Emerging technologies for systems vaccinology — multi-omics integration and single-cell (epi)genomic profiling

Current Opinion in Immunology - Tập 65 - Trang 57-64 - 2020
Florian Wimmers1, Bali Pulendran1,2,3,4
1Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
2Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
3Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
4Chemistry, Engineering & Medicine for Human Health, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA

Tài liệu tham khảo

Querec, 2009, Systems biology approach predicts immunogenicity of the yellow fever vaccine in humans, Nat Immunol, 10, 116, 10.1038/ni.1688 Gaucher, 2008, Yellow fever vaccine induces integrated multilineage and polyfunctional immune responses, J Exp Med, 205, 3119, 10.1084/jem.20082292 Monath, 2005, Yellow fever vaccine, Expert Rev Vaccines, 4, 553, 10.1586/14760584.4.4.553 Querec, 2006, Yellow fever vaccine YF-17D activates multiple dendritic cell subsets via TLR2,7, 8, and 9 to stimulate polyvalent immunity, J Exp Med, 203, 413, 10.1084/jem.20051720 Reif, 2009, Integrated analysis of genetic and proteomic data identifies biomarkers associated with adverse events following smallpox vaccination, Genes Immun, 10, 112, 10.1038/gene.2008.80 Vahey, 2010, Expression of genes associated with immunoproteasome processing of major histocompatibility complex peptides is indicative of protection with adjuvanted RTS,S malaria vaccine, J Infect Dis, 201, 580, 10.1086/650310 Kazmin, 2017, Systems analysis of protective immune responses to RTS,S malaria vaccination in humans, PNAS, 114, 2425, 10.1073/pnas.1621489114 van den Berg, 2017, Predicting RTS,S vaccine-mediated protection from transcriptomes in a malaria-challenge clinical trial, Front Immunol, 8, 10.3389/fimmu.2017.00557 Zak, 2012, Merck Ad5/HIV induces broad innate immune activation that predicts CD8+ T-cell responses but is attenuated by preexisting Ad5 immunity, PNAS, 109, E3503, 10.1073/pnas.1208972109 Li, 2014, Molecular signatures of antibody responses derived from a systems biology study of five human vaccines, Nat Immunol, 15, 195, 10.1038/ni.2789 Obermoser, 2013, Systems scale interactive exploration reveals quantitative and qualitative differences in response to influenza and pneumococcal vaccines, Immunity, 38, 831, 10.1016/j.immuni.2012.12.008 Li, 2017, Metabolic phenotypes of response to vaccination in humans, Cell, 169, 862, 10.1016/j.cell.2017.04.026 Qi, 2016, Defective T memory cell differentiation after varicella zoster vaccination in older individuals, PLoS Pathog, 12, 10.1371/journal.ppat.1005892 Centers for Disease Control and Prevention, 2020 Nakaya, 2011, Systems biology of vaccination for seasonal influenza in humans, Nat Immunol, 12, 786, 10.1038/ni.2067 Franco, 2013, Integrative genomic analysis of the human immune response to influenza vaccination, eLife, 2, 10.7554/eLife.00299 Furman, 2013, Apoptosis and other immune biomarkers predict influenza vaccine responsiveness, Mol Syst Biol, 9, 659, 10.1038/msb.2013.15 Tsang, 2014, Global analyses of human immune variation reveal baseline predictors of postvaccination responses, Cell, 157, 499, 10.1016/j.cell.2014.03.031 Nakaya, 2015, Systems analysis of immunity to influenza vaccination across multiple years and in diverse populations reveals shared molecular signatures, Immunity, 43, 1186, 10.1016/j.immuni.2015.11.012 Chaussabel, 2008, A modular analysis framework for blood genomics studies: application to systemic lupus erythematosus, Immunity, 29, 150, 10.1016/j.immuni.2008.05.012 Oh, 2014, TLR5-mediated sensing of gut microbiota is necessary for antibody responses to seasonal influenza vaccination, Immunity, 41, 478, 10.1016/j.immuni.2014.08.009 Hagan, 2019, Antibiotics-driven gut microbiome perturbation alters immunity to vaccines in humans, Cell, 178, 1313, 10.1016/j.cell.2019.08.010 Pulendran, 2019, Immunology taught by vaccines, Science, 366, 1074, 10.1126/science.aau6975 Guo, 2016, Bile acids control inflammation and metabolic disorder through inhibition of NLRP3 inflammasome, Immunity, 45, 802, 10.1016/j.immuni.2016.09.008 Villani, 2017, Single-cell RNA-seq reveals new types of human blood dendritic cells, monocytes, and progenitors, Science, 356, 10.1126/science.aah4573 See, 2017, Mapping the human DC lineage through the integration of high-dimensional techniques, Science, 356, 10.1126/science.aag3009 Alcántara-Hernández, 2017, High-dimensional phenotypic mapping of human dendritic cells reveals interindividual variation and tissue specialization, Immunity, 47, 10.1016/j.immuni.2017.11.001 Shalek, 2014, Single-cell RNA-seq reveals dynamic paracrine control of cellular variation, Nature, 510, 363, 10.1038/nature13437 Wimmers, 2018, Single-cell analysis reveals that stochasticity and paracrine signaling control interferon-alpha production by plasmacytoid dendritic cells, Nat Commun, 9, 10.1038/s41467-018-05784-3 Kadoki, 2017, Organism-level analysis of vaccination reveals networks of protection across tissues, Cell, 171, 398, 10.1016/j.cell.2017.08.024 Waickman, 2019, Dissecting the heterogeneity of DENV vaccine-elicited cellular immunity using single-cell RNA sequencing and metabolic profiling, Nat Commun, 10, 1, 10.1038/s41467-019-11634-7 Lau, 2017, Low CD21 expression defines a population of recent germinal center graduates primed for plasma cell differentiation, Sci Immunol, 2, 10.1126/sciimmunol.aai8153 Kazer, 2020, Integrated single-cell analysis of multicellular immune dynamics during hyperacute HIV-1 infection, Nat Med, 26, 511, 10.1038/s41591-020-0799-2 Brodin, 2015, Variation in the human immune system is largely driven by non-heritable influences, Cell, 160, 37, 10.1016/j.cell.2014.12.020 Cheung, 2018, Single-cell chromatin modification profiling reveals increased epigenetic variations with aging, Cell, 173, 10.1016/j.cell.2018.03.079 Bierne, 2012, Epigenetics and bacterial infections, Cold Spring Harb Perspect Med, 2, 10.1101/cshperspect.a010272 Martin, 2018, Environmental influences on the epigenome: exposure- associated DNA methylation in human populations, Annu Rev Public Health, 39, 309, 10.1146/annurev-publhealth-040617-014629 Allis, 2016, The molecular hallmarks of epigenetic control, Nat Rev Genet, 17, 487, 10.1038/nrg.2016.59 Klemm, 2019, Chromatin accessibility and the regulatory epigenome, Nat Rev Genet, 20, 207, 10.1038/s41576-018-0089-8 Schwartzman, 2015, Single-cell epigenomics: techniques and emerging applications, Nat Rev Genet, 16, 716, 10.1038/nrg3980 Kleinnijenhuis, 2012, Bacille Calmette-Guerin induces NOD2-dependent nonspecific protection from reinfection via epigenetic reprogramming of monocytes, Proc Natl Acad Sci U S A, 109, 17537, 10.1073/pnas.1202870109 Arts, 2018, BCG vaccination protects against experimental viral infection in humans through the induction of cytokines associated with trained immunity, Cell Host Microbe, 23, 89, 10.1016/j.chom.2017.12.010 Saeed, 2014, Epigenetic programming of monocyte-to-macrophage differentiation and trained innate immunity, Science, 345, 1251086, 10.1126/science.1251086 Higgins, 2014, Association of BCG, DTP, and measles containing vaccines with childhood mortality: systematic review, BMJ, 355 Netea, 2016, Trained immunity: a program of innate immune memory in health and disease, Science, 352, aaf1098, 10.1126/science.aaf1098 Akondy, 2017, Origin and differentiation of human memory CD8 T cells after vaccination, Nature, 552, 362, 10.1038/nature24633 Youngblood, 2017, Effector CD8 T cells dedifferentiate into long-lived memory cells, Nature, 552, 404, 10.1038/nature25144 Sen, 2016, The epigenetic landscape of T cell exhaustion, Science, 354, 1165, 10.1126/science.aae0491 Satpathy, 2018, Transcript-indexed ATAC-seq for precision immune profiling, Nat Med, 24, 580, 10.1038/s41591-018-0008-8 Satpathy, 2019, Massively parallel single-cell chromatin landscapes of human immune cell development and intratumoral T cell exhaustion, Nat Biotechnol, 37, 925, 10.1038/s41587-019-0206-z