Immune cell profiling in atherosclerosis: role in research and precision medicine

Dawn M Fernandez, Chiara Giannarelli, Dawn M Fernandez, Chiara Giannarelli

Abstract

Inflammation is intimately involved at all stages of atherosclerosis and remains a substantial residual cardiovascular risk factor in optimally treated patients. The proof of concept that targeting inflammation reduces cardiovascular events in patients with a history of myocardial infarction has highlighted the urgent need to identify new immunotherapies to treat patients with atherosclerotic cardiovascular disease. Importantly, emerging data from new clinical trials show that successful immunotherapies for atherosclerosis need to be tailored to the specific immune alterations in distinct groups of patients. In this Review, we discuss how single-cell technologies - such as single-cell mass cytometry, single-cell RNA sequencing and cellular indexing of transcriptomes and epitopes by sequencing - are ideal for mapping the cellular and molecular composition of human atherosclerotic plaques and how these data can aid in the discovery of new precise immunotherapies. We also argue that single-cell data from studies in humans need to be rigorously validated in relevant experimental models, including rapidly emerging single-cell CRISPR screening technologies and mouse models of atherosclerosis. Finally, we discuss the importance of implementing single-cell immune monitoring tools in early phases of drug development to aid in the precise selection of the target patient population for data-driven translation into randomized clinical trials and the successful translation of new immunotherapies into the clinic.

Conflict of interest statement

The authors declare no competing interests.

© 2021. Springer Nature Limited.

Figures

Fig. 1. Single-cell approaches to study human…
Fig. 1. Single-cell approaches to study human atherosclerosis.
a | After atherosclerotic tissue is dissociated into single cells, the sample is analysed using three approaches: cytometry by time of flight (CyTOF), cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq) and single-cell RNA sequencing (scRNA-seq). b | CyTOF can be used to analyse the broad cell types and frequencies of immune cells across patients, with the use of unbiased approaches such as Louvain clustering. c | CITE-seq accurately integrates proteomics and gene expression signatures. d | scRNA-seq can be used to characterize phenotypically the immune cells from patients and to compare profiles of different cell types. pDCs, plasmacytoid dendritic cells.
Fig. 2. Single-cell approaches to study atherosclerotic…
Fig. 2. Single-cell approaches to study atherosclerotic tissue.
Tissue dissociation methods and in situ approaches are complementary systems to understand cell properties at the single-cell level. Several methods exist for each approach, and both approaches can be used for proteomics and transcriptomics analyses. The integration of the resulting data can be useful for the identification of molecular targets for disease therapies and subsequent drug discovery. FFPE, formalin-fixed paraffin-embedded; MIBI, multiplexed ion beam imaging.
Fig. 3. Integration of single-cell methods for…
Fig. 3. Integration of single-cell methods for the discovery and validation of drug targets.
a | Single-cell studies in humans and mice provide information about the disease. Whereas studies in humans define the actual disease state, mechanistic studies in mice can aid in the understanding of how perturbations affect the disease. Integration and cross-species validation of these studies can be used to identify novel molecular targets. Understanding these molecular pathways in large clinical cohorts can be used as validation and then secondarily validated in animal models with the use of pooled CRISPR screening. b | When new targets are identified and validated, candidate drugs can be assessed for their specific effect in modulating these pathways. One evaluation method is phosphoproteomics with cytometry by time of flight (phospho-CyTOF). In vivo testing in animals can be used to investigate further the efficacy of the drug and to evaluate the go/no-go decisions to enter clinical phases of drug development. The adoption of immune monitoring in the early phases of clinical trials can provide crucial information on patient selection and efficacy for the design of future end point-driven clinical trials.

References

    1. Moore KJ, Sheedy FJ, Fisher EA. Macrophages in atherosclerosis: a dynamic balance. Nat. Rev. Immunol. 2013;13:709–721.
    1. Moore KJ, Tabas I. Macrophages in the pathogenesis of atherosclerosis. Cell. 2011;145:341–355.
    1. Tabas I, Lichtman AH. Monocyte-macrophages and T cells in atherosclerosis. Immunity. 2017;47:621–634.
    1. Ross R. Atherosclerosis–an inflammatory disease. N. Engl. J. Med. 1999;340:115–126.
    1. Pradhan AD, Aday AW, Rose LM, Ridker PM. Residual inflammatory risk on treatment with PCSK9 inhibition and statin therapy. Circulation. 2018;138:141–149.
    1. Ridker PM, et al. Antiinflammatory therapy with canakinumab for atherosclerotic disease. N. Engl. J. Med. 2017;377:1119–1131.
    1. Tardif JC, et al. Efficacy and safety of low-dose colchicine after myocardial infarction. N. Engl. J. Med. 2019;381:2497–2505.
    1. Ridker PM, et al. Low-dose methotrexate for the prevention of atherosclerotic events. N. Engl. J. Med. 2019;380:752–762.
    1. Nidorf SM, et al. Colchicine in patients with chronic coronary disease. N. Engl. J. Med. 2020;383:1838–1847.
    1. Fiolet ATL, et al. Short-term effect of low-dose colchicine on inflammatory biomarkers, lipids, blood count and renal function in chronic coronary artery disease and elevated high-sensitivity C-reactive protein. PLoS ONE. 2020;15:e0237665.
    1. Opstal TSJ, et al. Colchicine attenuates inflammation beyond the inflammasome in chronic coronary artery disease: a LoDoCo2 proteomic substudy. Circulation. 2020;142:1996–1998.
    1. Tong DC, et al. Colchicine in patients with acute coronary syndrome: the Australian COPS randomized clinical trial. Circulation. 2020;142:1890–1900.
    1. Zhao TX, et al. Low-dose interleukin-2 in patients with stable ischaemic heart disease and acute coronary syndromes (LILACS): protocol and study rationale for a randomised, double-blind, placebo-controlled, phase I/II clinical trial. BMJ Open. 2018;8:e022452.
    1. US National Library of Medicine. (2020)
    1. Zhao TX, Mallat Z. Targeting the immune system in atherosclerosis: JACC state-of-the-art review. J. Am. Coll. Cardiol. 2019;73:1691–1706.
    1. Charo IF, Taub R. Anti-inflammatory therapeutics for the treatment of atherosclerosis. Nat. Rev. Drug Discov. 2011;10:365–376.
    1. Choudhury RP, et al. Arterial effects of canakinumab in patients with atherosclerosis and type 2 diabetes or glucose intolerance. J. Am. Coll. Cardiol. 2016;68:1769–1780.
    1. Ridker PM. From C-reactive protein to interleukin-6 to Interleukin-1: moving upstream to identify novel targets for atheroprotection. Circ. Res. 2016;118:145–156.
    1. Wolf D, Ley K. Immunity and inflammation in atherosclerosis. Circ. Res. 2019;124:315–327.
    1. Stubbington MJT, Rozenblatt-Rosen O, Regev A, Teichmann SA. Single-cell transcriptomics to explore the immune system in health and disease. Science. 2017;358:58–63.
    1. Bandura DR, et al. Mass cytometry: technique for real time single cell multitarget immunoassay based on inductively coupled plasma time-of-flight mass spectrometry. Anal. Chem. 2009;81:6813–6822.
    1. Bendall SC, et al. Single-cell mass cytometry of differential immune and drug responses across a human hematopoietic continuum. Science. 2011;332:687–696.
    1. Olsen TK, Baryawno N. Introduction to single-cell RNA sequencing. Curr. Protoc. Mol. Biol. 2018;122:e57.
    1. Stoeckius M, et al. Simultaneous epitope and transcriptome measurement in single cells. Nat. Methods. 2017;14:865–868.
    1. Fernandez DM, et al. Single-cell immune landscape of human atherosclerotic plaques. Nat. Med. 2019;25:1576–1588.
    1. Witztum JL, Lichtman AH. The influence of innate and adaptive immune responses on atherosclerosis. Annu. Rev. Pathol. 2014;9:73–102.
    1. Burke AP, Taylor A, Farb A, Malcom GT, Virmani R. Coronary calcification: insights from sudden coronary death victims. Z. Kardiol. 2000;89:49–53.
    1. Virmani R, Kolodgie FD, Burke AP, Farb A, Schwartz SM. Lessons from sudden coronary death: a comprehensive morphological classification scheme for atherosclerotic lesions. Arterioscler. Thromb. Vasc. Biol. 2000;20:1262–1275.
    1. Sheedy FJ, et al. CD36 coordinates NLRP3 inflammasome activation by facilitating intracellular nucleation of soluble ligands into particulate ligands in sterile inflammation. Nat. Immunol. 2013;14:812–820.
    1. Rajamaki K, et al. Cholesterol crystals activate the NLRP3 inflammasome in human macrophages: a novel link between cholesterol metabolism and inflammation. PLoS ONE. 2010;5:e11765.
    1. Doran AC, Yurdagul A, Jr., Tabas I. Efferocytosis in health and disease. Nat. Rev. Immunol. 2020;20:254–267.
    1. Tabas I, Bornfeldt KE. Macrophage phenotype and function in different stages of atherosclerosis. Circ. Res. 2016;118:653–667.
    1. Koelwyn GJ, Corr EM, Erbay E, Moore KJ. Regulation of macrophage immunometabolism in atherosclerosis. Nat. Immunol. 2018;19:526–537.
    1. Saigusa R, Winkels H, Ley K. T cell subsets and functions in atherosclerosis. Nat. Rev. Cardiol. 2020;17:387–401.
    1. Taleb S, et al. Loss of SOCS3 expression in T cells reveals a regulatory role for interleukin-17 in atherosclerosis. J. Exp. Med. 2009;206:2067–2077.
    1. Simon T, et al. Circulating levels of interleukin-17 and cardiovascular outcomes in patients with acute myocardial infarction. Eur. Heart J. 2013;34:570–577.
    1. Poizeau F, et al. Association between early severe cardiovascular events and the initiation of treatment with the anti-interleukin 12/23p40 antibody ustekinumab. JAMA Dermatol. 2020;156:1208–1215.
    1. Lim H, et al. Proatherogenic conditions promote autoimmune T helper 17 cell responses in vivo. Immunity. 2014;40:153–165.
    1. Sage AP, Tsiantoulas D, Binder CJ, Mallat Z. The role of B cells in atherosclerosis. Nat. Rev. Cardiol. 2019;16:180–196.
    1. Getz GS, Reardon CA. Natural killer T cells in atherosclerosis. Nat. Rev. Cardiol. 2017;14:304–314.
    1. Winkels H, Ley K. Natural killer cells at ease: atherosclerosis is not affected by genetic depletion or hyperactivation of natural killer cells. Circ. Res. 2018;122:6–7.
    1. Silvestre-Roig C, Braster Q, Ortega-Gomez A, Soehnlein O. Neutrophils as regulators of cardiovascular inflammation. Nat. Rev. Cardiol. 2020;17:327–340.
    1. Nour-Eldine W, et al. Genetic depletion or hyperresponsiveness of natural killer cells do not affect atherosclerosis development. Circ. Res. 2018;122:47–57.
    1. von Scheidt M, et al. Applications and limitations of mouse models for understanding human atherosclerosis. Cell Metab. 2017;25:248–261.
    1. Abolins S, et al. The comparative immunology of wild and laboratory mice, Mus musculus domesticus. Nat. Commun. 2017;8:14811.
    1. Kolodgie FD, et al. Localization of apoptotic macrophages at the site of plaque rupture in sudden coronary death. Am. J. Pathol. 2000;157:1259–1268.
    1. Taylor AJ, et al. A comparison of the Framingham risk index, coronary artery calcification, and culprit plaque morphology in sudden cardiac death. Circulation. 2000;101:1243–1248.
    1. Virmani R, et al. Atherosclerotic plaque progression and vulnerability to rupture: angiogenesis as a source of intraplaque hemorrhage. Arterioscler. Thromb. Vasc. Biol. 2005;25:2054–2061.
    1. Yahagi K, et al. Pathology of human coronary and carotid artery atherosclerosis and vascular calcification in diabetes mellitus. Arterioscler. Thromb. Vasc. Biol. 2017;37:191–204.
    1. Vergallo R, Crea F. Atherosclerotic plaque healing. N. Engl. J. Med. 2020;383:846–857.
    1. Melamed MR. A brief history of flow cytometry and sorting. Methods Cell Biol. 2001;63:3–17.
    1. Bonanno E, Mauriello A, Partenzi A, Anemona L, Spagnoli LG. Flow cytometry analysis of atherosclerotic plaque cells from human carotids: a validation study. Cytometry. 2000;39:158–165.
    1. Feher K, et al. Multispectral flow cytometry: the consequences of increased light collection. Cytometry A. 2016;89:681–689.
    1. Parks DR. Multispectral flow cytometry: unaddressed issues and recommendations for improvement. Cytometry A. 2020;97:1276–1277.
    1. Mazza EMC, et al. Background fluorescence and spreading error are major contributors of variability in high-dimensional flow cytometry data visualization by t-distributed stochastic neighboring embedding. Cytometry A. 2018;93:785–792.
    1. Nettey L, Giles AJ, Chattopadhyay PK. OMIP-050: A 28-color/30-parameter fluorescence flow cytometry panel to enumerate and characterize cells expressing a wide array of immune checkpoint molecules. Cytometry A. 2018;93:1094–1096.
    1. Surre J, et al. Strong increase in the autofluorescence of cells signals struggle for survival. Sci. Rep. 2018;8:12088.
    1. Njoroge JM, et al. Characterization of viable autofluorescent macrophages among cultured peripheral blood mononuclear cells. Cytometry. 2001;44:38–44.
    1. Kockx MM, et al. Phagocytosis and macrophage activation associated with hemorrhagic microvessels in human atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2003;23:440–446.
    1. Walsh AJ, et al. Classification of T-cell activation via autofluorescence lifetime imaging. Nat. Biomed. Eng. 2021;5:77–88.
    1. Laifer LI, et al. Biochemical basis for the difference between normal and atherosclerotic arterial fluorescence. Circulation. 1989;80:1893–1901.
    1. Lee MW, et al. Comprehensive intravascular imaging of atherosclerotic plaque in vivo using optical coherence tomography and fluorescence lifetime imaging. Sci. Rep. 2018;8:14561.
    1. Htun NM, et al. Near-infrared autofluorescence induced by intraplaque hemorrhage and heme degradation as marker for high-risk atherosclerotic plaques. Nat. Commun. 2017;8:75.
    1. Spitzer MH, Nolan GP. Mass cytometry: single cells, many features. Cell. 2016;165:780–791.
    1. Bjornson ZB, Nolan GP, Fantl WJ. Single-cell mass cytometry for analysis of immune system functional states. Curr. Opin. Immunol. 2013;25:484–494.
    1. Bendall SC, Nolan GP, Roederer M, Chattopadhyay PK. A deep profiler’s guide to cytometry. Trends Immunol. 2012;33:323–332.
    1. Hartmann FJ, et al. Scalable conjugation and characterization of immunoglobulins with stable mass isotope reporters for single-cell mass cytometry analysis. Methods Mol. Biol. 2019;1989:55–81.
    1. Schulz AR, Stanislawiak S, Baumgart S, Grutzkau A, Mei HE. Silver nanoparticles for the detection of cell surface antigens in mass cytometry. Cytometry A. 2017;91:25–33.
    1. Han G, et al. Atomic mass tag of bismuth-209 for increasing the immunoassay multiplexing capacity of mass cytometry. Cytometry A. 2017;91:1150–1163.
    1. Hartmann FJ, Simonds EF, Bendall SC. A universal live cell barcoding-platform for multiplexed human single cell analysis. Sci. Rep. 2018;8:10770.
    1. Lai L, Ong R, Li J, Albani SA. CD45-based barcoding approach to multiplex mass-cytometry (CyTOF) Cytometry A. 2015;87:369–374.
    1. Zunder ER, et al. Palladium-based mass tag cell barcoding with a doublet-filtering scheme and single-cell deconvolution algorithm. Nat. Protoc. 2015;10:316–333.
    1. Yao Y, et al. CyTOF supports efficient detection of immune cell subsets from small samples. J. Immunol. Methods. 2014;415:1–5.
    1. Kotecha N, Krutzik PO, Irish JM. Web-based analysis and publication of flow cytometry experiments. Curr. Protoc. Cytom. 2010 doi: 10.1002/0471142956.cy1017s53.
    1. Kimball AK, et al. A beginner’s guide to analyzing and visualizing mass cytometry data. J. Immunol. 2018;200:3–22.
    1. Liu X, et al. A comparison framework and guideline of clustering methods for mass cytometry data. Genome Biol. 2019;20:297.
    1. Meehan S, et al. Automated subset identification and characterization pipeline for multidimensional flow and mass cytometry data clustering and visualization. Commun. Biol. 2019;2:229.
    1. Palit S, Heuser C, de Almeida GP, Theis FJ, Zielinski CE. Meeting the challenges of high-dimensional single-cell data analysis in immunology. Front. Immunol. 2019;10:1515.
    1. Weber LM, Robinson MD. Comparison of clustering methods for high-dimensional single-cell flow and mass cytometry data. Cytometry A. 2016;89:1084–1096.
    1. Morris MK, Chi A, Melas IN, Alexopoulos LG. Phosphoproteomics in drug discovery. Drug Discov. Today. 2014;19:425–432.
    1. Nassar AF, Ogura H, Wisnewski AV. Impact of recent innovations in the use of mass cytometry in support of drug development. Drug Discov. Today. 2015;20:1169–1175.
    1. Glassberg J, et al. Application of phospho-CyTOF to characterize immune activation in patients with sickle cell disease in an ex vivo model of thrombosis. J. Immunol. Methods. 2018;453:11–19.
    1. Gaudilliere B, et al. Clinical recovery from surgery correlates with single-cell immune signatures. Sci. Transl. Med. 2014;6:255ra131.
    1. Nair N, et al. Single-cell immune competency signatures associate with survival in phase II GVAX and CRS-207 randomized studies in patients with metastatic pancreatic cancer. Cancer Immunol. Res. 2020;8:609–617.
    1. Huang AC, et al. T-cell invigoration to tumour burden ratio associated with anti-PD-1 response. Nature. 2017;545:60–65.
    1. Deng M, et al. Apatinib exhibits anti-leukemia activity in preclinical models of acute lymphoblastic leukemia. J. Transl. Med. 2018;16:47.
    1. Lavin Y, et al. Innate immune landscape in early lung adenocarcinoma by paired single-cell analyses. Cell. 2017;169:750–765.e17.
    1. Fluidigm. Use of CyTOF technology in clinical research trials. (2021).
    1. Trussart M, et al. Removing unwanted variation with CytofRUV to integrate multiple CyTOF datasets. eLife. 2020;9:e59630.
    1. Amir ED, et al. Development of a comprehensive antibody staining database using a standardized analytics pipeline. Front. Immunol. 2019;10:1315.
    1. Chevrier S, et al. Compensation of signal spillover in suspension and imaging mass cytometry. Cell Syst. 2018;6:612–620.e5.
    1. Geanon D, et al. A streamlined CyTOF workflow to facilitate standardized multi-site immune profiling of COVID-19 patients. Preprint at. medRxiv. 2020 doi: 10.1101/2020.06.26.20141341.
    1. Sumatoh HR, Teng KW, Cheng Y, Newell EW. Optimization of mass cytometry sample cryopreservation after staining. Cytometry A. 2017;91:48–61.
    1. Hartmann FJ, et al. Comprehensive immune monitoring of clinical trials to advance human immunotherapy. Cell Rep. 2019;28:819–831.e4.
    1. Thomas GD, et al. Human blood monocyte subsets: a new gating strategy defined using cell surface markers identified by mass cytometry. Arterioscler. Thromb. Vasc. Biol. 2017;37:1548–1558.
    1. Hamers AAJ, et al. Human monocyte heterogeneity as revealed by high-dimensional mass cytometry. Arterioscler. Thromb. Vasc. Biol. 2019;39:25–36.
    1. Cole JE, et al. Immune cell census in murine atherosclerosis: cytometry by time of flight illuminates vascular myeloid cell diversity. Cardiovasc. Res. 2018;114:1360–1371.
    1. Cochain C, et al. Single-cell RNA-Seq reveals the transcriptional landscape and heterogeneity of aortic macrophages in murine atherosclerosis. Circ. Res. 2018;122:1661–1674.
    1. Winkels H, et al. Atlas of the immune cell repertoire in mouse atherosclerosis defined by single-cell RNA-sequencing and mass cytometry. Circ. Res. 2018;122:1675–1688.
    1. Jonasson L, Holm J, Skalli O, Bondjers G, Hansson GK. Regional accumulations of T cells, macrophages, and smooth muscle cells in the human atherosclerotic plaque. Arteriosclerosis. 1986;6:131–138.
    1. Depuydt MA, et al. Microanatomy of the human atherosclerotic plaque by single-cell transcriptomics. Circ. Res. 2020;127:1437–1455.
    1. Burke AP, et al. Morphologic findings of coronary atherosclerotic plaques in diabetics: a postmortem study. Arterioscler. Thromb. Vasc. Biol. 2004;24:1266–1271.
    1. Bjorkegren JL, Kovacic JC, Dudley JT, Schadt EE. Genome-wide significant loci: how important are they? Systems genetics to understand heritability of coronary artery disease and other common complex disorders. J. Am. Coll. Cardiol. 2015;65:830–845.
    1. Choi YH, Kim JK. Dissecting cellular heterogeneity using single-cell RNA sequencing. Mol. Cell. 2019;42:189–199.
    1. Hwang B, Lee JH, Bang D. Single-cell RNA sequencing technologies and bioinformatics pipelines. Exp. Mol. Med. 2018;50:1–14.
    1. See P, Lum J, Chen J, Ginhoux F. A single-cell sequencing guide for immunologists. Front. Immunol. 2018;9:2425.
    1. Ziegenhain C, et al. Comparative analysis of single-cell RNA sequencing methods. Mol. Cell. 2017;65:631–643.e4.
    1. Binek A, et al. Flow cytometry has a significant impact on the cellular metabolome. J. Proteome Res. 2019;18:169–181.
    1. Llufrio EM, Wang L, Naser FJ, Patti GJ. Sorting cells alters their redox state and cellular metabolome. Redox Biol. 2018;16:381–387.
    1. Nguyen QH, Pervolarakis N, Nee K, Kessenbrock K. Experimental considerations for single-cell RNA sequencing approaches. Front. Cell Dev. Biol. 2018;6:108.
    1. Valihrach L, Androvic P, Kubista M. Platforms for single-cell collection and analysis. Int. J. Mol. Sci. 2018;19:807.
    1. Gao C, Zhang M, Chen L. The comparison of two single-cell sequencing platforms: BD Rhapsody and 10x Genomics Chromium. Curr. Genomics. 2020;21:602–609.
    1. Satija R, Farrell JA, Gennert D, Schier AF, Regev A. Spatial reconstruction of single-cell gene expression data. Nat. Biotechnol. 2015;33:495–502.
    1. Wolf FA, Angerer P, Theis FJ. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 2018;19:15.
    1. Zappia L, Phipson B, Oshlack A. Exploring the single-cell RNA-seq analysis landscape with the scRNA-tools database. PLoS Comput. Biol. 2018;14:e1006245.
    1. Trapnell C, et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat. Biotechnol. 2014;32:381–386.
    1. La Manno G, et al. RNA velocity of single cells. Nature. 2018;560:494–498.
    1. Luecken MD, Theis FJ. Current best practices in single-cell RNA-seq analysis: a tutorial. Mol. Syst. Biol. 2019;15:e8746.
    1. Schadt EE. Molecular networks as sensors and drivers of common human diseases. Nature. 2009;461:218–223.
    1. Schadt EE, Bjorkegren JL. NEW: network-enabled wisdom in biology, medicine, and health care. Sci. Transl. Med. 2012;4:115rv111.
    1. Foroughi Asl H, et al. Expression quantitative trait loci acting across multiple tissues are enriched in inherited risk for coronary artery disease. Circ. Cardiovasc. Genet. 2015;8:305–315.
    1. Franzen O, et al. Cardiometabolic risk loci share downstream cis- and trans-gene regulation across tissues and diseases. Science. 2016;353:827–830.
    1. Talukdar HA, et al. Cross-tissue regulatory gene networks in coronary artery disease. Cell Syst. 2016;2:196–208.
    1. Kharchenko PV, Silberstein L, Scadden DT. Bayesian approach to single-cell differential expression analysis. Nat. Methods. 2014;11:740–742.
    1. Wagner A, Regev A, Yosef N. Revealing the vectors of cellular identity with single-cell genomics. Nat. Biotechnol. 2016;34:1145–1160.
    1. Pratapa A, Jalihal AP, Law JN, Bharadwaj A, Murali TM. Benchmarking algorithms for gene regulatory network inference from single-cell transcriptomic data. Nat. Methods. 2020;17:147–154.
    1. Armingol E, Officer A, Harismendy O, Lewis NE. Deciphering cell-cell interactions and communication from gene expression. Nat. Rev. Genet. 2021;22:71–88.
    1. Stoeckius M, et al. Cell Hashing with barcoded antibodies enables multiplexing and doublet detection for single cell genomics. Genome Biol. 2018;19:224.
    1. Mimitou EP, et al. Multiplexed detection of proteins, transcriptomes, clonotypes and CRISPR perturbations in single cells. Nat. Methods. 2019;16:409–412.
    1. Kim K, et al. Transcriptome analysis reveals nonfoamy rather than foamy plaque macrophages are proinflammatory in atherosclerotic murine models. Circ. Res. 2018;123:1127–1142.
    1. Lin JD, et al. Single-cell analysis of fate-mapped macrophages reveals heterogeneity, including stem-like properties, during atherosclerosis progression and regression. JCI Insight. 2019;4:e124574.
    1. Robbins CS, et al. Local proliferation dominates lesional macrophage accumulation in atherosclerosis. Nat. Med. 2013;19:1166–1172.
    1. Zernecke A, et al. Meta-analysis of leukocyte diversity in atherosclerotic mouse aortas. Circ. Res. 2020;127:402–426.
    1. Korsunsky I, et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods. 2019;16:1289–1296.
    1. Allahverdian S, Chehroudi AC, McManus BM, Abraham T, Francis GA. Contribution of intimal smooth muscle cells to cholesterol accumulation and macrophage-like cells in human atherosclerosis. Circulation. 2014;129:1551–1559.
    1. Wang Y, et al. Smooth muscle cells contribute the majority of foam cells in ApoE (apolipoprotein E)-deficient mouse atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2019;39:876–887.
    1. Shankman LS, et al. KLF4-dependent phenotypic modulation of smooth muscle cells has a key role in atherosclerotic plaque pathogenesis. Nat. Med. 2015;21:628–637.
    1. Rong JX, Shapiro M, Trogan E, Fisher EA. Transdifferentiation of mouse aortic smooth muscle cells to a macrophage-like state after cholesterol loading. Proc. Natl Acad. Sci. USA. 2003;100:13531–13536.
    1. Vengrenyuk Y, et al. Cholesterol loading reprograms the microRNA-143/145-myocardin axis to convert aortic smooth muscle cells to a dysfunctional macrophage-like phenotype. Arterioscler. Thromb. Vasc. Biol. 2015;35:535–546.
    1. Basatemur GL, Jorgensen HF, Clarke MCH, Bennett MR, Mallat Z. Vascular smooth muscle cells in atherosclerosis. Nat. Rev. Cardiol. 2019;16:727–744.
    1. Guo L, et al. CD163+ macrophages promote angiogenesis and vascular permeability accompanied by inflammation in atherosclerosis. J. Clin. Invest. 2018;128:1106–1124.
    1. Wirka RC, et al. Atheroprotective roles of smooth muscle cell phenotypic modulation and the TCF21 disease gene as revealed by single-cell analysis. Nat. Med. 2019;25:1280–1289.
    1. Newman AM, et al. Robust enumeration of cell subsets from tissue expression profiles. Nat. Methods. 2015;12:453–457.
    1. Wroblewska A, et al. Protein barcodes enable high-dimensional single-cell CRISPR screens. Cell. 2018;175:1141–1155.e16.
    1. Goltsev Y, et al. Deep profiling of mouse splenic architecture with CODEX multiplexed imaging. Cell. 2018;174:968–981.e15.
    1. Angelo M, et al. Multiplexed ion beam imaging of human breast tumors. Nat. Med. 2014;20:436–442.
    1. Giesen C, et al. Highly multiplexed imaging of tumor tissues with subcellular resolution by mass cytometry. Nat. Methods. 2014;11:417–422.
    1. Stahl PL, et al. Visualization and analysis of gene expression in tissue sections by spatial transcriptomics. Science. 2016;353:78–82.
    1. Xia C, Fan J, Emanuel G, Hao J, Zhuang X. Spatial transcriptome profiling by MERFISH reveals subcellular RNA compartmentalization and cell cycle-dependent gene expression. Proc. Natl Acad. Sci. USA. 2019;116:19490–19499.
    1. Rodriques SG, et al. Slide-seq: a scalable technology for measuring genome-wide expression at high spatial resolution. Science. 2019;363:1463–1467.
    1. Wang F, et al. RNAscope: a novel in situ RNA analysis platform for formalin-fixed, paraffin-embedded tissues. J. Mol. Diagn. 2012;14:22–29.
    1. Kulkarni MM. Digital multiplexed gene expression analysis using the NanoString nCounter system. Curr. Protoc. Mol. Biol. 2011 doi: 10.1002/0471142727.mb25b10s94.
    1. Coskun AF, Cai L. Dense transcript profiling in single cells by image correlation decoding. Nat. Methods. 2016;13:657–660.
    1. Liu Y, et al. High-spatial-resolution multi-omics sequencing via deterministic barcoding in tissue. Cell. 2020;183:1665–1681.e18.
    1. Dobnikar L, et al. Disease-relevant transcriptional signatures identified in individual smooth muscle cells from healthy mouse vessels. Nat. Commun. 2018;9:4567.
    1. Flores AM, et al. Pro-efferocytic nanoparticles are specifically taken up by lesional macrophages and prevent atherosclerosis. Nat. Nanotechnol. 2020;15:154–161.
    1. Wolf D, et al. Pathogenic autoimmunity in atherosclerosis evolves from initially protective apolipoprotein B100-reactive CD4(+) T-regulatory cells. Circulation. 2020;142:1279–1293.
    1. Dixit A, et al. Perturb-seq: dissecting molecular circuits with scalable single-cell RNA profiling of pooled genetic screens. Cell. 2016;167:1853–1866.e17.
    1. Replogle JM, et al. Combinatorial single-cell CRISPR screens by direct guide RNA capture and targeted sequencing. Nat. Biotechnol. 2020;38:954–961.
    1. Xie S, Duan J, Li B, Zhou P, Hon GC. Multiplexed engineering and analysis of combinatorial enhancer activity in single cells. Mol. Cell. 2017;66:285–299.e5.
    1. Datlinger P, et al. Pooled CRISPR screening with single-cell transcriptome readout. Nat. Methods. 2017;14:297–301.
    1. Rubin AJ, et al. Coupled single-cell CRISPR screening and epigenomic profiling reveals causal gene regulatory networks. Cell. 2019;176:361–376.e17.

Source: PubMed

3
Tilaa