key: cord-0019298-eqm0on4p authors: Zhao, Suhui; Tsibris, Athe title: Leveraging Novel Integrated Single-Cell Analyses to Define HIV-1 Latency Reversal date: 2021-06-22 journal: Viruses DOI: 10.3390/v13071197 sha: 626ae1348e65d6f6c2a860a0179b6fee9e597d89 doc_id: 19298 cord_uid: eqm0on4p While suppressive antiretroviral therapy can effectively limit HIV-1 replication and evolution, it leaves behind a residual pool of integrated viral genomes that persist in a state of reversible nonproductive infection, referred to as the HIV-1 reservoir. HIV-1 infection models were established to investigate HIV-1 latency and its reversal; recent work began to probe the dynamics of HIV-1 latency reversal at single-cell resolution. Signals that establish HIV-1 latency and govern its reactivation are complex and may not be completely resolved at the cellular and regulatory levels by the aggregated measurements of bulk cellular-sequencing methods. High-throughput single-cell technologies that characterize and quantify changes to the epigenome, transcriptome, and proteome continue to rapidly evolve. Combinations of single-cell techniques, in conjunction with novel computational approaches to analyze these data, were developed and provide an opportunity to improve the resolution of the heterogeneity that may exist in HIV-1 reactivation. In this review, we summarize the published single-cell HIV-1 transcriptomic work and explore how cutting-edge advances in single-cell techniques and integrative data-analysis tools may be leveraged to define the mechanisms that control the reversal of HIV-1 latency. HIV-1 persists in CD4 + T cells and rebounds after antiretroviral therapy (ART) is interrupted or stopped. Viral latency is defined as a state of reversible nonproductive infection that, in the case of HIV-1, is associated with the use of suppressive ART [1] . Previous work investigated which proportion of integrated sequences are intact, and thereby may contribute to the replication-competent HIV-1 reservoir [2] [3] [4] [5] . The vast majority, greater than 90%, of integrated proviruses are defective, accumulate during acute HIV-1 infection, and cannot support viral replication [2, 4] . However, these "defective" proviruses may still retain HIV-1 transcriptional and translation activity that is relevant to eradication efforts [6, 7] . Cells harboring latent HIV-1 are extremely rare, numbering in the range of 10 0 -10 3 per million CD4 + T cells in peripheral blood during treated suppressed infection, and may be heterogenous [8] [9] [10] [11] . As few as 100 CD4+ T cells harbor intact HIV-1 proviruses that may contribute to rapid HIV-1 rebound after ART is stopped [2, 4, 12] . Cells that comprise the HIV-1 reservoir express no known distinguishing markers, making them a challenge to identify [2, 7, [13] [14] [15] [16] [17] [18] . Several approaches were developed to identify cells harboring HIV-1 DNA and measure the frequencies, size, and phenotypes of the HIV-1 reservoir in ART-suppressed individuals. The tat/rev-induced limiting dilution assay (TILDA) can measure multiply spliced HIV-1 transcripts after stimulation and has the advantage of requiring fewer cells than other methods [19] [20] [21] , while the quantitative viral outgrowth assay (QVOA), a gold standard of estimation of the size of HIV-1 reservoir, quantifies replication-competent proviruses [22] [23] [24] [25] . Even the gold standard has limitations: repeated rounds of activation change the result, and a newer QVOA that preferentially differentiates replication-competent proviruses [22] [23] [24] [25] . Even the gold standard has limitations: repeated rounds of activation change the result, and a newer QVOA that preferentially differentiates T cells to an effector memory phenotype, dQVOA, may be more accurate [26] . The intact proviral DNA assay (IPDA) may offer an accurate and scalable alternative to the more labor-intensive QVOA [12, 27] . Full-length sequencing of the HIV-1 genome can quantify intact noninduced genomes of HIV-1 [3, 4] . Cells expressing the HIV-1 capsid protein, p24, that is produced during viral reactivation can be quantified by flow cytometry [28] [29] [30] . These approaches should be viewed as complementary; sequencing cannot determine translation or replication competence, and flow cytometry cannot assess proviral deletions and/or mutations. The rarity of latently HIV-infected cells in patients makes their reactivation a challenge to study at the single-cell level. To address this difficulty, in vitro and ex vivo models were developed to investigate HIV-1 latency, each with their own unique sets of advantages and disadvantages. In vitro models can provide sufficient numbers of cells for analyses, but may not recapitulate latency in vivo, and are rarely truly latent [31] . Primary ex vivo models that use CD4 + T cells are widely used in the investigation of HIV-1 latency and latency reversal [24, [32] [33] [34] [35] [36] . The availability of high-resolution single-cell RNA sequencing (scRNA-seq) in combination with novel technologies that assess the epigenome and proteome, such as the assay for transposase-accessible chromatin using sequencing (scATAC-seq) and Cellular Indexing of Transcriptomes and Epitopes by Sequencing (CITE-seq), provides a unique opportunity to study the heterogeneity of HIV-1 latency reversal, and more precisely define the mechanisms that control HIV-1 transcriptional reactivation ( Figure 1 ). In this review, we summarize current approaches to leverage single-cell sequencing technologies to study HIV-1 latency. We review downstream integrative-analysis approaches to identify new strategies that may shed light on the complexities of HIV-1 latency and its reactivation. 1 latency in model systems that use primary human CD4 + T lymphocytes. Generally, cells are activated, infected by reporter HIV-1 constructs, and allowed for returning to a quiescent state prior to reactivation with pharmacologic latency reversal agents (LRA). (B) Summary of single-cell technologies to characterize transcriptome, proteome, and regulome. (C) Integration approaches to single-cell datasets that assess transcription and chromatin accessibility. Tn5 transposases, coupled with DNA adapters, fragment and tag accessible genomic DNA. Resulting fragments are amplified and sequenced to generate chromatin accessibility data. Major contributions to our understanding of the HIV-1 reservoir and its latency come from the study of cells in bulk. This is true for how the reservoir is established, how it is maintained, and how its latency may be pharmacologically reversed [4, [22] [23] [24] [37] [38] [39] [40] . Based on these important discoveries, latency-reversal agents (LRA) were advanced into clinical studies but have, to date, underwhelmed [41] [42] [43] [44] . The general concept of latency reversal is that, during suppressive ART that prevents viral replication, HIV-1 proviruses are induced to transcribe and translate viral proteins, "marking" the cell for subsequent immune-system clearance. However, HIV-1 latency is not always effectively reversed and total reservoir size in clinical trials post-LRA exposure remains unchanged. Mathematicalmodeling studies suggest that a 3-4 log 10 reduction in HIV-1 reservoir size may be required to achieve a reasonable probability of viral eradication; an empirical study in stem-cell transplant participants supports this idea [45, 46] . To better understand the relationship between HIV-1 latency reversal and CD4 + T cells, recent studies have focused on the mechanisms that define latency at single-cell resolution (Table 1) . More insight into cell-to-cell variations that predict effective latency reversal may identify key nodes or checkpoints in cell function and further efforts to improve LRA performance. In general, the purpose of single-cell approaches is to more clearly resolve important cellular and regulatory signals that may be obscured in the aggregated and averaged data generated by bulk cell analyses. There are reasons to believe that the heterogeneity of the HIV-1 reservoir may justify single-cell approaches to study viral latency. Treatment with LRAs leads to the transcriptional reactivation of only a small subset of total proviruses, even with more than one round of stimulation [4, 47, 48] . Researchers hypothesized that heterogeneity in the HIV-1 integration site, CD4 + T-cell subsets, and their activation state, epigenetic modifications, and stochastic transcriptional noise [49, 50] may play a role in viral latency and justify experiments at the single-cell level that characterize transcriptional diversity and resolve heterogeneity in regulatory mechanisms. HIV-1 may integrate into euchromatin, heterochromatin, and gene "deserts", be present as intact replication-competent provirus, defective transcriptionally silent provirus, or replication-defective provirus that is still capable of some viral protein translation [7, 16] . This heterogeneity extends to T-cell subsets that are infected and their relative state of rest or activation, all of which can be studied with greater granularity and detail at singlecell resolution [39, 51, 52] . Studies published in the past 3 years, discussed below, probed these important questions in a primary HIV-1 latency model and in ex vivo samples from treated suppressed participants with HIV-1. Key experimental details of these studies are summarized in Table 1 . Due to the rarity of latent cells in ART-treated virologically suppressed participants with HIV-1, most studies leverage lab-adapted or primary-latency models, the scope of which is beyond this review. Two studies used a well-described primary-latency model to investigate how the host transcriptional program influences HIV-1 latency at the single-cell level [50, 53] . In this model, CD4 + T cells are activated, infected with a GFP-expressing reference HIV-1 strain NL4-3, and then cultured for 8-12 weeks on H80 cells. H80 cells are a lab-adapted adherent glioma cell line that rescues T cells from death and reduces their activation, as defined by HLA-DR and CD25 expression [54] . More recent work identified TGF-β and IL-8 as cytokines secreted by H80 cells that play a role in the induction of T-cell quiescence [34] . After 12 weeks of coculture, these CD4 + T cells display a heterogeneous range of GFP expression. To understand heterogeneity in HIV-1 latency and its reversal, HIV-1-infected CD4 + T-cell populations after 8 weeks of H80 cell coculture were incubated in the presence or absence of vorinostat (VOR), a histone deacetylase inhibitor, for 24 h or tetrameric anti-CD3/anti-CD28 antibody complexes (TCR) for 48 h [53] . A total of 224 cells, namely, 43 untreated, 90 VOR-treated, and 91 TCR-treated, were then subjected to Smart-seq single-cell analyses. Principal-component analysis (PCA) identified two main clusters: a well-demarcated Cluster 1 and a larger Cluster 2. Each cluster contained cells from all three treatment conditions, suggesting some commonality in transcription that did not depend on drug exposure. CD4 + T cells in this model therefore generally exist in at least two transcriptional states. Visually, TCR-treated cells in Cluster 2 segregated from untreated and VOR-treated cells, possibly indicating a TCR activation-specific effect on transcription. The two main clusters significantly differed in GFP intensity, as assessed by fluorescent microscopy, and suggested that Cluster 1 corresponded to noninduced cell phenotypes, and Cluster 2 to inducible cells. However, GFP fluorescence in VOR-treated cells was not observed. HIV-1 transcript levels were higher in Cluster 2 across treatment conditions, suggesting that perhaps a mixture of productively and latently HIV-1-infected cells were included in the analyses. This identifies a potential source of confounding: CD4 + T cells may have expressed HIV-1 transcripts either because they were productively infected pre-stimulation, or because HIV-1 was, in fact, latent and was reactivated by TCR-mediated signaling. A total of 134 differentially expressed genes were identified that distinguished Clusters 1 and 2, approximately half of which were ribosomal, and half were enriched in pathways specific for metabolism, translation, electron transport, splicing, and HIV-1 infection. To confirm and extend this signature, analysis of a total of 77 resting CD4 + T cells, defined as CD25-CD69-HLA-DR-, isolated from two treated suppressed participants with HIV-1 identified similar differential expression of these 134 genes. Whether this signature is a function of T-cell biology or the presence of integrated HIV-1 and its reactivation remains an important open question. In a study later that same year, the transcriptomes of CD4 + T cells from three donors, infected ex vivo with HIV-1 and maintained on H80 cells for 6-12 weeks, were assessed using a 3 10× Genomics sequencing approach [50] . Dimensionality reduction and visualization in t-distributed stochastic neighbor embedding (tSNE) plots of data generated from 4206 cells demonstrated an expected clustering by donor. To probe the association of host transcriptome with HIV-1 latency, differences in gene expression were assessed between cells with and without measurable GFP transcripts. Cells with latent GFP expressed a specific set of cellular genes, marked by higher CCR7, CD27, and SELL transcripts, when compared to highly GFP-expressing cells that demonstrated greater levels of IL2RA, HLA-DRA, CD38, TNFRSF4, and TNFRSF18 transcripts. Cells enriched for transcripts in cell death or survival and proliferation pathways, as identified in Ingenuity Pathway Analysis, were more likely to express GFP. After cell activation through T-cell receptor signaling with anti-CD3/anti-CD28 beads, however, it was GFP-cells that demonstrated greater proliferative capacity, preferentially expanding naïve and central memory T-cell subsets. When comparing these two published reports that used the H80 latency model, some similarities in gene expression were observed but also many differences that may in part be due to technical variations in the precise methodologies that the two groups used. To explore the gene-expression program associated with ex vivo HIV-1 latency reversal, investigators developed a method to isolate CD4 + T cells that express HIV-1 envelope (Env) after reactivation [55] . Purified CD4 + T cells isolated from 10 treated suppressed participants with HIV-1 were cultured for 36 h in PBMC-conditioned media that contained phytohemagglutinin (PHA), IL-2, and a pan-caspase inhibitor. Incubation with a cocktail of three broadly neutralizing antibodies (bnAb), which targeted the CD4 binding site and the V2 and V3 loops, enriched for cells that expressed HIV-1 mRNA. Env-expressing cells from three participants that coexpressed intracellular HIV-1 Gag underwent scRNAseq; a total of 85 such cells were sequenced to an average depth of 1500 expressed genes per cell. Approximately 4% of reads in Env + Gag + cells mapped to HIV-1 and, when combined with T-cell receptor-sequencing analysis, strongly suggested a clonal origin of these reactivated cells. Using a Seurat analysis approach that included comparator unfractionated PHAactivated cells and unrelated cells productively infected with the YU2 reference strain, a set of differentially expressed genes in PHA-activated, HIV-1 Env-and Gag-expressing cells were identified [56] . These genes were enriched in processes mostly related to immunesystem function. More recently, a method known as SortSeq was developed and used to assess cellular transcription during latency reversal [57] ( Table 1 ). The approach incubates resting CD4 + T cells, defined as CD25 − CD69 − HLA-DR − , isolated from participants with HIV-1 in the presence of phorbol 12-myristate 13-acetate (PMA) and ionomycin for 16 h. Cells are then fixed, permeabilized, and incubated with fluorescently labeled probes that are designed to bind the 5 and 3 regions of HIV-1 mRNA molecules. Probe-positive cells are isolated by flow cytometry and sequenced by SMART-seq. In this study, a total of 48 cells were analyzed from 14 treated virologically suppressed participants with HIV-1 (mean 3.4 cells, median 2 cells per participant). Differentially expressed genes in these SortSeq-identified cells were enriched for RNA binding proteins and pathways related to RNA processing and immune-system function. Taken together, these studies provide important insights into the transcriptional changes that associate with HIV-1 latency reversal. To advance the field further and facilitate comparisons across studies, subsequent experiments should verify the HIV-1 latency state of the cell prior to reactivation and downstream analyses. To evaluate the scientific rigor of primary latency model systems, the markers used to define latency, fluorescent or viral, should be quantified at the protein and RNA level and compared. Appropriate HIV-uninfected comparator conditions should be included and reactivation conditions should ideally be standardized. Corroborating the findings of these early pioneering single-cell studies should be a priority, along with further mechanism-driven work to determine how the identified pathways more specifically impact viral latency. To mitigate batch effects in experimental designs, and more accurately identify and remove cell multiplets, cell hashing can combine comparator conditions into one sequencing run [58] [59] [60] (Figure 2 ). Cell hashing is a process that labels cells with sequence-tagged monoclonal antibodies to common surface proteins such as the β3 Na + /K + ATPase and β2-microglobulin, permits the unambiguous identification of cell multiplets, and can assign cells from different conditions back to their original sample. Disentangling the transcriptome associated with T-cell activation biology from any changes specifically associated with the presence of integrated HIV-1 genomes should be prioritized, focusing on total CD4 + T-cell populations and participant-to-participant variations in transcriptome. While cost remains an issue in single-cell experimental design, technological advances permit the aggregation of experimental data across groups and platforms, increasing their power and value [56] . Sequencing a larger number of cells improves the reproducibility of results, a process made simpler with the wider availability of droplet-based single-cell sequencing methods to increase throughput. Importantly, future work should extend to myeloid cells, a potential HIV-1 reservoir, and rapidly developing spatial transcriptomics approaches to study latency in tissue. variations in transcriptome. While cost remains an issue in single-cell experimental design, technological advances permit the aggregation of experimental data across groups and platforms, increasing their power and value [56] . Sequencing a larger number of cells improves the reproducibility of results, a process made simpler with the wider availability of droplet-based single-cell sequencing methods to increase throughput. Importantly, future work should extend to myeloid cells, a potential HIV-1 reservoir, and rapidly developing spatial transcriptomics approaches to study latency in tissue. To confirm findings from single-cell HIV-1 analyses, orthogonal methods that leverage more classical testing approaches should be used. As the current literature highlights, there is not always commonality in the findings across single-cell studies of HIV-1 latency. While it is perhaps premature to suggest a standardized confirmatory approach, classical testing helps to disentangle which findings relate more to the used latency system or the employed precise experimental approach, and which elucidate the fundamental biology of HIV-1 latency reversal. Single-cell transcriptomics can characterize the cellular heterogeneity that is present in primary HIV-1 latency model systems and in samples from patients with HIV-1. To define the regulatory mechanisms that govern transcription and its downstream translation, single-cell RNA sequencing may be combined with assessments of the epigenome and proteome. Combining -omics approaches may help answer a more fundamental question: at which level-DNA, RNA, or protein-is the latency-reversal program encoded, if one exists? Multimodal profiling offers advantages during the dynamic cellular-transition states associated with viral latency reversal, when the correlation between transcript and protein levels may weaken [61] . We summarize these modalities and discuss how they may be applied to the study of HIV-1 latency reversal. Important work that incorporates RNA and protein measurements to study HIV-1 latency at single-cell resolution was developed, but it does not have the throughput of droplet-based single-cell profiling and is not enumerated in this review [12, 44, 51, [62] [63] [64] [65] . With combinatorial labelling and barcode approaches, single-cell sequencing may be widely applied to analyze epigenomic [66, 67] , transcriptomics [68] , chromatin accessibility [69] [70] [71] [72] [73] , cell-surface proteins [74, 75] , and chromosomal conformation [76, 77] . A variety of single-cell sequencing approaches were developed to characterize mRNA, including Drop-seq, InDrop, Smart-seq, MARS-seq, 10X Genomics, SPLiT-seq, sci-RNA-seq; surface-protein identification and cell phenotyping using CITE-seq, REAP-seq, FACS; and chromatin-accessibility measurements with scATAC-seq, sciATAC-seq, and scTHS-seq [78] ( Figure 1B and Table 2 ). Human DNA wraps into nucleosomes, which then condense into solenoids, forming the chromatin that comprises chromosomes. To assist in the discovery of chromatin modulators and regulators, the assay for transposase-accessible chromatin using sequencing (ATAC-Seq) was developed, first in bulk and now in single cells [71, [79] [80] [81] [82] . Recently, the approach was applied to joint measurements in the same cell [83] [84] [85] . In general, ATAC-seq measures the openness of DNA, a proxy for how easily transcription factors or other regulatory elements may bind. ATAC-seq is based on the use of a hyperactive Tn5 transposase that binds and cleaves open DNA before adding adaptors that tag accessible chromatin [79] . The identification of accessible DNA, or peak calling, is the core analysis function of ATACseq [86] . Gene regulatory assessments follow from peak calling to include differential peak analysis across cell types or experimental conditions, motif identification, nucleosome positioning, and transcription-factor footprint occupancy. The goal of these analyses is to infer gene regulatory networks and identify their key regulators. Analytical tools specifically designed for scATAC-seq datasets either with or without scRNAseq, including chromeVAR, Signac, and MAESTRO, were developed, although computational challenges remain [87] [88] [89] [90] [91] . To study HIV-1 latency, epigenetic profiles generated by ATAC-seq could be used to identify noncoding regulatory elements that associate with the transcriptome changes of CD4 + T-cell activation and/or HIV-1 latency reversal. At a minimum, multimodal interrogations of latency models can establish the relationship between epigenome and transcriptome during latency reversal. To facilitate this work in the context of HIV-1, RNA-seq and ATAC-seq datasets in bulk and single HIV-uninfected primary CD4 + T cells and their phenotypic subsets were published, providing a useful comparator framework to evaluate the specificity of observed changes in the presence of integrated HIV-1 [92] [93] [94] [95] [96] . The development of methods to simultaneously quantify surface-protein expression and mRNA transcription is a seminal advance in droplet-based single-cell sequencing techniques. These approaches digitize the abundance of cell-surface proteins into discrete sequence information that can be coupled to and analyzed along with transcriptome measurements in single-cell droplets. Monoclonal antibodies (mAb) that would traditionally be conjugated to fluorophores or metals for flow or mass cytometry are instead labelled with DNA barcodes. The multiplexed identification of surface proteins using barcodes significantly exceeds the spectral capacities of flow cytometry or the number of available metal isotopes; DNA-barcoded mAb panels that simultaneously interrogate all known cell-surface proteins have been commercialized. Two studies that were initially described in 2017, using similar methodologies, explored these approaches. Cellular Indexing of Transcriptomes and Epitopes by Sequencing or CITE-seq detects multiple proteins and simultaneously characterizes the unbiased transcriptomes at the single-cell level [74] . Qualitative and quantitative comparisons of CITE-seq and flow cytometry in this report demonstrate similar results in PBMC. To the best of our knowledge, a comparison of CITE-seq and mass cytometry has not yet been reported. The RNA expression and protein sequencing assay or REAP-seq analyzes protein and RNA levels in single cells using DNA-conjugated antibodies and droplet microfluidics [75] . Both REAP-seq and CITE-seq leverage the difference in size between amplified cDNAs and antibody-derived tags to generate independent sequencing libraries. REAP-seq was benchmarked against flow cytometry for the detection of B lymphocytes, T lymphocytes, natural-killer (NK) cells, and monocytes, and delivered comparable results. The CITE-seq approach was recently extended to an expanded CRISPR-compatible cellular indexing of transcriptomes and epitopes by sequencing (ECCITE-seq), an approach that was adapted to 5 capture-based scRNA-seq methods [97] . ECCITE-seq was used to perform CRISPR screens, reliably identify and quantify the heterogeneity present in single cells that received the same guide RNA (gRNA), and further define the mechanisms that control programmed death-ligand 1 (PD-L1) expression [98] . CITE-seq was applied to profile and compare HIV-1 infected cells during immediate and delayed antiretroviral therapy [99] . Single-nucleus-based CITE-seq, Intranuclear Cellular Indexing of Transcriptomes and Epitopes (inCITE-seq), was reported that measures quantitative intranuclear protein levels and transcriptomes in cells and tissues [100] . Some recently described approaches may hold promise for future studies of HIV-1 latency. Split-pool ligation-based transcriptome sequencing (SPLiT-seq) labels individual transcriptomes from cells or nuclei by combinatorial barcoding, generating uniquely barcoded cells; therefore, SPLiT-seq does not require cell partitioning workflows such as cell sorting, custom microfluidics, and microwells [101] . Taking advantages of scRNAseq, single-nucleus RNA-sequencing (snRNA-seq) was introduced to visualize the extremely low levels of mRNA in a single nucleus, including sNuc-DropSeq [102] and DroNc-seq [103, 104] . snRNA-seq is widely applied to recapitulate transcriptome and gene Viruses 2021, 13, 1197 9 of 17 expression responding to a certain stimulus in neuronal cells and tissues [105] . However, primary cells used for HIV-1 latency investigation are rich in proteases and RNases [106] , which may limit the application of these approaches in HIV-1 latency research. Split-pool ligation-based transcriptome sequencing SPLiT-seq 3 -end only [101] single-cell assay for transposase-accessible chromatin sequencing scATAC-seq Chromatin accessibility [79] Chromosome conformation capture (3C) coupled with sequencing Hi-C/3C-Seq/Capture-C Chromatin structure [114] Droplet-based single-cell chromatin immune-precipitation sequencing scChIP-seq/Drop-ChIP Chromatin fragments [115] Single-cell transposome hypersensitive site sequencing scTHS-seq Chromatin accessibility [116] Chromosome conformation capture sequencing combining chromatin immunoprecipitation HiChIP Chromasome capture [117] Single-cell combinatorial indexing ATAC-seq sciATAC-seq Chromatin accessibility [118] Cellular indexing of transcriptomes and epitopes by sequencing CITE-seq Multiomic [74] RNA expression and protein sequencing assay REAP-seq Multiomic [75] Expanded CRISPR-compatible cellular indexing of transcriptomes and epitopes by sequencing ECCITE-seq Multiomic [97] Intranuclear cellular indexing of transcriptomes and epitopes inCITE-seq Intranuclear protein and transcriptome [100] Single-cell high-throughput techniques characterize cell and tissue heterogeneity, and cell phenotype, fate, and function in a high-resolution manner. To increase the power of these datasets, single-cell sequencing data generated across different conditions, experiments, and samples could be integrated. With the rapid progression of machine-learning techniques, computational efforts focus on the integration of multiple sequencing modalities, including the integration of scRNA-seq data from multiple scRNA-seq experiments using different methods, the classification of multiple cell phenotypes from different studies, and joint analysis of transcriptional and spatial signals. The combination of multiple datasets generated by different high-throughput single-cell methods requires new computational methods that can integrate multiple types of single-cell data, detect correlations, and reveal relationships across modalities. While the single-cell genomic field continues to rapidly develop and evolve, we summarize some common and high-impact analysis strategies for these complex datasets. The discussed toolkits in this section are either in current widespread use in the field or have particular promise to advance the analysis of single-cell datasets in unique ways. Many statistical methods that leverage Poisson (P), negative binomial (NB), and Gaussian (G) distribution approaches were developed to integrate scRNA-seq datasets generated from different conditions or experimental platforms [119] . Multimodal Model Selection (M3S) is an R package that selects the most parsimonious model to fit the distribution of gene expression in a genewise manner, characterizes the transcriptional events, and detects differentially expressed genes for scRNA-seq data. Other approaches were developed to integrate datasets in multimodal single-cell screens. With the use of targeting guide RNAs (gRNAs) in ECCITE-seq, heterogeneities with no perturbation effects present high background noise into downstream analyses. The Satija group designed a novel computational method, mixscape, for reducing such noise and refining the characterization of multimodal perturbations and relevant transcript and surface-protein levels. Heterogeneity was observed in the independent analysis of control cells with nontargeting gRNAs (NT cells). To obtain the transcriptome that revealed the real genetic perturbation, the most similar mRNA expression profiles of NT were subtracted from the original RNA profiles of target cells. On the basis of the Mixture Discriminant Analysis (MDA) algorithm, ECCIT-seq grouped cells on the basis of their expression of gRNA. Each group had a mixture of perturbed and nonperturbed subpopulations. The Gaussian mixture model was used to assign data points in each group on the basis of their subclass identity. The nonperturbed group had similar perturbation to that of NT cells. Mixscape is capable of grouping cells to perturbed or nonperturbed group [98] . Originally developed as a computational method to generate spatial transcriptomes in zebrafish embryos, Seurat is an R package that is in widespread use to analyze scRNA-seq datasets and identify rare cellular subpopulations [120] . Subsequent versions of Seurat introduced strategies to integrate and identify cellular populations across multiple scRNA-seq datasets [56] and eventually extend these approaches to other modalities, such as scATAC-seq [121] . Most recently, Seurat version 4.0 incorporated a weighted nearest-neighbor statistical-analysis approach to analyze multimodal single-cell datasets and created Azimuth, a free web application that allows for users to map their datasets to a multimodal reference atlas that incorporates scRNA-seq and CITE-seq data [87] . Reference atlases are available for the peripheral blood immune system and some tissue types. In the context of HIV-1 research, Azimuth may be used to annotate previously published scRNAseq datasets, which did not simultaneously assess protein expression, with surfaceprotein expression information, particularly valuable in the context of CD4 + T-cell subsets that are notoriously similar in RNA transcriptomes. However, its operating characteristics in this regard require careful validation, particularly in the context of memory T-cell subsets and rare cell populations, e.g., stem-cell memory cells, which may be important in HIV-1 reservoir dynamics. The main challenge of computational tools for the flexible modeling of single-cell datasets is the immense data heterogeneity generated from multiple modalities. A recently introduced statistical algorithm, Linked Inference of Genomic Experimental Relationships (LIGER), identifies shared cell types across samples. LIGER integrates in situ and geneexpression data to reveal the accurate spatial location of subtypes of cells, and jointly defines cell types by coupled scRNA-seq and DNA methylation profiles, providing a tool to characterize epigenomic regulation in specific cell types. After a common process of a raw digital gene-expression (DGE) matrix and dataset normalization, LIGER generates a dimensionally reduced space by integrative non-negative matrix factorization (iNMF) [122, 123] , and bases joint clustering on a shared factor neighborhood network showing cells with similar factor-loading patterns [122] . Recent work introduced a flexible and scalable algorithm across datasets, Harmony, to integrate datasets that have experimental, biological, and technical differences [124] . Harmony uses an unsupervised scRNA-seq joint embedding strategy that projects cells into a common low-dimensional space. Cells in Harmony are clustered by a soft k-means algorithm instead of a specific condition or platform. Clusters of small subsets of datasets are penalized by an information theoretic metric. Multiple penalties are allowed for in Harmony, and datasets from different techniques or sample sources are thereby accommodated with soft clustering. Harmony creates the final cell cluster after several iterations of adjustments on the basis of the cell-specific linear factor and is available as part of established pipelines in several analytical packages. As the number of single-cell techniques increases, it is increasingly common that, while these analyses are performed on the same sample, they do not simultaneously interrogate all features from the same cell. Therefore, improved computational methods are needed to integrate the data matrices that each technology, e.g., scRNA-seq, scATAC-seq, CITE-seq, and spatial transcriptomics, generates, and align these features being characterized across cells. To generate accurate coembedding matrices from multimodal data, a computational data-integration toolkit called biorder integration of single-cell data (bindSC) was recently developed to integrate variant single-cell data [125] . On the basis of biorder canonical correlation analysis (bi-CCA), bindSC accurately identifies the simultaneous alignment of columns and rows between data matrices from multimodal technologies, such as scRNAseq and scATAC-seq, on the same biological sample. The bindSC method was tested for its ability to integrate different types of datasets to include epigenomic and transcriptomic, scRNA-seq, and spatial-transcriptomic data, as well as scRNA-seq and protein data. BindSC initially preprocesses individual datasets by selecting variable features and clustering cells, matching the initial feature across modalities, identifying correspondence on the basis of the bi-CCA algorithm, integrating clustered cells in a latent space, and constructing multiomic profiles. To integrate data from scRNA-seq and scATAC-seq datasets, the Shirley Liu lab developed the Model-based AnalysEs of Transcriptome and RegulOme (MAESTRO) workflow [89] . MAESTRO provides multiple functions to characterize the transcriptome and regulome, including cell-cluster integration and automated cell-type annotation. MAE-STRO is compatible with multiple scRNA-seq and scATAC-seq platforms and inputs of different sequence files that are generated across these platforms. The creators presented data that MAESTRO may perform statistically significantly better than SnapATAC, cicero, and Seurat can in some aspects of scATAC-seq and scRNA-seq dataset integration. Another approach to address the limitation that data integration of parallel RNAseq and ATAC-seq is asymmetric, relying more on scRNA-seq data, is the single-cell aggregation and integration (scAI) method [126] . scAI uses an unsupervised iterativelearning approach and, in the simulation modeling of three existing datasets, was shown to effectively integrate epigenomic and transcriptomic datasets. Some data were also presented that scAI may offer advantages when different cell types are coassayed. Single-cell sequencing approaches hold great promise to identify mechanisms that control viral latency reversal and define whether a cellular program associates with HIV-1 reactivation and under which cellular conditions. Recent advances in high-throughput techniques enable the integration of multiple single-cell methods, simultaneously profiling gene expression, epigenetics, and surface and intracellular protein content. Multimodal analyses are required to generate novel insights into the biological diversity and cellular heterogeneity that underlies HIV-1 latency and the regulatory networks that govern its reversal. While benchmarking studies are yet to reveal a consensus approach to the computational analysis of single-cell datasets, the development of a unified approach among HIV researchers should be considered to be a high priority. In the meantime, investigators that tailor their individual methods to their systems' biology and the data types they generate may help to most expeditiously explore this new research space and lay the foundation for a future consensus on an integrated single-cell analysis pipeline. Cold Spring Harb Defective proviruses rapidly accumulate during acute HIV-1 infection Identification of Genetically Intact HIV-1 Proviruses in Specific CD4 + T Cells from Effectively Treated Participants Replication-Competent Noninduced Proviruses in the Latent Reservoir Increase Barrier to HIV-1 Cure Clonal expansion of genome-intact HIV-1 in functionally polarized Th1 CD4+ T cells Defective HIV-1 proviruses produce novel protein-coding RNA species in HIV-infected patients on combination antiretroviral therapy Defective HIV-1 Proviruses Are Expressed and Can Be Recognized by Cytotoxic T Lymphocytes, which Shape the Proviral Landscape Levels of HIV-1 persistence on antiretroviral therapy are not associated with markers of inflammation or activation Determinants of HIV-1 reservoir size and long-term dynamics during suppressive ART The decay of the latent reservoir of replication-competent HIV-1 is inversely correlated with the extent of residual viral replication during prolonged anti-retroviral therapy Long-term follow-up studies confirm the stability of the latent reservoir for HIV-1 in resting CD4+ T cells A quantitative approach for measuring the reservoir of latent HIV-1 proviruses The role of CD32 during HIV-1 infection Evidence that CD32a does not mark the HIV-1 latent reservoir Conflicting evidence for HIV enrichment in CD32+ CD4 T cells Distinct viral reservoirs in individuals with spontaneous control of HIV-1 HIV-1 Integration in the Human Genome Favors Active Genes and Local Hotspots The single-cell landscape of immunological responses of CD4+ T cells in HIV versus severe acute respiratory syndrome coronavirus 2 Inducible HIV RNA transcription assays to measure HIV persistence: Pros and cons of a compromise HIV latency in isolated patient CD4+T cells may be due to blocks in HIV transcriptional elongation, completion, and splicing Identification of a Reservoir for HIV-1 in Patients on Highly Active Antiretroviral Therapy Recovery of Replication-Competent HIV Despite Prolonged Suppression of Plasma Viremia Quantification of latent tissue reservoirs and total body viral load in HIV-1 infection Rapid Quantification of the Latent Reservoir for HIV-1 Using a Viral Outgrowth Assay Effector memory differentiation increases detection of replication-competent HIV-l in resting CD4+ T cells from virally suppressed individuals Intact proviral DNA assay analysis of large cohorts of people with HIV provides a benchmark for the frequency and composition of persistent proviral DNA Detection of intracellular HIV in lymphocytes by flow cytometry Antibodies to p24 antigen do not specifically detect HIV-infected lymphocytes in AIDS patients Specificity of binding of HIV-1 anti-p24 antibodies to CD4+ lymphocytes from HIV-infected subjects A Flexible Model of HIV-1 Latency Permitting Evaluation of Many Primary CD4 T-Cell Reservoirs In vivo fate of HIV-1-infected T cells: Quantitative analysis of the transition to stable latency Establishment and Reversal of HIV-1 Latency in Naive and Central Memory CD4 + T Cells In Vitro Entry of Polarized Effector Cells into Quiescence Forces HIV Latency CD4+T Cell Subsets and Pathways to HIV Latency HIV-1-Infected CD4+ T Cells Facilitate Latent Infection of Resting CD4+ T Cells through Cell-Cell Contact Latent infection of CD4+ T cells provides a mechanism for lifelong persistence of HIV-1, even in patients on effective combination therapy Early establishment of a pool of latently infected, resting CD4+ T cells during primary HIV-1 infection HIV reservoir size and persistence are driven by T cell survival and homeostatic proliferation Expression of latent human immunodeficiency type 1 is induced by novel and selective histone deacetylase inhibitors Interval dosing with the HDAC inhibitor vorinostat effectively reverses HIV latency Panobinostat, a histone deacetylase inhibitor, for latent-virus reactivation in HIV-infected patients on suppressive antiretroviral therapy: A phase 1/2, single group, clinical trial The Depsipeptide Romidepsin Reverses HIV-1 Latency In Vivo Bryostatin-1 for latent virus reactivation in HIV-infected patients on antiretroviral therapy Antiretroviral-free HIV-1 remission and viral rebound after allogeneic stem cell transplantation: Report of 2 cases Predicting the outcomes of treatment to eradicate the latent reservoir for HIV-1 Quantification of HIV-1 latency reversal in resting CD4+ T cells from patients on suppressive antiretroviral therapy Proliferation of latently infected CD4+ T cells carrying replication-competent HIV-1: Potential role in latent reservoir dynamics Screening for noise in gene expression identifies drug synergies Single-Cell Analysis of Quiescent HIV Infection Reveals Host Transcriptional Profiles that Regulate Proviral Latency Naive CD4+ T Cells Harbor a Large Inducible Reservoir of Latent, Replication-competent Human Immunodeficiency Virus Type 1 Persistence of an intact HIV reservoir in phenotypically naive T cells Single-Cell RNA-Seq Reveals Transcriptional Heterogeneity in Latent and Reactivated HIV-Infected Cells A novel in vitro system to generate and study latently HIV-infected long-lived normal CD4+ T-lymphocytes Clonal CD4+ T cells in the HIV-1 latent reservoir display a distinct gene profile upon reactivation Integrating single-cell transcriptomic data across different conditions, technologies, and species Single-cell transcriptional landscapes reveal HIV-1-driven aberrant host gene transcription as a potential therapeutic target Cell Hashing with barcoded antibodies enables multiplexing and doublet detection for single cell genomics Computational and analytical challenges in single-cell transcriptomics Missing data and technical variability in single-cell RNA-sequencing experiments On the Dependency of Cellular Protein Levels on mRNA Abundance Defining total-body AIDS-virus burden with implications for curative strategies Latency reversal agents affect differently the latent reservoir present in distinct CD4+ T subpopulations Cellular Metabolism Is a Major Determinant of HIV-1 Reservoir Seeding in CD4+ T Cells and Offers an Opportunity to Tackle Infection Single-cell characterization and quantification of translation-competent viral reservoirs in treated and untreated HIV infection Tumour evolution inferred by single-cell sequencing Sequencing thousands of single-cell genomes with combinatorial indexing Simultaneous measurement of chromatin accessibility, DNA methylation, and nucleosome phasing in single cells Lineage-specific and single-cell chromatin accessibility charts human hematopoiesis and leukemia evolution Single-cell chromatin accessibility reveals principles of regulatory variation Multiplex single-cell profiling of chromatin accessibility by combinatorial cellular indexing Integrative single-cell analysis by transcriptional and epigenetic states in human adult brain Simultaneous epitope and transcriptome measurement in single cells Multiplexed quantification of proteins and transcripts in single cells Massively multiplex single-cell Hi-C Single-cell Hi-C reveals cell-to-cell variability in chromosome structure Integrative single-cell analysis Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position Single-cell and single-molecule epigenomics to uncover genome regulation at unprecedented resolution A rapid and robust method for single cell chromatin accessibility profiling Massively parallel single-cell mitochondrial DNA genotyping and chromatin profiling Joint profiling of chromatin accessibility and gene expression in thousands of single cells scNMT-seq enables joint profiling of chromatin accessibility DNA methylation and transcription in single cells Chromatin Potential Identified by Shared Single-Cell Profiling of RNA and Chromatin From reads to insight: A hitchhiker's guide to ATAC-seq data analysis Integrated analysis of multimodal single-cell data Inferring transcription-factor-associated accessibility from single-cell epigenomic data Integrative analyses of single-cell transcriptome and regulome using MAESTRO Assessment of computational methods for the analysis of single-cell ATAC-seq data Inference and effects of barcode multiplets in droplet-based single-cell assays Transcript-indexed ATAC-seq for precision immune profiling Single-cell transcriptomics of human T cells reveals tissue and activation signatures in health and disease Integrated Single-Cell Analysis Maps the Continuous Regulatory Landscape of Human Hematopoietic Differentiation Genetic determinants of co-accessible chromatin regions in activated T cells across humans T cell fate and clonality inference from single-cell transcriptomes Multiplexed detection of proteins, transcriptomes, clonotypes and CRISPR perturbations in single cells Characterizing the molecular regulation of inhibitory immune checkpoints with multimodal single-cell screens Single-cell immune profiling reveals the impact of antiretroviral therapy on HIV-1-induced immune dysfunction, T cell clonal expansion, and HIV-1 persistence in vivo Simultaneous single cell measurements of intranuclear proteins and gene expression Single-cell profiling of the developing mouse brain and spinal cord with split-pool barcoding Dissecting Cell-Type Composition and Activity-Dependent Transcriptional State in Mammalian Brains by Massively Parallel Single-Nucleus RNA-Seq Massively parallel single-nucleus RNA-seq with DroNc-seq RNA-sequencing from single nuclei Advantages of Single-Nucleus over Single-Cell RNA Sequencing of Adult Kidney: Rare Cell Types and Novel Cell States Revealed in Fibrosis PBMC fixation and processing for Chromium single-cell RNA sequencing mRNA-Seq whole-transcriptome analysis of a single cell Full-length mRNA-Seq from single-cell levels of RNA and individual circulating tumor cells Massively Parallel Single-Cell RNA-Seq for Marker-Free Decomposition of Tissues into Cell Types Highly Parallel Genome-wide Expression Profiling of Individual Cells Using Nanoliter Droplets Nuclear RNA-seq of single neurons reveals molecular signatures of activation Comprehensive single-cell transcriptional profiling of a multicellular organism Analysis of hundreds of cis-regulatory landscapes at high resolution in a single, high-throughput experiment Single-cell ChIP-seq reveals cell subpopulations defined by chromatin state Characterization of chromatin accessibility with a transposome hypersensitive sites sequencing (THS-seq) assay HiChIP: Efficient and sensitive analysis of protein-directed genome architecture M3S: A comprehensive model selection for multi-modal single-cell RNA sequencing data Spatial reconstruction of single-cell gene expression data Comprehensive Integration of Single-Cell Data Single-Cell Multi-omic Integration Compares and Contrasts Features of Brain Cell Identity A non-negative matrix factorization method for detecting modules in heterogeneous omics multi-modal data Fast, sensitive and accurate integration of single-cell data with Harmony Unbiased integration of single cell multi-omics data scAI: An unsupervised approach for the integrative analysis of parallel single-cell transcriptomic and epigenomic profiles