SARS-CoV-2 infection risk assessment in the endometrium: viral infection-related gene expression across the menstrual cycle

Ismael Henarejos-Castillo, Patricia Sebastian-Leon, Almudena Devesa-Peiro, Antonio Pellicer, Patricia Diaz-Gimeno, Ismael Henarejos-Castillo, Patricia Sebastian-Leon, Almudena Devesa-Peiro, Antonio Pellicer, Patricia Diaz-Gimeno

Abstract

Objective: To determine the susceptibility of the endometrium to infection by-and thereby potential damage from-SARS-CoV-2.

Design: Analysis of SARS-Cov-2 infection-related gene expression from endometrial transcriptomic data sets.

Setting: Infertility research department affiliated with a public hospital.

Patient(s): Gene expression data from five studies in 112 patients with normal endometrium collected throughout the menstrual cycle.

Intervention(s): None.

Main outcome measure(s): Gene expression and correlation between viral infectivity genes and age throughout the menstrual cycle.

Result(s): Gene expression was high for TMPRSS4, CTSL, CTSB, FURIN, MX1, and BSG; medium for TMPRSS2; and low for ACE2. ACE2, TMPRSS4, CTSB, CTSL, and MX1 expression increased toward the window of implantation. TMPRSS4 expression was positively correlated with ACE2, CTSB, CTSL, MX1, and FURIN during several cycle phases; TMPRSS2 was not statistically significantly altered across the cycle. ACE2, TMPRSS4, CTSB, CTSL, BSG, and MX1 expression increased with age, especially in early phases of the cycle.

Conclusion(s): Endometrial tissue is likely safe from SARS-CoV-2 cell entry based on ACE2 and TMPRSS2 expression, but susceptibility increases with age. Further, TMPRSS4, along with BSG-mediated viral entry into cells, could imply a susceptible environment for SARS-CoV-2 entry via different mechanisms. Additional studies are warranted to determine the true risk of endometrial infection by SARS-CoV-2 and implications for fertility treatments.

Keywords: ACE2; COVID-19; SARS-CoV-2; coronavirus; endometrial transcriptomics.

Copyright © 2020 American Society for Reproductive Medicine. Published by Elsevier Inc. All rights reserved.

Figures

Figure 1
Figure 1
Endometrial data set integration and menstrual cycle clock. (A) Data set experiment effect and correction. Each point of the principal component analysis (PCA) plots represents endometrial gene expression of one sample and is colored by the endometrial transcriptomic data sets to which it belongs. Principal component 1 (PC1) and principal component 2 (PC2) explain the percentage of variability due to these components for each PCA. Integration of the transcriptomic studies showed a clear batch effect by the nature of each experiment (left PCA plot). After correction (right PCA plot), all genes in common between data sets were retained, amounting to a total of 13,437 genes, as no differentially expressed genes were detected between experiments. (B) Menstrual cycle effect. The samples of the integrated data sets are colored by the menstrual cycle phase to show how they are grouped by phases of the menstrual cycle. ESE = early secretory endometrium; LSE = late secretory endometrium; MSE = midsecretory endometrium; PF = proliferative phase.
Figure 2
Figure 2
Gene expression of viral infection-related genes throughout the menstrual cycle. (A) Landscape of expression changes. Genes were located depending on their relative expression against the whole set. Low, medium, and high expression thresholds correspond to 1% to 10%, 11% to 50%, and 51% to 100% categories of gene expression values of the entire integrated data set, respectively. Analysis of variance results for overall change of expression during cycle are shown for each gene. ∗P<.05; ∗∗∗P<.0001. (B) Molecular scheme of SARS-CoV-2 endometrial infection. ACE2, TMPRSS4, FURIN, and BSG are shown in plasma membrane of an endometrial cell (lower left figure). CTSL and CTSB are represented outside the cell. MX1 is shown in cytoplasm. Expression of viral genes in comparison to whole transcriptomic set is represented as arrows next to their names: up = highly expressed; down = lowly expressed. Viral genes are positioned in their schematized cell locations as stablished by GeneCards database > Localization section (release 4.14) (56). Only maximum confidence levels (5 and 4) for compartments-derived cell locations were used. Proteins were grouped considering the highest coactivation values between pairs of viral genes during the menstrual cycle, which are shown in the lower right table in the figure. Discontinuous arrow shows less evidence according to our results, given that only FURIN showed high activation with BSG and that further studies are needed to understand BSG-related mechanisms of SARS-CoV-2 entry. ESE = early secretory endometrium; LSE = late secretory endometrium; MSE = midsecretory endometrium; PF = proliferative phase.
Figure 3
Figure 3
Impact of age on viral-related infectivity gene expression throughout the menstrual cycle. (A) Effect of age on ACE2 expression. Gene expression is represented for ACE2 in each phase of the cycle according to the age of the sample analyzed. The range of age from patients involved in this study was 23 to 50 years. (B) Effect of age on viral gene expression. Pearson correlation R2 values are shown for each gene studied through of the phases of the menstrual cycle. Gray scale represents the magnitude of the correlation of increase or decrease in expression with age. High values are colored darker, and low values are colored lighter. ESE = early secretory endometrium; LSE = late secretory endometrium; MSE = midsecretory endometrium; PF = proliferative phase.
Supplemental Figure 1
Supplemental Figure 1
Flowchart of the selection of transcriptomic studies evaluating the endometrium of control patients (without any known endometrial pathology). The selection of suitable individual transcriptomic studies at Gene Expression Omnibus (GEO) and the number of individual studies excluded and remaining after each filtering step are shown (n, number of studies; N, sample size; lncRNA, long noncoding RNA).
Supplemental Figure 2
Supplemental Figure 2
Excluded samples from endometrial data sets. (A) An early secretory sample in the Bradley 2011 data set was grouped to proliferative samples at the transcriptomic level due to mistaken labeling, as progesterone levels for that sample were the same as the samples from the proliferative phase. Given the doubt, this sample was removed from further analysis. (B) After correcting a batch effect due to different sequencing protocols in the data set of Altmäe et al. (63), two outliers were detected due to different transcriptomic behaviors and consequently removed. ESE = early secretory endometrium; LSE = late secretory endometrium; MSE = midsecretory endometrium; PF = proliferative phase.
Supplemental Figure 3
Supplemental Figure 3
Statistical tests for expression changes in viral genes throughout the menstrual cycle. (A) Analysis of variance P values and pairwise t-test P adjusted values are shown for each gene through the phases of the menstrual cycle. Pairwise t-test P values were adjusted by false discovery rate (FDR). Statistically significant values (FDR <0.05) are represented in red. (B) Fold change expression calculated for each gene from each phase to the phase immediately following in endometrial progression. Proliferative to early secretory, early secretory to midsecretory, and midsecretory to late secretory increases or decreases of expression are shown for each gene. ESE = early secretory endometrium; LSE = late secretory endometrium; MSE = midsecretory endometrium; PF = proliferative phase.
Supplemental Figure 4
Supplemental Figure 4
Expression changes across menstrual cycle phases. Gene expression distributions of each viral infectivity related gene are shown for each phase of the menstrual cycle in figures A to H. Statistically significant pairwise t-test results evaluating expression changes between menstrual cycle stages is indicated as ∗P<.1; ∗∗P<.01; ∗∗∗P<.001. ESE = early secretory; LSE, late secretory; MSE = medium secretory; PF = proliferative phase.

References

    1. Zhu N., Zhang D., Wang W., Li X., Yang B., Song J., et al. A novel coronavirus from patients with pneumonia in China, 2019. N Engl J Med. 2020;382:727–733.
    1. World Health Organization Novel coronavirus (n2019): situation report 124. Available at:
    1. Huang C., Wang Y., Li X., Ren L., Zhao J., Hu Y., et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 2020;395:497–506.
    1. Shang J., Wan Y., Luo C., Ye G., Geng Q., Auerbach A., Fang L. Cell entry mechanisms of SARS-CoV-2. Proc Natl Acad Sci USA. 2020;117:11727–11734.
    1. Wang C., Horby P.W., Hayden F.G., Gao G.F. A novel coronavirus outbreak of global health concern. Lancet. 2020;395:470–473.
    1. Liu C., Zhou Q., Li Y., Garner L.V., Watkins S.P., Carter L.J., et al. Research and development on therapeutic agents and vaccines for COVID-19 and related human coronavirus diseases. ACS Cent Sci. 2020;6:315–331.
    1. Butler MJ, Barrientos RM. The impact of nutrition on COVID-19 susceptibility and long-term consequences. Brain Behav Immunol. Published online April 18, 2020. Available at: 10.1016/j.bbi.2020.04.040.
    1. Xiong T.Y., Redwood S., Prendergast B., Chen M. Coronaviruses and the cardiovascular system: acute and long-term implications. Eur Heart J. 2020;41:1798–1800.
    1. Fiorillo A., Gorwood P. The consequences of the COVID-19 pandemic on mental health and implications for clinical practice. Eur Psychiatry. 2020;63:e32.
    1. Requena A., Cruz M., Vergara V., Prados N., Galliano D., Pellicer A. A picture of the covid-19 impact on IVIRMA fertility treatment clinics in Spain and Italy. Reprod Biomed Online. 2020;41:1–5.
    1. Monteleone P.A.A., Nakano M., Lazar V., Gomes A.P., de Martin H., Bonetti T.C. A review of initial data on pregnancy during the COVID-19 out-break: implications for assisted reproductive treatments. JBRA Assist Reprod. 2020;24:219–225.
    1. Laopaiboon M., Lumbiganon P., Intarut N., Mori R., Ganchimeg T., Vogel J.P., et al. Advanced maternal age and pregnancy outcomes: a multicountry assessment. BJOG. 2014;121:49–56.
    1. Zhang JJ, Dong X, Cao YY, Yuan YD, Yang YB, Yan YQ, et al. Clinical characteristics of 140 patients infected with SARS-CoV-2 in Wuhan, China. Allergy Eur J Allergy Clin Immunol. Published online February 27, 2020. Available at: 10.1111/all.14238.
    1. Huang Y, Tu M, Wang S, Chen S, Zhou W, Chen D, et al. Clinical characteristics of laboratory confirmed positive cases of SARS-CoV-2 infection in Wuhan, China: a retrospective single center analysis. Travel Med Infect Dis. Published online February 27, 2020. Available at: 10.1016/j.tmaid.2020.101606.
    1. Nikolich-Zugich J., Knox K.S., Rios C.T., Natt B., Bhattacharya D., Fain M.J. SARS-CoV-2 and COVID-19 in older adults: what we may expect regarding pathogenesis, immune responses, and outcomes. GeroScience. 2020;42:505–514.
    1. Chen H., Guo J., Wang C., Luo F., Yu X., Zhang W., et al. Clinical characteristics and intrauterine vertical transmission potential of COVID-19 infection in nine pregnant women: a retrospective review of medical records. Lancet. 2020;395:809–815.
    1. Di Mascio D., Khalil A., Saccone G., Rizzo G., Buca D., Liberati M., et al. Outcome of coronavirus spectrum infections (SARS, MERS, COVID-19) during pregnancy: a systematic review and meta-analysis. Am J Obstet Gynecol MFM. 2020;2
    1. Gajbhiye R, Modi D, Mahale S. Pregnancy outcomes, newborn complications and maternal-fetal transmission of SARS-CoV-2 in women with COVID-19: a systematic review. medRxiv. Published online May 5, 2020. Available at: 10.1101/2020.04.11.20062356.
    1. Hoffmann M., Kleine-Weber H., Schroeder S., Krüger N., Herrler T., Erichsen S., et al. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell. 2020;181:271–280.e8.
    1. Yan R., Zhang Y., Li Y., Xia L., Guo Y., Zhou Q. Structural basis for the recognition of SARS-CoV-2 by full-length human ACE2. Science. 2020;367:1444–1448.
    1. Kai H., Kai M. Interactions of coronaviruses with ACE2, angiotensin II, and RAS inhibitors—lessons from available evidence and insights into COVID-19. Hypertens Res. 2020;43:648–654.
    1. Benigni A., Cassis P., Remuzzi G. Angiotensin II revisited: new roles in inflammation, immunology and aging. EMBO Mol Med. 2010;2:247–257.
    1. Iwai M., Horiuchi M. Devil and angel in the renin-angiotensin system: ACE-angiotensin II-AT1 receptor axis vs. ACE2-angiotensin-(1–7)-Mas receptor axis. Hypertens Res. 2009;32:533–536.
    1. Wang X, Dhindsa RS, Povysil G, Zoghbi A, Joshua E, Hostyk JA, et al. Transcriptional inhibition of host viral entry proteins as a therapeutic strategy for SARS-CoV-2. Preprints. Published online April 28, 2020. Available at: .
    1. Wang K, Chen W, Zhou Y-S, Lian J-Q, Zhang Z, Du P, et al. SARS-CoV-2 invades host cells via a novel route: CD147-spike protein. bioRxiv. Published online March 14, 2020. Available at: 10.1101/2020.03.14.988345.
    1. Ou X., Liu Y., Lei X., Li P., Mi D., Ren L., et al. Characterization of spike glycoprotein of SARS-CoV-2 on virus entry and its immune cross-reactivity with SARS-CoV. Nat Commun. 2020;11:1620.
    1. Hussain M, Jabeen N, Amanullah A, Baig AA, Aziz B, Shabbir S, et al. Structural basis of SARS-CoV-2 spike protein priming by TMPRSS2. bioRxiv. Published online April 22, 2020. Available at: 10.1101/2020.04.21.052639.
    1. Zang R, Castro MFG, McCune BT, Zeng Q, Rothlauf PW, Sonnek NM, et al. TMPRSS2 and TMPRSS4 mediate SARS-CoV-2 infection of human small intestinal enterocytes. bioRxiv. April 23, 2020. Available at: 10.1101/2020.04.21.054015.
    1. Wang Q, Qiu Y, Li JY, Zhou ZJ, Liao CH, Ge XY. A unique protease cleavage site predicted in the spike protein of the novel pneumonia coronavirus (2019-nCoV) potentially related to viral transmissibility. Virol Sin. Published online March 20, 2020. Available at: 10.1007/s12250-020-00212-7.
    1. Mei Z, Bingpeng L, Hongbin G, Xinhong W, Kaibin W, Mingxiao L, et al. Significant expression of FURIN and ACE2 on oral epithelial cells may facilitate the efficiency of 2019-nCov entry. bioRxiv. Published online April 18, 2020. Available at: 10.1101/2020.04.18.047951.
    1. Bhattacharyya C, Das C, Ghosh A, Singh AK, Mukherjee S, Majumder PP, et al. Global spread of SARS-CoV-2 subtype with spike protein mutation D614G is shaped by human genomic variations that regulate expression of TMPRSS2 and MX1 genes. bioRxiv. Published online May 5, 2020. Available at: 10.1101/2020.05.04.075911.
    1. Guan W.J., Ni Z.Y., Hu Y., Liang W.H., Ou C.Q., He J.X., et al. Clinical characteristics of coronavirus disease 2019 in China. N Engl J Med. 2020;382:1708–1720.
    1. Pan L., Mu M., Yang P., Sun Y., Wang R., Yan J., et al. Clinical characteristics of COVID-19 patients with digestive symptoms in Hubei, China. Am J Gastroenterol. 2020;115:766–773.
    1. Fan C, Li K, Ding Y, Lu WL, Wang J. ACE2 expression in kidney and testis may cause kidney and testis damage after 2019-nCoV infection. medRxiv. Published online February 13, 2020. Available at: 10.1101/2020.02.12.20022418.
    1. Wang Z., Xu X. scRNA-seq Profiling of human testes reveals the presence of the ACE2 receptor, a target for SARS-CoV-2 infection in spermatogonia, Leydig and Sertoli cells. Cells. 2020;9:920.
    1. Li M.-Y., Li L., Zhang Y., Wang X.-S. Expression of the SARS-CoV-2 cell receptor gene ACE2 in a wide variety of human tissues. Infect Dis Poverty. 2020;9:45.
    1. Zhou L, Niu Z, Jiang X, Zhang Z, Zheng Y, Wang Z, et al. 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. Published online April 10, 2020. Available at: 10.1101/2020.04.06.028522.
    1. Stanley KE, Thomas E, Leaver M, Wells D. Coronavirus disease (COVID-19) and fertility: viral host entry protein expression in male and female reproductive tissues. Fertil Steril. Published online May 8, 2020. Available at: 10.1016/j.fertnstert.2020.05.001.
    1. Noyes R.W., Hertig A.T., Rock J. Dating the endometrial biopsy. Am J Obstet Gynecol. 1975;122:262–263.
    1. Sebastian-Leon P., Garrido N., Remohí J., Pellicer A., Diaz-Gimeno P. Asynchronous and pathological windows of implantation: two causes of recurrent implantation failure. Hum Reprod. 2018;33:626–635.
    1. Barrett T., Wilhite S.E., Ledoux P., Evangelista C., Kim I.F., Tomashevsky M., et al. NCBI GEO: archive for functional genomics data sets—update. Nucleic Acids Res. 2013;41:D991–D995.
    1. Navani S. The human protein atlas. J Obstet Gynecol India. 2011;61:27–31.
    1. Uhlén M., Fagerberg L., Hallström B.M., Lindskog C., Oksvold P., Mardinoglu A., et al. Tissue-based map of the human proteome. Science. 2015;347
    1. National Center for Biotechnology Information. Available at:
    1. Moher D., Liberati A., Tetzlaff J., Altman D.G., Altman D., Antes G., et al. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. PLoS Med. 2009;6
    1. Ritchie M.E., Phipson B., Wu D., Hu Y., Law C.W., Shi W., et al. Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43:e47.
    1. Durinck S., Spellman P.T., Birney E., Huber W. Mapping identifiers for the integration of genomic datasets with the R/Bioconductor package biomaRt. Nat Protoc. 2009;4:1184–1191.
    1. Villanueva R.A.M., Chen Z.J. 2nd ed. Springer-Verlag; New York: 2019. ggplot2: Elegant graphics for data analysis.
    1. Tajti F., Kuppe C., Antoranz A., Ibrahim M.M., Kim H., Ceccarelli F., et al. A functional landscape of CKD entities from public transcriptomic data. Kidney Int Reports. 2020;5:211–224.
    1. Benjamini Y., Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Ser B. 1995;57:289–300.
    1. Freedman D., Pisani R., Purves R. 4th ed. W.W. Norton; New York: 2007. Statistics.
    1. R Core Team . R Foundation for Statistical Computing; Vienna, Austria: 2014. R: A language and environment for statistical computing. Version 3.0.1. Available at:
    1. Talbi S., Hamilton A.E., Vo K.C., Tulac S., Overgaard M.T., Dosiou C., et al. Molecular phenotyping of human endometrium distinguishes menstrual cycle phases and underlying biological processes in normo-ovulatory women. Endocrinology. 2006;147:1097–1121.
    1. Altmäe S., Koel M., Võsa U., Adler P., Suhorutšenko M., Laisk-Podar T., et al. Meta-signature of human endometrial receptivity: a meta-analysis and validation study of transcriptomic biomarkers. Sci Rep. 2017;7:10077.
    1. Sigurgeirsson B., Åmark H., Jemt A., Ujvari D., Westgren M., Lundeberg J., et al. Comprehensive RNA sequencing of healthy human endometrium at two time points of the menstrual cycle. Biol Reprod. 2016;96:24–33.
    1. Stelzer G., Rosen N., Plaschkes I., Zimmerman S., Twik M., Fishilevich S., et al. The GeneCards suite: from gene data mining to disease genome sequence analyses. Curr Protoc Bioinformatics. 2016;2016:1.30.1–1.30.33.
    1. Carson D.D. Changes in gene expression during the early to mid-luteal (receptive phase) transition in human endometrium detected by high-density microarray screening. Mol Hum Reprod. 2002;8:871–879.
    1. Meng T, Cao H, Zhang H, Kang Z, Xu D, Gong H, et al. The insert sequence in SARS-CoV-2 enhances spike protein cleavage by TMPRSS. bioRxiv. Published online February 16, 2020. Available at: 10.1101/2020.02.08.926006.
    1. Walls A.C., Park Y.J., Tortorici M.A., Wall A., McGuire A.T., Veesler D. Structure, function, and antigenicity of the SARS-CoV-2 spike glycoprotein. Cell. 2020;181:281–292.e6.
    1. Shan T., Shang W., Zhang L., Zhao C., Chen W., Zhang Y., et al. Effect of angiotensin-(1–7) and angiotensin II on the proliferation and activation of human endometrial stromal cells in vitro. Int J Clin Exp Pathol. 2015;8:8948–8957.
    1. Li X.F., Ahmed A. Dual role of angiotensin II in the human endometrium. Hum Reprod. 1996;11:95–108.
    1. Vaz-Silva J., Carneiro M.M., Ferreira M.C., Pinheiro S.V.B., Silva D.A., Silva Filho A.L., et al. The vasoactive peptide angiotensin-(1–7), its receptor Mas and the angiotensin-converting enzyme type 2 are expressed in the human endometrium. Reprod Sci. 2009;16:247–256.
    1. Jing Y, Run-Qian L, Hao-Ran W, Hao-Ran C, Ya-Bin L, Yang G, et al. Potential influence of COVID-19/ACE2 on the female reproductive system. Mol Hum Reprod. Published online May 4, 2020. Available at: 10.1093/molehr/gaaa030.
    1. Richards B, ed. COVID-19 host genetics initiative. Quebec: McGill University. Available at: .

Source: PubMed

3
S'abonner