Single-cell transcriptomic atlas reveals immune and metabolism perturbation of depression in the pathogenesis of breast cancer

IF 20.1 1区 医学 Q1 ONCOLOGY
Lingling Wu, Junwei Liu, Yimeng Geng, Jianwen Fang, Xingle Gao, Jianbo Lai, Minya Yao, Shaojia Lu, Weiwei Yin, Peifen Fu, Wei Chen, Shaohua Hu
{"title":"Single-cell transcriptomic atlas reveals immune and metabolism perturbation of depression in the pathogenesis of breast cancer","authors":"Lingling Wu, Junwei Liu, Yimeng Geng, Jianwen Fang, Xingle Gao, Jianbo Lai, Minya Yao, Shaojia Lu, Weiwei Yin, Peifen Fu, Wei Chen, Shaohua Hu","doi":"10.1002/cac2.12603","DOIUrl":null,"url":null,"abstract":"<p>Epidemiological evidence indicates that major depressive disorder (MDD) may predispose the development and prognosis of breast cancer (BC) in females [<span>1</span>]. However, the mechanisms linking these phenotypes are not fully understood. Chronic stress, a hallmark of depression, has been underscored to affect anti-tumor immunity, tumor metabolic reprogramming, hormone synthesis in BC [<span>2, 3</span>], and increase tumor metastasis [<span>4</span>], but there is a lack of detailed cellular-level characterization of how MDD history affects the tumorigenesis of BC. This study explored the single-cell atlas of multiple tissues from BC patients with and without a history of MDD for characterizing the potential molecular alternations in their tumorigenesis (Figure 1A).</p>\n<figure><picture>\n<source media=\"(min-width: 1650px)\" srcset=\"/cms/asset/7dfea6ca-4ed3-4e4d-814d-bc3b80b4b013/cac212603-fig-0001-m.jpg\"/><img alt=\"Details are in the caption following the image\" data-lg-src=\"/cms/asset/7dfea6ca-4ed3-4e4d-814d-bc3b80b4b013/cac212603-fig-0001-m.jpg\" loading=\"lazy\" src=\"/cms/asset/5468c0bd-c08d-4563-8e5a-ddde12a7a3d9/cac212603-fig-0001-m.png\" title=\"Details are in the caption following the image\"/></picture><figcaption>\n<div><strong>FIGURE 1<span style=\"font-weight:normal\"></span></strong><div>Open in figure viewer</div>\n</div>\n<div>scRNA-seq reveals MDD-associated pathogenesis of breast cancer. (A) The experimental design of single-cell analyses in this study. (B) UMAP plot of single cells from tumor tissues, adjacent normal tissues, and peripheral blood samples, colored by major cell types. (C) Comparisons of the enrichment of OXPHOS (left) and glycolysis (right) pathways within aneuploidy cells from BC-Ctrl and BC-MDD groups. (D) Overall survival analyses of the contributions of top-50 highly-expressed gene signatures of LumSec-2 (left) and LumSec-3 (right) clusters in prognosis prediction of the estrogen receptor-positive patients in the TCGA-BRCA cohort. (E) Venn plot of the overlaps between upregulated differentially expressed genes of endothelial, fibroblast, and pericyte cells between BC-Ctrl and BC-MDD sample groups (left) and the pathway enrichment analysis of the shared upregulated genes (right). (F) Pathway enrichment analysis of differentially expressed genes of CD8<sup>+</sup> T cells (left) and macrophages (right) in the primary tumor tissue of BC-Ctrl and BC-MDD patients. (G) The number of interactions of ligands and receptors across major cell subtypes within BC-Ctrl (left) and BC-MDD (right) samples. (H) The changes of incoming and outgoing interactions across major cell subtypes between BC-MDD and BC-Ctrl primary tumor tissues. Abbreviations: scRNA-seq, single-cell RNA sequencing; BC, breast cancer; MDD, major depressive disorder; UMAP, uniform manifold approximation and projection; OXPHOS, oxidative phosphorylation; GSVA, gene set variation analysis; LumSec, luminal secretory cell; DEGs, differentially expressed genes.</div>\n</figcaption>\n</figure>\n<p>Paired primary tumor tissues (<i>n</i> = 10), adjacent normal tissues (<i>n</i> = 7), and peripheral blood samples (<i>n</i> = 10) were collected from a cohort of 10 BC patients, 5 of whom had a history of MDD (Supplementary Table S1). All BC patients had estrogen receptor (ER)-positive tumors and were further predicted as Luminal A (<i>n</i> = 9) and B subtypes (<i>n</i> = 1) (Supplementary Table S2). Further details on patient recruitment, sample handling, and single-cell data analysis are provided in Supplementary Methods. In total, we obtained 224,557 single cells and further annotated them into major cell subsets based on lineage markers and copy number variations [<span>5</span>] (Figure 1B, Supplementary Figure S1A-B). Aneuploid cells in primary tumor tissues were obtained (Supplementary Table S3) to characterize their phenotypic differences in BC patients with MDD history (BC-MDD) or not (BC-Ctrl). The Uniform Manifold Approximation and Projection (UMAP) of unintegrated aneuploid cells revealed intrinsic differences across individual patients (Supplementary Figure S1C). Downstream functional profiling analysis identified distinct immune response pathways in BC-MDD and BC-Ctrl groups and enrichment of the oxidative phosphorylation (OXPHOS) pathway in BC-Ctrl tumors (Supplementary Figure S1D-E). Cellular Gene Set Variation Analysis (GSVA) [<span>6</span>] confirmed the distinct metabolic phenotypes between BC-MDD and BC-Ctrl groups (Figure 1C). Additionally, utilizing predefined gene module (GM) signatures of BC tumor cells [<span>7</span>], we observed specific restraint of GM4 and GM6 in BC-MDD (Supplementary Figure S1F-G). GM6 encompasses various antigen presentation genes, aligning with the observed downregulation of major histocompatibility complex class I (MHC-I) class genes in BC-MDD tumor cells (Supplementary Figure S1H).</p>\n<p>Upon re-clustering 12,371 normal epithelial cells within primary breast tumor and adjacent normal tissues from the 10 patients, we identified 9 cell clusters, including luminal hormone-responsive (LumHR), luminal secretory (LumSec), and myoepithelial cells [<span>8</span>] (Supplementary Figure S2A-B). Distribution analysis suggested the potential enrichment of LumSec-2 and -3 clusters in primary tumor tissues of BC-Ctrl patients (Supplementary Figure S2C-E). Subsequently, we investigated the potential roles of these BC-Ctrl-enriched tumor epithelial cells in tumorigenesis and prognosis. We utilized the top 50 differentially expressed genes (DEGs) of these cell clusters to evaluate their prognostic roles using The Cancer Genome Atlas-Breast Invasive Carcinoma (TCGA-BRCA) datasets [<span>9</span>]. Notably, we revealed that the gene signatures of these cells could only predict the prognosis of ER<sup>+</sup> but not ER<sup>−</sup> patients (Figure 1D, Supplementary Figure S2F). Despite the high expression of basal and human epidermal growth factor receptor 2 (HER2) breast cancer phenotypes, the prognostic roles of these cells may be related to the downregulation of prostate-derived ETS factor (PDEF/SPDEF) [<span>10</span>] (Supplementary Figure S2G-H).</p>\n<p>We then integrated and characterized the functional profiling of 25,076 stromal cells within primary breast tumor and adjacent normal tissues, encompassing 7 endothelial cell, 10 fibroblast, and 5 pericyte clusters (Supplementary Figure S3A-E). Frequency distribution analyses identified the tumor-specific cell cluster enrichment but not across patient groups in both tissue sites (Supplementary Figure S3F-H). We then characterized the potential MDD-specific functional changes within stromal cells, and noted consistent functional changes across all major stromal subtypes. We identified 70 upregulated genes shared by all stromal subtypes in BC-MDD patients, with pathway analysis suggested their involvement in metabolic regulation and cellular stress response pathways, consistent with trends observed in individual patients (Figure 1E, Supplementary Figure S3I). Specifically, we found the potential up-regulation of solute carrier family 38 member 2 (<i>SLC38A2</i>) and heat shock protein family H member 1 (<i>HSPH1</i>) in primary breast tumor and adjacent normal tissues from BC-MDD patients (Supplementary Figure S3J), highlighting the potential roles of amino acid transport and stress responses in MDD-affected BC patients.</p>\n<p>To characterize the T cell immune response alterations in BC-MDD patients, we re-integrated 91,615 annotated T/natural killer (NK) cells from primary breast tumors, adjacent normal tissues, and peripheral blood samples. This analysis identified 15 distinct cell clusters (Supplementary Figure S4A-B). Notably, C-X-C motif chemokine ligand 13 (<i>CXCL13</i>)<sup>+</sup> CD4<sup>+</sup> T cells (CD4-Tem-2) and regulatory T cells were enriched in primary tumors, NK and γδT cells were enriched in peripheral blood (Supplementary Figure S4C). However, the frequency comparisons of T/NK cell clusters across patient groups revealed non-significant differences across all tissues (Supplementary Figure S4D). We then profiled the functional alternations in CD8<sup>+</sup> T cells in primary tumors with MDD history, which proposed the enrichment of interferon-related genes and cytotoxic genes in BC-Ctrl tumor tissues, validated with individual patients (Supplementary Figure S4E-F). This finding is consistent with the diminished activation of immune response and cytokine response pathways in CD8 T cells in the BC-MDD group (Figure 1F).</p>\n<p>We conducted a parallel analysis with 27,563 myeloid cells and identified 16 cell clusters within monocytes, macrophages, and dendritic cells (DCs) (Supplementary Figure S5A-B). Among those, the Macro-m1-1 cell cluster, characterized by high expression of chemokine genes, predominated in primary tumor tissues, alongside the G protein-coupled receptor 183 (<i>GPR183</i>)-positive cDC-2 cells (Supplementary Figure S5C). Conversely, we observed no significant distribution variations in myeloid cells across all tissues among patient groups (Supplementary Figure S5D). Exploring the functional modifications of macrophages in primary tumor tissues, we uncovered enrichment of fatty acid-binding genes in the BC-MDD group and interferon-related genes in the BC-Ctrl group, which were validated across individual patients (Supplementary Figure S5E-F). Similar to CD8<sup>+</sup> T cells, we revealed enrichment of stress response and metabolic-related pathways but with the impairment of innate immune response pathways in the BC-MDD group (Figure 1F).</p>\n<p>We next compared the ligand-receptor interactions within primary tumor tissues, and observed differences in interaction number among major cell types between BC-Ctrl and BC-MDD groups (Figure 1G). In the BC-MDD group, the signal strength for interactions showed a significant reduction in epithelial cells, followed by fibroblasts and tumor cells, while signals related to T/NK cells exhibited a slight enhancement (Figure 1H). We then focused on the interaction changes within tumor cells, epithelial cells, and T/NK cells. Within T/NK cells, the C-X-C chemokine receptor 4 and ligand 12 (<i>CXCR4</i>-<i>CXCL12</i>) axis and macrophage migration inhibitory factor (MIF) signal were upregulated in the BC-MDD group (Supplementary Figure S6A-B). Meanwhile, we identified the enrichment in signals of midkine (MDK), laminin, and fibronectin (FN1) in the BC-Ctrl group (Supplementary Figure S6C-D). These results suggest altered breast cancer tumor signal interactions associated with a history of MDD.</p>\n<p>In summary, our study offers preliminary insights regarding the impacts of MDD history in BC patients. We suggest a potential association between MDD history and a poorer prognosis in BC, characterized by a decrease in specific epithelial cells and impaired immune regulation signals within primary tumors. Furthermore, we propose potential alterations in the metabolic status of BC tumor cells in patients with MDD. Given the constraints of sample size and limited follow-up duration, larger patient cohorts, longitudinal data, and additional validation experiments are necessary to fully understand the complex interplay between MDD history and BC.</p>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":null,"pages":null},"PeriodicalIF":20.1000,"publicationDate":"2024-09-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Cancer Communications","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1002/cac2.12603","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

Abstract

Epidemiological evidence indicates that major depressive disorder (MDD) may predispose the development and prognosis of breast cancer (BC) in females [1]. However, the mechanisms linking these phenotypes are not fully understood. Chronic stress, a hallmark of depression, has been underscored to affect anti-tumor immunity, tumor metabolic reprogramming, hormone synthesis in BC [2, 3], and increase tumor metastasis [4], but there is a lack of detailed cellular-level characterization of how MDD history affects the tumorigenesis of BC. This study explored the single-cell atlas of multiple tissues from BC patients with and without a history of MDD for characterizing the potential molecular alternations in their tumorigenesis (Figure 1A).

Abstract Image
FIGURE 1
Open in figure viewer
scRNA-seq reveals MDD-associated pathogenesis of breast cancer. (A) The experimental design of single-cell analyses in this study. (B) UMAP plot of single cells from tumor tissues, adjacent normal tissues, and peripheral blood samples, colored by major cell types. (C) Comparisons of the enrichment of OXPHOS (left) and glycolysis (right) pathways within aneuploidy cells from BC-Ctrl and BC-MDD groups. (D) Overall survival analyses of the contributions of top-50 highly-expressed gene signatures of LumSec-2 (left) and LumSec-3 (right) clusters in prognosis prediction of the estrogen receptor-positive patients in the TCGA-BRCA cohort. (E) Venn plot of the overlaps between upregulated differentially expressed genes of endothelial, fibroblast, and pericyte cells between BC-Ctrl and BC-MDD sample groups (left) and the pathway enrichment analysis of the shared upregulated genes (right). (F) Pathway enrichment analysis of differentially expressed genes of CD8+ T cells (left) and macrophages (right) in the primary tumor tissue of BC-Ctrl and BC-MDD patients. (G) The number of interactions of ligands and receptors across major cell subtypes within BC-Ctrl (left) and BC-MDD (right) samples. (H) The changes of incoming and outgoing interactions across major cell subtypes between BC-MDD and BC-Ctrl primary tumor tissues. Abbreviations: scRNA-seq, single-cell RNA sequencing; BC, breast cancer; MDD, major depressive disorder; UMAP, uniform manifold approximation and projection; OXPHOS, oxidative phosphorylation; GSVA, gene set variation analysis; LumSec, luminal secretory cell; DEGs, differentially expressed genes.

Paired primary tumor tissues (n = 10), adjacent normal tissues (n = 7), and peripheral blood samples (n = 10) were collected from a cohort of 10 BC patients, 5 of whom had a history of MDD (Supplementary Table S1). All BC patients had estrogen receptor (ER)-positive tumors and were further predicted as Luminal A (n = 9) and B subtypes (n = 1) (Supplementary Table S2). Further details on patient recruitment, sample handling, and single-cell data analysis are provided in Supplementary Methods. In total, we obtained 224,557 single cells and further annotated them into major cell subsets based on lineage markers and copy number variations [5] (Figure 1B, Supplementary Figure S1A-B). Aneuploid cells in primary tumor tissues were obtained (Supplementary Table S3) to characterize their phenotypic differences in BC patients with MDD history (BC-MDD) or not (BC-Ctrl). The Uniform Manifold Approximation and Projection (UMAP) of unintegrated aneuploid cells revealed intrinsic differences across individual patients (Supplementary Figure S1C). Downstream functional profiling analysis identified distinct immune response pathways in BC-MDD and BC-Ctrl groups and enrichment of the oxidative phosphorylation (OXPHOS) pathway in BC-Ctrl tumors (Supplementary Figure S1D-E). Cellular Gene Set Variation Analysis (GSVA) [6] confirmed the distinct metabolic phenotypes between BC-MDD and BC-Ctrl groups (Figure 1C). Additionally, utilizing predefined gene module (GM) signatures of BC tumor cells [7], we observed specific restraint of GM4 and GM6 in BC-MDD (Supplementary Figure S1F-G). GM6 encompasses various antigen presentation genes, aligning with the observed downregulation of major histocompatibility complex class I (MHC-I) class genes in BC-MDD tumor cells (Supplementary Figure S1H).

Upon re-clustering 12,371 normal epithelial cells within primary breast tumor and adjacent normal tissues from the 10 patients, we identified 9 cell clusters, including luminal hormone-responsive (LumHR), luminal secretory (LumSec), and myoepithelial cells [8] (Supplementary Figure S2A-B). Distribution analysis suggested the potential enrichment of LumSec-2 and -3 clusters in primary tumor tissues of BC-Ctrl patients (Supplementary Figure S2C-E). Subsequently, we investigated the potential roles of these BC-Ctrl-enriched tumor epithelial cells in tumorigenesis and prognosis. We utilized the top 50 differentially expressed genes (DEGs) of these cell clusters to evaluate their prognostic roles using The Cancer Genome Atlas-Breast Invasive Carcinoma (TCGA-BRCA) datasets [9]. Notably, we revealed that the gene signatures of these cells could only predict the prognosis of ER+ but not ER patients (Figure 1D, Supplementary Figure S2F). Despite the high expression of basal and human epidermal growth factor receptor 2 (HER2) breast cancer phenotypes, the prognostic roles of these cells may be related to the downregulation of prostate-derived ETS factor (PDEF/SPDEF) [10] (Supplementary Figure S2G-H).

We then integrated and characterized the functional profiling of 25,076 stromal cells within primary breast tumor and adjacent normal tissues, encompassing 7 endothelial cell, 10 fibroblast, and 5 pericyte clusters (Supplementary Figure S3A-E). Frequency distribution analyses identified the tumor-specific cell cluster enrichment but not across patient groups in both tissue sites (Supplementary Figure S3F-H). We then characterized the potential MDD-specific functional changes within stromal cells, and noted consistent functional changes across all major stromal subtypes. We identified 70 upregulated genes shared by all stromal subtypes in BC-MDD patients, with pathway analysis suggested their involvement in metabolic regulation and cellular stress response pathways, consistent with trends observed in individual patients (Figure 1E, Supplementary Figure S3I). Specifically, we found the potential up-regulation of solute carrier family 38 member 2 (SLC38A2) and heat shock protein family H member 1 (HSPH1) in primary breast tumor and adjacent normal tissues from BC-MDD patients (Supplementary Figure S3J), highlighting the potential roles of amino acid transport and stress responses in MDD-affected BC patients.

To characterize the T cell immune response alterations in BC-MDD patients, we re-integrated 91,615 annotated T/natural killer (NK) cells from primary breast tumors, adjacent normal tissues, and peripheral blood samples. This analysis identified 15 distinct cell clusters (Supplementary Figure S4A-B). Notably, C-X-C motif chemokine ligand 13 (CXCL13)+ CD4+ T cells (CD4-Tem-2) and regulatory T cells were enriched in primary tumors, NK and γδT cells were enriched in peripheral blood (Supplementary Figure S4C). However, the frequency comparisons of T/NK cell clusters across patient groups revealed non-significant differences across all tissues (Supplementary Figure S4D). We then profiled the functional alternations in CD8+ T cells in primary tumors with MDD history, which proposed the enrichment of interferon-related genes and cytotoxic genes in BC-Ctrl tumor tissues, validated with individual patients (Supplementary Figure S4E-F). This finding is consistent with the diminished activation of immune response and cytokine response pathways in CD8 T cells in the BC-MDD group (Figure 1F).

We conducted a parallel analysis with 27,563 myeloid cells and identified 16 cell clusters within monocytes, macrophages, and dendritic cells (DCs) (Supplementary Figure S5A-B). Among those, the Macro-m1-1 cell cluster, characterized by high expression of chemokine genes, predominated in primary tumor tissues, alongside the G protein-coupled receptor 183 (GPR183)-positive cDC-2 cells (Supplementary Figure S5C). Conversely, we observed no significant distribution variations in myeloid cells across all tissues among patient groups (Supplementary Figure S5D). Exploring the functional modifications of macrophages in primary tumor tissues, we uncovered enrichment of fatty acid-binding genes in the BC-MDD group and interferon-related genes in the BC-Ctrl group, which were validated across individual patients (Supplementary Figure S5E-F). Similar to CD8+ T cells, we revealed enrichment of stress response and metabolic-related pathways but with the impairment of innate immune response pathways in the BC-MDD group (Figure 1F).

We next compared the ligand-receptor interactions within primary tumor tissues, and observed differences in interaction number among major cell types between BC-Ctrl and BC-MDD groups (Figure 1G). In the BC-MDD group, the signal strength for interactions showed a significant reduction in epithelial cells, followed by fibroblasts and tumor cells, while signals related to T/NK cells exhibited a slight enhancement (Figure 1H). We then focused on the interaction changes within tumor cells, epithelial cells, and T/NK cells. Within T/NK cells, the C-X-C chemokine receptor 4 and ligand 12 (CXCR4-CXCL12) axis and macrophage migration inhibitory factor (MIF) signal were upregulated in the BC-MDD group (Supplementary Figure S6A-B). Meanwhile, we identified the enrichment in signals of midkine (MDK), laminin, and fibronectin (FN1) in the BC-Ctrl group (Supplementary Figure S6C-D). These results suggest altered breast cancer tumor signal interactions associated with a history of MDD.

In summary, our study offers preliminary insights regarding the impacts of MDD history in BC patients. We suggest a potential association between MDD history and a poorer prognosis in BC, characterized by a decrease in specific epithelial cells and impaired immune regulation signals within primary tumors. Furthermore, we propose potential alterations in the metabolic status of BC tumor cells in patients with MDD. Given the constraints of sample size and limited follow-up duration, larger patient cohorts, longitudinal data, and additional validation experiments are necessary to fully understand the complex interplay between MDD history and BC.

单细胞转录组图谱揭示了乳腺癌发病机制中免疫和代谢对抑郁症的干扰
流行病学证据表明,重度抑郁障碍(MDD)可能导致女性乳腺癌(BC)的发生和预后[1]。然而,这些表型之间的关联机制尚不完全清楚。慢性应激是抑郁症的特征之一,已被强调会影响抗肿瘤免疫、肿瘤代谢重编程、乳腺癌激素合成[2, 3],并增加肿瘤转移[4],但目前还缺乏关于MDD病史如何影响乳腺癌肿瘤发生的详细细胞水平表征。本研究探索了有和无 MDD 史的 BC 患者多个组织的单细胞图谱,以表征其肿瘤发生过程中的潜在分子变化(图 1A)。(A)本研究中单细胞分析的实验设计。(B) 肿瘤组织、邻近正常组织和外周血样本单细胞的 UMAP 图,按主要细胞类型着色。(C) BC-Ctrl 组和 BC-MDD 组非整倍体细胞中 OXPHOS(左)和糖酵解(右)通路的富集比较。(D) LumSec-2(左)和 LumSec-3(右)簇前 50 个高表达基因特征对 TCGA-BRCA 队列中雌激素受体阳性患者预后预测贡献的总生存率分析。(E) BC-Ctrl和BC-MDD样本组之间内皮细胞、成纤维细胞和周细胞的上调差异表达基因重叠的维恩图(左)以及共享上调基因的通路富集分析(右)。(F)BC-Ctrl 和 BC-MDD 患者原发肿瘤组织中 CD8+ T 细胞(左)和巨噬细胞(右)差异表达基因的通路富集分析。(G)BC-Ctrl(左)和 BC-MDD(右)样本中主要细胞亚型的配体和受体相互作用的数量。(H)BC-MDD 和 BC-Ctrl 原发性肿瘤组织中各主要细胞亚型的传入和传出相互作用的变化。缩写:SCRNA-seq,单细胞 RNA 测序;BC,乳腺癌;MDD,重度抑郁症;UMAP,均匀流形近似和投影;OXPHOS,氧化磷酸化;GSVA,基因组变异分析;LumSec,管腔分泌细胞;DEGs,差异表达基因。研究人员收集了10名BC患者的配对原发肿瘤组织(n = 10)、邻近正常组织(n = 7)和外周血样本(n = 10),其中5名患者有MDD病史(补充表S1)。所有 BC 患者的肿瘤均为雌激素受体(ER)阳性,并被进一步预测为腔隙 A 亚型(9 例)和 B 亚型(1 例)(补充表 S2)。关于患者招募、样本处理和单细胞数据分析的更多详情,请参阅补充方法。我们总共获得了 224,557 个单细胞,并根据系谱标记和拷贝数变异[5]将它们进一步注释为主要的细胞亚群(图 1B,补充图 S1A-B)。我们还获得了原发肿瘤组织中的非整倍体细胞(补充表 S3),以确定它们在有 MDD 病史(BC-MDD)或无 MDD 病史(BC-Ctrl)的 BC 患者中的表型差异。未整合的非整倍体细胞的均匀簇逼近和投影(UMAP)显示了不同患者的内在差异(补充图 S1C)。下游功能谱分析发现,BC-MDD 组和 BC-Ctrl 组的免疫反应通路截然不同,氧化磷酸化(OXPHOS)通路在 BC-Ctrl 肿瘤中富集(补充图 S1D-E)。细胞基因组变异分析(GSVA)[6] 证实了 BC-MDD 组和 BC-Ctrl 组之间不同的代谢表型(图 1C)。此外,利用预定义的 BC 肿瘤细胞基因模块(GM)特征[7],我们观察到 BC-MDD 中 GM4 和 GM6 的特异性抑制(补充图 S1F-G)。GM6包括各种抗原递呈基因,与在BC-MDD肿瘤细胞中观察到的主要组织相容性复合体I类(MHC-I)类基因的下调相一致(补充图S1H)。在对 10 名患者的原发性乳腺肿瘤和邻近正常组织中的 12,371 个正常上皮细胞重新聚类后,我们发现了 9 个细胞群,包括管腔激素反应性细胞(LumHR)、管腔分泌性细胞(LumSec)和肌上皮细胞[8](补充图 S2A-B)。分布分析表明,LumSec-2 和 -3 簇可能在 BC-Ctrl 患者的原发肿瘤组织中富集(补充图 S2C-E)。随后,我们研究了这些 BC-Ctrl 富集的肿瘤上皮细胞在肿瘤发生和预后中的潜在作用。我们利用癌症基因组图谱--乳腺浸润性癌(TCGA-BRCA)数据集[9],对这些细胞集群的前 50 个差异表达基因(DEGs)进行了评估。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
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.
×
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
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信