转移性结直肠癌患者恶性腹水的单细胞图谱。

IF 20.1 1区 医学 Q1 ONCOLOGY Cancer Communications Pub Date : 2024-03-26 DOI:10.1002/cac2.12541
Haiyang Zhou, Jiahui Yin, Anqi Wang, Xiaomao Yin, Taojun Jin, Kai Xu, Lin Zhu, Jiexuan Wang, Wenqiang Wang, Wei Zhang, Xinxiang Li, Zhiqian Hu, Xinxing Li
{"title":"转移性结直肠癌患者恶性腹水的单细胞图谱。","authors":"Haiyang Zhou,&nbsp;Jiahui Yin,&nbsp;Anqi Wang,&nbsp;Xiaomao Yin,&nbsp;Taojun Jin,&nbsp;Kai Xu,&nbsp;Lin Zhu,&nbsp;Jiexuan Wang,&nbsp;Wenqiang Wang,&nbsp;Wei Zhang,&nbsp;Xinxiang Li,&nbsp;Zhiqian Hu,&nbsp;Xinxing Li","doi":"10.1002/cac2.12541","DOIUrl":null,"url":null,"abstract":"<p>The presence of malignant ascites in colorectal cancer (CRC) patients is associated with a poor prognosis, a high risk of recurrence, and resistance to chemotherapy and immune therapy [<span>1-3</span>]. Understanding the complex interactions among different kinds of cells and the ecosystem of peritoneal metastasized colorectal cancer (pmCRC) ascites may provide insights into effective treatment strategies.</p><p>We profiled the single-cell transcriptomes of 96,065 cells from ascites samples of 12 treatment-naïve patients with pmCRC using the 10× single-cell RNA-sequencing (scRNA-seq) (Supplementary Figure S1A, Supplementary Table S1). Eleven major cell types were identified by characteristic canonical cell markers, including epithelial cells, endothelial cells, fibroblasts, T cells, B cells, monocytes, macrophages, plasma cells, natural killer (NK) cells, dendritic cells (DCs), and mast cells (Figure 1A-B). The main cellular components of pmCRC ascites are T cells (40,095; 41.7%), macrophages (28,487; 29.7%), and fibroblasts (5,932; 6.2%). Compared with primary CRC, which showed 14.8% epithelial cells [<span>4</span>], only 0.3% (291) epithelial cells were found in the ascites. The low percentage of epithelial cells in pmCRC ascites was consistent with the scRNA-seq studies of another tumor ascites [<span>5-7</span>].</p><p>We classified the 12 patients into 2 groups according to their treatment response as follows: 8 patients (P02, P03, P04, P07, P08, P09, P11, and P12) had stable disease (SD), while 4 (P01, P05, P06, and P10) had progressive disease (PD). Single-cell transcriptomic analyses have revealed high heterogeneity of cell composition in 12 patients. The SD group exhibited a higher proportion of fibroblasts and epithelial cells (Figure 1B). Remarkably, fibroblasts had significantly different expression characteristics between the 2 groups (Figure 1C), and the top five upregulated/downregulated genes were visualized in 11 cell types (Figure 1D). We also found a significant increase in the frequency of macrophages in pmCRC ascites compared with the primary tumors [<span>4</span>] (Figure 1E). It hinted that significant inter-patient variability in the composition and functional programs of pmCRC ascites cells under different disease states.</p><p>To comprehensively study the cellular interactions within the pmCRC ascites ecosystem, we predicted cell-cell communication networks using CellChat. Overall, we identified 44 significant ligand-receptor pair interactions. Although T cells were the most abundant cell population (41.7%) in pmCRC ascites, fibroblasts and macrophages were the core of the cellular interaction network (Figure 1F), suggesting their important roles in recruiting and cross-talking with diverse cells in the pmCRC ascites ecosystem.</p><p>The result of cellular communications suggested that there was a complex interplay between various signaling molecule. Macrophage migration inhibitory factor (MIF), annexin, complement, and C-C chemokine ligand (CCL) were the most active outgoing/incoming signaling molecules in CRC ascites (Supplementary Figure S1B). Fibroblasts directly contacted with different types of cells via ligand-receptor interactions of the MIF-(CD74 + C-X-C chemokine receptor type 4 [CXCR4]) and MIF-(CD74 + CD44) axes and C3-(integrin alpha X [ITGAX] + integrin subunit beta 2 [ITGB2]) (Figure 1G). Notably, macrophage populations were more likely to interact with other cells through the adhesive ligand-receptor pairs galectin-9 (LGALS9)-CD44 and LGALS9-CD45, which were not observed in other cell populations (Figure 1G). CD74, LGALS9 were significantly associated with metastasis in The Cancer Genome Atlas (TCGA) CRC cohorts. We also found CD44 and ITGAX were survival (Figure 1H; CD44 and ITGAX showed no significant differential expression between metastatic and non-metastatic patients, so data are not shown). These results indicated that the entire cellular interaction network of pmCRC ascites contributed to establishing an immunosuppressive and metastatic microenvironment.</p><p>We observed that the abundance of fibroblasts in pmCRC ascites samples was significantly greater in SD patients than in PD patients (Figure 1B). The fibroblasts were partitioned into 7 distinct clusters (C0-C6) based on unsupervised clustering (Figure 1I). All sub-clusters of cancer-associated fibroblasts (CAfs) showed a high expression of extracellular matrix cancer-associated fibroblasts (eCAFs) signature (Figure 1J), while inflammatory CAF (iCAF), myofibroblast CAF (myCAF), matrix CAF (mCAF), and vascular CAF (vCAF) only presented in a small fraction of fibroblasts (Supplementary Figure S1C), supporting the role of eCAFs in enhancing the metastatic potential of pmCRC. A higher abundance of antigen-presenting cancer-associated fibroblasts (apCAFs) was observed in the PD cohort (<i>n</i> = 310) than in the SD cohort (<i>n</i> = 93) (Wilcoxon test, <i>P</i> = 0.049). These results indicated that the CAFs in pmCRC ascites have bidirectional associations with immune regulation function, serving as a favorable candidate for CRC treatment. Differentially expressed genes and gene ontology (GO) analyses showed that the “cell-cell adhesion”, “inflammatory response”, and “cytokine production” were differentially enriched between primary tumors and ascites (Supplementary Figure S1D), which implied that the liquid state of ascites changed the functions of the fibroblast populations.</p><p>Macrophages were significantly enriched in pmCRC ascites and categorized into 8 sub-clusters (C0-C7) (Figure 1K). Using the previously defined “M1” and “M2” signatures, C2 showed an “M1-like” pattern, and C5 showed “M2-like” patterns. We also identified a small sub-cluster of C5 co-expressed both “M1” and “M2” gene signatures (Figure 1L), which have been reported in previous studies on solid tumors [<span>8</span>]. We next examined the expression of a series of the previously reported immunosuppressive genes (leukocyte-associated immunoglobulin-like receptor 1 [LAIR1], hepatitis A virus cellular receptor 2 [HAVCR2; also known as T cell immunoglobulin and mucin domain-containing protein 3], LGALS9, and V-set immunoregulatory receptor [VSIR]) in macrophage sub-clusters. Because the expression pattern of the “M2” marker gene CD163 perfectly coincided with that of LAIR1 in all sub-clusters (Figure 1M), we postulated that the immunosuppressive function of tumor-associated macrophages (TAMs) might be exerted via LAIR1. Two other immunosuppressive genes, T cell immune receptor with Ig and ITIM domains (TIGIT) and programmed cell death 1 (PDCD1), were also identified highly expressed in C5. C4 highly expressed the key immunosuppressive phenotypic marker triggering receptor expressed on myeloid cells 2 (TREM2) (Figure 1N). In summary, the majority of macrophages in pmCRC ascites exhibited high immunosuppressive features.</p><p>We identified 11 sub-clusters of T cells according to the expression of their respective markers, including CD4<sup>+</sup> T cells (C1, C4, C5, and C6) and CD8<sup>+</sup> T cells (C0, C2, C3, C7, C8, C9, and C10) (Figure 1O). Most CD8<sup>+</sup> effector memory cells re-expressing CD45RA T (Temra/Teff) cells (C8) were from patient 5 (P05), and CD8<sup>+</sup> effector memory T (Tem) cells (C7) were mostly from patient 8 (P08); the remaining 10 patients exhibited high heterogeneity in 11 T-cell sub-clusters (Supplementary Figure S1E-F). Importantly, CD8<sup>+</sup> tissue-resident memory (Trm) cells (C7), which were reported to be associated with forming a tertiary lymphoid structure (TLS) [<span>9</span>], were less abundant in SD patients (P03, P08, P09, P11, and P12). We also observed that all sub-clusters expressed ferritin light chain (FTL) (Supplementary Figure S1G), which was reported to regulate chemoresistance and metastasis in CRC [<span>10</span>]. We calculated the cytotoxicity, proliferation, and exhaustion signatures for all CD8<sup>+</sup> T-cell sub-clusters (Figure 1P). Only one sub-cluster of CD8<sup>+</sup> T cells was not show exhaustion signature (C10). Clusters 9 and 10 exhibited slightly higher proliferation, which could recruit cytotoxic T cells. The abundance of C9 and C10 was low, suggesting that T cells may play a minor role in the immune microenvironment of ascites and may work synergistically with other cell populations. Moreover, we predicted immunomodulatory drugs targeting macrophages, where gene sets were extracted from the macrophages of the pmCRC ascites data (Figure 1Q).</p><p>In conclusion, we found that T cells, fibroblasts, and macrophages exhibited immunosuppressive features in pmCRC ascites (Figure 1R). The cellular landscape of pmCRC ascites has the significant indication of patients’ immune status, providing insights for prognosis and therapy selection.</p><p>Haiyang Zhou made contributions to the conceptualization, funding acquisition, investigation, resource acquisition, and writing original draft.</p><p>Jiahui Yin made contributions to the data curation, formal analysis, and software analysis.</p><p>Anqi Wang made contributions to the formal analysis, investigation, and resource acquisition.</p><p>Xiaomao Yin made contributions to the formal analysis.</p><p>Taojun Jin made contributions to the formal analysis and visualization.</p><p>Kai Xu made contributions to the resource acquisition and supervision.</p><p>Lin Zhu made contributions to the investigation and resource acquisition.</p><p>Jiexuan Wang made contributions to the investigation and resources acquisition.</p><p>Wenqiang Wang made contributions to the resource acquisition.</p><p>Wei Zhang made contributions to the resource acquisition.</p><p>Xinxiang Li made contributions to the conceptualization, visualization, and writing—review &amp; editing.</p><p>Zhiqian Hu made contributions to the funding acquisition, visualization, and writing—review &amp; editing.</p><p>Xinxing Li made contributions to the investigation, resource acquisition, visualization, writing—original draft, and writing—review &amp; editing.</p><p>All authors read and approved the final manuscript.</p><p>The authors declare that they have no competing interests.</p><p>The authors gratefully acknowledge the financial support from the National Key R&amp;D Program of China (No. 2019YFA0110601), National Natural Science Foundation of China (No. 81571827), Natural Science Foundation Project of Shanghai Science and Technology Commission (SKW2030), Excellent Discipline Reserve Talent Plan of Tongji Hospital Affiliated to Tongji University (HBRC2014), Clinical research Project of Tongji Hospital Affiliated to Tongji University (ITJ-ZD-2104), Key talent introduction project of Tongji Hospital Affiliated to Tongji University (RCQD2102), Talent project of Tongji Hospital Affiliated to Tongji University (GJPY2111), and Shanghai Tongji Hospital special disease database construction project (TJ-DB-2105).</p><p>All the investigation protocols were approved by the Institutional Ethics Committees of Shanghai Changzheng Hospital and Shanghai Tongji Hospital (SBKT-2022-155). All subjects provided informed consent to participate in the study and approved the use of their biological samples for analysis. All experiments were performed following institutional guidelines, in compliance with relevant laws. Data sharing mechanisms will ensure that the rights and privacy of individuals participating in research will be guaranteed.</p>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":null,"pages":null},"PeriodicalIF":20.1000,"publicationDate":"2024-03-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11260760/pdf/","citationCount":"0","resultStr":"{\"title\":\"Single-cell landscape of malignant ascites from patients with metastatic colorectal cancer\",\"authors\":\"Haiyang Zhou,&nbsp;Jiahui Yin,&nbsp;Anqi Wang,&nbsp;Xiaomao Yin,&nbsp;Taojun Jin,&nbsp;Kai Xu,&nbsp;Lin Zhu,&nbsp;Jiexuan Wang,&nbsp;Wenqiang Wang,&nbsp;Wei Zhang,&nbsp;Xinxiang Li,&nbsp;Zhiqian Hu,&nbsp;Xinxing Li\",\"doi\":\"10.1002/cac2.12541\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p>The presence of malignant ascites in colorectal cancer (CRC) patients is associated with a poor prognosis, a high risk of recurrence, and resistance to chemotherapy and immune therapy [<span>1-3</span>]. Understanding the complex interactions among different kinds of cells and the ecosystem of peritoneal metastasized colorectal cancer (pmCRC) ascites may provide insights into effective treatment strategies.</p><p>We profiled the single-cell transcriptomes of 96,065 cells from ascites samples of 12 treatment-naïve patients with pmCRC using the 10× single-cell RNA-sequencing (scRNA-seq) (Supplementary Figure S1A, Supplementary Table S1). Eleven major cell types were identified by characteristic canonical cell markers, including epithelial cells, endothelial cells, fibroblasts, T cells, B cells, monocytes, macrophages, plasma cells, natural killer (NK) cells, dendritic cells (DCs), and mast cells (Figure 1A-B). The main cellular components of pmCRC ascites are T cells (40,095; 41.7%), macrophages (28,487; 29.7%), and fibroblasts (5,932; 6.2%). Compared with primary CRC, which showed 14.8% epithelial cells [<span>4</span>], only 0.3% (291) epithelial cells were found in the ascites. The low percentage of epithelial cells in pmCRC ascites was consistent with the scRNA-seq studies of another tumor ascites [<span>5-7</span>].</p><p>We classified the 12 patients into 2 groups according to their treatment response as follows: 8 patients (P02, P03, P04, P07, P08, P09, P11, and P12) had stable disease (SD), while 4 (P01, P05, P06, and P10) had progressive disease (PD). Single-cell transcriptomic analyses have revealed high heterogeneity of cell composition in 12 patients. The SD group exhibited a higher proportion of fibroblasts and epithelial cells (Figure 1B). Remarkably, fibroblasts had significantly different expression characteristics between the 2 groups (Figure 1C), and the top five upregulated/downregulated genes were visualized in 11 cell types (Figure 1D). We also found a significant increase in the frequency of macrophages in pmCRC ascites compared with the primary tumors [<span>4</span>] (Figure 1E). It hinted that significant inter-patient variability in the composition and functional programs of pmCRC ascites cells under different disease states.</p><p>To comprehensively study the cellular interactions within the pmCRC ascites ecosystem, we predicted cell-cell communication networks using CellChat. Overall, we identified 44 significant ligand-receptor pair interactions. Although T cells were the most abundant cell population (41.7%) in pmCRC ascites, fibroblasts and macrophages were the core of the cellular interaction network (Figure 1F), suggesting their important roles in recruiting and cross-talking with diverse cells in the pmCRC ascites ecosystem.</p><p>The result of cellular communications suggested that there was a complex interplay between various signaling molecule. Macrophage migration inhibitory factor (MIF), annexin, complement, and C-C chemokine ligand (CCL) were the most active outgoing/incoming signaling molecules in CRC ascites (Supplementary Figure S1B). Fibroblasts directly contacted with different types of cells via ligand-receptor interactions of the MIF-(CD74 + C-X-C chemokine receptor type 4 [CXCR4]) and MIF-(CD74 + CD44) axes and C3-(integrin alpha X [ITGAX] + integrin subunit beta 2 [ITGB2]) (Figure 1G). Notably, macrophage populations were more likely to interact with other cells through the adhesive ligand-receptor pairs galectin-9 (LGALS9)-CD44 and LGALS9-CD45, which were not observed in other cell populations (Figure 1G). CD74, LGALS9 were significantly associated with metastasis in The Cancer Genome Atlas (TCGA) CRC cohorts. We also found CD44 and ITGAX were survival (Figure 1H; CD44 and ITGAX showed no significant differential expression between metastatic and non-metastatic patients, so data are not shown). These results indicated that the entire cellular interaction network of pmCRC ascites contributed to establishing an immunosuppressive and metastatic microenvironment.</p><p>We observed that the abundance of fibroblasts in pmCRC ascites samples was significantly greater in SD patients than in PD patients (Figure 1B). The fibroblasts were partitioned into 7 distinct clusters (C0-C6) based on unsupervised clustering (Figure 1I). All sub-clusters of cancer-associated fibroblasts (CAfs) showed a high expression of extracellular matrix cancer-associated fibroblasts (eCAFs) signature (Figure 1J), while inflammatory CAF (iCAF), myofibroblast CAF (myCAF), matrix CAF (mCAF), and vascular CAF (vCAF) only presented in a small fraction of fibroblasts (Supplementary Figure S1C), supporting the role of eCAFs in enhancing the metastatic potential of pmCRC. A higher abundance of antigen-presenting cancer-associated fibroblasts (apCAFs) was observed in the PD cohort (<i>n</i> = 310) than in the SD cohort (<i>n</i> = 93) (Wilcoxon test, <i>P</i> = 0.049). These results indicated that the CAFs in pmCRC ascites have bidirectional associations with immune regulation function, serving as a favorable candidate for CRC treatment. Differentially expressed genes and gene ontology (GO) analyses showed that the “cell-cell adhesion”, “inflammatory response”, and “cytokine production” were differentially enriched between primary tumors and ascites (Supplementary Figure S1D), which implied that the liquid state of ascites changed the functions of the fibroblast populations.</p><p>Macrophages were significantly enriched in pmCRC ascites and categorized into 8 sub-clusters (C0-C7) (Figure 1K). Using the previously defined “M1” and “M2” signatures, C2 showed an “M1-like” pattern, and C5 showed “M2-like” patterns. We also identified a small sub-cluster of C5 co-expressed both “M1” and “M2” gene signatures (Figure 1L), which have been reported in previous studies on solid tumors [<span>8</span>]. We next examined the expression of a series of the previously reported immunosuppressive genes (leukocyte-associated immunoglobulin-like receptor 1 [LAIR1], hepatitis A virus cellular receptor 2 [HAVCR2; also known as T cell immunoglobulin and mucin domain-containing protein 3], LGALS9, and V-set immunoregulatory receptor [VSIR]) in macrophage sub-clusters. Because the expression pattern of the “M2” marker gene CD163 perfectly coincided with that of LAIR1 in all sub-clusters (Figure 1M), we postulated that the immunosuppressive function of tumor-associated macrophages (TAMs) might be exerted via LAIR1. Two other immunosuppressive genes, T cell immune receptor with Ig and ITIM domains (TIGIT) and programmed cell death 1 (PDCD1), were also identified highly expressed in C5. C4 highly expressed the key immunosuppressive phenotypic marker triggering receptor expressed on myeloid cells 2 (TREM2) (Figure 1N). In summary, the majority of macrophages in pmCRC ascites exhibited high immunosuppressive features.</p><p>We identified 11 sub-clusters of T cells according to the expression of their respective markers, including CD4<sup>+</sup> T cells (C1, C4, C5, and C6) and CD8<sup>+</sup> T cells (C0, C2, C3, C7, C8, C9, and C10) (Figure 1O). Most CD8<sup>+</sup> effector memory cells re-expressing CD45RA T (Temra/Teff) cells (C8) were from patient 5 (P05), and CD8<sup>+</sup> effector memory T (Tem) cells (C7) were mostly from patient 8 (P08); the remaining 10 patients exhibited high heterogeneity in 11 T-cell sub-clusters (Supplementary Figure S1E-F). Importantly, CD8<sup>+</sup> tissue-resident memory (Trm) cells (C7), which were reported to be associated with forming a tertiary lymphoid structure (TLS) [<span>9</span>], were less abundant in SD patients (P03, P08, P09, P11, and P12). We also observed that all sub-clusters expressed ferritin light chain (FTL) (Supplementary Figure S1G), which was reported to regulate chemoresistance and metastasis in CRC [<span>10</span>]. We calculated the cytotoxicity, proliferation, and exhaustion signatures for all CD8<sup>+</sup> T-cell sub-clusters (Figure 1P). Only one sub-cluster of CD8<sup>+</sup> T cells was not show exhaustion signature (C10). Clusters 9 and 10 exhibited slightly higher proliferation, which could recruit cytotoxic T cells. The abundance of C9 and C10 was low, suggesting that T cells may play a minor role in the immune microenvironment of ascites and may work synergistically with other cell populations. Moreover, we predicted immunomodulatory drugs targeting macrophages, where gene sets were extracted from the macrophages of the pmCRC ascites data (Figure 1Q).</p><p>In conclusion, we found that T cells, fibroblasts, and macrophages exhibited immunosuppressive features in pmCRC ascites (Figure 1R). The cellular landscape of pmCRC ascites has the significant indication of patients’ immune status, providing insights for prognosis and therapy selection.</p><p>Haiyang Zhou made contributions to the conceptualization, funding acquisition, investigation, resource acquisition, and writing original draft.</p><p>Jiahui Yin made contributions to the data curation, formal analysis, and software analysis.</p><p>Anqi Wang made contributions to the formal analysis, investigation, and resource acquisition.</p><p>Xiaomao Yin made contributions to the formal analysis.</p><p>Taojun Jin made contributions to the formal analysis and visualization.</p><p>Kai Xu made contributions to the resource acquisition and supervision.</p><p>Lin Zhu made contributions to the investigation and resource acquisition.</p><p>Jiexuan Wang made contributions to the investigation and resources acquisition.</p><p>Wenqiang Wang made contributions to the resource acquisition.</p><p>Wei Zhang made contributions to the resource acquisition.</p><p>Xinxiang Li made contributions to the conceptualization, visualization, and writing—review &amp; editing.</p><p>Zhiqian Hu made contributions to the funding acquisition, visualization, and writing—review &amp; editing.</p><p>Xinxing Li made contributions to the investigation, resource acquisition, visualization, writing—original draft, and writing—review &amp; editing.</p><p>All authors read and approved the final manuscript.</p><p>The authors declare that they have no competing interests.</p><p>The authors gratefully acknowledge the financial support from the National Key R&amp;D Program of China (No. 2019YFA0110601), National Natural Science Foundation of China (No. 81571827), Natural Science Foundation Project of Shanghai Science and Technology Commission (SKW2030), Excellent Discipline Reserve Talent Plan of Tongji Hospital Affiliated to Tongji University (HBRC2014), Clinical research Project of Tongji Hospital Affiliated to Tongji University (ITJ-ZD-2104), Key talent introduction project of Tongji Hospital Affiliated to Tongji University (RCQD2102), Talent project of Tongji Hospital Affiliated to Tongji University (GJPY2111), and Shanghai Tongji Hospital special disease database construction project (TJ-DB-2105).</p><p>All the investigation protocols were approved by the Institutional Ethics Committees of Shanghai Changzheng Hospital and Shanghai Tongji Hospital (SBKT-2022-155). All subjects provided informed consent to participate in the study and approved the use of their biological samples for analysis. All experiments were performed following institutional guidelines, in compliance with relevant laws. Data sharing mechanisms will ensure that the rights and privacy of individuals participating in research will be guaranteed.</p>\",\"PeriodicalId\":9495,\"journal\":{\"name\":\"Cancer Communications\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":20.1000,\"publicationDate\":\"2024-03-26\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11260760/pdf/\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Cancer Communications\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://onlinelibrary.wiley.com/doi/10.1002/cac2.12541\",\"RegionNum\":1,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"ONCOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Cancer Communications","FirstCategoryId":"3","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/cac2.12541","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"ONCOLOGY","Score":null,"Total":0}
引用次数: 0

摘要

结直肠癌(CRC)患者出现恶性腹水与预后不良、复发风险高以及对化疗和免疫疗法的耐药性有关 [1-3]。我们利用10×单细胞RNA测序技术(scRNA-seq)分析了12名未经治疗的pmCRC患者腹水样本中96,065个细胞的单细胞转录组(补充图S1A,补充表S1)。通过特征性的典型细胞标记鉴定出11种主要细胞类型,包括上皮细胞、内皮细胞、成纤维细胞、T细胞、B细胞、单核细胞、巨噬细胞、浆细胞、自然杀伤(NK)细胞、树突状细胞(DC)和肥大细胞(图1A-B)。pmCRC 腹水的主要细胞成分是 T 细胞(40,095;41.7%)、巨噬细胞(28,487;29.7%)和成纤维细胞(5,932;6.2%)。原发性 CRC 的上皮细胞比例为 14.8%[4],而腹水中的上皮细胞比例仅为 0.3%(291 个)。pmCRC 腹水中上皮细胞的低比例与另一种肿瘤腹水的 scRNA-seq 研究一致[5-7]:根据治疗反应,我们将 12 名患者分为两组:8 名患者(P02、P03、P04、P07、P08、P09、P11 和 P12)病情稳定(SD),4 名患者(P01、P05、P06 和 P10)病情进展(PD)。单细胞转录组分析显示,12 名患者的细胞组成具有高度异质性。SD 组的成纤维细胞和上皮细胞比例较高(图 1B)。值得注意的是,成纤维细胞的表达特征在两组之间存在显著差异(图 1C),前五大上调/下调基因在 11 种细胞类型中均可视化(图 1D)。我们还发现,与原发肿瘤相比,pmCRC 腹水中巨噬细胞的频率明显增加[4](图 1E)。为了全面研究 pmCRC 腹水生态系统中的细胞相互作用,我们使用 CellChat 预测了细胞-细胞通讯网络。总体而言,我们发现了 44 对重要的配体-受体相互作用。虽然T细胞是pmCRC腹水中数量最多的细胞群(41.7%),但成纤维细胞和巨噬细胞是细胞相互作用网络的核心(图1F),这表明它们在pmCRC腹水生态系统中与不同细胞的招募和交叉对话中发挥着重要作用。巨噬细胞迁移抑制因子(MIF)、附件蛋白、补体和C-C趋化因子配体(CCL)是CRC腹水中最活跃的传出/传入信号分子(补充图S1B)。成纤维细胞通过MIF-(CD74 + C-X-C 趋化因子受体4型 [CXCR4])和MIF-(CD74 + CD44)轴的配体-受体相互作用以及C3-(整合素αX [ITGAX] + 整合素亚基β2 [ITGB2])直接与不同类型的细胞接触(图1G)。值得注意的是,巨噬细胞群更有可能通过粘附配体-受体对 galectin-9 (LGALS9)-CD44 和 LGALS9-CD45 与其他细胞群相互作用,而在其他细胞群中没有观察到这种情况(图 1G)。在癌症基因组图谱(TCGA)的 CRC 队列中,CD74、LGALS9 与转移显著相关。我们还发现 CD44 和 ITGAX 具有生存性(图 1H;CD44 和 ITGAX 在转移性和非转移性患者之间没有明显的表达差异,因此数据未显示)。这些结果表明,pmCRC 腹水的整个细胞相互作用网络有助于建立免疫抑制和转移微环境。我们观察到,在 pmCRC 腹水样本中,SD 患者成纤维细胞的丰度明显高于 PD 患者(图 1B)。根据无监督聚类法,成纤维细胞被划分为 7 个不同的集群(C0-C6)(图 1I)。所有癌症相关成纤维细胞(CAfs)亚簇都显示了细胞外基质癌症相关成纤维细胞(eCAFs)特征的高表达(图 1J),而炎症性 CAF(iCAF)、肌成纤维细胞 CAF(myCAF)、基质 CAF(mCAF)和炎症性 CAF(iCAF)的表达都很高、而炎性CAF(iCAF)、肌成纤维细胞CAF(myCAF)和血管CAF(vCAF)只出现在一小部分成纤维细胞中(补充图S1C),这支持了eCAFs在增强pmCRC转移潜能中的作用。在PD队列(n = 310)中观察到的抗原呈递癌症相关成纤维细胞(apCAFs)的丰度高于SD队列(n = 93)(Wilcoxon检验,P = 0.049)。 这些结果表明,pmCRC 腹水中的 CAFs 与免疫调节功能有双向关联,是治疗 CRC 的有利候选者。差异表达基因和基因本体(GO)分析表明,"细胞-细胞粘附"、"炎症反应 "和 "细胞因子产生 "在原发肿瘤和腹水中的富集程度不同(补充图 S1D),这意味着腹水的液态改变了成纤维细胞群的功能。利用之前定义的 "M1 "和 "M2 "特征,C2 显示出 "类 M1 "模式,C5 显示出 "类 M2 "模式。我们还发现 C5 中有一小部分同时表达 "M1 "和 "M2 "基因特征(图 1L),这在之前的实体瘤研究中已有报道[8]。接下来,我们检测了之前报道的一系列免疫抑制基因(白细胞相关免疫球蛋白样受体 1 [LAIR1]、甲型肝炎病毒细胞受体 2 [HAVCR2;又称 T 细胞免疫球蛋白和含粘蛋白结构域蛋白 3]、LGALS9 和 V 集免疫调节受体 [VSIR])在巨噬细胞亚簇中的表达情况。由于 "M2 "标记基因 CD163 的表达模式与 LAIR1 在所有亚簇中的表达模式完全一致(图 1M),我们推测肿瘤相关巨噬细胞(TAMs)的免疫抑制功能可能是通过 LAIR1 发挥的。另外两个免疫抑制基因,即具有 Ig 和 ITIM 结构域的 T 细胞免疫受体(TIGIT)和程序性细胞死亡 1(PDCD1),也在 C5 中高表达。C4 高表达关键的免疫抑制表型标志物--髓系细胞上表达的触发受体 2(TREM2)(图 1N)。总之,pmCRC 腹水中的大多数巨噬细胞表现出高度免疫抑制特征。我们根据其各自标记物的表达确定了 11 个 T 细胞亚群,包括 CD4+ T 细胞(C1、C4、C5 和 C6)和 CD8+ T 细胞(C0、C2、C3、C7、C8、C9 和 C10)(图 1O)。大多数重新表达CD45RA T(Temra/Teff)细胞的CD8+效应记忆细胞(C8)来自患者5(P05),而CD8+效应记忆T(Tem)细胞(C7)主要来自患者8(P08);其余10名患者的11个T细胞亚簇表现出高度异质性(补充图S1E-F)。重要的是,CD8+组织驻留记忆(Trm)细胞(C7)据报道与形成三级淋巴结构(TLS)有关[9],但在SD患者(P03、P08、P09、P11和P12)中含量较少。我们还观察到,所有亚簇均表达铁蛋白轻链(FTL)(补充图 S1G),据报道,铁蛋白轻链可调控 CRC 的化疗耐药性和转移[10]。我们计算了所有 CD8+ T 细胞亚簇的细胞毒性、增殖和衰竭特征(图 1P)。只有一个 CD8+ T 细胞亚簇未显示衰竭特征(C10)。簇 9 和簇 10 的增殖率略高,这可能会招募细胞毒性 T 细胞。C9 和 C10 的丰度较低,这表明 T 细胞在腹水的免疫微环境中可能扮演次要角色,并可能与其他细胞群协同作用。此外,我们还预测了以巨噬细胞为靶点的免疫调节药物,从 pmCRC 腹水数据的巨噬细胞中提取了基因集(图 1Q)。总之,我们发现 T 细胞、成纤维细胞和巨噬细胞在 pmCRC 腹水中表现出免疫抑制特征(图 1R)。pmCRC腹水的细胞图谱对患者的免疫状态有重要指示作用,为预后和治疗选择提供了启示。尹佳慧在数据整理、形式分析和软件分析方面做出了贡献。王安琪在形式分析、调查和资源获取方面做出了贡献。朱琳在调查和资源获取方面做出了贡献。王杰轩在调查和资源获取方面做出了贡献。王文强在资源获取方面做出了贡献。胡志谦在资金获取、可视化和写作-审阅-编辑方面做出了贡献。李新星在调查、资源获取、可视化、写作-原稿和写作-审阅-编辑方面做出了贡献。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

摘要图片

查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
Single-cell landscape of malignant ascites from patients with metastatic colorectal cancer

The presence of malignant ascites in colorectal cancer (CRC) patients is associated with a poor prognosis, a high risk of recurrence, and resistance to chemotherapy and immune therapy [1-3]. Understanding the complex interactions among different kinds of cells and the ecosystem of peritoneal metastasized colorectal cancer (pmCRC) ascites may provide insights into effective treatment strategies.

We profiled the single-cell transcriptomes of 96,065 cells from ascites samples of 12 treatment-naïve patients with pmCRC using the 10× single-cell RNA-sequencing (scRNA-seq) (Supplementary Figure S1A, Supplementary Table S1). Eleven major cell types were identified by characteristic canonical cell markers, including epithelial cells, endothelial cells, fibroblasts, T cells, B cells, monocytes, macrophages, plasma cells, natural killer (NK) cells, dendritic cells (DCs), and mast cells (Figure 1A-B). The main cellular components of pmCRC ascites are T cells (40,095; 41.7%), macrophages (28,487; 29.7%), and fibroblasts (5,932; 6.2%). Compared with primary CRC, which showed 14.8% epithelial cells [4], only 0.3% (291) epithelial cells were found in the ascites. The low percentage of epithelial cells in pmCRC ascites was consistent with the scRNA-seq studies of another tumor ascites [5-7].

We classified the 12 patients into 2 groups according to their treatment response as follows: 8 patients (P02, P03, P04, P07, P08, P09, P11, and P12) had stable disease (SD), while 4 (P01, P05, P06, and P10) had progressive disease (PD). Single-cell transcriptomic analyses have revealed high heterogeneity of cell composition in 12 patients. The SD group exhibited a higher proportion of fibroblasts and epithelial cells (Figure 1B). Remarkably, fibroblasts had significantly different expression characteristics between the 2 groups (Figure 1C), and the top five upregulated/downregulated genes were visualized in 11 cell types (Figure 1D). We also found a significant increase in the frequency of macrophages in pmCRC ascites compared with the primary tumors [4] (Figure 1E). It hinted that significant inter-patient variability in the composition and functional programs of pmCRC ascites cells under different disease states.

To comprehensively study the cellular interactions within the pmCRC ascites ecosystem, we predicted cell-cell communication networks using CellChat. Overall, we identified 44 significant ligand-receptor pair interactions. Although T cells were the most abundant cell population (41.7%) in pmCRC ascites, fibroblasts and macrophages were the core of the cellular interaction network (Figure 1F), suggesting their important roles in recruiting and cross-talking with diverse cells in the pmCRC ascites ecosystem.

The result of cellular communications suggested that there was a complex interplay between various signaling molecule. Macrophage migration inhibitory factor (MIF), annexin, complement, and C-C chemokine ligand (CCL) were the most active outgoing/incoming signaling molecules in CRC ascites (Supplementary Figure S1B). Fibroblasts directly contacted with different types of cells via ligand-receptor interactions of the MIF-(CD74 + C-X-C chemokine receptor type 4 [CXCR4]) and MIF-(CD74 + CD44) axes and C3-(integrin alpha X [ITGAX] + integrin subunit beta 2 [ITGB2]) (Figure 1G). Notably, macrophage populations were more likely to interact with other cells through the adhesive ligand-receptor pairs galectin-9 (LGALS9)-CD44 and LGALS9-CD45, which were not observed in other cell populations (Figure 1G). CD74, LGALS9 were significantly associated with metastasis in The Cancer Genome Atlas (TCGA) CRC cohorts. We also found CD44 and ITGAX were survival (Figure 1H; CD44 and ITGAX showed no significant differential expression between metastatic and non-metastatic patients, so data are not shown). These results indicated that the entire cellular interaction network of pmCRC ascites contributed to establishing an immunosuppressive and metastatic microenvironment.

We observed that the abundance of fibroblasts in pmCRC ascites samples was significantly greater in SD patients than in PD patients (Figure 1B). The fibroblasts were partitioned into 7 distinct clusters (C0-C6) based on unsupervised clustering (Figure 1I). All sub-clusters of cancer-associated fibroblasts (CAfs) showed a high expression of extracellular matrix cancer-associated fibroblasts (eCAFs) signature (Figure 1J), while inflammatory CAF (iCAF), myofibroblast CAF (myCAF), matrix CAF (mCAF), and vascular CAF (vCAF) only presented in a small fraction of fibroblasts (Supplementary Figure S1C), supporting the role of eCAFs in enhancing the metastatic potential of pmCRC. A higher abundance of antigen-presenting cancer-associated fibroblasts (apCAFs) was observed in the PD cohort (n = 310) than in the SD cohort (n = 93) (Wilcoxon test, P = 0.049). These results indicated that the CAFs in pmCRC ascites have bidirectional associations with immune regulation function, serving as a favorable candidate for CRC treatment. Differentially expressed genes and gene ontology (GO) analyses showed that the “cell-cell adhesion”, “inflammatory response”, and “cytokine production” were differentially enriched between primary tumors and ascites (Supplementary Figure S1D), which implied that the liquid state of ascites changed the functions of the fibroblast populations.

Macrophages were significantly enriched in pmCRC ascites and categorized into 8 sub-clusters (C0-C7) (Figure 1K). Using the previously defined “M1” and “M2” signatures, C2 showed an “M1-like” pattern, and C5 showed “M2-like” patterns. We also identified a small sub-cluster of C5 co-expressed both “M1” and “M2” gene signatures (Figure 1L), which have been reported in previous studies on solid tumors [8]. We next examined the expression of a series of the previously reported immunosuppressive genes (leukocyte-associated immunoglobulin-like receptor 1 [LAIR1], hepatitis A virus cellular receptor 2 [HAVCR2; also known as T cell immunoglobulin and mucin domain-containing protein 3], LGALS9, and V-set immunoregulatory receptor [VSIR]) in macrophage sub-clusters. Because the expression pattern of the “M2” marker gene CD163 perfectly coincided with that of LAIR1 in all sub-clusters (Figure 1M), we postulated that the immunosuppressive function of tumor-associated macrophages (TAMs) might be exerted via LAIR1. Two other immunosuppressive genes, T cell immune receptor with Ig and ITIM domains (TIGIT) and programmed cell death 1 (PDCD1), were also identified highly expressed in C5. C4 highly expressed the key immunosuppressive phenotypic marker triggering receptor expressed on myeloid cells 2 (TREM2) (Figure 1N). In summary, the majority of macrophages in pmCRC ascites exhibited high immunosuppressive features.

We identified 11 sub-clusters of T cells according to the expression of their respective markers, including CD4+ T cells (C1, C4, C5, and C6) and CD8+ T cells (C0, C2, C3, C7, C8, C9, and C10) (Figure 1O). Most CD8+ effector memory cells re-expressing CD45RA T (Temra/Teff) cells (C8) were from patient 5 (P05), and CD8+ effector memory T (Tem) cells (C7) were mostly from patient 8 (P08); the remaining 10 patients exhibited high heterogeneity in 11 T-cell sub-clusters (Supplementary Figure S1E-F). Importantly, CD8+ tissue-resident memory (Trm) cells (C7), which were reported to be associated with forming a tertiary lymphoid structure (TLS) [9], were less abundant in SD patients (P03, P08, P09, P11, and P12). We also observed that all sub-clusters expressed ferritin light chain (FTL) (Supplementary Figure S1G), which was reported to regulate chemoresistance and metastasis in CRC [10]. We calculated the cytotoxicity, proliferation, and exhaustion signatures for all CD8+ T-cell sub-clusters (Figure 1P). Only one sub-cluster of CD8+ T cells was not show exhaustion signature (C10). Clusters 9 and 10 exhibited slightly higher proliferation, which could recruit cytotoxic T cells. The abundance of C9 and C10 was low, suggesting that T cells may play a minor role in the immune microenvironment of ascites and may work synergistically with other cell populations. Moreover, we predicted immunomodulatory drugs targeting macrophages, where gene sets were extracted from the macrophages of the pmCRC ascites data (Figure 1Q).

In conclusion, we found that T cells, fibroblasts, and macrophages exhibited immunosuppressive features in pmCRC ascites (Figure 1R). The cellular landscape of pmCRC ascites has the significant indication of patients’ immune status, providing insights for prognosis and therapy selection.

Haiyang Zhou made contributions to the conceptualization, funding acquisition, investigation, resource acquisition, and writing original draft.

Jiahui Yin made contributions to the data curation, formal analysis, and software analysis.

Anqi Wang made contributions to the formal analysis, investigation, and resource acquisition.

Xiaomao Yin made contributions to the formal analysis.

Taojun Jin made contributions to the formal analysis and visualization.

Kai Xu made contributions to the resource acquisition and supervision.

Lin Zhu made contributions to the investigation and resource acquisition.

Jiexuan Wang made contributions to the investigation and resources acquisition.

Wenqiang Wang made contributions to the resource acquisition.

Wei Zhang made contributions to the resource acquisition.

Xinxiang Li made contributions to the conceptualization, visualization, and writing—review & editing.

Zhiqian Hu made contributions to the funding acquisition, visualization, and writing—review & editing.

Xinxing Li made contributions to the investigation, resource acquisition, visualization, writing—original draft, and writing—review & editing.

All authors read and approved the final manuscript.

The authors declare that they have no competing interests.

The authors gratefully acknowledge the financial support from the National Key R&D Program of China (No. 2019YFA0110601), National Natural Science Foundation of China (No. 81571827), Natural Science Foundation Project of Shanghai Science and Technology Commission (SKW2030), Excellent Discipline Reserve Talent Plan of Tongji Hospital Affiliated to Tongji University (HBRC2014), Clinical research Project of Tongji Hospital Affiliated to Tongji University (ITJ-ZD-2104), Key talent introduction project of Tongji Hospital Affiliated to Tongji University (RCQD2102), Talent project of Tongji Hospital Affiliated to Tongji University (GJPY2111), and Shanghai Tongji Hospital special disease database construction project (TJ-DB-2105).

All the investigation protocols were approved by the Institutional Ethics Committees of Shanghai Changzheng Hospital and Shanghai Tongji Hospital (SBKT-2022-155). All subjects provided informed consent to participate in the study and approved the use of their biological samples for analysis. All experiments were performed following institutional guidelines, in compliance with relevant laws. Data sharing mechanisms will ensure that the rights and privacy of individuals participating in research will be guaranteed.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
Cancer Communications
Cancer Communications Biochemistry, Genetics and Molecular Biology-Cancer Research
CiteScore
25.50
自引率
4.30%
发文量
153
审稿时长
4 weeks
期刊介绍: Cancer Communications is an open access, peer-reviewed online journal that encompasses basic, clinical, and translational cancer research. The journal welcomes submissions concerning clinical trials, epidemiology, molecular and cellular biology, and genetics.
期刊最新文献
Perioperative toripalimab plus neoadjuvant chemotherapy might improve outcomes in resectable esophageal cancer: an interim analysis of a phase III randomized clinical trial. Tryptophan 2,3-dioxygenase-positive matrix fibroblasts fuel breast cancer lung metastasis via kynurenine-mediated ferroptosis resistance of metastatic cells and T cell dysfunction. Targeting SRSF10 might inhibit M2 macrophage polarization and potentiate anti-PD-1 therapy in hepatocellular carcinoma. Unraveling the routes to distant metastases in colorectal cancer: Tumor deposits and lymph node metastases as the gateway. Issue Information
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1