留言板

尊敬的读者、作者、审稿人, 关于本刊的投稿、审稿、编辑和出版的任何问题, 您可以本页添加留言。我们将尽快给您答复。谢谢您的支持!

姓名
邮箱
手机号码
标题
留言内容
验证码

基于加权基因共表达网络鉴定肾移植术后排斥反应中巨噬细胞M1亚型相关基因

董博清, 李杨, 石玉婷, 等. 基于加权基因共表达网络鉴定肾移植术后排斥反应中巨噬细胞M1亚型相关基因[J]. 器官移植, 2023, 14(1): 83-92. doi: 10.3969/j.issn.1674-7445.2023.01.011
引用本文: 董博清, 李杨, 石玉婷, 等. 基于加权基因共表达网络鉴定肾移植术后排斥反应中巨噬细胞M1亚型相关基因[J]. 器官移植, 2023, 14(1): 83-92. doi: 10.3969/j.issn.1674-7445.2023.01.011
Dong Boqing, Li Yang, Shi Yuting, et al. Identification of M1 macrophage-related genes in rejection after kidney transplantation based on weighted gene co-expression network analysis[J]. ORGAN TRANSPLANTATION, 2023, 14(1): 83-92. doi: 10.3969/j.issn.1674-7445.2023.01.011
Citation: Dong Boqing, Li Yang, Shi Yuting, et al. Identification of M1 macrophage-related genes in rejection after kidney transplantation based on weighted gene co-expression network analysis[J]. ORGAN TRANSPLANTATION, 2023, 14(1): 83-92. doi: 10.3969/j.issn.1674-7445.2023.01.011

基于加权基因共表达网络鉴定肾移植术后排斥反应中巨噬细胞M1亚型相关基因

doi: 10.3969/j.issn.1674-7445.2023.01.011
基金项目: 

国家自然科学基金 81970668

陕西省重点研发计划 2022SF-148

西安交通大学基本科研业务基金 xzy012021060

详细信息
    作者简介:
    通讯作者:

    薛武军,博士,主任医师,研究方向为肾移植,Email: xwujun126@xjtu.edu.cn

  • 中图分类号: R617, R392.12

Identification of M1 macrophage-related genes in rejection after kidney transplantation based on weighted gene co-expression network analysis

More Information
  • 摘要:   目的  鉴定肾移植术后排斥反应中巨噬细胞M1亚型表达的相关基因并构建风险模型预测移植肾存活。  方法  在基因表达综合(GEO)数据库下载肾移植术后的GSE36059及GSE21374数据集。GSE36059包括发生排斥反应和稳定移植物的样本,使用该数据集进行加权基因共表达网络分析(WGCNA)和差异分析筛选差异表达的巨噬细胞M1亚型相关差异表达基因(M1-DEG)。随后将GSE21374数据集(包含了移植物丢失的随访数据)按照7∶3拆分为训练集以及验证集,在训练集中使用最小绝对收缩和选择算法(LASSO)筛选变量构建多因素Cox模型,并评估模型预测移植物存活的能力。使用CIBERSORT分析高、低风险组浸润的免疫细胞的差异,并分析两组间人类白细胞抗原(HLA)相关基因的分布,基因集富集分析(GSEA)用于进一步明确高风险组中富集的生物学过程以及通路。最后使用数据库预测与预后基因互作的微小核糖核酸(miRNA)。  结果  在GSE36059数据集中,筛选得到14个M1-DEG。在GSE21374数据集中,使用LASSO-Cox回归筛选出Toll样受体8(TLR8)、Fc γ受体1B(FCGR1B)、BCL2相关蛋白A1(BCL2A1)、组织蛋白酶S(CTSS)、鸟苷酸结合蛋白2(GBP2)及半胱氨酸天冬氨酸蛋白酶招募域家族成员16(CARD16),基于这6个M1-DEG构建多因素Cox模型。风险模型在训练集中预测1年及3年移植物存活的受试者工作特征曲线下面积(AUC)分别为0.918和0.877,在验证集中预测1年及3年移植物存活的AUC分别为0.765及0.736。免疫浸润分析表明,高风险组静息及活化的CD4+记忆T细胞、γδT细胞、巨噬细胞M1亚型浸润增多(均为P < 0.05)。高风险组HLA Ⅰ类基因表达上调。GSEA分析表明,高风险组免疫反应及移植物排斥反应富集。CTSS与8个miRNA相互作用、BCL2A1和GBP2与3个miRNA相互作用、FCGR1B与1个miRNA相互作用。  结论  本研究基于6个M1-DEG构建的预后风险模型对于预测移植肾存活具有良好的表现,可为早期对高风险受者干预提供依据。

     

  • 图  1  加权基因共表达网络分析

    注:A图为样本聚类图; B图为根据R2和平均连通性确定软阈值; C图为基因特征树图; D图为基因聚类树状图; E、F图为基因模块与临床特征相关性热图; G图为模块基因相关性散点图。

    Figure  1.  Analysis of weighted gene co-expression networks

    图  2  差异表达的火山图和韦恩图

    注:A图为火山图; B图为韦恩图。

    Figure  2.  Differentially expressed volcano plots and Wayne diagrams

    图  3  基于M1-DEG构建风险预后模型

    注:A图为在使用LASSO-Cox筛选构建的模型; B图为根据MMSE选择LASSO的惩罚系数(λ); C图为预测肾移植术后移植物存活的列线图模型。

    Figure  3.  Risk prognosis model based on M1-DEG

    图  4  6个基因模型在训练集的表现

    注:A图为根据风险评分将受者分为高风险和低风险组; B图为受者风险评分与移植物存活间的关系; C图为预测移植物存活的受试者工作特征曲线; D图为移植物存活的生存分析。

    Figure  4.  Six gene models performed in the training set

    图  5  6个基因模型在验证集的表现

    注:A图为根据风险评分将受者分为高风险和低风险组; B图为受者的风险评分和移植物存活间的关系; C图为预测移植物存活的受试者工作特征曲线; D图为移植物存活的生存分析。

    Figure  5.  Six gene models performed in the validation set

    图  6  高风险组和低风险组的免疫浸润及相关基因分布分析

    注:A图为免疫浸润分析; B图为预后基因和HLA基因分布。

    Figure  6.  Immune infiltration and related gene distribution analysis between high-risk group and low-risk group

    图  7  基因集富集分析

    注:A图为高风险组中富集的生物学过程; B图为高风险组中富集的KEGG通路。

    Figure  7.  Analysis of gene set enrichment

    图  8  miRNA和预后基因互作网络

    Figure  8.  miRNA and prognostic gene interaction network

  • [1] FLETCHER BR, DAMERY S, AIYEGBUSI OL, et al. Symptom burden and health-related quality of life in chronic kidney disease: a global systematic review and meta-analysis[J]. PLoS Med, 2022, 19(4): e1003954. DOI: 10.1371/journal.pmed.1003954.
    [2] THONGPRAYOON C, HANSRIVIJIT P, LEEAPHORN N, et al. Recent advances and clinical outcomes of kidney transplantation[J]. J Clin Med, 2020, 9(4): 1193. DOI: 10.3390/jcm9041193.
    [3] LAI X, ZHENG X, MATHEW JM, et al. Tackling chronic kidney transplant rejection: challenges and promises[J]. Front Immunol, 2021, 12: 661643. DOI: 10.3389/fimmu.2021.661643.
    [4] ORDIKHANI F, POTHULA V, SANCHEZ-TARJUELO R, et al. Macrophages in organ transplantation[J]. Front Immunol, 2020, 11: 582939. DOI: 10.3389/fimmu.2020.582939.
    [5] WANG J, LUO P, ZHAO J, et al. Profiling the resident and infiltrating monocyte/macrophages during rejection following kidney transplantation[J]. J Immunol Res, 2020: 5746832. DOI: 10.1155/2020/5746832.
    [6] LOCATI M, CURTALE G, MANTOVANI A. Diversity, mechanisms, and significance of macrophage plasticity[J]. Annu Rev Pathol, 2020, 15: 123-147. DOI: 10.1146/annurev-pathmechdis-012418-012718.
    [7] DEVRAJ VM, KALIDINDI K, GUDITI S, et al. Macrophage polarization in kidney transplant patients[J]. Transpl Immunol, 2022, 75: 101717. DOI: 10.1016/j.trim.2022.101717.
    [8] DOU M, DING C, ZHENG B, et al. Immune-related genes for predicting future kidney graft loss: a study based on GEO database[J]. Front Immunol, 2022, 13: 859693. DOI: 10.3389/fimmu.2022.859693.
    [9] XU J, HU J, XU H, et al. Long non-coding RNA expression profiling in biopsy to identify renal allograft at risk of chronic damage and future graft loss[J]. Appl Biochem Biotechnol, 2020, 190(2): 660-673. DOI: 10.1007/s12010-019-03082-2.
    [10] LIU S, WANG Z, ZHU R, et al. Three differential expression analysis methods for RNA sequencing: limma, EdgeR, DESeq2[J]. J Vis Exp, 2021(175). DOI: 10.3791/62528.
    [11] TIBSHIRANI R. The LASSO method for variable selection in the Cox model[J]. Stat Med, 1997, 16(4): 385-395.
    [12] RU Y, KECHRIS KJ, TABAKOFF B, et al. The multiMiR R package and database: integration of microRNA-target interactions along with their disease and drug associations[J]. Nucleic Acids Res, 2014, 42(17): e133. DOI: 10.1093/nar/gku631.
    [13] ZHOU H, LU H, SUN L, et al. Diagnostic biomarkers and immune infiltration in patients with T cell-mediated rejection after kidney transplantation[J]. Front Immunol, 2022, 12: 774321. DOI: 10.3389/fimmu.2021.774321.
    [14] SHEN Q, WANG Y, CHEN J, et al. Single-cell RNA sequencing reveals the immunological profiles of renal allograft rejection in mice[J]. Front Immunol, 2021, 12: 693608. DOI: 10.3389/fimmu.2021.693608.
    [15] LU J, ZHANG Y, SUN J, et al. The immune cell landscape in renal allografts[J]. Cell Transplant, 2021, 30: 963689721995458. DOI: 10.1177/0963689721995458.
    [16] HOLÁN V, KRULOVÁ M, ZAJÍCOVÁ A, et al. Nitric oxide as a regulatory and effector molecule in the immune system[J]. Mol Immunol, 2002, 38(12/13): 989-995. DOI: 10.1016/s0161-5890(02)00027-5.
    [17] BRAZA F, BROUARD S, CHADBAN S, et al. Role of TLRs and DAMPs in allograft inflammation and transplant outcomes[J]. Nat Rev Nephrol, 2016, 12(5): 281-290. DOI: 10.1038/nrneph.2016.41.
    [18] DESSING MC, BEMELMAN FJ, CLAESSEN N, et al. Intragraft Toll-like receptor profiling in acute renal allograft rejection[J]. Nephrol Dial Transplant, 2010, 25(12): 4087-4092. DOI: 10.1093/ndt/gfq589.
    [19] WANG N, LIANG H, ZEN K. Molecular mechanisms that influence the macrophage M1-M2 polarization balance[J]. Front Immunol, 2014, 5: 614. DOI: 10.3389/fimmu.2014.00614.
    [20] VAN LOON E, GAZUT S, YAZDANI S, et al. Development and validation of a peripheral blood mRNA assay for the assessment of antibody-mediated kidney allograft rejection: a multicentre, prospective study[J]. EBioMedicine, 2019, 46: 463-472. DOI: 10.1016/j.ebiom.2019.07.028.
    [21] TAN PS, GAVIN AL, BARNES N, et al. Unique monoclonal antibodies define expression of Fc gamma RI on macrophages and mast cell lines and demonstrate heterogeneity among subcutaneous and other dendritic cells[J]. J Immunol, 2003, 170(5): 2549-2556. DOI: 10.4049/jimmunol.170.5.2549.
    [22] MANNAM VK, LEWIS RE, CRUSE JM. The fate of renal allografts hinges on responses of the microvascular endothelium[J]. Exp Mol Pathol, 2013, 94(2): 398-411. DOI: 10.1016/j.yexmp.2012.06.002.
    [23] SHAATH H, VISHNUBALAJI R, ELKORD E, et al. Single-cell transcriptome analysis highlights a role for neutrophils and inflammatory macrophages in the pathogenesis of severe COVID-19[J]. Cells, 2020, 9(11): 2374. DOI: 10.3390/cells9112374.
    [24] KUBO K, KAWATO Y, NAKAMURA K, et al. Effective suppression of donor specific antibody production by cathepsin S inhibitors in a mouse transplantation model[J]. Eur J Pharmacol, 2018, 838: 145-152. DOI: 10.1016/j.ejphar.2018.09.007.
    [25] FUJⅡ H, IVISON SM, SHIMIZU H, et al. Inhibition of cathepsin S reduces allogeneic T cell priming but not graft-versus-host disease against minor histocompatibility antigens[J]. Biol Blood Marrow Transplant, 2012, 18(4): 546-556. DOI: 10.1016/j.bbmt.2011.11.027.
    [26] HUGHES CS, COLHOUN LM, BAINS BK, et al. Extracellular cathepsin S and intracellular caspase 1 activation are surrogate biomarkers of particulate-induced lysosomal disruption in macrophages[J]. Part Fibre Toxicol, 2016, 13: 19. DOI: 10.1186/s12989-016-0129-5.
    [27] REEVE J, EINECKE G, MENGEL M, et al. Diagnosing rejection in renal transplants: a comparison of molecular- and histopathology-based approaches[J]. Am J Transplant, 2009, 9(8): 1802-1810. DOI: 10.1111/j.1600-6143.2009.02694.x.
    [28] KOBAYASHI S, NAGANO H, MARUBASHI S, et al. Guanylate-binding protein 2 mRNA in peripheral blood leukocytes of liver transplant recipients as a marker for acute cellular rejection[J]. Transpl Int, 2010, 23(4): 390-396. DOI: 10.1111/j.1432-2277.2009.00991.x.
    [29] LARA-REYNA S, CASELEY EA, TOPPING J, et al. Inflammasome activation: from molecular mechanisms to autoinflammation[J]. Clin Transl Immunology, 2022, 11(7): e1404. DOI: 10.1002/cti2.1404.
    [30] KARASAWA T, KAWASHIMA A, USUI F, et al. Oligomerized CARD16 promotes Caspase-1 assembly and IL-1β processing[J]. FEBS Open Bio, 2015, 5: 348-356. DOI: 10.1016/j.fob.2015.04.011.
    [31] NANKIVELL BJ, ALEXANDER SI. Rejection of the kidney allograft[J]. N Engl J Med, 2010, 363(15): 1451-1462. DOI: 10.1056/NEJMra0902927.
    [32] KAMINSKI H, MARSÈRES G, COSENTINO A, et al. Understanding human γδ T cell biology toward a better management of cytomegalovirus infection[J]. Immunol Rev, 2020, 298(1): 264-288. DOI: 10.1111/imr.12922.
    [33] KIRK AD, IBRAHIM S, DAWSON DV, et al. Characterization of T cells expressing the gamma/delta antigen receptor in human renal allografts[J]. Hum Immunol, 1993, 36(1): 11-19. DOI: 10.1016/0198-8859(93)90003-j.
    [34] KAMINSKI H, GARRIGUE I, COUZI L, et al. Surveillance of γδ T cells predicts cytomegalovirus infection resolution in kidney transplants[J]. J Am Soc Nephrol, 2016, 27(2): 637-645. DOI: 10.1681/ASN.2014100985.
    [35] BACHELET T, COUZI L, PITARD V, et al. Cytomegalovirus-responsive γδ T cells: novel effector cells in antibody-mediated kidney allograft microcirculation lesions[J]. J Am Soc Nephrol, 2014, 25(11): 2471-2482. DOI: 10.1681/ASN.2013101052.
    [36] JENKINS SJ, ALLEN JE. The expanding world of tissue-resident macrophages[J]. Eur J Immunol, 2021, 51(8): 1882-1896. DOI: 10.1002/eji.202048881.
  • 加载中
图(9)
计量
  • 文章访问数:  301
  • HTML全文浏览量:  161
  • PDF下载量:  84
  • 被引次数: 0
出版历程
  • 收稿日期:  2022-09-14
  • 网络出版日期:  2023-01-17
  • 刊出日期:  2023-01-15

目录

    /

    返回文章
    返回