1、下载临床相关的表格以及原始数据(GDCdata-TCGA-LAML-CLINICDATA里面),这里我们以TCGA-LAML(急性髓细胞白血病)数据为例,下载其临床数据信息。

#####一、下载临床数据#####setwd("H:\\RDATA\\test/")# 临床数据# XML数据-全面信息library(TCGAbiolinks)query <- GDCquery(project = "TCGA-LAML", #癌症TCGA存储文件,可以在官网查询data.category = "Clinical", #需要下载的文件类型,可以在官网查看可以下在什么信息data.type = "Clinical Supplement", #需要下载的文件类型data.format = "BCR XML") #文件格式GDCdownload(query = query, #下载文件,下载过的话,后面直接将directory改到这个储存位置就可以了method = "api",directory = "GDCdata-TCGA-LAML-CLINICDATA",# 默认文件夹名称GDCdata,后面也要相应修改files.per.chunk = 3) # 网速慢、文件大可以设置files.per.chunk分开下载# 循环输出临床数据clinical.info<-c("patient","radiation")#可以提取的信息#clinical.info<-c("patient","drug","follow_up","radiation","stage_event","new_tumor_event","admin")# 循环写出for(i in clinical.info){clidata <- GDCprepare_clinic(query, clinical.info = i,directory = 'GDCdata-TCGA-LAML-CLINICDATA')#directory:临床文件储存的位置write.csv(clidata, file = paste0('TCGA-LAML_clinical_',i,'.csv'), row.names =F)}# 在excel中处理数据,也可以在R处理

Tips:

所有TCGA数据可以在官网查看,关于TCGA使用方法下面有个视频可以参考看看:

GDC (cancer.gov)https://portal.gdc.cancer.gov/TCGA-01-TCGA数据库介绍_哔哩哔哩_bilibilihttps://www.bilibili.com/video/BV1824y1H75N/?spm_id_from=333.999.0.0

2、下载矩阵数据(Count矩阵),这里我们下载转录组数据为例

#####二、下载矩阵数据###### counts下载# 以RNA-seq表达矩阵为例query <- GDCquery(project = "TCGA-LAML", data.category = "Transcriptome Profiling",data.type = 'Gene Expression Quantification',experimental.strategy = "RNA-Seq",workflow.type = "STAR - Counts")GDCdownload(query,directory = "GDCdata-TCGA-LAML-COUNTS") #保存的位置testdata <- GDCprepare(query = query, directory = 'GDCdata-TCGA-LAML-COUNTS')# SummarizedExperiment对象library(SummarizedExperiment)names(assays(testdata)) #可以进行提取的矩阵rowdata <- rowData(testdata)# 查看rowdata内容names(rowdata)table(rowdata$gene_type) # 统计基因类型,下面可以提取的选择就在这里head(rowdata$gene_name,10) # gene_name就是symbollength(rowdata$gene_name) #有多少genetest_miRNA <- testdata[rowdata$gene_type == "miRNA"]# 提取miRNAtest_mRNA <- testdata[rowdata$gene_type == "protein_coding",] # 提取mRNA

到这里位置我们有两个文件夹,GDCdata-TCGA-LAML-CLINICDATA和GDCdata-TCGA-LAML-COUNTS,分别存放着临床数据和数值矩阵两个原始文件,如果之后要重新分析,可以不用下载,直接将directory放到这两个文件对应的地方。

三、提取矩阵,这里我们使用counts矩阵为例

#####三、提取表达矩阵###### 提取表达矩阵# mRNA的counts矩阵test.mRNA.counts <- assay(test_mRNA,"unstranded") # counts矩阵用于后续差异分析# mRNA的tpm矩阵test.mRNA.tpm <- assay(test_mRNA,"tpm_unstrand")# mRNA的fpkm矩阵test.mRNA.fpkm <- assay(test_mRNA,"fpkm_unstrand")# 添加gene_symbol,基因名称mRNA.symbol <- rowData(test_mRNA)$gene_name# 合并test.mrna.frame <- cbind(as.data.frame(mRNA.symbol), as.data.frame(test.mRNA.counts))dim(test.mrna.frame) #基因/样品数量

四、初步处理并保存该count矩阵

#####四、初步处理###### 去重数据qc = as.matrix(test.mrna.frame)rownames(qc)=qc[,1] # gene_symbolexp=qc[,2:ncol(qc)] # matrixdimnames=list(rownames(exp),colnames(exp))data=matrix(as.numeric(as.matrix(exp)),nrow=nrow(exp),dimnames=dimnames)# 对重复的基因取平均值data = limma::avereps(data) # 去重,出现多行取平均值# 过滤表达量低的基因,可根据基因数目进行调整CHOL.test=data[rowMeans(data)>5,]output <- rbind(colnames(CHOL.test),CHOL.test)dim(output)write.table(output, file="LAML_mRNA矩阵去重.txt", sep="\t", quote=F, col.names=F)

以及根据“TCGA-LAML_clinical_patient”准备好了meta分组文件(怎么准备?用excel吧=-=):

五、RNAseq(使用的DESEQ2)

#####五、RNAseq##########5.1 读入文件并处理#####rt=read.table("LAML_mRNA矩阵去重.txt",sep="\t",header=T,check.names=F, row.names = 1) dim(rt)rt <- round(rt,0) #counts需要取整数rt <- rt[, substr(names(rt), 14, 16) == "03A"] #仅保留03A的数据#TCGA命名规则:https://zhuanlan.zhihu.com/p/564801425,这个一定要看一遍patient_ID <- read.csv("meta.csv") #读入meta文件(根据临床文件自己准备好,这里准备的是性别)patient_ID <- patient_ID[!duplicated(patient_ID$id), ]#去重复行名(有的病人有多个样本)patientIDs <- patient_ID[, 1] #提取ID向量rt <- rbind(colnames(rt), rt) #加一行格式为TCGA-XX-XXXX的行names(rt) <- substr(names(rt), 1, 12) #加一行格式为TCGA-XX-XXXX的行selected_rt <- rt[, colnames(rt) %in% patientIDs] #取矩阵和meta数据中都有的病例(因为counts矩阵有时候会有缺失的,或者说没有对应病人的)colnames(selected_rt) <- as.character(unlist(selected_rt[1,]))#列名改格式为TCGA-XX-XXXXselected_rt <- selected_rt[-1,]#列名改格式为TCGA-XX-XXXXnames(selected_rt) <- substr(names(selected_rt), 1, 12)#列名改格式为TCGA-XX-XXXXhas_duplicates <- any(duplicated(names(selected_rt)))# 看有无重复列名(一个病例多个样本)selected_rt <- selected_rt[, !duplicated(names(selected_rt))]#去重复列名A <- c(colnames(selected_rt)) #处理过的矩阵里面所有病例B <- c(patient_ID$id) #meta里面所有病例different_values <- setdiff(B, A) #有临床数据但是没有相对应的矩阵数据的病例patient_ID <- patient_ID[!patient_ID$id %in% different_values, ] #删掉这些没有对应数据的病例write.csv(selected_rt,file = "mRNA.csv")write.csv(patient_ID,file = "mymeta.csv")#####5.2 进行DESeQ2前的预处理#####mycounts <- read.csv("mRNA.csv",row.names = 1, check.names = F) #数据矩阵mymeta <- read.csv("mymeta.csv", stringsAsFactors = T, row.names = 1) #分组矩阵rownames(mymeta) <- mymeta$id #行名改为ID,但是忘了有啥用了colnames(mycounts) == mymeta$id #检查行名列名一致性,通常全为FALSE,数据少的时候在EXCEL里手动调一下#处理行列一致性mycounts <- as.data.frame(t(mycounts)) #数值矩阵转置mycounts$V1 <- NA mycounts$V1 <- rownames(mycounts) #行名(sample名放到最后一列,相比于直接处理行名bug更少)mycounts_1 <- mycounts[match(mymeta$id, mycounts$V1), ] #将数值矩阵的行顺序按照分组矩阵的顺序更改mycounts_1 <- mycounts_1[, -ncol(mycounts_1)] #删掉最后一列(行名列)mycounts_1 <- as.data.frame(t(mycounts_1)) #转置回来colnames(mycounts_1) == mymeta$id #全为TRUE可进行下面操作,这里也可以存一下矩阵,之后再做可以不用处理直接进行DESEQ2write.csv(mycounts_1,file = "mRNA.csv")#会直接覆盖之前的write.csv(mymeta, file = "mymeta.csv")#这个没覆盖,可以覆盖一下免得弄错#####5.3 DESEQ2一条龙#####library(DESeq2)mycounts_1 <- read.csv("mRNA.csv",row.names = 1, check.names = F) #数据矩阵mymeta <- read.csv("mymeta.csv", stringsAsFactors = T, row.names = 1) #分组矩阵dds <- DESeqDataSetFromMatrix(countData = mycounts_1, #数值矩阵colData = mymeta, #分组矩阵design = ~gender) #分组依据的那个列名ddsdds  5,] #过滤一遍,基因表达之和得大于5dds$gender<-relevel(dds$gender, ref="MALE")#谁作为参照组dds$gendertable(dds$gender) #样品计数,这里要记下来一下,后面会用dds <- DESeq(dds) #进行自动差异分析res <- results(dds) #提取结果文件head(res)class(res)res_1 %mutate(group = case_when(log2FoldChange >= 0 & pvalue <=0.05 ~"UP",log2FoldChange <= 0 & pvalue  res_2 #区分上下调基因,其中条件可自选,条件在列名里看head(res_2)table(res_2$group) #上下调基因数量res_2$symbol <- row.names(res_2) #加一列存下symbol后面会用write.csv(res_2, file = "diff_expr_results.csv") #保存差异基因文件,可以自己根据这个分析,也可以跟我这走走流程

TCGA命名规则:一文讲清TCGA数据库中样本编码信息 – 知乎一文讲清TCGA数据库中样本编码信息 TCGA的样本命名 Sample:其中编号01~09表示肿瘤,10~19表示正常对照。(区分正常和癌症样本的凭证)CodeDefinition 01Primary Soild Tumor(原发性实体肿瘤) 02Recurrent Soild …https://zhuanlan.zhihu.com/p/564801425

六、可视化

#####六、富集分析###########6.1 GO分析######采取方法up和down gene分别富集library(org.Hs.eg.db)library(clusterProfiler)library(stringr)library(ggplot2)upgene <- res_2[res_2$group == "UP", ]upggoBP <- enrichGO(upgene$symbol, org.Hs.eg.db, keyType = "SYMBOL",ont = "BP",pvalueCutoff = 0.05, pAdjustMethod = "BH", qvalueCutoff = 0.2,minGSSize = 10, maxGSSize = 500, readable = FALSE, pool = FALSE)upGOBP %mutate(GeneRatio_a = as.numeric(str_extract(GeneRatio, "\\d+")), BgRatio_a = as.numeric(str_extract(BgRatio, "\\d+")), RichFactor = GeneRatio_a / BgRatio_a)write.csv(upGOBP, 'UPGOBP.csv')downgene <- res_2[res_2$group == "DOWN", ]downggoBP <- enrichGO(downgene$symbol, org.Hs.eg.db, keyType = "SYMBOL",ont = "BP",pvalueCutoff = 0.05, pAdjustMethod = "BH", qvalueCutoff = 0.2,minGSSize = 10, maxGSSize = 500, readable = FALSE, pool = FALSE)downGOBP %mutate(GeneRatio_a = as.numeric(str_extract(GeneRatio, "\\d+")), BgRatio_a = as.numeric(str_extract(BgRatio, "\\d+")), RichFactor = GeneRatio_a / BgRatio_a)write.csv(downGOBP, 'downGOBP.csv')# GO分析的双侧柱状图upGOBP <- read.csv("UPGOBP.csv",row.names = 1)downGOBP <- read.csv("downGOBP.csv", row.names = 1)upGOBP$Group <- NAupGOBP$Group = 1downGOBP$Group <- NAdownGOBP$Group = -1upGOBP$Description<-sub("(.)", "\\U\\1",upGOBP$Description,perl=TRUE)downGOBP$Description<-sub("(.)", "\\U\\1",downGOBP$Description,perl=TRUE)upGOBP.sorted <- upGOBP[order(upGOBP$pvalue), ]# 根据pvalue从小到大排序upGOBP <- upGOBP.sorted[1:20, ]# 选择前20行downGOBP.sorted <- downGOBP[order(downGOBP$pvalue), ]# 根据pvalue从小到大排序downGOBP <- downGOBP.sorted[1:20, ]# 选择前20行dat=rbind(upGOBP,downGOBP)colnames(dat)dat$group 0)]='up'dat$group[which(dat$Group <0)]='down'dat$RichFactor = dat$RichFactor*dat$Group dat <- dat[dat$pvalue<0.05,]gk_plot 0),],aes(x=Description, y=-0.01, label=Description),hjust=1, size=5)+geom_text(data = dat[which(dat$RichFactor0),],aes(label=sprintf("%.2e", pvalue)),hjust=-0.1, size=5, color='red')+geom_text(data = dat[which(dat$RichFactor<0),],aes(label=sprintf("%.2e", pvalue)),hjust=1.1, size=5, color="red")+scale_fill_manual(values = c("#fb6a4a","#3182bd"), breaks = c("up", "down"))+scale_x_discrete(expand = expansion(mult = c(0,0)))+ylim(-1, 1)+labs(x='', y='RichFactor')+theme(legend.text = element_text(size = 12),axis.title.x = element_text(size = 14))+ggtitle("GO BP of The Top 20 Most Significant P-values")+theme(plot.title = element_text(hjust = 0.5, size = 20))gk_plot#####6.2 KEGG分析######唯一注意的一点就是把它换为ENTREZID,再换回去library(clusterProfiler)library(stringr)upgene <- res_2[res_2$group == "UP", ]genes <- bitr(upgene$symbol, fromType = "SYMBOL", toType = "ENTREZID", OrgDb = "org.Hs.eg.db")genes <- genes[,2]ekegg <- enrichKEGG(genes, organism = 'hsa',pvalueCutoff = 0.05,pAdjustMethod = 'BH',qvalueCutoff = 0.2,minGSSize=10,maxGSSize=500,use_internal_data=F)ekegg <- setReadable(ekegg, 'org.Hs.eg.db','ENTREZID')upKEGG %mutate(GeneRatio_a = as.numeric(str_extract(GeneRatio, "\\d+")), BgRatio_a = as.numeric(str_extract(BgRatio, "\\d+")), RichFactor = GeneRatio_a / BgRatio_a)write.csv(upKEGG, 'UPKEGG.csv')downgene <- res_2[res_2$group == "DOWN", ]genes <- downgene$symbolgenes <- bitr(genes, fromType = "SYMBOL", toType = "ENTREZID", OrgDb = "org.Hs.eg.db")genes <- genes[,2]ekegg <- enrichKEGG(genes, organism = 'hsa',pvalueCutoff = 0.05,pAdjustMethod = 'BH',qvalueCutoff = 0.2,minGSSize=10,maxGSSize=500,use_internal_data=F)ekegg <- setReadable(ekegg, 'org.Hs.eg.db','ENTREZID')downKEGG %mutate(GeneRatio_a = as.numeric(str_extract(GeneRatio, "\\d+")), BgRatio_a = as.numeric(str_extract(BgRatio, "\\d+")), RichFactor = GeneRatio_a / BgRatio_a)write.csv(downKEGG, 'downKEGG.csv')#画图偷懒直接移花接木#upGOBP <- read.csv("UPKEGG.csv",row.names = 1)#downGOBP <- read.csv("downKEGG.csv", row.names = 1)#####七、单个基因分析######基因表达箱线图pTSPAN6d <- plotCounts(dds, gene="TSPAN6", intgroup="gender", returnData=TRUE) #提取数据library(ggsignif)pTSPAN6 <- ggplot(pTSPAN6d, aes(x=gender, y=count)) + geom_boxplot(aes(fill=gender)) +scale_y_log10() +theme_bw()+labs(x = " ", y = "Relative Counts", fill = "Gender") +ggtitle("TSPAN6")+#theme(legend.position = "none")+#加上就是不要图例geom_signif(comparisons = list(c("MALE", "FEMALE")),annotations = c("***"),#这里查看差异基因表手动改相关系数星星y_position = c(3, 3), #这里调p值横线线的高低tip_length = c(0, 0)) + #这里调小撇撇的长短annotate("text", x = 1, y = 0, label = "(n=13)", #数量看前面那个记住的东西 vjust = 4, hjust = 0.5, size = 4, fontface = "bold") +annotate("text", x = 2, y = 0, label = "(n=14)", #数量看前面那个记住的东西 vjust = 4, hjust = 0.5, size = 4, fontface = "bold") +coord_cartesian(clip = "off") +theme(panel.grid.major = element_blank(), panel.grid.minor = element_blank(),axis.title.x = element_text(size = 18, face = "bold"),axis.title.y = element_text(size = 18, face = "bold"),axis.text = element_text(size = 18, face = "bold"),plot.title = element_text(size = 18, face = "bold.italic"),legend.title = element_text(size = 10, face = "bold"))pTSPAN6#其余同理还可以拼图library(patchwork)(pTSPAN6 + pa) / (pb + pc) +plot_layout(guides = 'collect')#####八、火山图#####library(ggrepel)library(ggplot2)library(pheatmap)##筛选条件设置log2FC_cutoff = 0.5pvalue_cutoff = 0.05##选取差异分析结果need_DEG <- res_1[,c(2,5)] #选取log2FoldChange, pvalue信息colnames(need_DEG) <- c('log2FoldChange','pvalue') need_DEG$significance<- as.factor(ifelse(need_DEG$pvalue  log2FC_cutoff, ifelse(need_DEG$log2FoldChange > log2FC_cutoff ,'UP','DOWN'),'NOT_CHANGE'))title = ',round(log2FC_cutoff,3))ggplot(data=need_DEG,aes(x=log2FoldChange, y=-log10(pvalue),color=significance)) +#点和背景geom_point(alpha=0.4, size=1) +theme_classic()+ #无网格线#坐标轴xlab("log2 ( FoldChange )") + ylab("-log10 ( pvalue)") +#标题文本ggtitle( title ) +#分区颜色scale_colour_manual(values = c('blue','grey','red'))+ #辅助线geom_vline(xintercept = c(-log2FC_cutoff,log2FC_cutoff),lty=4,col="grey",lwd=0.8) +geom_hline(yintercept = -log10(pvalue_cutoff),lty=4,col="grey",lwd=0.8) +#图例标题间距等设置theme(plot.title = element_text(hjust = 0.5), plot.margin=unit(c(2,2,2,2),'lines'), #上右下左legend.title = element_blank(), #不显示图例标题legend.position="right") +#图例位置#添加标签geom_text_repel(data = filter(res_2, abs(log2FoldChange) > 5 & -log10(pvalue) > 5),max.overlaps = getOption("ggrepel.max.overlaps", default = 20),aes(label = symbol, color = group),size = 2)#####九、差异表达热图######needDEG见上gene_up log2FC_cutoff & pvalue<pvalue_cutoff),])gene_down <- rownames(need_DEG[with(need_DEG,log2FoldChange< -log2FC_cutoff & pvalue<pvalue_cutoff),])df02 <- c(head(gene_up, 10), #取前10 padj上下调基因名head(gene_down, 10))#df02 <- c(gene_up,gene_down) #取所有#用counts来画图diff_expr <- mycounts_1[intersect(rownames(mycounts_1),df02),] ## 用标准化(标准化Counts值) 来画图#vsd <- vst(dds, blind = FALSE)#normalizeExp <- assay(vsd)## 差异基因的标准化Count#diff_expr <- normalizeExp[intersect(rownames(normalizeExp),df02),]#head(diff_expr)## 差异热图library(pheatmap)annotation_col <- data.frame(Group = factor(c(rep("MALE",78), rep("FEMALE",59))))#前面记住的rownames(annotation_col) <- colnames(diff_expr)p <- pheatmap(diff_expr,annotation_col = annotation_col,color = colorRampPalette(c("#8854d0", "#ffffff","#fa8231"))(50),cluster_cols = F,show_rownames = F,show_colnames = F,scale = "row", ## none, row, columnfontsize = 12,fontsize_row = 12,fontsize_col = 6,border = FALSE)p