review of medical records Coronavirus disease 2019 in pregnant women: a report based on 116 cases No SARS-CoV-2 detected in amniotic fluid in mid-pregnancy Severe SARS-CoV-2 placenta infection can impact neonatal outcome 1115 in the absence of vertical transmission Vertical transmission of coronavirus disease 2019: a systematic 1117 review and meta-analysis Mom knows best: the universality of maternal 1120 microbial transmission A critical assessment 1123 of the "sterile womb" and "in utero colonization" hypotheses: implications for research 1124 on the pioneer infant microbiome Does the human placenta delivered at term have a microbiota? Results 1127 of cultivation, quantitative real-time PCR, 16S rRNA gene sequencing, and 1128 metagenomics Human placenta has no microbiome but can contain potential 1131 pathogens No evidence for a placental microbiome in human pregnancies at term No Consistent Evidence for Microbiota in Murine Placental and Fetal 1135 Tissues Deep microbial analysis of multiple placentas shows no evidence 1137 for a placental microbiome Evidence for Microbiota in the Placental and Fetal Tissues of Rhesus Macaques. mSphere 1140 5 Hypertension and Preeclampsia: ACOG Practice Bulletin, Number 1142 222 Sampling and Definitions of Placental Lesions: Amsterdam Placental 1144 Workshop Group Consensus Statement Acute chorioamnionitis and funisitis: definition, pathologic features, and 1147 clinical significance Classification of placental lesions Chronic inflammation of the 1152 placenta: definition, classification, pathogenesis, and clinical significance The frequency and type of placental histologic lesions in term 1155 pregnancies with normal outcome Robust relationship inference in genome-wide association studies Near-optimal probabilistic RNA-seq 1160 quantification Modular and efficient pre-processing of single-cell RNA-seq. bioRxiv Enhancing droplet-based single-nucleus RNA-seq resolution using the 1164 semi-supervised machine learning classifier DIEM STAR: ultrafast universal RNA-seq aligner Souporcell: robust clustering of single-cell RNA-seq data by genotype 1169 without reference genotypes Multiplexed droplet single-cell RNA-sequencing using natural genetic 1172 variation Normalization and variance stabilization of single-cell RNA-1174 seq data using regularized negative binomial regression Comprehensive Integration of Single-Cell Data Fast, sensitive and accurate integration of single-cell data with 1179 UMAP: Uniform Manifold Approximation and 1181 Projection for Dimension Reduction Dimensionality reduction for visualizing single-cell data using UMAP Reference-based analysis of lung single-cell sequencing reveals a 1186 transitional profibrotic macrophage Moderated estimation of fold change and dispersion 1189 for RNA-seq data with DESeq2 Controlling the false discovery rate: a practical and 1192 powerful approach to multiple testing clusterProfiler: an R package for comparing 1194 biological themes among gene clusters Gene set enrichment analysis: a knowledge-based approach for 1197 interpreting genome-wide expression profiles The STRING database in 2017: quality-controlled protein-protein 1200 association networks, made broadly accessible viruSITE-integrated database for viral genomics Changes in the lung microbiome following lung transplantation 1205 include the emergence of two distinct Pseudomonas species with distinct clinical 1206 associations Development of a dual-index sequencing strategy and curation pipeline for analyzing 1209 amplicon sequence data on the MiSeq Illumina sequencing platform DADA2: High-resolution sample inference from Illumina amplicon 1212 data R: A language and environment for statistical computing (R Foundation for Statistical 1214 Computing Simple 1216 statistical identification and removal of contaminant sequences in marker-gene and 1217 metagenomics data 1222 We thank the physicians, nurses, and research assistants from the Center for Advanced