Abstract
Purpose of review
CD4+ T cell loss is the hallmark of uncontrolled HIV-1 infection. Strikingly, CD4+ T cell depletion is a strong indicator for disease severity in the recently emerged coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. We reviewed recent single-cell immune profiling studies in HIV-1 infection and COVID-19 to provide critical insight in virus-induced immunopathogenesis.
Recent findings
Cytokine dysregulation in HIV-1 leads to chronic inflammation, while severe SARS-CoV-2 infection induces cytokine release syndrome and increased mortality. HIV-1-specific CD4+ T cells are dysfunctional, while SARS-CoV-2-specific CD4+ T cells exhibit robust Th1 function and correlate with protective antibody responses. In HIV-1 infection, follicular helper T cells (TFH) are susceptible to HIV-1 infection and persist in immune-sanctuary sites in lymphoid tissues as an HIV-1 reservoir. In severe SARS-CoV-2 infection, TFH are absent in lymphoid tissues and are associated with diminished protective immunity. Advancement in HIV-1 DNA, RNA, and protein-based single-cell capture methods can overcome the rarity and heterogeneity of HIV-1-infected cells and identify mechanisms of HIV-1 persistence and clonal expansion dynamics.
Summary
Single-cell immune profiling identifies a high-resolution picture of immune dysregulation in HIV-1 and SARS-CoV-2 infection and informs outcome prediction and therapeutic interventions.
Keywords: antigen-specific CD4+ T cells, coronavirus disease 2019, marker for HIV-1 latent reservoir, severe acute respiratory syndrome coronavirus 2, single-cell RNAseq
INTRODUCTION
The emergence of unknown and widespread infections, both HIV-1 in the 1980s and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in 2019, creates a global public health crisis. Researchers race at breathtaking speed to study viral pathogenesis and stop surging deaths and socioeconomic distress during the pandemic. Through the past 30 years, the HIV research community has built infrastructure such as advanced biosafety-level laboratories, competence to work on small-volume and infectious clinical samples, multidisciplinary approaches, and collaborating platforms and provided critical contributions to rapid scientific advancement in viral pathogenesis, immune responses, outcome prediction, treatment, and vaccine design for coronavirus disease 2019 (COVID-19).
CD4+ T cells are the central orchestrator of cellular and humoral adaptive immunity. In cellular immunity, cytotoxic T lymphocytes (CTLs) recognize and kill infected cells, and block exponential viral production. In humoral immunity, B cells undergo rounds of affinity maturation and class switching to produce antibodies that neutralize viral particles and block new rounds of infection. CD4+ T cells provide critical help for the development of CTL and B cell function and memory [1–3]. Both HIV-1 [4] and SARS-CoV-2 (reviewed in [5▪]), two RNA viruses causing widespread global infections, impair CD4+ T cell function and induce morbidity and mortality. The two viral infections are distinct in nature: acute HIV infection is characterized by virus-induced CD4+ T cell depletion, while chronic HIV infection, despite antiretroviral therapy (ART), still shows efficient immune escape, a lack of protective immunity, and hallmarks of chronic immune activation. COVID-19, on the other hand, shows greatest severity in acute infection when the innate immune system misfires. While there can be prolonged periods of virus shedding or rapid fading of protective immune responses in rare cases, most SARS-CoV-2 infected individuals develop protective immunity and do not develop chronic infection. The commonality between both diseases is the fact that a thorough understanding of immune cells is needed to advance treatment.
Recent advances in single-cell profiling of heterogeneous populations of immune cells, from flow cytometry and mass cytometry to single-cell RNAseq and multiomics profiling, have enabled in-depth understanding of virus-induced immune dysfunction in multiple dimensions including frequency, differentiation, function, and exhaustion phenotypes. While flow cytometry and mass cytometry provide high-dimensional protein profiling on a targeted selection of up to 50 proteins of interest, single-cell RNAseq provides genome-wide profiling of more than 20 000 genes in individual single cells. Yet, RNA content in single cell is sparse and hard to capture. It is estimated that a single cell contains ~10 pg of RNA. Among them, ~1–5% are mRNA [6]. Primary CD4+ T cells contain even less RNA. Recent advances in single-cell multiomic profiling methods (reviewed in [6]), their performance [7▪▪], batch correction methods [8▪▪], trajectory inference [9▪▪], and bioinformatic analysis (reviewed in [10▪▪]) have evolved rapidly to overcome the heterogeneity of immune cells, the rarity of specific cellular populations in different biological states, and the sparse RNA content in primary cells.
By contrasting the differences in HIV-1 versus SARS-CoV-2-induced immune dysfunction at the single-cell level (Fig. 1, Table 1) and by summarizing recent advances in single-cell profiling in HIV-1-infected cells (Fig. 2), we provide here mechanisms of immunopathogenesis and potential targets for therapeutic interventions.
FIGURE 1.
CD4+ T cell dysfunction in SARS-CoV-2 versus HIV-1 infection. (a) Both SARS-CoV-2 and HIV-1 infection cause CD4+ T cell depletion. CD4+ T cell counts are restored after SARS-CoV-2 clearance while untreated HIV-1 infection continues to deplete CD4+ T cells and eventually causes AIDS. Antiretroviral therapy controls HIV viral load to clinically undetectable levels and maintains relatively normal CD4+ T cell levels. The proliferation capacity of CD4+ T cells is relatively high in recovered COVID-19 patients. In contrast, the proliferation capacity is impaired during acute HIV-1 infection but can be partially restored with long-term antiretroviral therapy. (b) Cytokine dysregulation induces CD4+ T cell dysfunction during viral infection. SARS-CoV-2 infection is characterized by delayed IFNα response, cytokine release syndrome or cytokine storm (IL-6, IL-10, TNF-α), and misfiring of type 2 immune responses. SARS-CoV-2-specific CD4+ T cells remain functional, showing Th1 effector function (IFN-γ, IL-2, TNF-α), cytotoxic signatures, and robust proliferation potential. Acute HIV-1 infection is characterized by robust IFNα responses but impaired Th1 responses. HIV-1-specific CD4+ T cells are dysfunctional, exhibiting impaired IFN-γ and IL-2 secretion, exhaustion phenotypes, and decreased proliferation capacity. Despite antiretroviral therapy, CD4+ T cells remain dysfunctional and exhausted during chronic HIV-1 infection, secondary to chronic inflammation. HIV-1-infected cells are heterogeneous, upregulating cellular programs involving antiviral chemokines and cellular survival. A few surface markers have been reported to enrich for HIV-1-infected cells.
Table 1.
Cytokine production and CD4+ T cell dysfunction in SARS-CoV-2 versus HIV-1 infection
SARS-CoV-2 | HIV-1 (during viremia) | HIV-1 (after ART suppression) | HIV-1-infected cells | |
---|---|---|---|---|
Cytokine response | ↑ IL-1α, IL-1β, IL-6, IL-10, IL-18, TNF-α, IFNα [18▪▪,19,20,22] | ↑ IFNγ, TNF-α, MCP-1, IL-2R, IL-6, IL-8, IL-10, IL-12 [11,12▪,13] | ↑ IFNγ, TNF-α, TGF-β [13,14] | IRF4, JAK1, LTA, CCL3, CCL4, XCL1 [106▪,125▪] |
CD4 depletion | Lymphopenia [19,39▪,40] Negatively correlated with IL-6 and TNF-α levels [39▪] ↑ Apoptotic programs [23▪,41,42] |
Fas/FasL-mediated apoptosis through caspase 1 [4,27] Abortive infection-mediated pyroptosis through caspase 3 [28] GALT CD4+ T cell depletion and microbial translocation [30–34] |
Lymphoid tissue fibrosis prevents CD4+ T cell homeostasis and reconstitution [35–38] | Survival-associated genes: MIR155HG and ZNF217 [106▪,125▪] |
CD4 responses | ↑ ISG expression [48,49,50▪▪] ↑ TNF, IL-1β [52▪▪] ↑ TFH [80▪] ↑ Activation, proliferation [46▪] ↑ Type 1 and type 3 responses [18▪▪] ↑ Misfiring of type 2 responses (IL-5, IL-13) in severe COVID-19 [18▪▪] ↑ Cytotoxic CD4 [47] |
↑ ISG expression [44▪▪] ↑TNF-α, IL-1β response [44▪▪] ↑ TFH [57,90,91] |
↑CTLA4, TIGIT [45▪] | ↑Th1 [106▪] ↑TIGIT, HLA-DR [125▪] ↑ During viremia: activation, proliferation, exhaustion markers, CXCR5, α4β7, α4β1 [114,115] ↑ During suppression: exhaustion markers, α4β1 [114,115] ↑ CD2, CD30, CD161 enrichment [116–118] ↑Tem [114] |
Antigen-specific CD4+ T cells | Frequency ~0.2–2% [73▪,74▪▪] ↑ IL-2, IFNγ, TNF-α, proliferation [23▪,39▪,41,42,73▪, 76▪▪,78,79] ↑ cytotoxic effectors [80▪] |
Frequency ~0.1–0.7% [55,57] IFNγ but not IL-2 production [60,61] ↓ proliferation [69–71] ↑ exhaustion markers [66–68] |
↓TFH signature [57] ↓IFNγ production [55,60] | HIV-specific CD4+ T cells are enriched in HIV-1 infection [55] |
ART, antiretroviral therapy; COVID-19, coronavirus disease 2019; GALT, gut-associated lymphoid tissue; ISG, interferon-stimulated gene; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2; TNF, tumor necrosis factor.
FIGURE 2.
Single-cell methods characterize HIV-1-infected CD4+ T cells. (a–d) Targeted capture of HIV-1-infected cells from HIV-1-infected individuals. (a) The PCR-activated cell sorting by the Abate group [128]: using a microfluidics device, droplets of single cells that are HIV-1 DNA PCR positive are sorted for single-cell RNAseq or single-cell DNAseq. This method is currently under development. (b) HIV-1 SortSeq by the Ho group [106▪] identifies HIV-1 RNA+ cells using a combination of 192 fluorescently labeled probes targeting 5′ and 3′ HIV-1 RNA for single-cell RNAseq. HIV-1 integration site may be identified by mapping HIV-1-host chimeric RNA junction. (c) LURE by the Nussenzweig group [125▪] identifies HIV-1 Env expressing cells using broadly neutralizing antibody-based magnetic enrichment and fluorescence-based cytometric sorting. (d) HIV-1 RNA flow-FISH by the Kaufmann group [115] identifies HIV-1 Gag expressing cells using a combination of HIV-1 RNA fluorescent in-situ hybridization (FISH) (targeting gag-pol) and HIV-1 p24 protein staining. (e and f) Large-scale single-cell profiling with paired HIV-1 RNA mapping. (e) Microwell-based profiling, such as Seq-Well by the Shalek group, encloses single cells with barcoded beads in microwells for single-cell RNAseq. (f) Droplet-based profiling, such as that commercialized by 10× Genomics, and single-cell multiomic method ECCITEseq developed by the Smibert group [134], was applied to HIV-1 clinical samples by the Ho group [137] and profiled surface protein expression, cellular transcriptome, T cell receptor, and HIV-1 RNA in the same cell. AAAA, poly(A) tail. TCR, T cell receptor. TSO, template switch oligonucleotide. rGrGrG, 3 riboguanosines (rGrGrG). UMI, unique molecular identifiers, are molecular tags used to detect and quantify unique RNA transcripts.
CYTOKINE DYSREGULATION DRIVES CD4+ T CELL RESPONSES
Acute viral infection induces innate immune responses through pattern recognition receptors, type I interferon, proinflammatory cytokines, and interferon-stimulated gene (ISG) expression. While these antiviral programs hinder viral replication, prolonged cytokinemia induces overactivation of immune cells and tissue damage. Acute HIV-1 infection induces a surge in type I interferon responses within the first 2 weeks of infection, along with increased IFNγ, tumor necrosis factor (TNF), monocyte chemoattractant protein-1 (MCP-1), soluble IL-2 receptor, IL-6, IL-8, IL-10, and IL-12 plasma levels [11,12▪,13]. Early ART substantially reduces cytokinemia to basal levels during acute infection [11,12▪]. During untreated chronic HIV-1 infection, IFNγ, TNF-α, TGF-β, IL-4, IL-6, and IL-10 plasma levels are elevated. After 12 months of ART suppression, IL-4, IL-6, and IL-10 plasma levels return to normal, but IFNγ, TNF-α, and TGF-β levels remain elevated [13,14]. Therefore, although HIV-1-infected individuals do not experience cytokine storms involving multiorgan dysfunction, chronic inflammation contributes to long-term complications such as accelerated atherosclerosis [15].
Unlike HIV-1 infection, cytokine dysregulation in SARS-CoV-2 infection induces cytokine release syndrome (or cytokine storm), an acute systemic inflammatory syndrome that involves multiorgan dysfunction [16,17]. SARS-CoV-2-induced cytokine release syndrome, involving increased IL-1α, IL-1β, IL-6, IL-10, IL-18, and TNF plasma levels, is strongly associated with disease severity [18▪▪,19,20]. Unlike HIV-1 and influenza infections in which IFNα peaks early and initiates antiviral programs, IFNα peaks relatively late (around 2 weeks after symptom onset) in severe SARS-CoV-2-infected patients [18▪▪,21]. This is consistent with the observation that the late IFNα peak fails to limit viral infection and induces CD4+ and CD8+ T cell depletion in SARS-CoV-1 infection [22]. In another study, IFNα levels are decreased in severe SARS-CoV-2-infected patients: the lower plasma level of IFNα in these severe patients is associated with lower ISG expression and higher levels of SARS-CoV-2 viremia [23▪], potentially due to inborn errors in type I interferon immunity [24] or the development of autoantibodies against type I interferon [25]. Overall, SARS-CoV-2 infection drives a decrease in antiviral cytokines (such as IFNα) and an increase in proinflammatory cytokines (such as IL-6) as the hallmark of COVID-19 [20].
MECHANISMS OF CD4+ T CELL DEPLETION
Both SARS-CoV-2 and HIV-1 infection lead to CD4+ T cell depletion. However, the underlying causes are very different. CD4+ T cell loss is the hallmark of untreated HIV-1 infection. A peripheral CD4+ T cell count less than 200/μl defines AIDS in which overt opportunistic infections occur [26]. In HIV-1 infection, CD4+ T cells are profoundly depleted due to Fas/Fas-ligand mediated apoptosis during direct infections [4,27] and pyroptosis during abortive viral infections [28]. The depletion of CD4+ T cells in the gut-associated lymphoid tissue [29] compromises gut epithelial barrier integrity, increases bacterial translocation, and causes persistent immune activation [30–34]. Chronic immune activation induces lymphoid tissue fibrosis [35,36], limits T cell access to homeostatic cytokine IL-7, and causes CD4+ T cell homeostasis failure. As the structural damage to lymphoid tissues is irreversible, HIV-induced CD4+ T cell depletion can be restored in most but not all infected individuals despite ART [37,38].
Mechanisms of CD4+ T cell depletion remain incompletely understood in SARS-CoV-2 infection. In severe COVID-19 patients, the level of CD4+ T cell depletion can be as low as ~200/μl in the peripheral blood, approaching the level seen in advanced HIV-1 infection [19,39▪,40]. The CD4+ T cell count is significantly lower in severe COVID-19 cases than in mild and moderate cases, indicating that CD4+ T cell reduction is associated with disease severity [19,39▪]. Preliminary studies suggest that increased apoptosis [23▪,41,42] secondary to systemic inflammation may be a cause of SARS-CoV-2-induced CD4+ T cell depletion. Unlike HIV-1 infection, CD4+ T cell depletion resolves after recovery in SARS-CoV-2 infection [39▪,43].
SINGLE-CELL IMMUNE LANDSCAPE OF IMMUNOLOGIC RESPONSES OF HIV-1 VERSUS SARS-COV-2 INFECTION
Advancement in single-cell RNAseq methods allows genome-wide profiling of heterogeneous immune cells down to the single-cell level, including detailed characterization of novel or previously unknown cell types. Bioinformatic and computational analysis enables integration and correction of interindividual variabilities and identification of differentially expressed genes and immune signaling pathways. Thus, single-cell data sets from very few study participants from carefully designed clinical studies may provide unbiased discovery of the landscape of immune responses and disease mechanisms.
Consistent with the IFNα surge in the plasma during acute HIV-1 infection, single-cell RNAseq of peripheral blood mononuclear cells (PBMCs) reveals upregulation of an interferon response signature in multiple cell types in acute HIV-1 infection [44▪▪]. CD4+ T cells upregulate a gene module of ISGs (such as ISG15, IFI44, IFI44L, IFI6, IFIT1, IFIT3, MX1, XAF1, S100A8, S100A9). Upstream regulator analysis reveals that the transcriptional program in CD4+ T cells is driven by IFNα, IFNγ, TNF, TGF-β, CD3, KRAS, IL-1β, IL-2, IL-4, IL-6, and oncostatin [44▪▪], reflecting that plasma cytokines and T cell activation drive CD4+ T cell responses during acute HIV-1 infection. Of note, during acute HIV-1 infection, CD4+ T cells are the major mediators of TNF-α and IL-1β responses in the peripheral blood [44▪▪]. During chronic HIV-1 infection, a subset of CD4+ T cells express exhaustion markers (TIGIT, CTLA4), although studies from more study participants may be needed to identify the upstream regulators of such immune exhaustion phenotypes [45▪].
High-dimensional profiling of PBMCs from COVID-19 patients using flow cytometry reveals increased frequency of activated and proliferating CD4+ T cells (expressing human leukocyte antigen – DR isotype (HLA-DR), CD38, and Ki67) [46▪]. By incorporating plasma cytokine levels in high-dimensional flow cytometry-based PBMC profiling, a longitudinal analysis showed that moderate COVID-19 patients exhibit type 1 antiviral immune responses which decrease as the patient recovers [18▪▪]. However, severe COVID-19 patients not only exhibit type 1 (IFNα) and type 3 (IFNλ) immune responses but also type 2 immune responses (IL-5 and IL-13) [18▪▪], suggesting that misfiring of type 2 immune responses directs the clinical trajectory toward severe COVID-19.
Single-cell profiling of PBMCs from COVID-19 patients reveals increased frequency of clonally expanded Th1-like cytotoxic CD4+ T lymphocytes (expressing GZMA, GZMB, GZMH, and GNLY) [47]. CD4+ T cells upregulate ISGs similar to those seen in acute HIV-1 infection, such as ISG15, IFI44L, IFIT3, MX1, XAF1, S100A8, and S100A9 [48,49,50▪▪], which were not seen after recovery [51]. By comparing the single-cell transcriptome profile between COVID-19 patients and severe influenza-infected patients, TNF/IL-1β-driven inflammation characterizes COVID-19 but not severe influenza infection [52▪▪]. Treatment with IL-6R blockade tocilizumab reduces S100A8 and S100A9 expression and potentially breaks the IL-6/S100A8/9 feedback loop [50▪▪]. Overall, single-cell profiling identifies the unique immune effectors (cytotoxic CD4+ T cells) and drivers of immune dysfunction in COVID-19.
ANTIGEN-SPECIFIC CD4+ TH1 RESPONSES
Antigen-specific CD4+ T cells provide help to rescue exhausted CD8+ T cells during chronic viral infection [53] and correlate with viral control [54]. Antigen-specific CD4+ T cells are typically isolated by flow cytometry using effector cytokines (using intra-cellular IL-2, IFNγ, or TNF staining) [55], surface activation-induced markers (AIM) such as CD69, CD154 (CD40L), and CD137 (4-1BB) [56–58], or cellular proliferation [59] upon ex vivo antigen peptide stimulation. The frequency of HIV-1-specific CD4+ T cells in HIV-1-infected individuals is ~0.1–0.7% measured by AIM, and only 0.2% express IFNγ [55,57]. Viremia increases CD4+ T cell production of IFNγ but not IL-2, stunting antigen-specific T cell proliferation despite high antigen levels [60,61]. The frequency of IFNγ-secreting HIV-specific CD4+ T cells further declines after ART [55,60]. In contrast, the same HIV-1-infected individuals have a higher frequency of cytomegalovirus (CMV)-specific CD4+ T cells (1.5%) [57]. Although HIV-1-specific CD4+ T cells can last for decades due to persistent antigen stimulation [62–64], these HIV-1-specific CD4+ T cells are dysfunctional [65], expressing exhaustion markers [66–68] with impaired proliferation capacity [69–71]. Immune checkpoint inhibitors, such as program cell death-1 (PD-1) blockade, reverse immune exhaustion, and restore HIV-1-specific CD4+ T cell proliferation capacity [62,72].
The frequency of SARS-CoV-2-specific CD4+ T cells in the recovery phase is ~0.2–2% when defined by cytokine production [73▪] and AIM [74▪▪]. SARS-CoV-2-specific CD4+ T cells exhibit robust Th1 function, express IL-2, IFNγ, and TNFα, and maintain proliferative capacities [73▪,74▪▪,75,76▪▪,77▪,78,79]. In particular, SARS-CoV-2-specific CD4+ T cells are enriched in cytotoxic CD4+ T cells expressing PRF1, GZMB, GNLY, NKG7, XCL1, and XCL2 [80▪]. SARS-CoV-2-specific CD4+ T cells from unexposed individuals, likely the result of cross-reactive stimulation from prior common cold coronavirus infection (such as OC43, HKU1, NL63, and 229E), are functional in IFNγ production and are inferred as a potential reason for cross-protection [77▪,81]. Importantly, the frequency of SARS-CoV-2 Spike-specific CD4+ T cells correlates with antibody titers (anti-Spike receptor binding domain IgG) [74▪▪,82▪▪]. While it is unclear how long SARS-CoV-2-specific CD4+ T cells will last, long-term studies from SARS-CoV-1 infection showed that, although SARS-CoV-1-specific IgG vanished, 60% of infected individuals have SARS-CoV-1-specific T cell responses for up to 6 years [83] or even 17 years [77▪]. One caveat is that while SARS-CoV-1 [84] specific CD4+ T cells can be polyfunctional, dysregulation of SARS-CoV-1-specific responses toward Th2 but not Th1 can be seen in some fatal cases [84]. This skewing from Th1 toward Th2 immune responses is concerning, as some SARS-CoV-1 vaccines induces immunopathologic Th2 responses instead of immune protective Th1 responses [85,86].
CD4+ T CELL HELP FOR B CELL-MEDIATED HUMORAL IMMUNE RESPONSES
Follicular helper CD4+ T cells (TFH) provide critical help for B cell-mediated humoral immunity, particularly in the germinal center, by facilitating B cell proliferation, differentiation, antibody affinity maturation, and class-switching [87]. Although TFH express CXCR5 and costimulatory molecules such as PD-1 and inducible T cell costimulator (ICOS), the definition of TFH relies on transcription factor Bcl6 expression [88] and IL-21 production [89]. HIV-1 viremia drives the expansion of TFH cells mainly in the lymphoid tissue [90], although TFH-like CD4+ T cells in the peripheral blood can sometimes be identified [57,91]. HIV-1-specific TFH are associated with protective antibody responses [91] and enhanced HIV-specific CD8+ T cell function measured by perforin production [92]. However, since CD8+ cytotoxic T cells lack the homing marker CXCR5 to reach TFH in the B cell follicle of lymphoid tissues [93], TFH become a source of the HIV-1 reservoir at the immune sanctuary site [94].
In COVID-19 patients who recovered, the frequency of Spike-specific TFH cells in peripheral blood correlates with neutralizing antibody titer, suggesting protective immunity [95]. A remarkable proportion of SARS-CoV-2-specific CD4+ T cells in the peripheral blood exhibit TFH phenotypes, such as IL21 expression [80▪]. However, in COVID-19 patients who died early (within 10 days of infection), there is an absence of germinal center formation, a block of TFH differentiation, and a remarkable reduction of Bcl6+ germinal center B cells in the lymphoid tissues [96▪▪]. This suggests that a lack of germinal centers and TFH is associated with deficient antibody responses and high mortality in COVID-19.
UNDERSTANDING HIV-1-INFECTED CD4+ T CELLS AT SINGLE-CELL RESOLUTION
To determine the transcriptional state induced by HIV-1 and to overcome the sparsity of in vivo samples, in vitro models and single-genome sequencing methods have been developed. Two groups applied single-cell RNA sequencing to in vitro-infected primary cells [97,98]. However, the findings are hampered by the heterogeneity associated between different individuals.
Studying HIV-1-infected cells from HIV-1-infected individuals provides the most clinically relevant understanding of HIV-1 persistence [99–105,106▪]. However, this translational approach has been extremely challenging. First, HIV-1-infected cells are extremely rare: ~100/million (0.01%) CD4+ T cells harbor an inducible HIV-1 provirus, and ~1000/million (0.1%) CD4+ T cells harbor a defective HIV-1 provirus [103,107]. Second, no cellular marker that can distinguish the rare HIV-1-infected cells from the 99.9% uninfected cells has been identified [108–110]. Third, HIV-1-infected cells are extremely heterogeneous, each having an HIV-1 provirus at a different integration site [99,111] and a different T cell functional state [112,113]. An advancement in single-cell methods is required to capture these rare HIV-1-infected cells.
Flow cytometry-based approaches have enabled some inferences into the HIV-1-infected cell landscape. During viremia, HIV-1 primarily infects activated CD4+ T cells. These cells are more likely to express activation markers (CD25, HLA-DR), proliferation markers (Ki67), immune checkpoint molecules (PD-1, LAG-3, TIGIT, Tim-3), CXCR5, and integrins α4β7 and α4β1 [114,115]. After ART suppression, HIV-1-infected cells may upregulate PD-1, TIGIT, and integrin α4β1 [114]. Other markers, such as CD2 [116], CD30 [117], CD161 [118], and CCR5 [119], have been shown to be enriched in HIV-1-infected cells, although these markers are insufficient to distinguish HIV-1-infected cells from uninfected cells. The Roan group developed a machine learning algorithm PP-SLIDE and a 37-parameter mass cytometry technique to identify a combination of protein markers to enrich for latently infected cells [120,121]. Using this model, a combination of protein markers (such as CD45RA−, CD45RO+, CD62L−, CCR7+, PD1+, TIGIT+, CD49D+ (integrin α4), CD28+, CCR5+ cells) (likely reflecting memory CD4+ T cells expressing activation, exhaustion, and homing markers) may enrich HIV-1-infected cells up to 100 fold in two out of three study participants examined [121]. Of note, the combination of protein markers is individual-dependent. One caveat is that this protein-based detection and algorithms may not be sufficient to recapitulate the heterogeneity of cellular states related to the breadth of tens of thousands of HIV-1 integration sites and the T cell antigen specificity in vivo.
To identify drivers for the proliferation of HIV-1-infected cells [122], several groups have developed flow cytometric sorting-based methods to determine the T cell antigen specificity and clonality [using T cell receptor (TCR) sequencing] and HIV-1 integration site, inducibility, and genome integrity (using single-genome sequencing) [121,123,124]. These studies demonstrated that latently infected cells are predominantly in the central and effector memory cells [123], and that antigen is a major driver for the clonal expansion of HIV-1-infected cells [124].
Since HIV-1-infected cells lack a specific cellular marker for effective enrichment, a breakthrough in the field is to use HIV-1 DNA (Fig. 2a), HIV-1 viral RNA (Fig. 2b and c), or protein expression (Fig. 2c and d) for sorting and subsequent single-cell profiling. HIV-1 SortSeq uses fluorescent in-situ hybridization (FISH)-based HIV-1 RNA staining after 16 h of 13-phorbol-12-myristate acetate (PMA)/ionomycin induction of HIV-1 RNA expression (Fig. 2b) [106▪]. Using a method similar to single-molecule FISH, cells are fixed and permeabilized for FISH probes to enter the cells and hybridize to HIV-1 RNA. The benefit of this approach is the use of RNA-preserving procedures and a total of 192 fluorophore-tagged probes to increase HIV-1 RNA signal intensity and to overcome HIV-1 sequence diversity in clinical samples. The caveat is that the fixation process reduces RNA quality. The use of dead cell staining and colocalization of both 5′ and 3′ HIV-1 RNA probes reduces nonspecific staining. Using SMARTseq-based library preparation, which involves DNase treatment, total RNA amplification, and ribosomal RNA depletion, HIV-1 SortSeq sequences a small number of HIV-1-infected cells but in depth, detecting early events during latency reversal such as HIV-1-driven aberrant transcription of cancer-related genes as a mechanism for integration site-related proliferation [106▪].
LURE uses broadly neutralizing antibody (bNAb)-based magnetic capture and fluorescence-tagged antibody staining of HIV-1 Env after 30 h of phytohemagglutinin (PHA) activation to induce Env expression (Fig. 2c) [125▪]. The protein capture-based LURE captures late activation markers such as TIGIT and HLA-DRB [125▪]. The benefit of LURE is that the cells remain viable for downstream cell culture. The caveat is that the capture efficiency depends on whether the diverse HIV-1 Env can be captured by bNAbs, as up to 18% of HIV-1-infected individuals may have HIV-1 strains that are resistant to bNAbs [126].
In both HIV-1 SortSeq and LURE, a unique transcriptional profile differentiates HIV-1-induced cells from uninduced and likely uninfected cells from the same HIV-1-infected individuals. Although each study reports more than 400 differentially expressed genes, only 30 genes were identified in both studies, highlighting the heterogeneity of CD4+ T cells and different gene expression during early versus late reactivation. Among them, genes associated with antiviral responses (IRF4, CCL3, CCL4, and XCL1) and cell survival (MIR155HG and ZNF217) are significantly upregulated in HIV-1-infected cells in both studies [106▪,125▪].
By a combination of HIV-1 gag-pol RNA expression (as detected by branched DNA-based RNA FISH) and HIV-1 Gag protein expression on flow cytometry, HIV-1 RNA Flow-FISH [115] can isolate HIV-1-infected cells for HIV-1 DNA genome targeted amplification and TCR sequencing [123] (Fig. 2d) using a method similar to RNAScope [127]. The benefit is that the fluorescent signal intensity is enhanced by branched DNA amplification. The caveat is that the branched DNA amplification step, which involves repeated cycles of heating and denature, damages RNA integrity and prevents subsequent RNAseq.
Of note, these methods require a stimulation step to induce HIV-1 RNA or protein expression and may not reflect the cellular state of truly transcriptionally silent, HIV-1 latently infected cells. A method that can detect the cellular transcriptome in transcriptionally silent HIV-1-infected cells remains unavailable. A potentially feasible solution may involve PCR-amplification of HIV-1 DNA in single-cell droplets and sorting of these HIV-1 DNA-positive cells for transcriptome analysis proposed by Clark and Abate et al. (Fig. 2d) [128]. While this method is still under development, Clark et al. [128] have provided a custom-built platform allowing sorting of single-cell droplets that are positive for HIV-1 DNA PCR for single-cell RNAseq or DNAseq. Whether the RNA integrity after the DNA PCR amplification step is sufficient for genome-wide transcriptome profiling remains to be examined.
As opposed to targeted HIV-1-infected cell capture, high-throughput and large-scale single-cell profiling without the enrichment step may identify HIV-1-infected cells by mapping the transcriptome to HIV-1 RNA (Fig. 2e and f). Current methods include microwell-based [129], droplet-based [130], and split-pool based [131] platforms for profiling tens of thousands of cells. Among them, two platforms have been used in blood samples from HIV-1-infected individuals. A microwell-based platform Seq-Well [129] (and a subsequent and improved version Seq-Well S^3) [132] encloses individual single cells with barcoded beads (Fig. 2e). With a unique barcode on each bead (which serves as a cellular barcode) and unique molecular identifiers (which denote each copy of RNA and measure RNA count), small amounts of blood and cerebrospinal fluid samples from HIV-1-infected individuals can be profiled for single-cell RNAseq and TCR profiling [44▪▪,133]. This platform allows HIV-1 RNA mapping in single cells, and the method is currently being developed.
Using a droplet-based platform, ECCITEseq [134] (improved from the original version of CITEseq [135]) applies the template-switching activity of Moloney murine leukemia virus (M-MLV) reverse transcriptase for 5′-based RNA synthesis. M-MLV reverse transcriptase switches the template from the polyadenylated RNA to template-switching oligonucleotides. This allows cDNA synthesis, 5′ RNA capture, and, most importantly, the addition of a PCR handle and increased PCR amplification efficiency at the same time [136]. After staining cell surface protein markers with DNA-barcoded surface protein antibodies, ECCITEseq captures surface protein expression, single-cell transcriptome, TCR sequence, and HIV-1 RNA sequence at the same time within the same single cell. Using ECCITEseq, our group identified HIV-1 RNA positive cells from HIV-1-infected individuals for surface protein expression, single-cell RNAseq, and T cell clonal expansion dynamics tracking [137].
CONCLUSION
In summary, both HIV-1 and SARS-CoV-2 infection drive CD4+ T cell dysfunction and evade adaptive immune responses. Advancement in single-cell methods enabled us to identify different mechanisms of virus-induced immune dysfunction in unique immune cell subsets, to nominate indicators of disease severity, and to design immunotherapeutic strategies to revert HIV-1 and SARS-CoV-2-induced immune dysfunction.
KEY POINTS.
HIV-1-specific CD4+ T cells are few and dysfunctional, with impaired IFNγ and IL-2 expression, increased immune exhaustion, and decreased proliferation capacity.
SARS-CoV-2-specific CD4+ T cells are functional and are enriched in a cytotoxic phenotype, expressing GZMA, GZMB, GZMH, and GNLY.
The frequency of SARS-CoV-2 Spike-specific CD4+ T cells correlates with antibody responses as a potential indicator for protective immunity.
Misfiring of SARS-CoV-2 immunity toward type 2 instead of type 1 (antiviral) immunity is associated with a clinical trajectory toward severe COVID-19.
Single-cell RNAseq of HIV-1-infected cells using HIV-1 SortSeq and LURE identified upregulation of antiviral response genes (IRF4, CCL3, CCL4, and XCL1) and cell survival genes (MIR155HG and ZNF217) in HIV-1-infected cells upon latency reversal.
Acknowledgements
We thank all study participants in HIV-1 and SARS-CoV-2 studies.
Financial support and sponsorship
The current work is supported by Yale Top Scholar, Rudolf J. Anderson Fellowship, NIH R01 AI141009, NIH R61 DA047037, NIH R37 AI147868, NIH R01DA051906, NIH UM1 DA051410, NIH CHEETAH P50 AI150464, NIH BEAT-HIV Delaney Collaboratory UM1 AI126620, Gilead HIV Research Scholar Grant, American Foundation for AIDS Research (amfAR 110029-67-RGRL), Lupus Research Alliance – Celgene, and NIH T32 AI055403 (J.A.C. and K.A.).
Footnotes
Conflicts of interest
There are no conflicts of interest.
REFERENCES AND RECOMMENDED READING
Papers of particular interest, published within the annual period of review, have been highlighted as:
▪ of special interest
▪▪ of outstanding interest
- 1.Nakanishi Y, Lu B, Gerard C, et al. CD8+ T lymphocyte mobilization to virus-infected tissue requires CD4+ T cell help. Nature 2009; 462:510–513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Zajac AJ, Blattman JN, Murali-Krishna K, et al. Viral immune evasion due to persistence of activated T cells without effector function. J Exp Med 1998; 188:2205–2213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Sette A, Moutaftsi M, Moyron-Quiroz J, et al. Selective CD4+ T cell help for antibody responses to a large viral pathogen: deterministic linkage of specificities. Immunity 2008; 28:847–858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Brenchley JM, Schacker TW, Ruff LE, et al. CD4+ T cell depletion during all stages of HIV disease occurs predominantly in the gastrointestinal tract. J Exp Med 2004; 200:749–759. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.▪.Vabret N, Britton GJ, Gruber C, et al. Immunology of COVID-19: current state of the science. Immunity 2020; 52:910–941. [DOI] [PMC free article] [PubMed] [Google Scholar]; The timely review article summarized coronavirus disease 2019 (COVID-19) immunology publications and preprints up to June 2020.
- 6.Picelli S Single-cell RNA-sequencing: the future of genome biology is now. RNA Biol 2017; 14:637–650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.▪▪.Ding J, Adiconis X, Simmons SK, et al. Systematic comparison of single-cell and single-nucleus RNA-sequencing methods. Nat Biotechnol 2020; 38:737–746. [DOI] [PMC free article] [PubMed] [Google Scholar]; The study provides head-to-head comparison of the performance of different single-cell platforms. This is a good technical reference regarding the quality of new single-cell methods, such as gene count and unique molecular identifier expectations in cell lines and in primary cells.
- 8.▪▪.Tran HTN, Ang KS, Chevrier M, et al. A benchmark of batch-effect correction methods for single-cell RNA sequencing data. Genome Biol 2020; 21:12. [DOI] [PMC free article] [PubMed] [Google Scholar]; The study provides a guide for choosing integration methods to remove or normalize batch effects between different samples.
- 9.▪▪.Saelens W, Cannoodt R, Todorov H, et al. A comparison of single-cell trajectory inference methods. Nat Biotechnol 2019; 37:547–554. [DOI] [PubMed] [Google Scholar]; The study provides a guide and selections for different single-cell trajectory inference methods, so-called pseudostate or pseudotime.
- 10.▪▪.Luecken MD, Theis FJ. Current best practices in single-cell RNA-seq analysis: a tutorial. Mol Syst Biol 2019; 15:e8746. [DOI] [PMC free article] [PubMed] [Google Scholar]; The study provides a standard protocol and step-by-step reasoning for bioinformatic processing and quality control of single-cell datasets.
- 11.Lama JR, Ignacio RAB, Alfaro R, et al. Clinical and immunologic outcomes after immediate or deferred antiretroviral therapy initiation during primary HIV infection: the sabes randomized clinical study. Clin Infect Dis 2020. doi: 10.1093/cid/ciaa167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.▪.Muema DM, Akilimali NA, Ndumnego OC, et al. Association between the cytokine storm, immune cell dynamics, and viral replicative capacity in hyperacute HIV infection. BMC Med 2020; 18:81. [DOI] [PMC free article] [PubMed] [Google Scholar]; The study reported plasma cytokine profiles, including IFNα surge, during hyperacute HIV-1 infection.
- 13.Sereti I, Krebs SJ, Phanuphak N, et al. Persistent, albeit reduced, chronic inflammation in persons starting antiretroviral therapy in acute HIV infection. Clin Infect Dis 2017; 64:124–131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Osuji FN, Onyenekwe CC, Ahaneku JE, et al. The effects of highly active antiretroviral therapy on the serum levels of pro-inflammatory and anti-inflammatory cytokines in HIV infected subjects. J Biomed Sci 2018; 25:88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Kearns A, Gordon J, Burdo TH, et al. HIV-1-associated atherosclerosis: unraveling the missing link. J Am Coll Cardiol 2017; 69:3084–3098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Moore JB, June CH. Cytokine release syndrome in severe COVID-19. Science 2020; 368:473–474. [DOI] [PubMed] [Google Scholar]
- 17.Pedersen SF, Ho YC. SARS-CoV-2: a storm is raging. J Clin Invest 2020; 130:2202–2205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.▪▪.Lucas C, Wong P, Klein J, et al. Longitudinal analyses reveal immunological misfiring in severe COVID-19. Nature 2020; 584:463–469. [DOI] [PMC free article] [PubMed] [Google Scholar]; The longitudinal studies examines plasma cytokine profiles and peripheral blood mononuclear cell (PBMC) surface markers using high-dimensional flow cytometry, characterized type 1, 2, and 3 immune responses, and correlated these parameters with clinical trajectories of mild-to-severe COVID-19.
- 19.Chen G, Wu D, Guo W, et al. Clinical and immunological features of severe and moderate coronavirus disease 2019. J Clin Invest 2020; 130:2620–2629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Blanco-Melo D, Nilsson-Payant BE, Liu WC, et al. Imbalanced host response to SARS-CoV-2 drives development of COVID-19. Cell 2020; 181:1036–1045.e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Park A, Iwasaki A. Type I and type III interferons – induction, signaling, evasion, and application to combat COVID-19. Cell Host Microbe 2020; 27:870–878. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Channappanavar R, Fehr AR, Vijay R, et al. Dysregulated type I interferon and inflammatory monocyte-macrophage responses cause lethal pneumonia in SARS-CoV-infected mice. Cell Host Microbe 2016; 19:181–193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.▪.Hadjadj J, Yatim N, Barnabei L, et al. Impaired type I interferon activity and inflammatory responses in severe COVID-19 patients. Science 2020; 369:718–724. [DOI] [PMC free article] [PubMed] [Google Scholar]; The study identified the correlation between low IFNα, reduced interferon-stimulated gene (ISG), high levels of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viremia, and COVID-19 severity.
- 24.Zhang Q, Bastard P, Liu Z, et al. Inborn errors of type I IFN immunity in patients with life-threatening COVID-19. Science 2020; 370:eabd4570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Bastard P, Rosen LB, Zhang Q, et al. Auto-antibodies against type I IFNs in patients with life-threatening COVID-19. Science 2020; 370:eabd4585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Masur H, Ognibene FP, Yarchoan R, et al. CD4 counts as predictors of opportunistic pneumonias in human immunodeficiency virus (HIV) infection. Ann Intern Med 1989; 111:223–231. [DOI] [PubMed] [Google Scholar]
- 27.Li Q, Duan L, Estes JD, et al. Peak SIV replication in resting memory CD4+ T cells depletes gut lamina propria CD4+ T cells. Nature 2005; 434:1148–1152. [DOI] [PubMed] [Google Scholar]
- 28.Doitsh G, Galloway NL, Geng X, et al. Cell death by pyroptosis drives CD4 T cell depletion in HIV-1 infection. Nature 2014; 505:509–514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Mattapallil JJ, Douek DC, Hill B, et al. Massive infection and loss of memory CD4+ T cells in multiple tissues during acute SIV infection. Nature 2005; 434:1093–1097. [DOI] [PubMed] [Google Scholar]
- 30.Brenchley JM, Paiardini M, Knox KS, et al. Differential Th17 CD4 T cell depletion in pathogenic and nonpathogenic lentiviral infections. Blood 2008; 112:2826–2835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Estes JD, Harris LD, Klatt NR, et al. Damaged intestinal epithelial integrity linked to microbial translocation in pathogenic simian immunodeficiency virus infections. PLoS Pathog 2010; 6:e1001052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Marchetti G, Bellistrì GM, Borghi E, et al. Microbial translocation is associated with sustained failure in CD4+ T cell reconstitution in HIV-infected patients on long-term highly active antiretroviral therapy. AIDS 2008; 22:2035–2038. [DOI] [PubMed] [Google Scholar]
- 33.Nazli A, Chan O, Dobson-Belaire WN, et al. Exposure to HIV-1 directly impairs mucosal epithelial barrier integrity allowing microbial translocation. PLoS Pathog 2010; 6:e1000852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Nowroozalizadeh S, Månsson F, da Silva Z, et al. Microbial translocation correlates with the severity of both HIV-1 and HIV-2 infections. J Infect Dis 2010; 201:1150–1154. [DOI] [PubMed] [Google Scholar]
- 35.Schacker TW, Nguyen PL, Beilman GJ, et al. Collagen deposition in HIV-1 infected lymphatic tissues and T cell homeostasis. J Clin Invest 2002; 110:1133–1139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Zeng M, Smith AJ, Wietgrefe SW, et al. Cumulative mechanisms of lymphoid tissue fibrosis and T cell depletion in HIV-1 and SIV infections. J Clin Invest 2011; 121:998–1008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Zeng M, Southern PJ, Reilly CS, et al. Lymphoid tissue damage in HIV-1 infection depletes naïve T cells and limits T cell reconstitution after antiretroviral therapy. PLoS Pathog 2012; 8:e1002437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Le T, Wright EJ, Smith DM, et al. Enhanced CD4+ T cell recovery with earlier HIV-1 antiretroviral therapy. N Engl J Med 2013; 368:218–230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.▪.Diao B, Wang C, Tan Y, et al. Reduction and functional exhaustion of T cells in patients with coronavirus disease 2019 (COVID-19). Front Immunol 2020; 11:827. [DOI] [PMC free article] [PubMed] [Google Scholar]; The study identified the correlation between low IFNα, reduced ISG, high levels of SARS-CoV-2 viremia, and COVID-19 severity.
- 40.Giamarellos-Bourboulis EJ, Netea MG, Rovina N, et al. Complex immune dysregulation in COVID-19 patients with severe respiratory failure. Cell Host Microbe 2020; 27:992–1000.e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Chen Y, Feng Z, Diao B, et al. The novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) directly decimates human spleens and lymph nodes. medRxiv 2020; 10.1101/2020.03.27.20045427. [DOI] [Google Scholar]
- 42.Laing AG, Lorenc A, del Molino del Barrio I, et al. A dynamic COVID-19 immune signature includes associations with poor prognosis. Nat Med 2020; 26:1623–1635. [DOI] [PubMed] [Google Scholar]
- 43.Liu J, Li S, Liu J, et al. Longitudinal characteristics of lymphocyte responses and cytokine profiles in the peripheral blood of SARS-CoV-2 infected patients. EBioMedicine 2020; 55:102763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.▪▪.Kazer SW, Aicher TP, Muema DM, et al. Integrated single-cell analysis of multicellular immune dynamics during hyperacute HIV-1 infection. Nat Med 2020; 26:511–518. [DOI] [PMC free article] [PubMed] [Google Scholar]; The study demonstrated the single-cell landscape of PBMC in HIV-infected individuals during acute infection at different time points.
- 45.▪.Wang S, Zhang Q, Hui H, et al. An atlas of immune cell exhaustion in HIV-infected individuals revealed by single-cell transcriptomics. Emerg Microbes Infect 2020; 19:1–77. [DOI] [PMC free article] [PubMed] [Google Scholar]; The study profiles PBMC from HIV-1-infected individuals across a heterogeneous clinical spectrum.
- 46.▪.Mathew D, Giles JR, Baxter AE, et al. Deep immune profiling of COVID-19 patients reveals distinct immunotypes with therapeutic implications. Science 2020; 369:eabc8511. [DOI] [PMC free article] [PubMed] [Google Scholar]; The study profiles PBMC from HIV-1-infected individuals with heterogeneous clinical spectrum.
- 47.Zhang JY, Wang XM, Xing X, et al. Single-cell landscape of immunological responses in patients with COVID-19. Nat Immunol 2020; 21:1107–1118. [DOI] [PubMed] [Google Scholar]
- 48.Wilk AJ, Rustagi A, Zhao NQ, et al. A single-cell atlas of the peripheral immune response in patients with severe COVID-19. Nat Med 2020; 26:1070–1076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Zhu L, Yang P, Zhao Y, et al. Single-cell sequencing of peripheral mononuclear cells reveals distinct immune response landscapes of COVID-19 and influenza patients. Immunity 2020; 53:685–696.e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.▪▪.Unterman A, Sumida TS, Nouri N, et al. Single-cell omics reveals dyssynchrony of the innate and adaptive immune system in progressive COVID-19. medRxiv 2020; 10.1101/2020.07.16.20153437. [DOI] [PMC free article] [PubMed] [Google Scholar]; The study involves single-cell multiomics profiling of COVID-19 patients with and without IL-6R blockade (tocilizumab).
- 51.Wen W, Su W, Tang H, et al. Immune cell profiling of COVID-19 patients in the recovery stageby single-cell sequencing. Cell Discov 2020; 6:31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.▪▪.Lee JS, Park S, Jeong HW, et al. Immunophenotyping of COVID-19 and influenza highlights the role of type I interferons in development of severe COVID-19. Sci Immunol 2020; 5:eabd1554. [DOI] [PMC free article] [PubMed] [Google Scholar]; The study compared the immunological differences between SARS-CoV-2 and severe influenza infection and identified the TNF/IL-1β-driven inflammation in COVID-19.
- 53.Aubert RD, Kamphorst AO, Sarkar S, et al. Antigen-specific CD4 T cell help rescues exhausted CD8 T cells during chronic viral infection. Proc Natl Acad Sci U S A 2011; 108:21182–21187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Rosenberg ES, Billingsley JM, Caliendo AM, et al. Vigorous HIV-1-specific CD4+ T cell responses associated with control of viremia. Science 1997; 278:1447–1450. [DOI] [PubMed] [Google Scholar]
- 55.Pitcher CJ, Quittner C, Peterson DM, et al. HIV-1-specific CD4+ T cells are detectable in most individuals with active HIV-1 infection, but decline with prolonged viral suppression. Nat Med 1999; 5:518–525. [DOI] [PubMed] [Google Scholar]
- 56.Dan JM, Lindestam Arlehamn CS, Weiskopf D, et al. A cytokine-independent approach to identify antigen-specific human germinal center T follicular helper cells and rare antigen-specific CD4+ T cells in blood. J Immunol 2016; 197:983–993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Morou A, Brunet-Ratnasingham E, Dubé M, et al. Altered differentiation is central to HIV-specific CD4+ T cell dysfunction in progressive disease. Nat Immunol 2019; 20:1059–1070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Reiss S, Baxter AE, Cirelli KM, et al. Comparative analysis of activation induced marker (AIM) assays for sensitive identification of antigen-specific CD4 T cells. PLoS One 2017; 12:e0186998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Becattini S, Latorre D, Mele F, et al. Functional heterogeneity of human memory CD4+ T cell clones primed by pathogens or vaccines. Science 2015; 347:400–406. [DOI] [PubMed] [Google Scholar]
- 60.Harari A, Petitpierre SP, Vallelian F, et al. Skewed representation of functionally distinct populations of virus-specific CD4 T cells in HIV-1-infected subjects with progressive disease: changes after antiretroviral therapy. Blood 2004; 103:966–972. [DOI] [PubMed] [Google Scholar]
- 61.Tilton JC, Luskin MR, Johnson AJ, et al. Changes in paracrine interleukin-2 requirement, CCR7 expression, frequency, and cytokine secretion of human immunodeficiency virus-specific CD4+ T cells are a consequence of antigen load. J Virol 2007; 81:2713–2725. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Porichis F, Hart MG, Massa A, et al. Immune checkpoint blockade restores HIV-specific CD4 T cell help for NK cells. J Immunol 2018; 201:971–981. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Pourgheysari B, Khan N, Best D, et al. The cytomegalovirus-specific CD4+ T cell response expands with age and markedly alters the CD4+ T cell repertoire. J Virol 2007; 81:7759–7765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Douek DC, Brenchley JM, Betts MR, et al. HIV preferentially infects HIV-specific CD4+ T cells. Nature 2002; 417:95–98. [DOI] [PubMed] [Google Scholar]
- 65.Lane HC, Depper JM, Greene WC, et al. Qualitative analysis of immune function in patients with the acquired immunodeficiency syndrome. Evidence for a selective defect in soluble antigen recognition. N Engl J Med 1985; 313:79–84. [DOI] [PubMed] [Google Scholar]
- 66.Kassu A, Marcus RA, D’Souza MB, et al. Regulation of virus-specific CD4+ T cell function by multiple costimulatory receptors during chronic HIV infection. J Immunol 2010; 185:3007–3018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Kaufmann DE, Kavanagh DG, Pereyra F, et al. Upregulation of CTLA-4 by HIV-specific CD4+ T cells correlates with disease progression and defines a reversible immune dysfunction. Nat Immunol 2007; 8:1246–1254. [DOI] [PubMed] [Google Scholar]
- 68.Day CL, Kaufmann DE, Kiepiela P, et al. PD-1 expression on HIV-specific T cells is associated with T cell exhaustion and disease progression. Nature 2006; 443:350–354. [DOI] [PubMed] [Google Scholar]
- 69.Wilson JD, Imami N, Watkins A, et al. Loss of CD4+ T cell proliferative ability but not loss of human immunodeficiency virus type 1 specificity equates with progression to disease. J Infect Dis 2000; 182:792–798. [DOI] [PubMed] [Google Scholar]
- 70.Palmer BE, Boritz E, Blyveis N, et al. Discordance between frequency of human immunodeficiency virus type 1 (HIV-1)-specific gamma interferon-producing CD4+ T cells and HIV-1-specific lymphoproliferation in HIV-1-infected subjects with active viral replication. J Virol 2002; 76:5925–5936. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.McNeil AC, Shupert WL, Iyasere CA, et al. High-level HIV-1 viremia suppresses viral antigen-specific CD4+ T cell proliferation. Proc Natl Acad Sci U S A 2001; 98:13878–13883. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.D’Souza M, Fontenot AP, Mack DG, et al. Programmed death 1 expression on HIV-specific CD4+ T cells is driven by viral replication and associated with T cell dysfunction. J Immunol 2007; 179:1979–1987. [DOI] [PubMed] [Google Scholar]
- 73.▪.Peng Y, Mentzer AJ, Liu G, et al. Broad and strong memory CD4+ and CD8+ T cells induced by SARS-CoV-2 in UK convalescent individuals following COVID-19. Nat Immunol 2020; 21:1336–1345. [DOI] [PMC free article] [PubMed] [Google Scholar]; The study characterized the SARS-CoV-2-specific CD4+ T cell responses and found that SARS-CoV-2-specific CD4+ T cell actually functional.
- 74.▪▪.Grifoni A, Weiskopf D, Ramirez SI, et al. Targets of T cell responses to SARS-CoV-2 coronavirus in humans with COVID-19 disease and unexposed individuals. Cell 2020; 181:1489–1501.e15. [DOI] [PMC free article] [PubMed] [Google Scholar]; This is the first study that mapped SARS-CoV-2-specific T cell responses. Importantly, they found that the frequency of SARS-CoV-2 spike-specific CD4+ T cells correlates with anti-Spike receptor binding domian (RBD) antibody titer.
- 75.Ni T, Yang W, Han M, et al. Global intron retention mediated gene regulation during CD4+ T cell activation. Nucleic Acids Res 2016; 44:6817–6829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.▪▪.Braun J, Loyal L, Frentsch M, et al. SARS-CoV-2-reactive T cells in healthy donors and patients with COVID-19. Nature 2020. doi: 10.1038/s41586-020-2598-9. [DOI] [PubMed] [Google Scholar]; This is the first study that mapped SARS-CoV-2-specific T cell responses. Importantly, they found that the frequency of SARS-CoV-2 spike-specific CD4+ T cells correlate with anti-spike RBD antibody titer.
- 77.▪.Le Bert N, Tan AT, Kunasegaran K, et al. SARS-CoV-2-specific T cell immunity in cases of COVID-19 and SARS, and uninfected controls. Nature 2020; 584:457–462. [DOI] [PubMed] [Google Scholar]; The study mapped SARS-CoV-2-specific CD4+ T cells using antigens other than the Spike region. This study also reported SARS-CoV-1-specific CD4+ T cells up to 17 years after infection.
- 78.Weiskopf D, Schmitz KS, Raadsen MP, et al. Phenotype and kinetics of SARS-CoV-2-specific T cells in COVID-19 patients with acute respiratory distress syndrome. Sci Immunol 2020; 5:eabd2071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Neidleman J, Luo X, Frouard J, et al. SARS-CoV-2-specific T cells exhibit phenotypic features of robust helper function, lack of terminal differentiation, and high proliferative potential. Cell Rep Med 2020; 1:100081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.▪.Meckiff BJ, Ramírez-Suástegui C, Fajardo V, et al. Single-cell transcriptomic analysis of SARS-CoV-2 reactive CD4+ T cells. bioRxiv 2020; 10.1101/2020.06.12.148916. [DOI] [PMC free article] [PubMed] [Google Scholar]; The study used single-cell profiling on SARS-CoV-2-specific CD4+ T cells and identified a remarkable proportion of cells exhibiting cytotoxic CD4+ T cell phenotypes (expressing granzyme-related genes).
- 81.Mateus J, Grifoni A, Tarke A, et al. Selective and cross-reactive SARS-CoV-2 T cell epitopes in unexposed humans. Science 2020; 370:89–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.▪▪.Rydyznski Moderbacher C, Ramirez SI, Dan JM, et al. Antigen-specific adaptive immunity to SARS-CoV-2 in acute COVID-19 and associations with age and disease severity. Cell 2020. doi: 10.1016/j.cell.2020.09.038. [DOI] [PMC free article] [PubMed] [Google Scholar]; The study demonstrated that the frequency of SARS-CoV-2-specific CD4+ T cells correlates with anti-Spike RBD antibody titer and highlighted the role of CD4+ T cell in immune control.
- 83.Tang F, Quan Y, Xin ZT, et al. Lack of peripheral memory B cell responses in recovered patients with severe acute respiratory syndrome: a six-year follow-up study. J Immunol 2011; 186:7264–7268. [DOI] [PubMed] [Google Scholar]
- 84.Li CK, Wu H, Yan H, et al. T cell responses to whole SARS coronavirus in humans. J Immunol 2008; 181:5490–5500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Tseng CT, Sbrana E, Iwata-Yoshikawa N, et al. Immunization with SARS coronavirus vaccines leads to pulmonary immunopathology on challenge with the SARS virus. PLoS One 2012; 7:e35421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Bolles M, Deming D, Long K, et al. A double-inactivated severe acute respiratory syndrome coronavirus vaccine provides incomplete protection in mice and induces increased eosinophilic proinflammatory pulmonary response upon challenge. J Virol 2011; 85:12201–12215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Crotty S A brief history of T cell help to B cells. Nat Rev Immunol 2015; 15:185–189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Nurieva RI, Chung Y, Martinez GJ, et al. Bcl6 mediates the development of T follicular helper cells. Science 2009; 325:1001–1005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Vogelzang A, McGuire HM, Yu D, et al. A fundamental role for interleukin-21 in the generation of T follicular helper cells. Immunity 2008; 29:127–137. [DOI] [PubMed] [Google Scholar]
- 90.Lindqvist M, van Lunzen J, Soghoian DZ, et al. Expansion of HIV-specific T follicular helper cells in chronic HIV infection. J Clin Invest 2012; 122:3271–3280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Schultz Bruce T, Teigler Jeffrey E, Pissani F, et al. Circulating HIV-specific interleukin-21+CD4+ T cells represent peripheral Tfh cells with antigen-dependent helper functions. Immunity 2016; 44:167–178. [DOI] [PubMed] [Google Scholar]
- 92.Chevalier MF, Jülg B, Pyo A, et al. HIV-1-specific interleukin-21+ CD4+ T cell responses contribute to durable viral control through the modulation of HIV-specific CD8+ T cell function. J Virol 2011; 85:733–741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Fukazawa Y, Lum R, Okoye AA, et al. B cell follicle sanctuary permits persistent productive simian immunodeficiency virus infection in elite controllers. Nat Med 2015; 21:132–139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Perreau M, Savoye AL, De Crignis E, et al. Follicular helper T cells serve as the major CD4 T cell compartment for HIV-1 infection, replication, and production. J Exp Med 2013; 210:143–156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Juno JA, Tan HX, Lee WS, et al. Humoral and circulating follicular helper T cell responses in recovered patients with COVID-19. Nat Med 2020; 26:1428–1434. [DOI] [PubMed] [Google Scholar]
- 96.▪▪.Kaneko N, Kuo HH, Boucau J, et al. Loss of Bcl-6-expressing T follicular helper cells and germinal centers in COVID-19. Cell 2020; 183:143–157.e13. [DOI] [PMC free article] [PubMed] [Google Scholar]; This is an important study showing the lymph node histopathology in COVID-19 patients who died early (within 10 days) of infection. The lack of germinal center TFH cells suggests lack of low antibody titer, lack of protective immunity, and early fatality in severe COVID-19.
- 97.Golumbeanu M, Cristinelli S, Rato S, et al. Single-cell RNA-Seq reveals transcriptional heterogeneity in latent and reactivated HIV-infected cells. Cell Rep 2018; 23:942–950. [DOI] [PubMed] [Google Scholar]
- 98.Bradley T, Ferrari G, Haynes BF, et al. Single-cell analysis of quiescent HIV infection reveals host transcriptional profiles that regulate proviral latency. Cell Rep 2018; 25:107–117.e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Jiang C, Lian X, Gao C, et al. Distinct viral reservoirs in individuals with spontaneous control of HIV-1. Nature 2020; 585:261–267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Bruner KM, Wang Z, Simonetti FR, et al. A quantitative approach for measuring the reservoir of latent HIV-1 proviruses. Nature 2019; 566:120–125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Bender AM, Simonetti FR, Kumar MR, et al. The landscape of persistent viral genomes in ART-treated SIV, SHIV, and HIV-2 infections. Cell Host Microbe 2019; 26:73–85.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Wang Z, Gurule EE, Brennan TP, et al. Expanded cellular clones carrying replication-competent HIV-1 persist, wax, and wane. Proc Natl Acad Sci U S A 2018; 115:E2575–E2584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Pollack RA, Jones RB, Pertea M, et al. Defective HIV-1 proviruses are expressed and can be recognized by cytotoxic T lymphocytes, which shape the proviral landscape. Cell Host Microbe 2017; 21:494–506.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Ho YC, Shan L, Hosmane NN, et al. Replication-competent noninduced proviruses in the latent reservoir increase barrier to HIV-1 cure. Cell 2013; 155:540–551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Yeh YJ, Jenike KM, Calvi RM, et al. Filgotinib suppresses HIV-1-driven gene transcription by inhibiting HIV-1 splicing and T cell activation. J Clin Invest 2020; 130:4969–4984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.▪.Liu R, Yeh YJ, Varabyou A, et al. Single-cell transcriptional landscapes reveal HIV-1-driven aberrant host gene transcription as a potential therapeutic target. Sci Transl Med 2020; 12:eaaz0802. [DOI] [PMC free article] [PubMed] [Google Scholar]; The study describes HIV-1 SortSeq which captures HIV-1-infected cells upon early latency reversal. This study also identified HIV-1-driven aberrant cancer gene expression at the integration site as a mechanism for integration site-dependent proliferation of HIV-1-infected cells.
- 107.Bruner KM, Murray AJ, Pollack RA, et al. Defective proviruses rapidly accumulate during acute HIV-1 infection. Nat Med 2016; 22:1043–1049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Bertagnolli LN, White JA, Simonetti FR, et al. The role of CD32 during HIV-1 infection. Nature 2018; 561:E17–E19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Osuna CE, Lim SY, Kublin JL, et al. Evidence that CD32a does not mark the HIV-1 latent reservoir. Nature 2018; 561:E20–E28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Perez L, Anderson J, Chipman J, et al. Conflicting evidence for HIV enrichment in CD32(+) CD4 T cells. Nature 2018; 561:E9–E16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Schroder AR, Shinn P, Chen H, et al. HIV-1 integration in the human genome favors active genes and local hotspots. Cell 2002; 110:521–529. [DOI] [PubMed] [Google Scholar]
- 112.Hiener B, Horsburgh BA, Eden JS, et al. Identification of genetically intact HIV-1 proviruses in specific CD4+ T cells from effectively treated participants. Cell Rep 2017; 21:813–822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Lee GQ, Orlova-Fink N, Einkauf K, et al. Clonal expansion of genome-intact HIV-1 in functionally polarized Th1 CD4+ T cells. J Clin Invest 2017; 127:2689–2696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Pardons M, Baxter AE, Massanella M, et al. Single-cell characterization and quantification of translation-competent viral reservoirs in treated and untreated HIV infection. PLoS Pathog 2019; 15:e1007619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Baxter AE, Niessl J, Fromentin R, et al. Single-cell characterization of viral translation-competent reservoirs in HIV-infected individuals. Cell Host Microbe 2016; 20:368–380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Iglesias-Ussel M, Vandergeeten C, Marchionni L, et al. High levels of CD2 expression identify HIV-1 latently infected resting memory CD4+ T cells in virally suppressed subjects. J Virol 2013; 87:9148–9158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Hogan LE, Vasquez J, Hobbs KS, et al. Increased HIV-1 transcriptional activity and infectious burden in peripheral blood and gut-associated CD4+ T cells expressing CD30. PLoS Pathog 2018; 14:e1006856. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Li X, Liu Z, Li Q, et al. CD161(+) CD4+ T cells harbor clonally expanded replication-competent HIV-1 in antiretroviral therapy-suppressed individuals. mBio 2019; 10:e02121–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Shan L, Deng K, Gao H, et al. Transcriptional reprogramming during effector-to-memory transition renders CD4+ T cells permissive for latent HIV-1 infection. Immunity 2017; 47:766–775.e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Cavrois M, Banerjee T, Mukherjee G, et al. Mass cytometric analysis of HIV entry, replication, and remodeling in tissue CD4+ T cells. Cell Rep 2017; 20:984–998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Neidleman J, Luo X, Frouard J, et al. Phenotypic analysis of the unstimulated in vivo HIV CD4 T cell reservoir. Elife 2020; 9:e60933. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Liu R, Simonetti FR, Ho YC. The forces driving clonal expansion of the HIV-1 latent reservoir. Virol J 2020; 17:4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Gantner P, Pagliuzza A, Pardons M, et al. Single-cell TCR sequencing reveals phenotypically diverse clonally expanded cells harboring inducible HIV proviruses during ART. Nat Commun 2020; 11:4089. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Simonetti FR, Zhang H, Soroosh H, et al. Antigen-driven clonal selection shapes the fate of HIV-infected CD4+ T cells in vivo. Conferences on Retroviruses and Opportunistic Infections. Boston, MA, USA. 2020. [Google Scholar]
- 125.▪.Cohn LB, da Silva IT, Valieris R, et al. Clonal CD4+ T cells in the HIV-1 latent reservoir display a distinct gene profile upon reactivation. Nat Med 2018; 24:604–609. [DOI] [PMC free article] [PubMed] [Google Scholar]; The study described the LURE assay involving the use of broadly neutralizing antibodies to capture HIV-1-infected cells after 30 h of ex-vivo activation. This method identified HIV-1-infected cells for single-cell RNAseq.
- 126.Mendoza P, Gruell H, Nogueira L, et al. Combination therapy with anti-HIV-1 antibodies maintains viral suppression. Nature 2018; 561:479–484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Wang F, Flanagan J, Su N, et al. RNAscope: a novel in situ RNA analysis platform for formalin-fixed, paraffin-embedded tissues. J Mol Diagn 2012; 14:22–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Clark IC, Abate AR. Finding a helix in a haystack: nucleic acid cytometry with droplet microfluidics. Lab Chip 2017; 17:2032–2045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Gierahn TM, Wadsworth MH, Hughes TK, et al. Seq-Well: portable, low-cost RNA sequencing of single cells at high throughput. Nat Methods 2017; 14:395–398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Macosko Evan Z, Basu A, Satija R, et al. Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell 2015; 161:1202–1214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Rosenberg AB, Roco CM, Muscat RA, et al. Single-cell profiling of the developing mouse brain and spinal cord with split-pool barcoding. Science 2018; 360:176–182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Hughes TK, Wadsworth MH 2nd, Gierahn TM, et al. Second-strand synthesis-based massively parallel scRNA-Seq reveals cellular states and molecular features of human inflammatory skin pathologies. Immunity 2020; 53:878–894.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Farhadian SF, Mehta SS, Zografou C, et al. Single-cell RNA sequencing reveals microglia-like cells in cerebrospinal fluid during virologically suppressed HIV. JCI Insight 2019; 3:e121718. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Mimitou EP, Cheng A, Montalbano A, et al. Multiplexed detection of proteins, transcriptomes, clonotypes and CRISPR perturbations in single cells. Nat Methods 2019; 16:409–412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Stoeckius M, Hafemeister C, Stephenson W, et al. Simultaneous epitope and transcriptome measurement in single cells. Nat Methods 2017; 14:865–868. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Zhu YY, Machleder EM, Chenchik A, et al. Reverse transcriptase template switching: a SMART approach for full-length cDNA library construction. Biotechniques 2001; 30:892–897. [DOI] [PubMed] [Google Scholar]
- 137.Collora JA, Pinto-Santini D, Pasalar S, et al. Single-cell atlas and clonal expansion dynamics of CD4+ T cells during HIV infection. In Conferences on Retroviruses and Opportunistic Infections Boston, MA, USA. 2020. [Google Scholar]