Integration of human organoids single-cell transcriptomic profiles and human genetics repurposes critical cell type-specific drug targets for severe COVID-19

Yunlong Ma, Yijun Zhou, Dingping Jiang, Wei Dai, Jingjing Li, Chunyu Deng, Cheng Chen, Gongwei Zheng, Yaru Zhang, Fei Qiu, Haojun Sun, Shilai Xing, Haijun Han, Jia Qu, Nan Wu, Yinghao Yao, Jianzhong Su

PDF
Cell Proliferation ›› 2024, Vol. 57 ›› Issue (3) : e13558. DOI: 10.1111/cpr.13558
ORIGINAL ARTICLE

Integration of human organoids single-cell transcriptomic profiles and human genetics repurposes critical cell type-specific drug targets for severe COVID-19

Author information +
History +

Abstract

Human organoids recapitulate the cell type diversity and function of their primary organs holding tremendous potentials for basic and translational research. Advances in single-cell RNA sequencing (scRNA-seq) technology and genome-wide association study (GWAS) have accelerated the biological and therapeutic interpretation of trait-relevant cell types or states. Here, we constructed a computational framework to integrate atlas-level organoid scRNA-seq data, GWAS summary statistics, expression quantitative trait loci, and gene–drug interaction data for distinguishing critical cell populations and drug targets relevant to coronavirus disease 2019 (COVID-19) severity. We found that 39 cell types across eight kinds of organoids were significantly associated with COVID-19 outcomes. Notably, subset of lung mesenchymal stem cells increased proximity with fibroblasts predisposed to repair COVID-19-damaged lung tissue. Brain endothelial cell subset exhibited significant associations with severe COVID-19, and this cell subset showed a notable increase in cell-to-cell interactions with other brain cell types, including microglia. We repurposed 33 druggable genes, including IFNAR2, TYK2, and VIPR2, and their interacting drugs for COVID-19 in a cell-type-specific manner. Overall, our results showcase that host genetic determinants have cellular-specific contribution to COVID-19 severity, and identification of cell type-specific drug targets may facilitate to develop effective therapeutics for treating severe COVID-19 and its complications.

Cite this article

Download citation ▾
Yunlong Ma, Yijun Zhou, Dingping Jiang, Wei Dai, Jingjing Li, Chunyu Deng, Cheng Chen, Gongwei Zheng, Yaru Zhang, Fei Qiu, Haojun Sun, Shilai Xing, Haijun Han, Jia Qu, Nan Wu, Yinghao Yao, Jianzhong Su. Integration of human organoids single-cell transcriptomic profiles and human genetics repurposes critical cell type-specific drug targets for severe COVID-19. Cell Proliferation, 2024, 57(3): e13558 https://doi.org/10.1111/cpr.13558

References

[1]
Wu Z, McGoogan JM. Characteristics of and important lessons from the coronavirus disease 2019 (COVID-19) outbreak in China: summary of a report of 72314 cases from the Chinese Center for Disease Control and Prevention. Jama. 2020;323:1239-1242.
[2]
Xu L, Ma Y, Yuan J, et al. COVID-19 quarantine reveals that behavioral changes have an effect on myopia progression. Ophthalmology. 2021;128(11):1652-1654.
[3]
Zhang B, Zhang Z, Koeken VA, et al. Altered and allele-specific open chromatin landscape reveals epigenetic and genetic regulators of innate immunity in COVID-19. Cell Genomics. 2023;3(2):100232.
[4]
Gaziano L, Giambartolomei C, Pereira AC, et al. Actionable druggable genome-wide mendelian randomization identifies repurposing opportunities for COVID-19. Nat Med. 2021;27(4):668-676.
[5]
Chen KG, Park K, Spence JR. Studying SARS-CoV-2 infectivity and therapeutic responses with complex organoids. Nat Cell Biol. 2021;23(8):822-833.
[6]
Hirt CK, Booij TH, Grob L, et al. Drug screening and genome editing in human pancreatic cancer organoids identifies drug-gene interactions and candidates for off-label therapy. Cell Genomics. 2022;2(2):100095.
[7]
Xiang B, Deng C, Qiu F, et al. Single cell sequencing analysis identifies genetics-modulated ORMDL3+ cholangiocytes having higher metabolic effects on primary biliary cholangitis. J Nanobiotechnol. 2021;19(1):1-21.
[8]
Li K, Ouyang M, Zhan J, Tian R. CRISPR-based functional genomics screening in human-pluripotent-stem-cell-derived cell types. Cell. Genomics. 2023;3(5):100300.
[9]
Han Y, Duan X, Yang L, et al. Identification of SARS-CoV-2 inhibitors using lung and colonic organoids. Nature. 2021;589(7841):270-275.
[10]
Luo C, Liu H, Xie F, et al. Single nucleus multi-omics identifies human cortical cell regulatory genome diversity. Cell Genomics. 2022;2(3):100107.
[11]
Monteil V, Kwon H, Prado P, et al. Inhibition of SARS-CoV-2 infections in engineered human tissues using clinical-grade soluble human ACE2. Cell. 2020;181(4):905-913. e907.
[12]
Mallard TT, Linnér RK, Grotzinger AD, et al. Multivariate GWAS of psychiatric disorders and their cardinal symptoms reveal two dimensions of cross-cutting genetic liabilities. Cell Genomics. 2022;2(6):100140.
[13]
Pairo-Castineira E, Clohisey S, Klaric L, et al. Genetic mechanisms of critical illness in COVID-19. Nature. 2021;591(7848):92-98.
[14]
Ellinghaus D, Degenhardt F, Bujanda L, et al. Genomewide association study of severe Covid-19 with respiratory failure. N Engl J Med. 2020;383(16):1522-1534.
[15]
Ma Y, Huang Y, Zhao S, et al. Integrative genomics analysis reveals a 21q22. 11 locus contributing risk to COVID-19. Hum Mol Genet. 2021;30(13):1247-1258.
[16]
Zheng J, Zhang Y, Zhao H, et al. Multi-ancestry mendelian randomization of omics traits revealing drug targets of COVID-19 severity. EBioMedicine. 2022;81:104112.
[17]
Zhou S, Butler-Laporte G, Nakanishi T, et al. A neanderthal OAS1 isoform protects individuals of European ancestry against COVID-19 susceptibility and severity. Nat Med. 2021;27(4):659-667.
[18]
Jagadeesh KA, Dey KK, Montoro DT, et al. Identifying disease-critical cell types and cellular processes by integrating single-cell RNA-sequencing and human genetics. Nat Genet. 2022;54(10):1479-1492.
[19]
Ma Y, Qiu F, Deng C, et al. Integrating single-cell sequencing data with GWAS summary statistics reveals CD16+ monocytes and memory CD8+ T cells involved in severe COVID-19. Genome Med. 2022;14(1):1-21.
[20]
Delorey TM, Ziegler CG, Heimberg G, et al. COVID-19 tissue atlases reveal SARS-CoV-2 pathology and cellular targets. Nature. 2021;595(7865):107-113.
[21]
Clough E, Barrett T. The Gene Expression Omnibus database. Methods Mol Biol. 2016;1418:93-110.
[22]
Athar A, Füllgrabe A, George N, et al. ArrayExpress update–from bulk to single-cell expression data. Nucleic Acids Res. 2019;47(D1):D711-D715.
[23]
Leinonen R, Sugawara H, Shumway M; Collaboration INSD. The sequence read archive. Nucleic Acids Res. 2010;39(Suppl 1):D19-D21.
[24]
Quinlan AR, Hall IM. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics. 2010;26(6):841-842.
[25]
Zheng GX, Terry JM, Belgrader P, et al. Massively parallel digital transcriptional profiling of single cells. Nat Commun. 2017;8(1):1-12.
[26]
Dobin A, Davis CA, Schlesinger F, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29(1):15-21.
[27]
1000 Genomes Project Consortium, Auton A, Brooks LD, Durbin RM, Garrison EP, Kang HM. A global reference for human genetic variation. Nature. 2015;526(7571):68-74.
[28]
Liao Y, Smyth GK, Shi W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics. 2013;30(7):923-930.
[29]
Wolf FA, Angerer P, Theis FJ. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 2018;19:1-5.
[30]
Butler A, Hoffman P, Smibert P, Papalexi E, Satija R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat Biotechnol. 2018;36(5):411-420.
[31]
Korsunsky I, Millard N, Fan J, et al. Fast, sensitive and accurate integration of single-cell data with harmony. Nat Methods. 2019;16(12):1289-1296.
[32]
Lotfollahi M, Naghipourfar M, Luecken MD, et al. Mapping single-cell data to reference atlases by transfer learning. Nat Biotechnol. 2022;40(1):121-130.
[33]
The COVID-19 host genetics initiative, a global initiative to elucidate the role of host genetic factors in susceptibility and severity of the SARS-CoV-2 virus pandemic. Eur J Hum Genet. 2020;28(6):715-718.
[34]
Turner SD. qqman: an R package for visualizing GWAS results using QQ and Manhattan plots. Biorxiv 2014:005165.
[35]
Ma Y, Deng C, Zhou Y, et al. Polygenic regression uncovers trait-relevant cellular contexts through pathway activation transformation of single-cell RNA sequencing data. Cell Genom. 2023;3(9):100383.
[36]
Kanehisa M, Goto S. KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 2000;28(1):27-30.
[37]
Finucane HK, Reshef YA, Anttila V, et al. Heritability enrichment of specifically expressed genes identifies disease-relevant tissues and cell types. Nat Genet. 2018;50(4):621-629.
[38]
DeTomaso D, Jones MG, Subramaniam M, Ashuach T, Ye CJ, Yosef N. Functional interpretation of single cell similarity maps. Nat Commun. 2019;10(1):4376.
[39]
Geary RC. The contiguity ratio and statistical mapping. Incorporated Stat. 1954;5(3):115-146.
[40]
Barbeira AN, Dickinson SP, Bonazzola R, et al. Exploring the phenotypic consequences of tissue specific gene expression variation inferred from GWAS summary statistics. Nat Commun. 2018;9(1):1825.
[41]
Barbeira AN, Pividori M, Zheng J, Wheeler HE, Nicolae DL, Im HK. Integrating predicted transcriptome from multiple tissues improves association detection. PLoS Genet. 2019;15(1):e1007889.
[42]
de Leeuw CA, Mooij JM, Heskes T, Posthuma D. MAGMA: generalized gene-set analysis of GWAS data. PLoS Comput Biol. 2015;11(4):e1004219.
[43]
Ma X, Wang P, Xu G, Yu F, Ma Y. Integrative genomics analysis of various omics data and networks identify risk genes and variants vulnerable to childhood-onset asthma. BMC Med Genomics. 2020;13(1):123.
[44]
Dong Z, Ma Y, Zhou H, et al. Integrated genomics analysis highlights important SNPs and genes implicated in moderate-to-severe asthma based on GWAS and eQTL datasets. BMC Pulm Med. 2020;20(1):270.
[45]
Chung NC, Miasojedow B, Startek M, Gambin A. Jaccard/Tanimoto similarity test and estimation methods for biological presence-absence data. BMC Bioinformatics. 2019;20(15):1-11.
[46]
Wang J, Duncan D, Shi Z, Zhang B. WEB-based GEne SeT AnaLysis toolkit (WebGestalt): update 2013. Nucleic Acids Res. 2013;41(Web Server issue):W77-W83.
[47]
Gene Ontology Consortium: going forward. Nucleic Acids Res. 2015;43(Database issue):D1049-D1056.
[48]
Bulik-Sullivan BK, Loh P-R, Finucane HK, et al. LD score regression distinguishes confounding from polygenicity in genome-wide association studies. Nat Genet. 2015;47(3):291-295.
[49]
Ma Y, Zhou Y, Su J. https://github.com/x-burner-ux/scDrugHunter. Github 2023.
[50]
Barbeira AN, Bonazzola R, Gamazon ER, et al. Exploiting the GTEx resources to decipher the mechanisms at GWAS loci. Genome Biol. 2021;22(1):49.
[51]
Freshour SL, Kiwala S, Cotto KC, et al. Integration of the drug–gene interaction database (DGIdb 4.0) with open crowdsource efforts. Nucleic Acids Res. 2021;49(D1):D1144-D1151.
[52]
Fang H, De Wolf H, Knezevic B, et al. A genetics-led approach defines the drug target landscape of 30 immune-related traits. Nat Genet. 2019;51(7):1082-1091.
[53]
Mountjoy E, Schmidt EM, Carmona M, et al. An open approach to systematically prioritize causal variants and genes at all published human GWAS trait-associated loci. Nat Genet. 2021;53(11):1527-1533.
[54]
Borkowski B, Wiliński A, Szczesny W, Binderman Z. Mathematical analysis of synthetic measures based on radar charts. Math Model Anal. 2020;25(3):473-489.
[55]
Jin S, Guerrero-Juarez CF, Zhang L, et al. Inference and analysis of cell-cell communication using CellChat. Nat Commun. 2021;12(1):1088.
[56]
Holloway EM, Wu JH, Czerwinski M, et al. Differentiation of human intestinal organoids with endogenous vascular endothelial cells. Dev Cell. 2020;54(4):516-528. e517.
[57]
Cruceanu C, Dony L, Krontira AC, et al. Cell-type-specific impact of glucocorticoid receptor activation on the developing brain: a cerebral organoid study. Am J Psychiatry. 2022;179(5):375-387.
[58]
Zhang MJ, Hou K, Dey KK, et al. Polygenic enrichment distinguishes disease associations of individual cells in single-cell RNA-seq data. Nat Genet. 2022;54:1572-1580.
[59]
Wang R, Lin D-Y, Jiang Y. EPIC: inferring relevant cell types for complex traits by integrating genome-wide association studies and single-cell RNA sequencing. PLoS Genet. 2022;18(6):e1010251.
[60]
Zhu R, Yan T, Feng Y, et al. Mesenchymal stem cell treatment improves outcome of COVID-19 patients via multiple immunomodulatory mechanisms. Cell Res. 2021;31(12):1244-1262.
[61]
Xu R, Feng Z, Wang F-S. Mesenchymal stem cell treatment for COVID-19. EBioMedicine. 2022;77:103920.
[62]
Ding S, Liang TJ. Is SARS-CoV-2 also an enteric pathogen with potential fecal–oral transmission? A COVID-19 virological and clinical review. Gastroenterology. 2020;159(1):53-61.
[63]
Xu E, Xie Y, Al-Aly Z. Long-term gastrointestinal outcomes of COVID-19. Nat Commun. 2023;14(1):983.
[64]
Sashindranath M, Nandurkar HH. Endothelial dysfunction in the brain: setting the stage for stroke and other cerebrovascular complications of COVID-19. Stroke. 2021;52(5):1895-1904.
[65]
Fullard JF, Lee H-C, Voloudakis G, et al. Single-nucleus transcriptome analysis of human brain immune response in patients with severe COVID-19. Genome Med. 2021;13(1):1-13.
[66]
Yuan J, Fan D, Xue Z, Qu J, Su J. Co-expression of mitochondrial genes and ACE2 in cornea involved in COVID-19. Invest Ophthalmol Vis Sci. 2020;61(12):13.
[67]
Horowitz JE, Kosmicki JA, Damask A, et al. Genome-wide analysis provides genetic evidence that ACE2 influences COVID-19 risk and yields risk scores associated with severe disease. Nat Genet. 2022;54(4):382-392.
[68]
Pathak GA, Singh K, Miller-Fleming TW, et al. Integrative genomic analyses identify susceptibility genes underlying COVID-19 hospitalization. Nat Commun. 2021;12(1):1-11.
[69]
Roberts GH, Partha R, Rhead B, et al. Expanded COVID-19 phenotype definitions reveal distinct patterns of genetic association and protective effects. Nat Genet. 2022;54(4):374-381.
[70]
Downes DJ, Cross AR, Hua P, et al. Identification of LZTFL1 as a candidate effector gene at a COVID-19 risk locus. Nat Genet. 2021;53(11):1606-1615.
[71]
Kousathanas A, Pairo-Castineira E, Rawlik K, et al. Whole-genome sequencing reveals host factors underlying critical COVID-19. Nature. 2022;607(7917):97-103.
[72]
Ma Y, Li MD. Establishment of a strong link between smoking and cancer pathogenesis through DNA methylation analysis. Sci Rep. 2017;7(1):1-13.
[73]
Menche J, Sharma A, Kitsak M, et al. Uncovering disease-disease relationships through the incomplete interactome. Science. 2015;347(6224):1257601.
[74]
Auwul MR, Rahman MR, Gov E, Shahjaman M, Moni MA. Bioinformatics and machine learning approach identifies potential drug targets and pathways in COVID-19. Brief Bioinform. 2021;22(5):bbab120.
[75]
Guan W-j, Liang W-h, Zhao Y, et al. Comorbidity and its impact on 1590 patients with COVID-19 in China: a nationwide analysis. Eur Respir J. 2020;55(5):2000547.
[76]
Zhou F, Yu T, Du R, et al. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. The Lancet. 2020;395(10229):1054-1062.
[77]
Samuel RM, Majd H, Richter MN, et al. Androgen signaling regulates SARS-CoV-2 receptor levels and is associated with severe COVID-19 symptoms in men. Cell Stem Cell. 2020;27(6):876-889. e812.
[78]
Kathiriya JJ, Wang C, Zhou M, et al. Human alveolar type 2 epithelium transdifferentiates into metaplastic KRT5+ basal cells. Nat Cell Biol. 2022;24(1):10-23.
[79]
Gulati GS, Sikandar SS, Wesche DJ, et al. Single-cell transcriptional diversity is a hallmark of developmental potential. Science. 2020;367(6476):405-411.
[80]
Qiu X, Mao Q, Tang Y, et al. Reversed graph embedding resolves complex single-cell trajectories. Nat Methods. 2017;14(10):979-982.
[81]
Rendeiro AF, Ravichandran H, Bram Y, et al. The spatial landscape of lung pathology during COVID-19 progression. Nature. 2021;593(7860):564-569.
[82]
Li H, Li X, Wu Q, et al. Plasma proteomic and metabolomic characterization of COVID-19 survivors 6 months after discharge. Cell Death Dis. 2022;13(3):1-12.
[83]
Sun J-K, Liu Y, Zou L, et al. Acute gastrointestinal injury in critically ill patients with COVID-19 in Wuhan, China. World J Gastroenterol. 2020;26(39):6087-6097.
[84]
Zang R, Castro MFG, McCune BT, et al. TMPRSS2 and TMPRSS4 promote SARS-CoV-2 infection of human small intestinal enterocytes. Sci Immunol. 2020;5(47):eabc3582.
[85]
Wilen CB, Lee S, Hsieh LL, et al. Tropism for tuft cells determines immune promotion of norovirus pathogenesis. Science. 2018;360(6385):204-208.
[86]
Baldridge MT, Nice TJ, McCune BT, et al. Commensal microbes and interferon-λ determine persistence of enteric murine norovirus infection. Science. 2015;347(6219):266-269.
[87]
Jyothula SS, Peters A, Liang Y, et al. Fulminant lung fibrosis in non-resolvable COVID-19 requiring transplantation. EBioMedicine. 2022;86:104351.
[88]
Leng L, Ma J, Zhang P-P, et al. Spatial region-resolved proteome map reveals mechanism of COVID-19-associated heart injury. Cell Rep. 2022;39(11):110955.
[89]
Melms JC, Biermann J, Huang H, et al. A molecular single-cell lung atlas of lethal COVID-19. Nature. 2021;595(7865):114-119.
[90]
Li Y, Wei L, He L, Sun J, Liu N. Interferon-induced transmembrane protein 3 gene polymorphisms are associated with COVID-19 susceptibility and severity: a meta-analysis. J Infect. 2022;84:825-833.
[91]
Rofeal M, El-Malek FA. Ribosomal proteins as a possible tool for blocking SARS-COV 2 virus replication for a potential prospective treatment. Med Hypotheses. 2020;143:109904.
[92]
Yang B, Fan J, Huang J, et al. Clinical and molecular characteristics of COVID-19 patients with persistent SARS-CoV-2 infection. Nat Commun. 2021;12(1):1-13.
[93]
Boldrini M, Canoll PD, Klein RS. How COVID-19 affects the brain. JAMA Psychiatry. 2021;78(6):682-683.
[94]
Ng J-H, Sun A, Je HS, Tan E-K. Unravelling pathophysiology of neurological and psychiatric complications of COVID-19 using brain organoids. Neuroscientist. 2021;29:30-40.
[95]
Meinhardt J, Radke J, Dittmayer C, et al. Olfactory transmucosal SARS-CoV-2 invasion as a port of central nervous system entry in individuals with COVID-19. Nat Neurosci. 2021;24(2):168-175.
[96]
Cantuti-Castelvetri L, Ojha R, Pedro LD, et al. Neuropilin-1 facilitates SARS-CoV-2 cell entry and infectivity. Science. 2020;370(6518):856-860.
[97]
Daly JL, Simonetti B, Klein K, et al. Neuropilin-1 is a host factor for SARS-CoV-2 infection. Science. 2020;370(6518):861-865.
[98]
Liddelow SA, Guttenplan KA, Clarke LE, et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature. 2017;541(7638):481-487.
[99]
Varga Z, Flammer AJ, Steiger P, et al. Endothelial cell infection and endotheliitis in COVID-19. The Lancet. 2020;395(10234):1417-1418.
[100]
Wang J, Liu J, Luo M, et al. Rational drug repositioning for coronavirus-associated diseases using directional mapping and side-effect inference. Iscience. 2022;25(11):105348.
[101]
Reay WR, Cairns MJ. Advancing the use of genome-wide association studies for drug repurposing. Nat Rev Genet. 2021;22(10):658-671.
[102]
Anderson AG, Rogers BB, Loupe JM, et al. Single nucleus multiomics identifies ZEB1 and MAFB as candidate regulators of Alzheimer's disease-specific cis-regulatory elements. Cell. Genomics. 2023;3(3):100263.
[103]
Jeong R, Bulyk ML. Blood cell traits' GWAS loci colocalization with variation in PU. 1 genomic occupancy prioritizes causal noncoding regulatory variants. Cell Genomics. 2023;3:100327.
[104]
Wu Y, Qi T, Wray NR, Visscher PM, Zeng J, Yang J. Joint analysis of GWAS and multi-omics QTL summary statistics reveals a large fraction of GWAS signals shared with molecular phenotypes. Cell Genomics. 2023;3:100344.
[105]
Hung IF-N, Lung K-C, Tso EY-K, et al. Triple combination of interferon beta-1b, lopinavir–ritonavir, and ribavirin in the treatment of patients admitted to hospital with COVID-19: an open-label, randomised, phase 2 trial. Lancet. 2020;395(10238):1695-1704.
[106]
Abdo-Cuza A, Castellanos-Gutiérrez R, Treto-Ramirez J, et al. Safety and efficacy of intranasal recombinant human interferon alfa 2b as prophylaxis for COVID-19 in patients on a hemodialysis program. J Renal Endocrinol. 2020;7(1):e05.
[107]
Dey KK, Gazal S, van de Geijn B, et al. SNP-to-gene linking strategies reveal contributions of enhancer-related and candidate master-regulator genes to autoimmune disease. Cell Genomics. 2022;2(7):100145.

RIGHTS & PERMISSIONS

2023 2023 The Authors. Cell Proliferation published by Beijing Institute for Stem Cell and Regenerative Medicine and John Wiley & Sons Ltd.
PDF

Accesses

Citations

Detail

Sections
Recommended

/