Skip to main content

A comprehensive assessment of exome capture methods for RNA sequencing of formalin-fixed and paraffin-embedded samples

Abstract

RNA-Seq analysis of Formalin-Fixed and Paraffin-Embedded (FFPE) samples has emerged as a highly effective approach and is increasingly being used in clinical research and drug development. However, the processing and storage of FFPE samples are known to cause extensive degradation of RNAs, which limits the discovery of gene expression or gene fusion-based biomarkers using RNA sequencing, particularly methods reliant on Poly(A) enrichment. Recently, researchers have developed an exome targeted RNA-Seq methodology that utilizes biotinylated oligonucleotide probes to enrich RNA transcripts of interest, which could overcome these limitations. Nevertheless, the standardization of this experimental framework, including probe designs, sample multiplexing, sequencing read length, and bioinformatic pipelines, remains an essential requirement. In this study, we conducted a comprehensive comparison of three main commercially available exome capture kits and evaluated key experimental parameters, to provide the overview of the advantages and limitations associated with the selection of library preparation protocols and sequencing platforms. The results provide valuable insights into the best practices for obtaining high-quality data from FFPE samples.

Peer Review reports

Introduction

RNA sequencing has been developed as one of the most sensitive tools for gene expression analysis. Among the library preparation methods, the standard Poly(A) enrichment protocol provides a comprehensive and accurate view of polyadenylated RNAs. This method allows for simultaneous quantification of a multitude of RNA transcripts, enabling unbiased annotation of splicing variants, novel transcripts, and non-coding RNAs. It’s widely used in the investigation of human diseases, as well as the identification of novel drug targets and biomarkers. However, when working with human tissue specimens from biobanks, hospitals, and other clinical research facilities, the quality and yield of RNA can often be compromised due to several factors, including sampling techniques and preservation conditions, affecting the downstream bioinformatic analysis [1].

Fusion genes play a crucial role in tumorigenesis and are involved in approximately 20% of human cancer cases. The rapid and accurate identification of fusion genes holds significant promise for understanding cancer pathogenesis, enabling precise therapeutic interventions, and targeting drugs that can effectively inhibit these abnormal gene fusions [2]. There is great potential to fully assess the performance characteristics, including accuracy, reproducibility, and analytical sensitivity, of RNA-Seq for detecting the fusion events.

In recent years, RNA-Seq library preparation protocols specifically designed for FFPE samples have been developed [3], such as the RiboZero and TruSeq RNA Exome kit (Illumina). RNA capture is a novel approach used to profile RNA samples of low integrity [4]. This method employs capture probes that target known exons, allowing for the enrichment of coding RNAs. Biotech brands, including Illumina, Agilent, and Nanodigmbio have developed commercial products, each utilizing distinct mechanisms and technologies. These products offer standardized, reproducible, and user-friendly protocols, making them suitable for gene expression studies conducted in various research settings. However, there is a lack of studies that investigate the differences of their applications and provide insights into fundamental technical questions [5]. To address this gap, we designed the study to evaluate the performance of three exome capture-based library preparation kits on human reference RNA from the Sequencing Quality Control consortium [6] and commercially available FFPE sections (Fig. 1). To our knowledge, this research is the first to compare the exome capture-based kits with the well-established rRNA depletion protocols specifically on FFPE samples. Additionally, we investigated the gene expression measurement in comparison to the TaqMan standard data and assessed the detection of fusion genes engineered in the FFPE reference RNA. The results aim to provide the scientific community with a comprehensive assessment of exome capture methods for RNA sequencing of FFPE samples.

Fig. 1
figure 1

Experimental design of this study. 100 ng of heat-degraded Universal Human Reference RNA (UHRR) and one commercially available Formalin-Fixed Paraffin-Embedded (FFPE) sample were utilized as input materials. Three commercially available exome capture kits, namely Illumina (IL), Agilent (AG), and NadPrep (NP/NS), were employed. The rRNA depletion method (RD) was employed as the benchmark for bioinformatics analysis

Methods

Samples

To emulate the effects of formalin fixation-induced degradation, the human reference RNA (UHRR, #740000, Agilent Technologies) was incubated at 94 °C for 60 minutes to obtain fragmented RNA [7]. The peak observed in the Bioanalyzer (Agilent Technologies) trace of the fragmented sample was below 200 nt, and the peaks corresponding to 18S and 28S rRNA were absent.

The Onco Fusion FFPE RNA Reference Standard (#GW-OPSM001, GeneWell Biotechnology) containing multiple engineered clinically relevant fusion genes (Table 1) was procured, and total RNA was extracted using the RNeasy FFPE Kit (#73504, Qiagen) following the manufacturer’s instructions. The RNA Integrity Number (RIN) of the sample was 2.2, indicating significant degradation during the FFPE sections preparation [8].

Table 1 The list of fusion genes validated in the FFPE sample

Library preparation

rRNA depleted RNA was generated with the Hieff NGS MaxUp Human rRNA Depletion Kit (#12257ES96, Yeasen Biotechnology), then strand-specific libraries were prepared using the Hieff NGS Ultima Dual-mode RNA Library Prep Kit (#12310ES96, Yeasen Biotechnology). For Illumina libraries, the TruSeq RNA Library Prep and Enrichment kit (#20020189, Illumina) was employed. Agilent and NadPrep libraries were generated using the Hieff NGS Ultima Dual-mode RNA Library Prep Kit, followed by the exome capture procedure using either SureSelect Human All Exon V6 (#5190–8864, Agilent Technologies) or Exome Plus Panel v2.0 (#1001841, Nanodigmbio). Multiplexing is a term used to describe the experimental strategy of pooling individual libraries together before probe hybridization. This approach is employed in Agilent and NadPrep kits to reduce the cost of the capture experiment, in which 2-plex means to pool two DNA libraries into one single tube. Illumina kits need to increase the consumption of the probes and hybridization reagent when pooling the libraries, multiplexing was not used in this study. The NadPrep libraries were divided into strand-specific and non-strand-specific groups. All protocols were conducted in accordance with the manufacturer’s instructions, with the recommended starting material of 100 ng of input RNA. The quality and yield of the prepared libraries were assessed using an Agilent 2100 Bioanalyzer.

Sequencing

All the prepared DNBSEQ libraries were sequenced on the MGISEQ-2000 platform (MGI Tech) with PE150 cycles, except for the NadPrep libraries, which underwent an additional run with PE100 cycles. The Illumina libraries, on the other hand, were sequenced on the Novaseq 6000 platform (Illumina). Each library generated more than 30 million paired end reads.

Data processing and quality control

Ribosomal RNAs were removed by bowtie2 (v2.4.5) alignment with the parameter “--very-sensitive-local --no-unal -I 1 -X 1000”. Duplication rate was calculated using fastp (v0.23.2). SOAPnuke (v1.5.6) was employed to filter out low quality reads based on the following conditions: 1) Reads containing over 50% of the length of 5′ or 3′ sequencing adapters; 2) Reads consisting of more than 1% of ambiguous bases; 3) Reads encompassing more than 20% of low-quality bases with quality score below 15. These filtering steps were carried out prior to any further analysis.

Alignment and quantification

The high-quality reads obtained from each sample were aligned to the human reference genome (NCBI version GCF_000001405.39_GRCh38.p13) using HISAT2 (v2.2.1) with the parameter “--sensitive --no-discordant --no-mixed -I 1 -X 1000 --rna-strandness RF”. Sense rate and distribution across genome features were calculated by RSeQC (v4.0.0). Simultaneously, the reads were also mapped to the reference mRNA sequences using bowtie2 (v2.4.5) with the parameter: “--sensitive --dpad 0 --gbar 300 --mp 1,1 --np 1 --score-min L,0,-0.1 -I 1 -X 1000 --no-mixed --no-discordant -k 200”. RSEM (v1.3.1) was utilized to estimate the normalized gene abundances, represented as Fragments Per Kilobase Million (FPKM).

Differential expressed gene (DEG) identification

The DEseq2 package (v1.31.16) was employed for the identification of DEGs. Two replicated libraries were considered as a single group for each sample. DEGs were determined based on the criteria of having a fold change greater than or equal to 1 and an adjusted p-value less than or equal to 0.05 when comparing the two groups.

Fusion gene detection

Three distinct approaches were employed for the identification of fusion genes. The first one was EricScript (v0.5.5), which used the BWA aligner (v0.7.17) for mapping against the transcriptome reference. Samtools (v0.1.19) was utilized to handle the SAM/BAM files. BLAT (v35) was utilized for the recalibration of exon junction references. The second method was FusionCatcher (v1.33) which used two aligners (Bowtie v1.2.3 and STAR v2.7.2b) for read mapping and candidate fusion gene finding. FusionCatcher can filter out likely false positive candidate fusion genes of several conditions, including pseudogene, paralog gene or miRNA genes. The last method employed was STAR-Fusion (v1.12.0), which leveraged the output generated by the STAR aligner (v2.7.8a) to map both junction reads and spanning reads. These reads were mapped against a reference annotation set GRCh38_gencode_v37_CTAT_lib_Mar012021.plug-n-play from the Trinity Cancer Transcriptome Analysis Toolkit (CTAT) genome lib. All the methods were implemented using default parameter settings.

Results

In order to evaluate the performance of exome capture-based RNA-Seq methods in profiling FFPE samples, we conducted a meticulous technical assessment of the three distinct protocols mentioned above, namely Illumina (IL), Agilent (AG), NadPrep with both stand-specific treatment (NP) and non-strand-specific treatment (NS) on human reference RNA (UHRR) and commercially available FFPE RNA (FFPE). The schematic of the workflow along with statistical information at each library preparation step are presented in Table S1. We utilized the dataset to evaluate and assess the performance of RNA-Seq library preparation protocols. First, we examined the protocols’ ability to maintain consistent alignment rates. Second, we assessed the accuracy of these protocols in calculating gene expression by comparing the results to TaqMan data. At last, we explored the protocols’ capability to identify fusion genes that have been experimentally validated within the samples.

Alignment statistics

Overall, we prepared a total of 20 libraries for sequencing, both the DNBSEQ and Novaseq platforms generated comparable high-quality reads. The Novaseq libraries exhibited a significantly higher duplication rate compared to the DNBSEQ libraries (Table S1). This can be attributed to the exponential amplification during the clustering process [9], which may have an impact on the saturation of data, particularly when sequencing resources are limited. We initially examined the overall alignment rates to the human genome (Table 2). The results demonstrated that all the protocols exhibited good performance, indicated by the high alignment rates greater than 90%. The AG libraries exhibited relatively higher rRNA contamination, suggesting lower specificity of the exome capture procedure in this protocol.

Table 2 The alignment statistics to the human genome

The sense rate is a metric that calculates the percentage of aligned forward reads mapped to the antisense gDNA strand and the percentage of aligned reverse reads mapped to the sense gDNA strand. Strand-specific RNA-Seq protocols offer the advantage of resolving read ambiguity in cases where overlapping genes are transcribed from the opposite strands. This specificity enhances the accuracy of gene quantification and prediction of fusion genes. The IL libraries showed the highest sense rate, indicating a more efficient strand-specific library preparation process [10]. Meanwhile, the IL libraries yielded the lowest count of identified genes and transcripts, potentially be attributed to the different design of targets and exome panels among these kits. We can make a preliminary inference about the genes for which Illumina kits lack probes, by noting a particular gene that exhibits consistent expression across all other libraries, yet in IL libraries the FPKM value is recorded as zero. Most of the selected genes are non-annotated, while some of them, such as gene C4orf48 may be disease relevant and important for clinical studies [11]. The NP libraries exhibited a higher genome mapping rate, but lower number of genes detected than the NS libraries. These findings align with previous studies based on Poly(A) RNA sequencing [12].

In addition, the protocols showed notable distinctions in the proportions of reads aligned to exons, introns, and other intergenic regions (Table S2). For NS and NP libraries, the percentages of reads that aligned to exons were above 94%, indicating the highest efficiency of the exome pull down by the capture approach. As anticipated [13], with the RD libraries, we observed that approximately half of the reads mapped to exons, while the remaining reads predominantly mapped to intronic regions (ranging from 32 to 42%).

Transcript coverage

We proceeded to assess the coverage across the full length of the genes and observed that all the protocols demonstrated broad and uniform transcript coverage (Fig. 2a). Notably, the exome capture-based protocols exhibited a slight 5′ bias, which can be attributed to the second structure of the transcripts and the mechanism of reverse transcription [14]. In other words, a higher proportion of reads mapped to the 5′ region of the transcripts compared to other methods, particularly those utilizing Poly(A) enrichment procedures. It is well established that with Poly(A) selected mRNA, the sensitivity of fusion detection is determined by the level of coverage at the position where the gene fusion resides, and this coverage decreases with the distance from the 3′ end of the mRNA, when the input RNA is in degradation [15]. Furthermore, the proportion of reads mapping to coding regions was significantly elevated comparing to RD libraries, making the exome capture-based protocols ideal for gene fusion detection in FFPE derived or other highly degraded samples.

Fig. 2
figure 2

Statistics of alignment and expression of the experiments. a Normalized transcript coverage of UHRR libraries. b Expression level of genes detected in UHRR libraries. Correlation heatmap c between UHRR libraries and TaqMan data, d of fold-change values of genes between the two samples (FFPE vs UHRR), and e among NP libraries with different read lengths

Gene expression

We first investigated the abundance distribution of all the genes detected by different protocols [16] and categorized them into groups based on their respective expression levels (Fig. 2b). Notably, the IL libraries exhibited the largest proportion of genes with low FPKM values (less than 1), when detecting the smallest total number of genes. This observation highlights a potential concern since clinical studies often establish thresholds for gene expression levels, like FPKM greater than 0.3 or 0.5, due to the methodology’s high sensitivity and inherent false-positive rate [17]. The mechanical filtering process may lead to the exclusion of clinical-relevant targets with low abundance or insufficient full-length coverage. This may further compromise the ability of IL libraries to identify rare fusion events.

We then assessed the agreement between the protocols by calculating the Pearson coefficient with all the UHRR libraries and the TaqMan reference data [18]. The exome capture-based libraries exhibited lower correlation (ranging from 0.66 to 0.76) than the RD libraries (exceeding 0.87). When comparing the different protocols against each other, the gene expression values exhibited significant protocol-specific biases, leading to a reduced agreement (correlations ranging from 0.46 to 0.88). However, the consistency within replicates and multiplexing was ideal, with correlations greater than 0.99 (Fig. 2c).

Finally, we assessed the agreement between the protocols by calculating the Pearson coefficient of the fold-change values of the differentially expressed genes between the two samples (FFPE vs UHRR, Fig. 2d). The results demonstrated that the fold-change values correlated acceptably across the entire dynamic range of expression between the protocols (correlations ranging from 0.68 to 0.73), suggesting the possibility of comparing the DEGs of paired samples using different methods when the consistency of the whole study cannot be guaranteed.

Fusion gene detection

Multiple bioinformatic pipelines have been developed to identify candidate fusion genes from RNA-Seq data [19]. Predicted fusions is typically supported by fragments found as junction reads that directly overlap the splicing site, or as spanning reads where each pair of reads maps to the opposite partner of the fusion genes (Fig. 3a). In this study, we assessed three most cited methods for fusion detection: EricScript [20], FusionCatcher, and Star-Fusion. EricScript and FusionCatcher called more fusion genes than Star-Fusion (Fig. 3b), but for FFPE samples, EricScript reported only 1 of the 6 validated transcripts, while FusionCatcher and Star-Fusion reported all the 6 events (Table S3). Although it could be argued that these fusions represent a limited target reference for method comparison, EricScript exhibited lower sensitivity and higher false-positive rate. Notably, EricScript also reported fusion transcripts involving opposite partners of HLA-C and HLA-A with EricScore greater than 0.95, indicating a poor performance in distinguishing highly homologous genes. On the other hand, FusionCatcher reported more than 300 fusion candidates per library, although the majority of which were remarked as exonic or in-frame, researchers would face significant challenges in filtering and validating all these predictions.

Fig. 3
figure 3

Statistics of fusion genes detection. a Diagram of COSF1196 transcript, depicting spanning and junction reads used to identify fusion genes. b Statistics of fusion events and the corresponding supporting read counts of all FFPE libraries by different pipelines. The circular diameter represents the total read counts for the cumulative fusion events, while the color corresponds to the mean read counts supporting each individual fusion event. Enrichment of 6 fusions between exome capture-based and rRNA depleted libraries calculated by c Star-Fusion, d FusionCatcher, and e using reads of different lengths

We then examined the target enrichment rate of different protocols by comparing the Fusion Fragments Per Million mapped reads (FFPM) from exome capture-based and rRNA depleted data (Table S3 and Fig. 3c, d), finding that Star-Fusion exhibited better reproducibility in replicate or multiplexed libraries, and achieved a mean 1.75, 5.03, 5.63 and 4.65-fold enrichment for the IL, AG, NS and NP libraries respectively, illustrating the advantage of exome capture-based protocols in fusion genes detection compared to the conventional methods, particularly for highly degraded samples [21].

Impact of read length

The length of the sequencing reads is a crucial factor to consider when conducting RNA-Seq experiments. Previous studies suggest that for generating a list of DEGs, 50 bp single-end reads are generally sufficient [22]. However, for isoform detection, longer reads are preferred to capture comprehensive information, while longer reads are not necessarily significantly better than shorter reads for differential expression analysis.

The DNBSEQ series of sequencers, including DNBSEQ-T7, MGISEQ-2000, and DNBSEQ-G99, offer a range of sequencing throughputs and read lengths for various research applications. In clinical occasions, researchers often face the decision between PE100 and PE150 run cycles, considering factors like sample quality, experimental costs, turnaround time requirements, and the challenges of pooling adequate libraries to fulfill a sequencing run. To investigate the impact of read length on FFPE RNA-Seq outcomes, we performed an additional MGISEQ-2000 PE100 run for NP libraries, made up three groups of NP data with different read lengths: PE150, PE150 trimmed into PE100 (PE150Trim) and PE100. The overall statistics of the sequencing results are presented in Table S4.

As expected, the PE100 data exhibited improved performance in terms of adapter contamination, reference alignment and number of genes detected. Nevertheless, it did not show significant changes in the accuracy of expression measurement (Fig. 2e) and the target enrichment rate of fusion genes (Table S3 and Fig. 3e). These findings suggest that while PE100 data improvements are beneficial for certain aspects, they may not provide additional advantages for clinical study applications. At the meantime, when compare FFPE_PE150_1 vs FFPE_PE100_1 (Table S5), there were 8904 specifically detected transcripts in PE150 data compared to PE100 data, hundreds of longer isoforms of clinically relevant transcripts can only be detected in PE150 data with low to median abundance (FPKM≥1), suggesting the potential of longer reads in disease studies.

Conclusions

In this study, we present a comprehensive assessment that encompasses all crucial factors to be considered in RNA-Seq experiments to optimize the utilization of clinical FFPE samples. Researchers are provided with the opportunity to make informed decisions regarding the selection of capture probes, library preparation and multiplexing, sequencing parameters, and bioinformatic pipelines, especially when sample quality is severely compromised (Table 3). For fusion genes detection, prudent filtering techniques and critical experimental validations are imperative to ensure accuracy and reliability of the results [23]. We recommend the adoption of exome capture-based RNA-Seq protocols when the input sample are not suitable for conventional methods. Although the commercial kits offer advantages in the sequencing depth of coding regions, the considerations of inconsistent capture efficiency and rRNA residue merit careful attention and pre-experiment contemplation to ensure the optimal application of the selected protocols. We look forward that exome capture-based RNA-Seq methodologies will experience growing adoption in clinical settings for the diagnosis of fusion genes, further advancing our understanding of fusion gene biology, and enhancing cancer diagnostics.

Table 3 Summary of the decisions in exome capture-based protocols

Availability of data and materials

All sequencing data have been deposited in the CNGB Nucleotide Sequence Archive (https://db.cngb.org/cnsa) under the accession number CNP0004621.

References

  1. Boneva S, Schlecht A, Böhringer D, Mittelviefhaus H, Reinhard T, Agostini H, et al. 3′ MACE RNA-sequencing allows for transcriptome profiling in human tissue samples after long-term storage. Lab Invest; a J Technical Methods Pathol. 2020;100(10):1345–55. https://doi.org/10.1038/s41374-020-0446-z.

    Article  CAS  Google Scholar 

  2. Winters JL, Davila JI, McDonald AM, Nair AA, Fadra N, Wehrs RN, et al. Development and verification of an RNA sequencing (RNA-Seq) assay for the detection of gene fusions in tumors. The J Mol Diag JMD. 2018;20(4):495–511. https://doi.org/10.1016/j.jmoldx.2018.03.007.

    Article  CAS  Google Scholar 

  3. Jacobsen SB, Tfelt-Hansen J, Smerup MH, Andersen JD, Morling N. Comparison of whole transcriptome sequencing of fresh, frozen, and formalin-fixed, paraffin-embedded cardiac tissue. PLoS One. 2023;18(3):e0283159. https://doi.org/10.1371/journal.pone.0283159.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Heyer EE, Deveson IW, Wooi D, Selinger CI, Lyons RJ, Hayes VM, et al. Diagnosis of fusion genes using targeted RNA sequencing. Nat Commun. 2019;10(1):1388. https://doi.org/10.1038/s41467-019-09374-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Endrullat C, Glökler J, Franke P, Frohme M. Standardization and quality management in next-generation sequencing. Appl Transl Genom. 2016;10:2–9. https://doi.org/10.1016/j.atg.2016.06.001.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Wang C, Gong B, Bushel PR, Thierry-Mieg J, Thierry-Mieg D, Xu J, et al. The concordance between RNA-seq and microarray data depends on chemical treatment and transcript abundance. Nat Biotechnol. 2014;32(9):926–32. https://doi.org/10.1038/nbt.3001.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Schuierer S, Carbone W, Knehr J, Petitjean V, Fernandez A, Sultan M, et al. A comprehensive assessment of RNA-seq protocols for degraded and low-quantity samples. BMC Genomics. 2017;18(1):442. https://doi.org/10.1186/s12864-017-3827-y.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Decruyenaere P, Verniers K, Poma-Soto F, Van Dorpe J, Offner F, Vandesompele J. RNA extraction method impacts quality metrics and sequencing results in formalin-fixed, paraffin-embedded tissue samples. Lab Invest; a J Technical Methods Pathol. 2023;103(2):100027. https://doi.org/10.1016/j.labinv.2022.100027.

    Article  Google Scholar 

  9. Li Q, Zhao X, Zhang W, Wang L, Wang J, Xu D, et al. Reliable multiplex sequencing with rare index mis-assignment on DNB-based NGS platform. BMC Genomics. 2019;20(1):215. https://doi.org/10.1186/s12864-019-5569-5.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Harrington CA, Fei SS, Minnier J, Carbone L, Searles R, Davis BA, et al. RNA-Seq of human whole blood: evaluation of globin RNA depletion on Ribo-zero library method. Sci Rep. 2020;10(1):6271. https://doi.org/10.1038/s41598-020-62801-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Endele S, Nelkenbrecher C, Bördlein A, Schlickum S, Winterpacht A. C4ORF48, a gene from the Wolf-Hirschhorn syndrome critical region, encodes a putative neuropeptide and is expressed during neocortex and cerebellar development. Neurogenetics. 2011;12(2):155–63. https://doi.org/10.1007/s10048-011-0275-8.

    Article  CAS  PubMed  Google Scholar 

  12. Zhao S, Zhang Y, Gordon W, Quan J, Xi H, Du S, et al. Comparison of stranded and non-stranded RNA-seq transcriptome profiling and investigation of gene overlap. BMC Genomics. 2015;16(1):675. https://doi.org/10.1186/s12864-015-1876-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Zhao S, Zhang Y, Gamini R, Zhang B, von Schack D. Evaluation of two main RNA-seq approaches for gene quantification in clinical RNA sequencing: polyA+ selection versus rRNA depletion. Sci Rep. 2018;8(1):4781. https://doi.org/10.1038/s41598-018-23226-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Wang Z, Gerstein M, Snyder M. RNA-Seq: a revolutionary tool for transcriptomics. Nat Rev Genet. 2009;10(1):57–63. https://doi.org/10.1038/nrg2484.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Davila JI, Fadra NM, Wang X, McDonald AM, Nair AA, Crusan BR, et al. Impact of RNA degradation on fusion detection by RNA-seq. BMC Genomics. 2016;17(1):814. https://doi.org/10.1186/s12864-016-3161-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Piovesan A, Caracausi M, Antonaros F, Pelleri MC, Vitale L. GeneBase 1.1: a tool to summarize data from NCBI gene datasets and its application to an update of human gene statistics. Database. 2016;2016:baw153. https://doi.org/10.1093/database/baw153.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Adiconis X, Borges-Rivera D, Satija R, DeLuca DS, Busby MA, Berlin AM, et al. Comparative analysis of RNA sequencing methods for degraded or low-input samples. Nat Methods. 2013;10(7):623–9. https://doi.org/10.1038/nmeth.2483.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Kumar A, Kankainen M, Parsons A, Kallioniemi O, Mattila P, Heckman CA. The impact of RNA sequence library construction protocols on transcriptomic profiling of leukemia. BMC Genomics. 2017;18(1):629. https://doi.org/10.1186/s12864-017-4039-1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Haas BJ, Dobin A, Li B, Stransky N, Pochet N, Regev A. Accuracy assessment of fusion transcript detection via read-mapping and de novo fusion transcript assembly-based methods. Genome Biol. 2019;20(1):213. https://doi.org/10.1186/s13059-019-1842-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Benelli M, Pescucci C, Marseglia G, Severgnini M, Torricelli F, Magi A. Discovering chimeric transcripts in paired-end RNA-seq data by using EricScript. Bioinformatics (Oxford, England). 2012;28(24):3232–9. https://doi.org/10.1093/bioinformatics/bts617.

    Article  CAS  PubMed  Google Scholar 

  21. Byron SA, Van Keuren-Jensen KR, Engelthaler DM, Carpten JD, Craig DW. Translating RNA sequencing into clinical diagnostics: opportunities and challenges. Nat Rev Genet. 2016;17(5):257–71. https://doi.org/10.1038/nrg.2016.10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Chhangawala S, Rudy G, Mason CE, Rosenfeld JA. The impact of read length on quantification of differentially expressed genes and splice junction detection. Genome Biol. 2015;16(1):131. https://doi.org/10.1186/s13059-015-0697-y.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Liu Y, Bhagwate A, Winham SJ, Stephens MT, Harker BW, McDonough SJ, et al. Quality control recommendations for RNASeq using FFPE samples based on pre-sequencing lab metrics and post-sequencing bioinformatics metrics. BMC Med Genet. 2022;15(1):195. https://doi.org/10.1186/s12920-022-01355-0.

    Article  CAS  Google Scholar 

Download references

Acknowledgments

We would like to thank the BGI sequencing team for the generous technical advice. We would like to thank Xiaohui Li and Lina Wang for the help in managing this project.

Funding

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

L.Z. and Y.Z. contributed equally to this work. L.Z. and Y.Z. conceived the study. L.Z., Y.J., Y.X. and Q.L. performed sample extraction, library preparation and sequencing experiments. Y.Z. and L.Z. performed bioinformatic analysis and wrote the initial draft. S.J. oversaw the project and reviewed the paper. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Sanjie Jiang.

Ethics declarations

Ethics approval and consent to participate

This Study was approved by the Institutional Review Board on Bioethics and Bio-safety of BGI (BGI-IRB) under the approval number 20230713791.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zong, L., Zhu, Y., Jiang, Y. et al. A comprehensive assessment of exome capture methods for RNA sequencing of formalin-fixed and paraffin-embedded samples. BMC Genomics 24, 777 (2023). https://doi.org/10.1186/s12864-023-09886-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12864-023-09886-1

Keywords