ABSTRACT The monkeypox (MPXV) outbreak in 2022 is more prevalent among individuals with human immunodeficiency virus (HIV). While it is plausible that HIV-induced immunosuppression could result in a more severe progression, the exact mechanisms remain undetermined. To better understand the immunopathology of MPXV in patients with and without HIV infection, we employed single-cell RNA sequencing (scRNA-seq) to analyse peripheral blood mononuclear cells (PBMCs) from six patients hospitalized for MPXV, three of whom had HIV infection (HIV antibody positive and HIV RNA level below the detection limit), and three patients only infected with MPXV (HIV-). We map the peripheral immune response in both the acute phase and the recovery period, showing the reconfiguration of peripheral immune cell phenotypes in acute stage compared with recovery stage, characterized by disturbed cell subsets and intense cell interactions mediated by monocytes and neutrophils. Importantly, we also found obviously dysregulated gene expression and cell subsets in HIV+ patients proposing mechanism underlying their serious condition. Our findings provide a comprehensive cell atlas of MPXV patients, shed light on the mechanisms underlying the severe disease progression and longer recovery time in HIV+ individuals. KEYWORDS: Monkeypox, HIV, single-cell RNA sequencing, immune response, antiviral innate immunity Introduction The monkeypox virus (MPXV) causes a human disease that resembles smallpox and has become the most prevalent Ortho pox virus affecting humans since the eradication of smallpox in 1980 [[41]1]. The 2022 global outbreaks, spanning over 100 countries, have raised significant concerns [[42]2]. The clinical presentation of this virus typically includes fever, rash, lymphadenopathy, and susceptibility to secondary bacterial infections. However, unlike previous outbreaks, recent study indicates that the 2022 MPXV shows epidemiological changes, lineage B.1 drives the outbreak, aided by APOBEC3 in viral evolution and lesion-based transmission fostering variants [[43]3]. Monkeypox infection can occur simultaneously with HIV and SARS-CoV-2 infections [[44]4], and its incidence among men who have sex with men (MSM) is higher, leading to predominantly localized lesions rather than extensive disseminated lesions [[45]5,[46]6]. Actually, among monkeypox cases with HIV, up to about 50% are HIV positive [[47]7]. Emerging data has indicated that such co-infections may lead to worse clinical outcomes and higher mortality rates [[48]8], and the immune response to monkeypox virus has attracted tremendous attention [[49]9]. Understanding the immune profiles of patients with and without HIV is essential for uncovering the shared pathogenesis and developing evidence-based strategies for managing co-infections. Following MPXV infection, the interactions between host immune cells and the pathogen evolve over time, significantly influencing the pathogenesis. Cytokine profiling has indicated an overproduction of certain cytokines in patients with severe MPXV disease, highlighting the critical role of an imbalanced immune response in clinical severity [[50]10]. Immune characteristics are intricately linked to the pathogenesis and disease progression of patients infected with viruses [[51]11]. HIV selectively infects and destroys CD4 T cells, with the number of circulating CD4 T cells in HIV+ individuals predicting the onset of overt immunodeficiency [[52]12]. HIV infection causes persistent depletion of CD4^+ T cells and severe immunodeficiency. Emerging data has indicated that such co-infections may lead to worse clinical outcomes, higher rates of secondary bacterial infection and higher mortality rates [[53]8,[54]13]. The relative study is still limited, and the information available regarding the in-vivo kinetics and crosstalk of immune cells in MPXV-infected individuals is limited. Given the prevalence of co-infections, there is an urgent need to elucidate the kinetics of the immune cells atlas in MPXV patients with or without HIV, to decipher the possible impact of HIV-associated immunocompromise on the progression of MPXV. Single-cell RNA sequencing (scRNA-seq) studies on diverse immune cells across different cohorts significantly contributed to our understanding of immune dysfunction in virus-related physiopathology. It could identify novel cell types and subtypes, discover the intricate diversity of immune cell functions. For instance, it provides in-depth understanding of the molecular and cellular dynamics underlying COVID-19, thereby offering crucial insights for the development of diagnostic tools, therapeutic strategies, and public health interventions [[55]14–16]. scRNA-seq has been utilized for the rapid acquisition of monoclonal antibodies during early outbreaks of MPXV, showing its versatility in pandemic response [[56]17]. However, there has been a notable absence of comprehensive investigations into the immune responses of patients afflicted with MPXV, particularly those co-infected with HIV. Here, we examined the pathological characteristics of patients with MPXV infection and those co-infected with HIV. We compared the peripheral immune cells proportion and gene expression of MPXV patients from acute phase to recovery phase using scRNA-seq, six hospitalized patients in a monocentre from January 2023 to June 2023 were included, three of them were co-infected with HIV. Cell subsets and gene expression were also compared between HIV+ and HIV– patients to decipher the impact of HIV-induced immune dysfunction to MPXV immunopathogenic. We aim to discuss our current understanding of the immune landscape of MPXV-infected patients and the impact of HIV on MPXV immunopathogenesis. Specifically, our findings identify cell type-specific gene expression changes in acute phase compared with recovery phase. We proposed mechanisms underlying the enhanced hospitality of HIV+ patients, including disturbed Mono/DC dynamic decreased NK cells and CD4 T cells, expansion of some CD8 T-cell subsets, and dysregulated gene expression in B cells. Results Characteristics and treatment outcome of patients in the present study Patients diagnosed with monkeypox in the Tianjin Second People’s Hospital since July 2023 according to the Chinese Monkeypox Diagnosis and Treatment Guidelines (2022 Edition). Patients were diagnosed by detecting monkeypox virus DNA through real-time PCR testing of specimens taken from the rash site, and recovery was determined when all rashes had scabbed off. All patients were male, with an average age of 38.17 ± 7.08. Among them, five patients reported a history of homosexual activity within 2 weeks before the onset of symptoms and only one patient had been vaccinated against smallpox. Patients were grouped based on whether the HIV virus was detectable at the time of enrolment. Among them, three cases of HIV+ patients who had been receiving regular highly active antiretroviral therapy (HAART) for over 2 years with undetectable HIV RNA levels and positive HIV antibodies at enrolment. Their clinical characteristics including CD4 counts are listed in [57]Table 1. There is a sharp decrease in CD4 counts of HIV+ patients. Rash areas in two groups were indistinctively observed at genitals, anus, extremities, torso, oropharynx, face and neck, as well as at inguinal lymph nodes. However, a higher incidence of gonococcal and purulent infections, as well as conditions such as anal fissures, was observed among HIV-positive patients. Since there are no specific antiviral drugs for monkeypox currently, besides the HAART drugs required for HIV patients, no other antiviral drugs were used. Treatment mainly focused on symptomatic support and management of complications, with emphasis on local disinfection using povidone-iodine type III, and antibacterial drugs were administered to patients with bacterial infections. The average duration of hospitalization was 15.5 days, with 13 days for the HIV– group and 18 days for the HIV+ group ([58]Table 1). Table 1. Characteristics of patients. Clinical features All patients (n = 6) HIV positive (n = 3) HIV negative (n = 3) Age (years) 38.17 ± 7.08 39.67 ± 9.07 36.67 ± 6.03 Smallpox vaccination [n (%)] 1(16.7) 1(33.3) 0(0) Homosexual activity [n (%)] 5(83.3) 3(100) 2(66.7) Fever time(day) 3.50 ± 2.43 3 ± 2.65 4 ± 2.65 Rash area  Genitals/anus [n (%)] 5(83.3) 2(66.7) 3(100)  Extremities [n (%)] 4(66.7) 1(33.3) 3(100)  Torso [n (%)] 2(33.3) 0(0) 2(66.7)  oropharynx [n (%)] 1(16.7) 0(0) 1(33.3)  Face and neck [n (%)] 5(83.3) 2(66.7) 3(100)  Inguinal Lymphadenopathy [n (%)] 4(66.7) 2(66.7) 2(66.7) Complications  Gonococcal infection [n (%)] 3(50.0) 3(100) 0(0)  Purulent infection [n (%)] 3(50.0) 3(100) 0(0)  Anal fissure [n (%)] 1(16.7) 1(33.3) 0(0)  Perianitis [n (%)] 1(16.7) 1(33.3) 0(0)  Syphilis [n (%)] 2(33.3) 1(33.3) 1(33.3)  Penile contusion [n (%)] 1(16.7) 0(0) 1(33.3)  Hypocythemia [n (%)] 1(16.7) 0(0) 1(33.3) Ct value of Vesicular fluid (Median) 25.57 25.13 26.00 ALT (U/L) 25.5 ± 14.9 13.8 ± 6.6 37.2 ± 9.9 AST (U/L) 23.0 ± 6.9 17.8 ± 3.2 28.2 ± 5.4 GGT (U/L) 35.2 ± 14.8 28.5 ± 9.2 41.8 ± 18.2 ALB (g/L) 42.7 ± 2.9 43.3 ± 1.4 42.2 ± 4.3 CD45 + (n/μL) 1989.9 ± 769.2 1811.1 ± 1065.2 2168 ± 298.8 CD3 + (n/μL) 1435.4 ± 541.1 1270.4 ± 685.1 1600.4 ± 425.5 CD4 + (n/μL) 737.1 ± 316.8 485.3 ± 100.6 988.9 ± 224.6 CD8 + (n/μL) 667.6 ± 421.2 744.8 ± 613.4 590.4 ± 222.1 CD19 + (n/μL) 117.7 ± 42.1 133.8 ± 60.1 101.6 ± 7.1 CD16+/CD56(n/μL)+ 337.2 ± 144.3 235.5 ± 112.9 439.0 ± 90.8 CD4/CD8 1.31 ± 0.6 0.9 ± 0.4 1.7 ± 0.4 HIV-RNA(copies/mL) <20 Negaitive HIV IgM Positive Negative [59]Open in a new tab Note: Data for age and fever days are shown as mean ± SD. Cellular profiles show difference between HIV+ and HIV– groups through MPXV progression To profile the peripheral immune response to MPXV in HIV+ and HIV– patients, we performed scRNA-seq on PBMCs from MPXV individuals in acute and recovery phases. A total of 253,397 cells were retained for the following analysis by removing cells with abnormal RNA count and too many mitochondrial genes. We created a cells-by-genes expression matrix and performed dimensionality reduction by uniform manifold approximation and projection (UMAP) and graph-based clustering, and identified 41 clusters. No cell cluster shows patient individual bias ([60]Figure 1(a)), but cells from patients of different MPXV stage ([61]Figure 1(b)) or HIV infection status ([62]Figure 1(c)) exhibit somewhat different distribution in the embedding space, indicates potential influence of MPXV PBMC composition, and the impact of HIV infection on MPXV immunopathogenesis. Manual annotation based on the canonical markers obtained total of eight cell types, including B cell, CD4 T cell, CD8 T cell, megakaryocyte (Mega), monocyte-derived dendritic cell (MonoDC), neutrophil (Neu), NK cell, and undifferentiated T cells (undiff T), and the distance between the same cell type in the UMAP embedding is obviously close than those between distinct cell types ([63]Figure 1(d)). We next quantified the cells to identify MPXV-derived cell type proportions changes and impact of HIV on them. In MPXV patients, B cells, NK cells, and a subset of T cells showed an obvious increase and CD4 T cells, neutrophil and MonoDC showed significant decrease in the recovery stage compared to the acute stage ([64]Figure 1(e)). Strikingly, though the decrease of CD4 T and CD8 T cell in HIV+ patients was observed, not all cell subset proportions show significant difference (p value >0.05) between HIV+ and HIV– patients ([65]Figure 1(f)). To find the possible impact of HIV on MPXV patients’ PBMC dynamic composition, we further compared the cell compositions between HIV+ and HIV– patients stratified by the MPXV infection status. A significant decrease in CD4+ T cells and expansion of neutrophils were observed in HIV-positive patients during the acute stage of MPXV infection ([66]Figure 1(g)). In the recovery phase, the proportion CD8^+ T cell, Mono/DC and neutrophil was higher in HIV+ patients, while NK cell and undifferentiated T cell (undiff T cell) were still largely inhibited in HIV+ patients ([67]Figure 1(h)). To further represent the enrichment of each cell type, we calculated the ratio observed to the expected proportion of each of the 41 cell clusters ([68]Figure 1(i)). As indicated in [69]Figure 1(j), each cell type shows enrichment or depletion of different subsets, both consistent and contrasting changes in PBMC composition were observed in HIV-positive patients. For example, five out of the nine CD8^+ T-cell subclusters were significantly enriched in PBMC samples from the acute stage of MPXV infection, but two of those five CD8^+ T cells were significantly depleted in HIV+ PBMC samples ([70]Figure 1(j)). These data collectively indicated that MPXV infection causes comprehensive immune cells alteration, and HIV patients showed CD4 defects in acute phage, followed by dysregulated NK cells and neutrophils in recovery phase. Figure 1. [71]Figure 1. [72]Open in a new tab Cell clustering analysis. (a–d) UMAP visualization of cell clusters coloured by patient cohorts, MPXV infection stage, HIV infection status and cell identity. (e) Cell composition comparison between acute and recovery stage after MPXV infection. (f–h) Cell composition comparison between HIV– and HIV+ samples from both acute and recovery stages of MPXV infection, acute stage of MPXV infection, and recovery stage of MPXV infection. (i) UMAP visualization of all cells coloured by cell subclusters. (j) Ratio of observed/expected (Roe) of each cell subcluster in different patients to analyse depletion or enrichment status of each subcluster in the corresponding patient group. P values in e–h were determined by Wilcoxon test. The Roe of each subcluster was calculated through Chi-square test. Changes of B lymphocytes and gene expressions upon MPXV infection B cells secrete antibodies and mediate the humoral immune response, making them extremely important in protective immunity against virus [[73]18]. We first assessed the B-cell response in MPXV infection by UMAP embedding of B cells and five clusters were identified based on canonical marker gene expressions ([74]Figure 2(a)). We assessed the expression of genes associated with activation, such as CD79A and CD79B, and plasma markers such as MZB1, XBP1 in different B-cell subsets. CD79A and CD79B belong to the immunoglobulin superfamily, they covalently bound together serve as the main signal transduction molecules during antigen binding B-cell receptor (BCR). CD79A were mainly enriched in the subclusters 1, 3, and 5, while MZB1 and XBP1 were mainly expressed in the subclusters 2 and 4 ([75]Figure 2(b)), indicate that they might represent plasma cells for antibody production. Meanwhile, the CD79B expression was absent from all the subclusters, which prompted the dysfunction of BCR organization and thus impacting B-cell function. Proportions of plasma cell subclusters 2 and 4 exhibited significantly higher proportions in the acute than the recovery stage of patients ([76]Figure 2(c)). As expected, the expression of cell proliferation marker, MKI67, is mainly enriched in subclusters 2 and 4 and nearly absent from subclusters 1, 3, and 5, showing the proliferation of antibody-secreting cells in acute stage ([77]Figure 2(d)), and subclusters 2 and 4 were decreased in recovery stage ([78]Figure 2(c)). To explore the molecular perturbation upon MPXV infection, we performed differential gene expression analysis of B cells, including plasma cell, between patients that at acute and recovery stage of MPXV infection. Compared with recovery samples, the top 50 differently expressed genes (DEGs) in acute stage compared with recovery stage in at least 5 samples were shown in heatmap ([79]Figure 2(e)) (FDR adjusted, p value <0.05 and absolute log2-based fold change >0.25 as the significant thresholds). Plasma cell marker (CD38) and memory cell marker (CD27) are more obviously upregulated in non-HIV patients ([80]Figure 2(e)), indicating that HIV infection may limit cell function and differentiation after monkeypox infection. KEGG pathway analysis based on clusterProfiler ([81]Figure 2(f)) and GSEA ([82]Figure 2(g)) pathway enrichment analysis for DEGs indicated significant activation of some neurodegenerative disease and metabolism related pathways. Notably, BCR pathway was significantly repressed in acute stage samples, which is consistent with the absent CD79B expression. Additionally, several immune response-related pathways, such as Leishmania infection, Graft versus host disease, and Intestinal immune network for IgA production, were repressed in the B cell of acute stage patients, indicated the inhibition role of MPXV on host B-cell immune defence. Moreover, oxidative phosphorylation pathway was consistently activated in B cell ([83]Figure 2(h)) as well as CD8^+ T cell and NK cell (the following section) in the acute stage, illustrated the influence of MPXV infection on lymphocyte metabolism, which should be closely associated with their immune function as previously reported. Mitogen activate protein kinase (MAPK) and WNT signalling pathways were significantly repressed in B cell of acute stage patients ([84]Figure 2(i and j)), indicating the defect in BCR signalling pathway [[85]19]. In addition, the MAPK pathway contributes to effector functions of BCR activation, including proliferation and cell survival [[86]20], so MPXV may decrease B-cell function by inhibiting MAPK pathway. Figure 2. [87]Figure 2. [88]Open in a new tab Compositional analyses of B lymphocytes illustrate the expansion of subclusters. (a) UMAP visualization of B cells based on gene expression and different cell types represented by different colours. (b) UMAP visualization of B cells coloured by the expression of BCR related gene CD79A, CD79B, and antibody secreting related XBP1, MZB1. (c) Analysis of proportions of B-cell subsets in acute and recovery stages. (d) Violin plot representation of KI67 expression in different B-cell subsets (up) and different stage (down); each dot represents a cell, and horizontal lines display the mean expression value. (e) Heatmap of the most upregulated and downregulated 50 DEGs in acute stage compared to recovery stage coloured by the log2 (fold change). (f) Dot plot illustrating the top 20 significantly enriched KEGG pathways of the DEGs in acute stage. (g) The top 10 most activated and the top 10 most repressed KEGG pathway in acute stage of MPXV infection obtained through GSEA. (h–j) The enrichment plot of oxidative phosphorylation, MAPK and Wnt signalling pathways. Characterization of CD8+ T-cell perturbation upon MPXV infection CD8^+ T cells play critical roles in viral clearance during infectious disease [[89]16]. Our clustering analysis identified nine subclusters for CD8^+ T cells based on canonical marker gene expressions ([90]Figure 3(a)). The observation of low expressions of the effective markers of T cells, such as IFNG, GZMB, TNF, IL2, in most of these subclusters, suggesting the sustained T-cell dysfunction of CD8^+ T cells in all the patients ([91]Figure 3(b)). We found the expanded CD8 T cells in recovery phase ([92]Figure 1(h)), and our further analysis suggests that subclusters 1, 2, 8, 9 were expanded in the recovery phase, but subclusters 3, 4, 5, 6, 7 were decreased ([93]Figure 3(c)), along with the reduction in expression of GZMB and KI67 ([94]Figure 3(d)), indicative of their less cytotoxic and proliferative potency after the virus were eliminated. HIV infection did not exhibit many influences on CD8^+ T-cell subcluster proportion in MPXV patients, only subclusters 1 and 9 proportions were augmented in HIV patients ([95]Figure 3(e)). Additionally, HIV patients exhibited slightly higher expression of GZMB, while expression of KI67 had no appreciable changes ([96]Figure 3(f)). To further determine the impact of MPXV infection on CD8^+ T-cell characterization, we next analysed genes and their related pathways involving MPXV infection progression. By using the same strategy as in B-cell analysis, we compared the CD8^+ T cells of acute phase with recovery phase, and [97]Figure 3(g) illustrated 50 DEGs along with their log2-based fold change. Notably, we found that proliferation related genes KI67, MT2A, and HMGB2, which promote differentiation and maintenance of precursor exhausted T cells (Tpex) during chronic viral infection [[98]21] were consistently highly expressed in all the acute patients, including HIV-negative and positive patients; on the contrary, FOS and JUNB, that belong to transcription factor activator protein-1 (AP-1) family and associated with CD8^+ T cells effector function and cytokine production [[99]22], were inhibited ([100]Figure 3(g)). In addition, KEGG pathway analysis based on cluster Profiler enclosed these DEGs were enriched in biological processes concerning virus infection, such as coronavirus infection and Epstein–Barr virus infection ([101]Figure 3(h)). The KEGG pathway analysis through GSEA showed that anti-virus-related pathways were almost all repressed in the acute patients, which was consistent with that of B cell ([102]Figure 3(i)). We also noted the activation of oxidative phosphorylation in CD8^+ T cells ([103]Figure 3(j)), which was reported to limit cell effector function during chronic antigenic stimulation [[104]23]. Additionally, the toll-like receptor (TLR) signalling pathway ([105]Figure 3(k)), and the nod-like receptor (NLR) signalling pathway ([106]Figure 3(l)), which were both closely associated with type I IFN and inflammasome signalling pathways, leading to the production of corresponding proinflammatory or antiviral cytokines and chemokines [[107]24], were both significantly repressed in the acute stage patient, suggesting the comprehensive inhibition influence of MPXV on CD8^+ T-cell cytotoxicity. Collectively, our results demonstrated that CD8^+ T cells showed inhibited effector function in MPXV-infected patients. Figure 3. [108]Figure 3. [109]Open in a new tab Heterogeneous CD8 T cell in MPXV patients. (a) UMAP embedding of CD8+ T cells coloured by subcluster. (b) TCR and effector genes expression (IFNG, GZMB, TNF, IL2) in different clusters. (c) Proportion of different cell subsets in acute and recovery stage. (d) GZMB and KI67 expression in acute and recovery stages. (e) Proportion of different cell subsets in HIV+ and HIV– samples. (f) GZMB and KI67 expressions in HIV+ and HIV– patients. (g) Heatmap of top enriched 50 DEGs in CD8 T cells in acute stage compared with recovery stage, significantly upregulated genes KI67, MT2A, and HMGB2 were highlighted in red, and significantly downregulated gene FOS and JUNB were highlighted in blue. (h) The top 20 most significantly enriched KEGG pathways of the DEGs. (i) The top 10 most significantly activated and repressed KEGG pathways in the acute stage samples after MPXV infection obtained through GSEA. (j–l) Enrichment plot of oxidative phosphorylation, toll-like receptor and nod-like receptor signalling pathways. Alterations of NK cells in the peripheral blood of patients Natural killer (NK) cells are key elements of innate immunity and impaired NK-cell function were found in MPXV patients [[110]9]. To further explore the changes of NK cells in the disease progression, we conducted NK-cell composition and molecular comparison analysis between the acute and recovery stages of the MPXV-infected patients. NK cells were divided into nine distinct subclusters based on marker genes using UMAP ([111]Figure 4(a)). The absence of effector gene expressions was observed in almost all subclusters, including IFNG and TNF ([112]Figure 4(b)), consistent with the impaired function of NK cells in previous study [[113]25]. Notably, subclusters 1, 2, 3, which expressed relatively high level of effector genes, like NKG7 (Natural Killer Cell Granule Protein 7), GNLY (Granulysin), and PRF1 (Perforin 1), showed a higher proportion during the recovery stage, indicating the re-establishment of the antiviral immune response in NK cells. Conversely, the proportions of subsets 4, 5, 6, 7, and 8 were decreased during the recovery stage. ([114]Figure 4(c)). PRF1 and GNLY code essential effector molecules for NK-cell cytotoxicity, we noted that only the expression of MKI67, but not PRF1 and GNLY, was decreased in the recovery stage, indicating sustaining NK-cell cytotoxicity even after the elimination of virus ([115]Figure 4(d)). DEG analysis identified a total of 214 DEGs in the acute stage of MPXV-infected patients. Heatmaps highlight the top 50 most significantly upregulated and downregulated DEGs in the acute phase compared to the recovery phase, genes associated with chronic inflammation (CLTA) [[116]26], immune cell dysfunction (CENPF) [[117]27] and activation (CD38) were all highly expressed in almost all patients ([118]Figure 4(e)). Similarly, KEGG pathway analysis using clusterProfiler revealed that the DEGs were predominantly enriched in virus-related pathways. ([119]Figure 4(f)). GSEA indicated that oxidative phosphorylation, DNA replication, cell cycle, glycolysis were significantly activated, but MAPK, EGFR, and WNT signalling pathways were inhibited ([120]Figure 4(g–j)). In virus infection such as COVID-19, the expression of MAPK pathway genes in NK cells was also downregulated in the cured patients, suggesting that decreasing NK-cell function by inhibition of MAPK pathway could be a common mechanism in virus infection [[121]28]. Our analysis suggests that NK cells play an anti-MPXV role by expressing cytotoxic molecules. However, HIV+ patients exhibit a decreased proportion of NK cells, which may hinder their recovery from MPXV infection. Figure 4. [122]Figure 4. [123]Open in a new tab Heterogeneous NK cells in MPXV patients. (a) UMAP embedding of all NK cells coloured by cell subcluster. (b) UMAP projection of NK cells coloured by expressions of canonical markers, including NKG7, KI67 and effector genes expression (GNLY, PRF1, IFNG, TNF) in different cell subclusters. (c) Proportion of different NK cell subsets in acute and recovery stages. (d) Violin plot illustrating GNLY, PRF1 and KI67 expressions in acute and recovery stage samples. (e) Heatmap showing the log2 (fold change) of the 50 most upregulated and downregulated DEGs of NK cells in acute stage compared with recovery stage. (f) Dot plot illustrating the top 20 most significantly enriched KEGG pathways. (g) The top 10 most activated and repressed KEGG pathways in the acute stage samples obtained by GSEA (h–j). Enrichment plot of oxidative phosphorylation, MAPK and WNT signalling pathways. Characteristics of monocytes and dendritic cells Monocytes and dendritic cells (DC) are crucial for antiviral immune responses. They sense virus by multiple pathways, secret IFN and present antigen [[124]29]. We next deciphered their subset characteristics in MPXV patients by UMAP embedding of all samples and identified three different subclusters ([125]Figure 5(a)). Compared with other major histocompatibility complex (MHC) molecules, the absence of HLA-DMA expression indicated the inhibition of antigen presentation by virus ([126]Figure 5(b)). As to the proportion of different subclusters in different stages, subcluster 1 decreased in the recovery stage, but subclusters 2 and 3 showed opposite change, showing that MPXV may impact their function differentiation ([127]Figure 5(c)). Notably, when stratified by HIV negative or positive, we found in HIV+ patients, subcluster 1 was elevated and subset 2 decreased ([128]Figure 5(d)), indicating the critical impact of HIV on the innate immune cells. Additionally, most of the antigen presenting related genes (APGs) exhibited obviously higher mRNA level in MPXV infection acute stage compared with recovery stage ([129]Figure 5(e)), indicating increased antigen presentation, but HIV+ patients did not show obvious change of their expression ([130]Figure 5(f)). For the significant influence of MPXV infection on APGs, we next analysed gene expressions of acute phase compared with recovery phase of MPXV infection and identified 42 DEGs in at least 5 acute stage patients ([131]Figure 5(g)). We found EGR1 is among the top upregulated genes in monocytes. EGR1 binds enhancers of genes governing monocytic development, as well as a large set of inflammatory genes [[132]30]. We observed genes correlated with anti-virus function were upregulated in acute phase compared with recovery phase ([133]Figure 5(g)). For instance, CD14 is a pattern recognition receptor (PRR) that enhances innate immune responses and CD14 upregulation on monocytes is associated with effective control and clearance of viral and bacterial [[134]31]. IFI27, a classical type-1 IFN related gene and is involved in several types of viral infections [[135]11], and proinflammatory cytokines CCL3, which was previously reported to be produced by circulating monocytes in severe COVID-19 [[136]27], were also upregulated in acute stage ([137]Figure 5(g)). We also analysed the enrichment of KEGG pathways of DEGs through cluster Profiler ([138]Figure 5(h)) and found several pathways related to infection and inflammation were all significantly activated in acute stage of MPXV infection ([139]Figure 5(i–k)). Our results suggest that mono/DCs were activated during anti-MPXV function, while HIV changes mono/DCs dynamics in monkeypox infection, which may inhibit the anti-MPXV immunity. Figure 5. [140]Figure 5. [141]Open in a new tab Heterogeneous monocytes and dendritic cells in MPXV patients. (a) UMAP embedding of all mono/DCs cells coloured by cell subcluster. (b) UMAP projection of mono/DCs coloured by canonical antigen presentation genes expression. (c) Proportion of different mono/DCs subsets in acute and recovery stage, or (d) HIV+ and HIV– patients. (e) Antigen presentation genes expression in acute and recovery stages, or (f) HIV+ and HIV– patients. (g) Heatmap of the log2 (fold change) of significantly upregulated and downregulated DEGs in mono/DCs between in acute stage compared with recovery stage samples. IF127 was identified as type-1 IFN gene correlated with MPXV infection and was highlighted in red. (h) Dot-plot illustrating the top 20 most significantly enriched KEGG pathways of the DEGs. (i) The significantly activated or repressed KEGG pathways in the acute stage samples identified through GSEA. (j–k) Enrichment plot of leishmania infection and systemic lupus erythematosus. Neutrophils in the peripheral blood of patients Neutrophils play critical roles in viral clearance during infections, they release neutrophil extracellular traps and activate adaptive immune response, while the overactivation of neutrophils may cause tissue damage and lead to poor outcomes [[142]32,[143]33]. We extracted the neutrophil UMAP embedding from the whole UMAP space which contained eight neutrophil subclusters ([144]Figure 6(a)). The visualization of several neutrophil developmental markers illustrates highly heterogeneous developmental stages of neutrophil in our samples ([145]Figure 6(b)). Specifically, FCGR3A and SELL, two markers of neutrophil maturation, exhibited obviously higher mRNA levels in acute stage of MPXV infection, illustrating the acceleration of neutrophil maturation induced by MPXV, which might be associated with viral elimination. Additionally, CXCR4, a marker of neutrophil senescence, was inhibited in the acute stage of MPXV infection, further validating the enhanced function of neutrophil upon MXPV infection ([146]Figure 6(c)). In terms of the subcluster composition dynamic, we found that subcluster 6, which represent the only one with MKI67 expression, was significantly decreased in the recovery stage ([147]Figure 6(d)), indicating that neutrophil should be at quiescent state after the elimination of MPXV. In addition, with the recovery of the MPXV infection, subsets 2, which highly expressed IFI27 was also decreased ([148]Figure 6(d)). DEGs analysis obtained 17 genes in at least 5 acute stage patients. We observed that the expressions of MK167 and STMN1, which control cell proliferation and cell cycle [[149]34], were upregulated in neutrophils in nearly all patients in acute phase compared to recovery phase. We also found that MPXV infection triggered a robust IFN-I response in all patients, characterized by interferon-stimulated genes (ISGs) IFI27 and IFIT3 expression, but the upregulation was more significant in neutrophils from non-HIV patients compared to HIV patients ([150]Figure 6(e)). So, in HIV+ patients IFI27 mediate ISGs expression and IFN-I production could be inhibited during MPXV infection ([151]Figure 6(e)). Some inflammatory responses as well as cytotoxic related genes, including IL32, GZMA, GZMB, were consistently upregulated. On the contrary, CCL3, which has been previously reported to inhibit cell differentiation, was significantly suppressed in acute stage ([152]Figure 6(e)), which was predictive of the induction of differentiation and cytotoxicity of neutrophil. CD83 was a marker of polymorphonuclear neutrophils (PMN) with characteristics of dendritic cells and served as a marker of acute inflammation [[153]35]. Previous study indicated that herpesviruses could acquire immune evasion by targeting CD83 [[154]36,[155]37]. Similarly, in our study, the expression of CD83 was also inhibited in all patients ([156]Figure 6(e)). KEGG pathway analysis of the 17 DEGs based on cluster Profiler revealed significant enrichment of strong antiviral pathways, such as COVID-19 signalling, cytokine signalling, and neutrophil extracellular trap formation ([157]Figure 6(f)). What’s more, GSEA also uncovered significant activation of cell cycle and immune response-related pathways ([158]Figure 6(g)) in the acute stage of MPXV infection. Collectively, we illustrated the neutrophil differentiation and ISGs expression could be inhibited in HIV+ patients. Figure 6. [159]Figure 6. [160]Open in a new tab Heterogeneous neutrophils in MPXV patients. (a) UMAP embedding of all neutrophils coloured by cell subcluster. (b) UMAP projection of all neutrophils coloured by canonical neutrophils marker genes expression. (c) Expression of neutrophils maturation related genes in acute and recovery stage samples. (d) Proportion of different neutrophil subsets in acute and recovery stage samples. (e) Heatmap illustrating the log2 (fold change) of the most upregulated and downregulated DEGs in neutrophils between the acute and recovery stage samples. (f) Dot plot illustrating the top 20 most significant KEGG pathways of the DEGs. (g) KEGG pathways that were significantly activated or repressed in the acute stage samples obtained through GSEA. Interactions of immune cells Cell interactions are known to play an important role in infectious diseases. Using CellphoneDB analyses with scRNA-seq data from monkeypox patients, we found intense cell interactions between mono/DC and neutrophils with other cells, and the interactions were more obvious in the acute stage, indicating a critical role of mono/DC and neutrophils in disease progression ([161]Figure 7(a)). When we analysed communication network, we found strong interactions between mono/DC and CD4 T cells, as well as neutrophils and CD4 T cells both in acute and recovery stages ([162]Figure 7(b)). Ligand–Receptor (L–R) pairs between each immune cell type were also illustrated in both acute and recovery stage samples ([163]Figure 7(c)). Notably, LGALS9-HAVCR2 mediated interaction was only found between mono/DC and CD4 T cells, as well as CD4 T cells and B cells in the acute stage of MPXV infection, indicating the unique role of HAVCR2 in CD4 cell communication ([164]Figure 7(c)). We also noted strong interaction of mono/DC and neutrophils with several cells by LGALS9-CD44/CD45, which were responsible for interactions of many types of cells in this study. Notably, LGALS9-CD44/CD45 pairs were related to the interactions between CD4+ T and CD8+ T cells in only recovery but not acute samples. LGALS9 was reported to induce CD8+ T-cell death [[165]38] and by interacting with CD44, it could promote Treg function [[166]39], thus playing critical role in limiting T-cell response and considered as an immune checkpoint, so we could speculate that LGALS9 could be a critical factor for limiting T-cell anti-virus response after MPXV elimination. Additionally, the outgoing and incoming interaction profile analysis of each cell type also supported the stronger interactions exist in acute stage ([167]Figure 7(d)). Mono/DC and neutrophils mediated strongest interactions both in acute and recovery stages, there was nearly no difference between acute and recovery samples in terms of the incoming and outgoing signals of cell interactions ([168]Figure 7(e,f)). In brief, galectin, annexin, IL16, and CCL represented the top four molecules that were responsible for cell interactions ([169]Figure 7(e,f)). This suggests innate immune cells are critical for MPXV elimination, and our results shed light on identifying potential targeting cell interactions to improve viral elimination. Figure 7. [170]Figure 7. [171]Open in a new tab Cell interaction analysis of immune cells in patients. (a) Heatmap showing the number of potential ligand-receptor pairs among the predicted cell types. (b) Interaction network constructed by cellchat. Each line colour indicates the ligands expressed by the cell population represented by the same colour (labelled). The lines connect to the cell types that express the cognate receptors. The line thickness is proportional to the number of ligands when cognate receptors are present in the recipient cell type. (c) Dot plot showing the ligand–receptor pairs that link the interacted cells. The x-axis and y-axis represent the cell pairs and ligand–receptor pairs, respectively. The colour from blue to red indicates a gradient stronger interaction. (d) Scatter plot illustrates the number of outgoing signals, i.e. ligand, and incoming signal, i.e. receptor, for each cell type in cell–cell interactions. (e) and (f) illustrate the number of incoming and outgoing signals in cell–cell interactions that each pathway contributed in acute stage and recovery stage samples. Influences of MPXV and HIV infections on immune cell differentiation To investigate the potential influence of MPXV on immune cell differentiation and assess whether HIV infection might affect this process, we performed pseudotime analysis of B, CD4^+ T, CD8^+ T, NK, Mono/DC, and neutrophil cells (see Supplemental Figures 1–6). Expression patterns of key genes associated with cell differentiation and maturation were analysed along the pseudotime trajectory to detect the cell differentiation process. Significant enrichment was observed in antibody-secreting B cells, marked by the upregulation of MZB1 (see Supplemental Figure 1a); proliferating and cytotoxic CD8+ T cells with high expression of GZMB and MKI67 (see Supplemental Figure 3a); Mono/DC subsets with elevated expression of MHC molecules (see Supplemental Figure 5a); and neutrophils showing increased expression of CXCR4, FCGR3A, and MKI67 (see Supplemental Figure 6a) in PBMC samples during the acute MPXV infection stage. The influence of HIV infection on immune cell differentiation appeared to be minimal, possibly due to the clearance of HIV, as the distribution of cells from HIV– and HIV+ patients was nearly even across the pseudotime trajectory. These findings suggest that MPXV accelerates immune cell activation and maturation. Discussion In this study, we utilized single-cell RNA sequencing (scRNA-seq) to analyse the immune cell atlas of peripheral blood from monkeypox patients at both acute and recovery stages, the potential impact of HIV was also proposed ([172]Figure 8). Our analysis revealed dysregulated cell subsets, function, and robust cell interactions during MPXV progression. Furthermore, our study elucidated the possible and complex effect of HIV infection on the dynamics of several cell subsets in monkeypox infection, which may contribute to their decreased antiviral response and longer recovery time. Figure 8. [173]Figure 8. [174]Open in a new tab Diagram illustrating the proposed mechanism underlying the severe condition of HIV-infected patients following MPXV infection. HIV induces CD4+ cell loss and dysfunction, impairs BCR signalling and humoral immunity in B cells, and reduces type 1 IFN signalling in neutrophils. The complex and diverse cellular interactions further exacerbate deficiencies in anti-MPXV immunity, contributing to prolonged hospitalization in HIV+ patients. The deficiency of CD4^+ T cells is a hallmark of HIV infection. There is a greater chance of severe manifestation of MPXV infection in immunosuppressed patients [[175]40], while the mechanism is unclear. Besides infecting T cells, HIV also induces the death of uninfected CD4 T cells. The mechanisms include the overexpression of death ligands, activation-induced cell death, and other processes, which have been extensively reviewed elsewhere [[176]41,[177]42]. According to U.S. Centers for Disease Control and Prevention (CDC), “Of the people with severe manifestations of monkeypox for whom CDC has been consulted, the majority have had HIV with CD4 counts <200 cells/ml, indicating substantial immunosuppression”. Indeed, in our study, we found HIV patients have lower CD4 T cell proportion compared with HIV– ones, especially in acute phase. So, in the current study, we mainly discussed the possible role of other cell subsets’ role in HIV-induced decreased anti-MPXV immune. Previous study indicates that HIV induces alterations of B-cell subpopulations, most B cells in healthy individuals are naive and memory B cells, whereas in HIV+ individuals, immature transitional B cells, exhausted B cells and plasma blasts are over-represented. In addition, HIV causes B-cell exhaustion [[178]43]. Notably, as shown in the heatmap of DEGs in acute phase versus recovery phase, the perturbations in B cells subsets is most significant in our study, upregulation of many genes critical in B-cell function in HIV– patients in acute phase was not seen in HIV+ patients, such as CD38, which is critical for B-cell activation, proliferation, and differentiation [[179]44]. Moreover, we found the absence of CD79B in all the patients which indicates the dysregulation of BCR signalling. Receptors initiate signalling through pathways and the central role of BCR in humoral immunity has been acknowledged [[180]45,[181]46]. High levels of many B-cell inhibitory receptors, including CD22, CD72 were reported to be expressed by tissue-like memory B cells from HIV+ individuals, and tissue-like memory B cells were recognized as an exhausted B-cell counterpart [[182]47], so we postulate that HIV impair humoral responses and long-term immune memory, which may responsible for severe condition of HIV+ patients after MPXV infection. Our analysis of total PBMCs identified decreased levels of NK cells in HIV+ patients during both the acute and recovery phases compared to HIV– patients. NK cells are important mediators of the innate immune defenders against viruses, mediate suppression of viral in many ways. The crucial role of NK cells and their dysfunction in antiviral immunity has been well established [[183]2,[184]48], including in monkeypox [[185]9,[186]49]. MPXV impairing NK-cell function by preventing its IFN-γ production [[187]9], activation, chemotaxis, and antibody-dependent cell-mediated cytotoxicity (ADCC) are also defective in HIV-infected individuals [[188]50,[189]51], which may further compromises host immune defences against MPXV. Our study provides additional evidence that HIV-induced NK-cell dysfunction may contribute to a decrease in protective immunity against the monkeypox virus. For CD8 T cells, several studies have documented their impaired function in HIV infection, these cells become non-cytolytic, exhibit lower levels of perforin, became exhausted and apoptosis [[190]52,[191]53]. While in our study, we found CD8 T cells increase in recovery phase in HIV+ patients compared with HIV– ones. Studies have also indicated that naïve and memory CD8 T cells turnover increases during HIV infection and differentiate towards senescence state [[192]53]. So the increased CD8 in HIV+ patients may be senescent T cells. Another critical question that remains to be answered is when T-cell exhaustion starts during HIV infection and how anti-virus treatment affects T-cell subsets, these may also cause the variation of CD8 T cells number. Since the severe condition and longer hospitality of HIV+ patients, the increased CD8 T cells may not promote the clearance of MPXV. Moreover, it is important to note that the MPXV/HIV co-infected patients included in our study had received prolonged antiviral therapy, achieving viral suppression. This could potentially explain why the presence of HIV does not significantly affect the immune cell subsets during the acute and recovery phases of MPXV infection, to elucidate the mechanism and role of the increased CD8 T cells in HIV+ patients upon MPXV infection, more mechanism study is needed. Recent studies have highlighted the potential of interferon-responsive neutrophil subsets in combating infections and patrolling the bloodstream. Following viral infections, there is often a sharp increase in the number of neutrophils in the local microenvironment. However, these neutrophils have been reported to produce Reactive Oxygen Species (ROS) that can damage the epithelial–endothelial barrier and impair virus elimination, thereby contributing to detrimental inflammation and increased morbidity in mice [[193]54,[194]55]. In our study, we observed a decrease in the proportion of neutrophils in the recovery stage among HIV-negative patients, whereas their percentage remained higher in both stages among HIV-positive patients. Additionally, the expression of IFGs in neutrophils was inhibited in HIV-positive patients, potentially exacerbating the condition of monkeypox patients. Nevertheless, the precise pathogenic roles of this neutrophil subset require further exploration through future studies investigating its phenotype and role in antiviral responses. Mono/DCs is the first line of defence against viral infections and are primary targets for viral assault [[195]2]. Innate immune cells response to HIV during the early stages of infection is an important contributor to persistent immune activation and chronic inflammation, causing the activation or immunosenescence of immune cells, and activated CD16^+ monocytes can disseminate HIV replication through transferring virions and effective infection of CD4 T cells [[196]56]. Though no obvious changes in HLA gene expression were observed in HIV+ and HIV– patients, dynamic change of Mono/DC subsets 1 and 2 showed differences between HIV-positive and HIV-negative patients from the acute phase to the recovery phase, indicating the persistent impact of HIV on Mono/DC function in MPXV infection, beyond its role in causing abnormal phagocytic function. In addition, we identified IFI27 as the most significantly upregulated ISG in both monocytes and neutrophils in Monkeypox infection. This highlights the pivotal role of IFI27 in the immune response against viral infections. Interestingly, IFI27 has been reported to interact with RIG-I, thereby impairing RIG-I activation and subsequently downregulating innate immune responses. Consequently, IFI27 provides a negative feedback mechanism that counteracts excessive inflammatory responses to RNA viral infections [[197]57]. Our results suggest that besides contributing to the cytokine production and inflammation, peripheral monocytes may also counteract excessive inflammatory monkeypox infection. We observed the upregulation of the oxidative phosphorylation pathway in several cell subsets in acute phase, alongside the inhibition of anti-virus-related signalling pathways such as MAPK and WNT compared to recovery phase. Metabolism correlates with cell subsets and activation states. For instance, T cells transit from predominantly oxidative metabolism to aerobic glycolysis upon activation. Alfredo et al. indicated that B cells chronically exposed TLR4 ligation showed enhanced and more rapid metabolic reprogramming towards aerobic glycolysis [[198]58]. The increased oxidative phosphorylation in our study may result from the difference of dominance B-cell subsets or the different stimulation type, while the detailed role of mitochondrial metabolism in B-cell activation and function, especially in virus infection remains to be further detected. MAPK cascade participates in regulating the immune response, but its role in viral is controversial [[199]59]. For example, in SARS-CoV-2 infection, TNF-α (tumour necrosis factor alpha), an important cytokine for proliferation and function of NK cells exerts its effects through MAPK pathways [[200]28]. Given its roles in type I IFN production, which is crucial in early innate responses against viral infections, the inhibition of the MAPK pathway genes indicates the significant impact of Monkeypox against host immune system. On the contrary, viruses extensively use MAPK cascade also implies that this pathway may be a good target for developing broad-spectrum antiviral drugs, for instance, p38 MAPK inhibitor, SB203580, inhibited the effective phosphorylation of HSP-27, CREB, and eIF4E in SARS-CoV-infected cells, showing promise as a new class of antiviral agents [[201]60]. Although disturbed MAPK signalling was observed in NK and B cells during the acute phase, we cannot definitively conclude the role of MAPK or the potential of its inhibitors as antiviral drugs without further research. The interaction of monocytes with T cells in Monkeypox patients might compromise cell anti-virus function. Interaction analysis indicates that monocytes interact with other cells largely through LGALS9. LGALS9 is a widely expressed protein involved in immune regulation, affecting prognosis of various types of cancer as an immune checkpoint inhibiting T-cell function [[202]61]. Notably, in the SARS-CoV-2 infection, the serum level of LGALS9 was elevated and positively correlated with inflammation and tissue injury [[203]62]. Recent studies indicate that targeting monocytes infiltration could alleviate virus-related syndrome in COVID-19 infection [[204]63]. Our study suggests that LGALS9 may be the primary molecule mediating cell interactions in monkeypox infection, offering novel insights into understanding the dynamic interplay of immune cells in this context. Inhibiting LGALS9 holds promise for decreasing overactivated inflammation in such infections. Our study also has some limitations, first, the limited sample size and sequencing depth is a major shortcoming of this study, potentially affecting the robustness of the results and leading to biased or less representative conclusions. For example, although we observed decreased NK cells in HIV-positive patients compared to HIV-negative ones in acute phase of MPXV, we were unable to identify a statistically significant difference in our analysis. In addition, B cells subset 4 shows the highest Ki67 expression and may exert unique anti-virus function, and biological processes and signalling pathways associated with T-cell dysfunction should be further analysed. Further study is needed to provide more sufficient resolution into the various cell compartments and interaction. In addition, our conclusion in the current study is mainly drawn from scRNA-seq data, clinical validations will be carried out in our further research. Overall, our work proposes the potential mechanisms underlying the severe condition of HIV-bearing MPXV patients ([205]Figure 8) and provides understanding of pathogenesis in HIV patients co-infected with MPXV, thus deepening current understanding of the MPXV infection. Our data identifies potential mechanisms of immunocompromised individuals, such as HIV-infected patients, being more probably to experience severe manifestations. Individuals with acquired immunodeficiency may require more active medical intervention rather than spontaneous resolution. Material and methods Subjects and specimen collection From January to June 2023, peripheral blood samples were collected from six monkeypox patients at Tianjin Second People’s Hospital after obtaining written informed consent from the patients (Tianjin Second People’s Hospital Ethics Committee Approval No. 2023-075). The sample collection criteria for this study are defined as follows: during the acute phase, samples were obtained in the febrile and eruptive stages, while in the recovery phase, samples were collected on the day complete detachment of rash scabs. All scRNA-seq samples from the enrolled patients were collected during the acute phase of infection, prior to the initiation of antibiotic treatment. Samples collected during the recovery phase were obtained at least 1 week after the completion of antibiotic therapy to avoid the impact of antibiotics on the immune profiles in our study. Eligibility criteria included age >18 years and hospitalization at Tianjin Second People’s Hospital with a positive monkeypox virus RT–PCR test from vesicular fluid. All enrolled patients provided informed consent. Blood samples were processed by isolating PBMCs using Ficoll-Paque Plus medium (GE Healthcare) through density gradient centrifugation and washed with Ca/Mg-free PBS. All blood samples were processed within 4 h of collection, with the majority processed within 1 h. scRNA sequencing by Seq-Well The Seq-Well platform for scRNA-seq was utilized as described previously11,12,42. Immediately after Ficoll separation, 50,000 PBMCs were resuspended in RPMI + 10% FCS at a concentration of 75,000 cells per ml. A 200-μl volume of this cell suspension (15,000 cells) was then loaded onto Seq-Well arrays pre-loaded with mRNA capture beads (ChemGenes). Following four washes with Dulbecco’s phosphate-buffered saline (DPBS) to remove serum, the arrays were sealed with a polycarbonate membrane (pore size of 0.01 μm) for 30 min at 37°C and then frozen at −80°C for no less than 24 h and no more than 14 days to allow batching of samples processed at irregular hours. Next, arrays were thawed, cells lysed, transcripts hybridized to the mRNA capture beads, and beads recovered from the arrays and pooled for downstream processing. Immediately after bead recovery, mRNA transcripts were reverse-transcribed using Maxima H-RT (ThermoFisher, EPO0753) in a template-switching-based rapid amplification of cDNA ends (RACE) reaction, excess unhybridized bead-conjugated oligonucleotides were removed with exonuclease I (NEB M0293L) and second-strand synthesis was performed with Klenow fragment (NEB M0212L) to enhance transcript recovery in the event of failed template switching. The whole transcriptome amplification (WTA) was performed with KAPA HiFi PCR Mastermix (Kapa Biosystems KK2602) using ∼6000 beads per 50-μl reaction volume. Resulting libraries were then pooled in sets of six (∼36,000 beads per pool) and products purified by Agencourt AMPure XP beads (Beckman Coulter, A63881) with a 0.6× volume wash followed by a 0.8× volume wash. The quality and concentration of WTA products were determined using an Agilent Fragment Analyser (Stanford Functional Genomics Facility), with a mean product size of >800 bp and a non-existent primer peak indicating successful preparation. Library preparation was performed with a Nextera XT DNA library preparation kit (Illumina FC-131-1096) with 1 ng of pooled library using dual-index primers. Tagmented and amplified libraries were again purified by Agencourt AMPure XP beads with a 0.6× volume wash followed by a 1.0× volume wash, and quality and concentration were determined by fragment analysis. Libraries between 400 and 1000 bp with no primer peaks were considered successful and pooled for sequencing. The sequencing was performed on a NovaSeq S2 instrument (Illumina; Chan Zuckerberg Biohub). The read structure was paired-end with read 1 beginning from a custom read 1 primer11 containing a 12-bp cell barcode and an 8-bp UMI, and with read 2 containing 50 bp of mRNA sequence. Bioinformatics analysis The raw sequencing data were preprocessed by CellRanger v8.0, mainly including the removal of low-quality reads, alignment to the reference genome, i.e. GRCh38 in this study, and generation of gene-cell expression matrix. Seurat v5 was then used for the integration of data from different samples, dimension reduction, cell clustering and differential expression analysis by using the raw expression matrix as input. In detail, quality control of the raw data, mainly including removal of cells that contained less than 200 or more than 4000 genes as well as those with more than 25% mitochondrial genes. The genes that expressed in no more than three cells were also removed. Scaled normalization of gene expression was conducted by using the NormalizeData function. The normalized data from different samples were integrated by anchors screened by FindIntegrationAnchors function to remove the batch effect. Data dimension reduction was performed by three methods, i.e. PCA, UMAP and TSNE, and cell clustering was performed based on the low-dimension data by using the FindClusters function with the most optimal cluster number identified by cluster R package. Differential expression analysis between different cell clusters or patient groups was conducted based on the FindMarkers function with the thresholds of FDR adjusted p value <0.05 and absolute log2-based fold change >0.25. Besides, only genes that expressed in at least 10% cells of each sample group were retained for differential expression analysis. Cell identity was manually annotated based on the canonical markers as well as the specifically expressed genes of each cell cluster. Both CellPhenoDb and cellchat were applied for the cell-cell interaction analysis with the default parameters. Supplementary Material supplementary.docx [206]TEMI_A_2459136_SM6310.docx^ (1.7MB, docx) Acknowledgements