Skip to main content

A PCR-independent approach for mtDNA enrichment and next-generation sequencing: comprehensive evaluation and clinical application

Abstract

Background

Sequencing the mitochondrial genome has been increasingly important for the investigation of primary mitochondrial diseases (PMD) and mitochondrial genetics. To overcome the limitations originating from PCR-based mtDNA enrichment, we set out to develop and evaluate a PCR-independent approach in this study, named Pime-Seq (PCR-independent mtDNA enrichment and next generation Sequencing).

Results

By using the optimized mtDNA enrichment procedure, the mtDNA reads ratio reached 88.0 ± 7.9% in the sequencing library when applied on human PBMC samples. We found the variants called by Pime-Seq were highly consistent among technical repeats. To evaluate the accuracy and reliability of this method, we compared Pime-Seq with lrPCR based NGS by performing both methods simultaneously on 45 samples, yielding 1677 concordant variants, as well as 146 discordant variants with low-level heteroplasmic fraction, in which Pime-Seq showed higher reliability. Furthermore, we applied Pime-Seq on 4 samples of PMD patients retrospectively, and successfully detected all the pathogenic mtDNA variants. In addition, we performed a prospective study on 192 apparently healthy pregnant women during prenatal screening, in which Pime-Seq identified pathogenic mtDNA variants in 4 samples, providing extra information for better health monitoring in these cases.

Conclusions

Pime-Seq can obtain highly enriched mtDNA in a PCR-independent manner for high quality and reliable mtDNA deep-sequencing, which provides us an effective and promising tool for detecting mtDNA variants for both clinical and research purposes.

Introduction

Mitochondria are eukaryotic organelles playing essential roles in a series of cellular processes, including bioenergy production, calcium handling, and intrinsic apoptosis regulation [1, 2]. Each mitochondrion contains multiple copies of their own genome in the mitochondrial matrix, known as mitochondrial DNA (mtDNA). Human mtDNA is a circular double-stranded DNA with 16,569 bp in size, encoding 37 genes, including 13 oxidative phosphorylation (OXPHOS) related polypeptides, 22 tRNAs and 2 rRNAs [3, 4]. Different from the nuclear genome, the mtDNA has a much higher mutation rate due to the exposure to reactive oxygen species and the lack of nucleosome protection. In addition, each human cell contains thousands of mtDNA molecules [5]. As a result, the mtDNA variants often co-exists with the wildtype mtDNA within a cell, known as a state of heteroplasmy [6].

Pathogenic mtDNA variants are potentially related to primary mitochondrial diseases (PMDs). The clinical phenotype of PMDs is highly variable, often affecting the central nervous and musculoskeletal system, leading to severe birth defects and early mortality [7]. In general population, the PMDs caused by pathogenic mtDNA variants present in approximately 1 in 10,000 adults [8]. This is much more prevalent than the PMDs caused by nuclear pathogenic variants, which is in approximately 2.9 per 100,000 adults [9], although more than 1000 mitochondrial proteins are encoded from the nuclear genome [10]. Moreover, most severe pathogenic mtDNA variants tend to be heteroplasmy in nature, where the severity of PMD symptoms correlates with the heteroplasmic fraction (HF) [11, 12]. In this way, it is important to accurately detect mtDNA variants, especially heteroplasmic ones, for both clinical and research purposes.

Currently, there are several major molecular methods for detecting mtDNA variants. Sanger sequencing is often used to reliably detect homoplasmic variants and heteroplasmic variants with high HF [13]. However, this strategy is commonly used to screen known pathogenic variants without providing quantitative information. For purposes of investigating causative rare variants in human diseases or novel mutations on cell biology, whole mtDNA sequencing is required, which is mostly achieved utilizing next generation sequencing (NGS) after PCR-based mtDNA enrichment [14,15,16,17]. However, this PCR-based approach can lead to false positive and false negative results of heteroplasmic variants due to mutations introduced by polymerase error and amplification bias [18,19,20,21,22,23,24], and the sequencing results can be further distorted when the amplicons lie inside nuclear-mitochondrial sequence (NUMTs) regions. In addition, polymorphisms at primer regions would potentially lead to amplification failure for these mtDNA molecues. Furthermore, due to its relatively long turn-around time, long-range PCR based NGS (lrPCR-NGS) is less preferable for large-scale screening studies. Therefore, an accurate, reliable and effective strategy for whole mitochondrial genome deep-sequencing is urgently needed.

Here, we developed a novel high-throughput mtDNA sequencing method based on mtDNA isolation and Tn5 tagmentation. Our new method can achieve PCR-independent mtDNA enrichment while minimizing the presence of nuclear DNA in the sequencing library, and show its reliability and accuracy for the detection of mtDNA variants, particularly when HF is low. The clinical assessment of this method, conducted on both retrospective and prospective samples, demonstrate the effectiveness of this PCR-independent method in detecting pathogenic variants and heteroplasmic variants with potential biological value, making it a promising approach for prenatal screening and other clinical or research applications in the future.

Materials and methods

Sample source

In this study, we utilized 1 mL peripheral blood sample for mtDNA sequencing, which were obtained from leftover blood samples of involved pregnant woman who underwent prenatal cell-free DNA screening at the Department of Prenatal Diagnosis in Nanjing Maternity and Child Health Care Hospital. For the retrospective study, peripheral blood sample was obtained from 4 patients who were previously diagnosed with mitochondrial diseases.

mtDNA isolation and sequencing library construction

Peripheral blood mononuclear cells (PBMC) were separated from 1 mL fresh peripheral blood samples (within 8 h of blood draw) using Lymphoprep Lymphocyte Isolation Solution (Stemcell, Canada) and SepMate Centrifuge Tube (Stemcell, Canada), and approximately 40,000 fresh PBMC were collected for further steps. Urine derived cells were harvested from 60 mL urine sample by centrifugating at 500 × g for 12 min followed by washing with 0.9% NaCl and centrifugating at 500 × g for 3 min. PBMC or urine derived cells were resuspended in 15 μL pre-chilled resuspension buffer (0.1 mM Tris–HCl pH 7.4, 20 μM NaCl, 9 μM MgCl2) containing 1% NP40 (Sigma-Aldrich, Missouri, USA), keeping in 4 °C for 3 min. After centrifugation at 10,000 × g for 5 min at 4 °C, 9.5 μL supernatant was collected, which contains a majority of circular mtDNA and a minority of linear nuclear DNA. Next, 1 μL Exonuclease V, 1.2 μL Adenosine 5'-Triphosphate, 1.2 μL NEBuffer™ 4 (NEB, Nebraska, USA) was added to remove any residual linear nuclear DNA in the supernatant, which was incubated at 37 ℃ for 30 min followed by heat inactivation. At this time, mtDNA isolation and enrichment has been completed. The isolated mtDNA was then added with 12.4 μL 2 × TD buffer (0.4 mM Tris–HCl pH 7.6, 0.1 mM MgCl2, 4% DMF) and 0.5 μL TTE Mix V50 Tn5 (Vazyme, China), incubating at 37 ℃ for 30 min to obtain mtDNA fragments through tagmentation. The tagmented mtDNA was then recovered using the DNA Clean & Concentrator kit (ZYMO Research, California, USA) and amplified with index primers to construct sequencing library. We established an optional quality control step by performing PCR assay of amplifying nuclear genes (18S, 28S) and mitochondrial genes (MT-TK, MT-ND5) to validate the efficacy of mtDNA enrichment and nuclear DNA elimination in the library. The libraries were then purified using DNA Clean Beads (Beckman Coulter, Germany) by 0.5 × /0.3 × double size selection and the quantification was measured by Qubit 3.0 (Thermo Fisher Scientific, Massachusetts, USA). Indexed DNA libraries were pooled and sequenced on Illumina NovaSeq 6000, generating paired-end 150 bp reads at 1G data per sample.

LrPCR-based mtDNA enrichment and sequencing library construction

Full length mtDNA is split into two overlapping segments, each approximately 8.5 kb in length. The primer pairs used were 5’-GCAAATCTTACCCCGCCTG-3’/5’-AATTAGGCTGTGGGTGGTTG-3’ and 5’-GCCATACTAGTCTTTGCCGC-3’/5’-GGCAGGTCAATTTCACTGG-3’. LrPCR was performed using Phanta Max Super-Fidelity DNA Polymerase (Vazyme, China) and 50 ng of total genome DNA. LrPCR reaction condition was 95 °C for 30 s, 25 cycles of 95 °C for 15 s, 60 °C for 15 s, 72 °C for 8 min with a final extension of 72 °C for 5 min. PCR products were then purified using 1% gel and FastPure Gel DNA Extraction Mini Kit (Vazyme, China), and measured using Qubit3.0 (Thermo Fisher Scientific, Massachusetts, USA). The two segments were then pooled with equal moles and tagmented using Tn5 (5 ng amplicon1, 5 ng amplicon2, 2 μL 5 × TTBL, 1 μL TTE Mix V50), incubating at 55 °C for 10 min. The tagments were retrieved using 1 × DNA Clean Beads (Beckman Coulter, Germany), and sequencing library was then constructed using TruePrep DNA Library Prep Kit V2 for Illumina (Vazyme, China). The libraries were purified using DNA Clean Beads by 0.5 × /0.3 × double size selection and the quantification was assessed by Qubit3.0 (Thermo Fisher Scientific, Massachusetts, USA). Libraries were pooled and sequenced on Illumina NovaSeq 6000.

Quality control and data analysis

The raw sequencing reads were trimmed using trim_galore (v0.6.6) (https://www.bioinformatics.babraham.ac.uk/projects/trim_galore) with following parameters “–paired -q 20 –phred33 –stringency 4 –length 20 –trim-n”. Reads quality control was performed with FastQC (v0.11.9) (https://www.bioinformatics.babraham.ac.uk/projects/fastqc). All the clean reads were aligned to the source genome (Nuclear, GRCh38 and Mitochondrial, NC_012920.1) (http://ftp.ensembl.org/pub/current_fasta/homo_sapiens/dna) using bowtie2 (v2.4.4) (https://github.com/BenLangmead/bowtie2) with default parameters to generate BAM files. Mitochondrial genome reads and nuclear genome reads extraction were performed by Samtools (version 1.7.0) (https://github.com/samtools/samtools/releases). MT reads percentage refers to the percentage of the reads mapped to the mitochondrial genome to all reads mapped to the source genome. The mtDNA variant calling was performed based on GATK Best Practices for SNP/Indel Variant Calling in Mitochondria (https://github.com/gatk-workflows/gatk4-mitochondria-pipeline), with optimization during the variants calling process by setting the downsampling parameter to "–max-reads-per-alignment-start 100,000". The pathogenic variants were visualized in the Integrative Genomics Viewer (IGV) program with BAM files.

PCR assay

PCR was used to verify the efficiency of mtDNA enrichment. The specific gene regions in the mitochondrial and nuclear genome were amplified and detected with specific primers (Additional file 1: Table S1) and 2 × Taq Master Mix (Vazyme, China). The total reaction volume was 50 μL, containing 25 μL of 2 × Taq Master Mix, 2 μL of 10 μM forward and reverse primer respectively, and DNA 100 ng, add nuclease-free water to 50 μL. The cycle conditions were as follows: an initial denaturation at 98 °C for 30 s, 35 cycles of 98 °C for 10 s, 63 °C for 30 s, 72 °C for 1 min, and a final extension at 72 °C for 5 min. The results were analyzed by visualizing the PCR product using 2% gel electrophoresis.

Sanger sequencing

We used 40 μL PCR products, amplified from the region containing candidate mtDNA variants, for Sanger sequencing to verify the pathogenic variants. Sanger sequencing reagents and instruments were provided by Applied Biosystems.

Pyrosequencing

Prepare PCR products according to pyrosequencing requirements. Biotin labeling is applied to one of the primers of PCR for the subsequent single-chain separation and purification, to ensure that there is no free biotin in the primer. PCR system refers to the instructions of PyroMark PCR Kit (QIAGEN, Germany). The samples were then sequenced on PyroMark Q96 Autoprep (QIAGEN, Germany).

Results

The mtDNA-isolation-based procedure can achieve highly enriched mtDNA for NGS

To develop and optimize a PCR-independent approach for mtDNA enrichment in human cells, we combined classic cell biology techniques with previously described method for subcellular fractionation to achieve highly efficient separation of mtDNA from nuclear genome [25]. We tested different conditions for mtDNA isolation and enrichment in human PBMC samples, including the use of NP40 and Tween-20 in resuspension buffer and the use of exonuclease after cell lysis. To evaluate the efficacy of mtDNA isolation and enrichment in different conditions, we used PCR assay to amplify and detect nuclear genes (18S, 28S) and mitochondrial genes (MT-TK, MT-ND5) in enriched DNA (Additional file 8: Fig. S1), which showed an optimal condition involving: (a) the use of resuspension buffer containing 1% NP40 to lyse the cells and isolate mitochondria, and (b) the use of 0.8 unit/μL exonuclease V to degrade the linear nuclear DNA in supernatant. We further performed a PCR assay on the enriched mtDNA from 4 PBMC samples, which revealed the mtDNA was convincingly isolated while the nuclear genome was hardly detectable under the given enrichment conditions (Fig. 1C). The circular mtDNA was then tagmented by Tn5 for sequencing library preparation followed by NGS, which is the entire procedure of Pime-Seq (Fig. 1A). The clean sequencing reads were aligned to the mitochondrial genome reference sequence (rCRS, or NC_012920.1). Finally, we called homoplasmic or heteroplasmic variants by GATK Best Practices for SNP/Indel Variant Calling in Mitochondria. We defined that MT reads percentage refers to the percentage of the reads mapped to the mitochondrial genome to all reads mapped to the whole genome reference sequence (GRCh38, mitochondrial genome + nuclear genome) (Fig. 1B). To evaluate this strategy, we performed our new method on 20 human PBMC samples and analyzed the MT reads percentage to determine the relative amount of mtDNA to nuclear DNA in the mtDNA enriched library. The results showed an average of 88.0% ± 7.9% of MT reads. Consistently, the average sequence depth was as high as 16,538 ± 8,987 × (Fig. 1D, Additional file 2: Table S2). These results demonstrated that our PCR-independent approach can achieve efficient mtDNA enrichment before sequencing on PBMC samples, and we thus named this method as PCR-independent mtDNA enrichment-based Sequencing (Pime-Seq). In addition, we tested this approach on 3 urine samples, which showed an average MT reads ratio of 0.5% in the sequencing results with relatively low sequence depth, indicating the current experimental procedure still needs optimization for sample types other than blood (Additional file 3: Table S3).

Fig. 1
figure 1

Overview and assessment of Pime-Seq. A Workflow of PCR-independent mtDNA enrichment-based sequencing; B Flowchart of bioinformatics analysis pipeline for Pime-Seq; C Analysis of mtDNA purity after different mtDNA isolation conditions; D The MT reads percentage obtained by Pime-Seq on 20 human PBMC samples

Pime-Seq can yield results with high reproducibility and accuracy

In order to evaluate the reproducibility of the variant identification using Pime-Seq, we randomly selected 3 human PBMC samples, and performed 3 technical repeats on each sample (Additional file 4: Table S4). The results showed that homoplasmic variants were consistently detected in 3 repeats across all the 3 samples. Similarly, results of variants called with HF greater than 1.4% were consistent in at least two repeats, such as the m.6068C > T in 22TS0001 and the m.5894A > AC in 22TS0003. However, heteroplasmic variants resulting in HF below 1.4% were called without consistency, such as the m. 6510G > A and m.8014A > G variants in 22TS0002 and the m.9576C > A in 22TS0003, indicating potential false positive or false negative results (Fig. 2). These results suggested a detection limit of Pime-Seq for reliable HF, and we therefore set 1.4% as the minimal HF threshold for heteroplasmic variant calling in subsequent studies.

Fig. 2
figure 2

The distribution of all variants identified by Pime-Seq across the mitochondrial genome in 3 technical repeats on 3 human PBMC samples

As lrPCR-based NGS is currently used as the most common strategy for mtDNA sequencing, we set out to compare Pime-Seq and lrPCR-NGS in terms of the experimental procedures and the accuracy. We performed both sequencing methods on 45 human PBMC samples in parallel (Additional file 5: Table S5). On average, Pime-Seq took 5 h to complete library construction starting from peripheral blood, whereas lrPCR-NGS required approximately 10 h for the same procedure. From the sequencing results, 1737 variants were called by Pime-Seq and 1763 variants were called by lrPCR-NGS (Fig. 3A). Out of these variants, 1677 were concordant in both methods with high correlation in HF (R2 = 0.9932, P < 0.0001, Spearman’s correlation analysis), including 1612 homoplasmic variants and 65 heteroplasmic variants (Fig. 3B). On the other side, a total of 146 discordant variants were only called by one method but not the other in the same sample, which were all heteroplasmic with low HF (Additional file 6: Table S6). A closer analysis of the 146 discrepancies revealed that 80% (41/51) of the putative variants called exclusively by Pime-Seq were found to be recorded in public databases including MitoMap, Human Mitochondrial Genome Database or HelixMTdb, suggesting the detection of these variants by Pime-Seq could reflect a genuine existence. On the other hand, 76% (42/55) of the putative variants called exclusively by lrPCR-NGS were not recorded in any of the three databases, indicating possible false positives in calling these putative variants, probably caused by PCR amplification errors during lrPCR-based enrichment (Fig. 3C). Furthermore, we utilized pyrosequencing to testify the existence of m.9540 T > C in sample 22MS0027, which was called with 4.1% HF by Pime-Seq, but not called by lrPCR-NGS. The pyrosequencing result validated the detection of m.9540 T > C in 22MS0027 (Fig. 3D). Whereas m.9540 T > C was not detected by pyrosequencing in negative control samples (22MS0038 and 22MS0043), in which m.9540 T > C was not called in both Pime-Seq and lrPCR-NGS results (Additional file 9: Fig. S2). These findings demonstrated the accuracy of Pime-Seq results, which is comparable to lrPCR-NGS when calling homoplasmic variants and heteroplasmic variants with high HF, and more reliable when calling heteroplasmic variants with low HF.

Fig. 3
figure 3

Comparison of Pime-Seq and lrPCR-NGS. A flow diagram of the comparison study; B HF correlation of the 1677 concordant variants detected by Pime-Seq and lrPCR-based NGS in 45 human PBMC samples. The HF is indicated by the numbers on both the horizontal and vertical axes. Correlations and P values are based on Spearman’s correlation; C Evaluating the reliability of the 146 discordant variants by analyzing them in human mitochondrial genome databases; D Verification of the discordant result of m.9540T > C in sample 22MS0027 using pyrosequencing

Evaluation of Pime-Seq in a retrospective study

To evaluate its clinical application, we retrospectively conducted Pime-Seq on PBMC samples of 4 previously diagnosed patients carrying pathogenic mtDNA variants, including 1 with homoplasmic m.11778A > G, 2 with homoplasmic m.4300A > G and 1 with heteroplasmic m.8344A > G. The results of Pime-Seq revealed that all the pathogenic variants were accurately detected (Table 1 and Additional file 10: Fig. S3). We further verified all the pathogenic variants using Sanger sequencing, which showed consistent results. It is worth noting that the m.11778A > G variant was recorded as homoplasmic, as well as from our Sanger sequencing result, while the Pime-Seq revealed a heteroplasmic result, with HF at about 94% (Additional file 10: Fig. S3). Above all, our results demonstrated the reliability of Pime-Seq when applied for detecting pathogenic mtDNA variants and PMD genetic diagnosis.

Table 1 Retrospective evaluation of Pime-Seq in 4 PMD patients

Evaluation of Pime-Seq in a prospective study

As a practical strategy for screening pathogenic mtDNA variants in clinical settings are currently lacking, we assessed the application of Pime-Seq in a prenatal screening setting, by utilizing the leftover blood samples from prenatal cell-free DNA screening. A total of 192 healthy pregnant women were randomly selected and included in this study (Table 2). In the 192 prospective samples, Pime-Seq results revealed 7196 variants, including 6743 homoplasmic variants and 453 heteroplasmic variants (Table 3). According to variants with confirmed pathogenic status in Mitomap database, and we identified 4 potential pathogenic variants, including 2 cases of homoplasmic m.1555A > G, 1 case of heteroplasmic m.1555A > G with HF of 2.4%, and 1 case of heteroplasmic m.3243A > G with HF of 13.4% (Table 4). We further verified these variants using Sanger sequencing (Additional file 11: Fig. S4). The homoplasmic m.1555A > G is associated with aminoglycosides-induced hearing loss, and the m.3243 A > G is associated with MELAS and other clinical features when HF is high. As the variant HFs in the 2 cases of heteroplasmic m.1555A > G and heteroplasmic m.3243A > G were relatively low, we therefore only informed the women in the 2 homoplasmic m.1555A > G cases, and planned to add PMD-related questions in the regular postnatal follow-up in all the 4 cases for better health monitoring.

Table 2 Clinical information of the 192 apparently healthy pregnant women
Table 3 Information of the variants called by Pime-Seq in the 192 apparently healthy pregnant women
Table 4 Pathogenic mtDNA variants identified by Pime-Seq from the 192 apparently healthy pregnant women

We next characterized the 453 heteroplasmic variants identified by Pime-Seq in these prospective samples, which were classified into high-level HF group (higher than 10%) and low-level HF group (lower than 10%). Most of these heteroplasmic variants were found to localize in the D-LOOP and non-coding region of mtDNA (Fig. 4A), while similar distribution patterns were shown in both high-level HF group and low-level HF group (Fig. 4B), which are both comparable to previous report [26]. Furthermore, the ratio of low-level HF variants in different regions of mitochondrial genome revealed highly consistent, accounting for 74.6% to 91.7% (Fig. 4C). These results also implied the low-level HF variants identified by Pime-Seq are unlikely to be sequencing artifacts, which are expected to distribute the mitochondrial genome uniformly. Interestingly, we found 65.4% (140/214) variants with low-level HF in mtDNA protein gene coding region were non-synonymous variants, while only 37.8% (17/45) variants in high HF group were non-synonymous (Fig. 4D). A possible explanation for this interesting phenomenon is that non-synonymous variants with high-level HF could become extremely detrimental and would be eliminated by selective pressure, suggesting distinct characters with these variants with low-level HF. The detail information of all the 453 heteroplasmic variants can be found in Additional file 7: Table S7. The results from Pime-Seq in this cohort revealed characters of human mtDNA variants in a comprehensive way, which would potentially advance our knowledge of human mitochondrial genome.

Fig. 4
figure 4

Analysis of the 453 heteroplasmic variants called by Pime-Seq in the 192 prospective samples. A Overall distribution of the 453 heteroplasmic variants on the mitochondrial genome; B The distribution of the variants with low-level HF and high-level HF; C The ratio of variants with low-level HF to high-level HF in different mtDNA locations; D The ratio of synonymous variants to non-synonymous in variants with low-level HF and high-level HF

Discussion

Pime-Seq is a novel method for mtDNA deep-sequencing. The key of Pime-Seq strategy is the mtDNA enrichment achieved by mitochondrial isolation, which has revealed satisfactory performance when applied to human PBMC samples. Comparing to other PCR dependent methods, Pime-Seq can achieve a high ratio of mtDNA fragments in the sequencing library through a PCR-independent manner, minimizing the impact of NUMTs contamination, which avoids false positive variants or heteroplasmy shifts introduced by PCR amplification. Additionally, our PCR-independent mtDNA enrichment ensures high sequencing depth of the mitochondrial genome at a relatively low cost and short turn-around time, making it beneficial for large-scale screening or population-wide studies. Similarly, several recent studies have attempted to develop a PCR-independent mtDNA sequencing method to avoid amplification errors and bias introduced by PCR pre-amplification. Legati et al. utilized Mitochondrial DNA Isolation Kit (Abcam, Cambridge, UK) for mtDNA enrichment in their deep-sequencing protocol, resulting in MT reads percentage ranging from 0.3% to 6.8% [27]. Another study by Keraite et al. reported the use of RNA-guided DNA endonuclease Cas9 to specifically enrich the linearized mitochondrial genome for sequencing on long-read sequencing platforms, achieving an average of 54% full-length mtDNA reads when applying on 4 cell lines (HEK293, A549, Capan-2, SH-SY5Y) [28]. Walsh et al. described their PCR-free method to yield purified mtDNA for NGS using differential centrifugation and alkaline lysis, which achieved an average of 75 ± 20% MT reads on fresh mouse tissue. However, this method has not been tested using human cells, and the authors mentioned they were not able to yield adequate mtDNA enrichment using commercial mtDNA enrichment methods on human cells [29]. In comparison, our results showed that Pime-Seq can achieve an average of 88.0% MT reads when applied to human PBMC samples, demonstrating its potential for accurate mtDNA sequencing on human samples with highly enriched mtDNA libraries.

Currently, prenatal screening mainly focused in detecting fetal common aneuploidies, including trisomy 21, trisomy 18, and trisomy 13. Prenatal cell-free DNA screening, one of the most successful clinical applications of NGS, has an excellent performance for this purpose and is broadly adopted worldwide [30]. However, prenatal screening strategy targeting severe genetic disease types other than aneuploidies are limited. In our study, we evaluated the clinical incorporation of Pime-Seq into prenatal screening using a cohort of 192 pregnant women, which revealed a detection rate of 2% (4/192) for pathogenic mtDNA variants. Due to the fact these pathogenic variants would be passed to the fetuses, possibly with increased heteroplasmic level [31, 32], it would be useful for the couple to obtain this information for appropriate genetic counseling and corresponding health monitoring. Furthermore, Pime-Seq and prenatal cell-free DNA screening used PBMC and plasma respectively, which allows one blood draw for both tests. In addition, as the library preparation of Pime-Seq could take as short as 5 h, the total turn-around time could be managed within 7 days, which is similar or shorter than that of prenatal cell-free DNA screening. In this way, our initial assessment revealed the incorporation of Pime-Seq in prenatal screening for pathogenic mtDNA variants would act as an important supplement for current prenatal screening strategy.

It is important to detect heteroplasmic mitochondrial variants with a high sensitivity and accuracy for PMDs, including clinical diagnosis, genetic causes, prevalence, and mtDNA editing evaluation [33,34,35,36]. In addition, detection of the heteroplasmic variants can contribute in cancer research [37], population genetics [38], and forensic genetics [39]. Currently, the commercially available lrPCR-NGS is widely used for mtDNA sequencing, but the detection efficacy for variants with low-level HF is limited, and variants with HF lower than a threshold, typically 5% to 10%, are often overlooked due to substantial false-positive calls [26, 40,41,42]. As Pime-Seq based on PCR independent mtDNA enrichment, it can theoretically achieve an advanced performance when calling heteroplasmic variants with low-level HF, which is also supported by our comparison data. In addition, our initial results showed that variants with low-level HF may have unique characters, such as high ratio of non-synonymous variants. These previously overlooked variants may not be able to accumulate to high-level HF due to their harmfulness to critical respiratory chain proteins and cell survival, and investigation into these variants could potentially lead to discover new variants with extreme pathogenicity with lower HF threshold. Therefore, the reliable detection of variants with low-level HF may provide an informative research angle for mtDNA studies.

It is widely accepted that the mitochondria evolved from α-proteobacterium to form endosymbiosis in the eukaryote history. During this prokaryote-to-eukaryote transition process, the gene-transfer from organelles to the nucleus took place progressively to shape the mitochondrial genome and the nuclear genome, leading to the creation of a number of nuclear copies of pseudo mitochondrial genes, NUMTs [43]. Over 500 NUMTs have been recorded in human reference genome [44], ranging from 39 bp to the entire mitochondrial genome [45], and are found to have population polymorphism and variation among human siblings [46, 47]. These features of NUMTs can lead to serious challenges in identification of pathogenic mtDNA variants [48, 49]. Multiple studies provided evidence that the presence of NUMTs can lead to mis-reported mitochondrial variants when using probe hybridization or PCR amplification to enrich the mtDNA [50,51,52]. It is worth noting that there have been some interesting reports of paternal mtDNA inheritance [53], but this phenomenon has yet to be confirmed as it may be due to ‘autosomal dominant-like inheritance mode’ actually derived from nuclear elements of mtDNA (NUMTs) [54]. In this way, Pime-Seq may provide an important platform for accurate mtDNA sequencing, even in the presence of NUMTs.

It is clinically critical to choose proper tissue types for genetic testing during PMD diagnosis and management. According to consensus statement from the Mitochondrial Medicine Society and UK best practice guidelines, blood and/or urine DNA are typically acting as the first sample types for genetic analyses, and invasive samples obtained from a diagnostic tissue biopsy are also preferred and informative [36, 55]. Pime-Seq requires to use alive cell samples to prevent degradation of the nuclear genome, and tissues are theoretically suitable when most cells are alive. In this study, we investigated the performance of Pime-Seq on blood samples, as well as a small number of urine samples. We found Pime-Seq on 40,000 PBMCs, isolated from 400 μL to 1 mL fresh peripheral blood, can achieve reliable results. However, although Pime-Seq was able to generate results from 60 mL urine samples with current procedure, the mtDNA enrichment efficiency was less ideal than that on blood samples, possibly due to the high proportion of dead cells, indicating the need for further optimization with regard to urine samples. In addition, we haven’t tested Pime-Seq on clinically relevant tissues such as muscle biopsy, and the performance of Pime-Seq on these precious samples would be investigated in the future. Furthermore, it is also beneficial to apply Pime-Seq in detecting mtDNA copy number variations (CNV) such as single large-scale deletions, but the reliability of CNV results called by Pime-Seq still needs to be validated using mtDNA CNV positive samples, which would be conducted in our future study.

In conclusion, our study here presents a novel low-cost and concise approach, Pime-Seq, that achieves high sensitivity and specificity in analyzing the whole mitochondrial genome on human PBMC samples. Compared to the traditional methods with PCR-based mtDNA enrichment approach, Pime-Seq showed advanced accuracy, especially in identifying variants with low HF. Our study provides evidence that Pime-Seq has a good potential for clinical applications in detecting pathogenic mtDNA variants, particularly in the field of prenatal screening.

Availability of data and materials

The datasets analyzed during the current study are available from the corresponding author on reasonable request.

References

  1. Suen DF, Norris KL, Youle RJ. Mitochondrial dynamics and apoptosis. Genes Dev. 2008;22:1577–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Mishra P, Chan DC. Metabolic regulation of mitochondrial dynamics. J Cell Biol. 2016;212:379–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Anderson S, Bankier AT, Barrell BG, de Bruijn MH, Coulson AR, Drouin J, Eperon IC, Nierlich DP, Roe BA, Sanger F, et al. Sequence and organization of the human mitochondrial genome. Nature. 1981;290:457–65.

    Article  CAS  PubMed  Google Scholar 

  4. Andrews RM, Kubacka I, Chinnery PF, Lightowlers RN, Turnbull DM, Howell N. Reanalysis and revision of the Cambridge reference sequence for human mitochondrial DNA. Nat Genet. 1999;23:147.

    Article  CAS  PubMed  Google Scholar 

  5. Kelly RD, Mahmud A, McKenzie M, Trounce IA, St John JC. Mitochondrial DNA copy number is regulated in a tissue specific manner by DNA methylation of the nuclear-encoded DNA polymerase gamma A. Nucleic Acids Res. 2012;40:10124–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Stefano GB, Bjenning C, Wang F, Wang N, Kream RM. Mitochondrial Heteroplasmy. Adv Exp Med Biol. 2017;982:577–94.

    Article  CAS  PubMed  Google Scholar 

  7. Ghaoui R, Sue CM. Movement disorders in mitochondrial disease. J Neurol. 2018;265:1230–40.

    Article  CAS  PubMed  Google Scholar 

  8. Chinnery PF, Elliott HR, Hudson G, Samuels DC, Relton CL. Epigenetics, epidemiology and mitochondrial DNA diseases. Int J Epidemiol. 2012;41:177–87.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Gorman GS, Schaefer AM, Ng Y, Gomez N, Blakely EL, Alston CL, Feeney C, Horvath R, Yu-Wai-Man P, Chinnery PF, et al. Prevalence of nuclear and mitochondrial DNA mutations related to adult mitochondrial disease. Ann Neurol. 2015;77:753–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Pagliarini DJ, Calvo SE, Chang B, Sheth SA, Vafai SB, Ong SE, Walford GA, Sugiana C, Boneh A, Chen WK, et al. A mitochondrial protein compendium elucidates complex I disease biology. Cell. 2008;134:112–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Gorman GS, Chinnery PF, DiMauro S, Hirano M, Koga Y, McFarland R, Suomalainen A, Thorburn DR, Zeviani M, Turnbull DM. Mitochondrial diseases. Nat Rev Dis Primers. 2016;2:16080.

    Article  PubMed  Google Scholar 

  12. Klopstock T, Priglinger C, Yilmaz A, Kornblum C, Distelmaier F, Prokisch H. Mitochondrial Disorders. Dtsch Arztebl Int. 2021;118:741–8.

    PubMed  PubMed Central  Google Scholar 

  13. Rohlin A, Wernersson J, Engwall Y, Wiklund L, Björk J, Nordling M. Parallel sequencing used in detection of mosaic mutations: comparison with four diagnostic DNA screening techniques. Hum Mutat. 2009;30:1012–20.

    Article  CAS  PubMed  Google Scholar 

  14. Cui H, Li F, Chen D, Wang G, Truong CK, Enns GM, Graham B, Milone M, Landsverk ML, Wang J, et al. Comprehensive next-generation sequence analyses of the entire mitochondrial genome reveal new insights into the molecular diagnosis of mitochondrial DNA disorders. Genet Med. 2013;15:388–94.

    Article  CAS  PubMed  Google Scholar 

  15. Fendt L, Zimmermann B, Daniaux M, Parson W. Sequencing strategy for the whole mitochondrial genome resulting in high quality sequences. BMC Genomics. 2009;10:139.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Zhang W, Cui H, Wong LJ. Comprehensive one-step molecular analyses of mitochondrial genome by massively parallel sequencing. Clin Chem. 2012;58:1322–31.

    Article  CAS  PubMed  Google Scholar 

  17. Wang J, Balciuniene J, Diaz-Miranda MA, McCormick EM, Aref-Eshghi E, Muir AM, Cao K, Troiani J, Moseley A, Fan Z, et al. Advanced approach for comprehensive mtDNA genome testing in mitochondrial disease. Mol Genet Metab. 2022;135:93–101.

    Article  CAS  PubMed  Google Scholar 

  18. Duan M, Tu J, Lu Z: Recent advances in detecting mitochondrial DNA heteroplasmic variations. Molecules 2018, 23.

  19. Zhou K, Mo Q, Guo S, Liu Y, Yin C, Ji X, Guo X, Xing J. A novel next-generation sequencing-based approach for concurrent detection of mitochondrial DNA copy number and mutation. J Mol Diagn. 2020;22:1408–18.

    Article  CAS  PubMed  Google Scholar 

  20. He Y, Wu J, Dressman DC, Iacobuzio-Donahue C, Markowitz SD, Velculescu VE, Diaz LA Jr, Kinzler KW, Vogelstein B, Papadopoulos N. Heteroplasmic mitochondrial DNA mutations in normal and tumour cells. Nature. 2010;464:610–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Duan M, Chen L, Ge Q, Lu N, Li J, Pan X, Qiao Y, Tu J, Lu Z. Evaluating heteroplasmic variations of the mitochondrial genome from whole genome sequencing data. Gene. 2019;699:145–54.

    Article  CAS  PubMed  Google Scholar 

  22. Lareau CA, Ludwig LS, Muus C, Gohil SH, Zhao T, Chiang Z, Pelka K, Verboon JM, Luo W, Christian E, et al. Massively parallel single-cell mitochondrial DNA genotyping and chromatin profiling. Nat Biotechnol. 2021;39:451–61.

    Article  CAS  PubMed  Google Scholar 

  23. Santibanez-Koref M, Griffin H, Turnbull DM, Chinnery PF, Herbert M, Hudson G. Assessing mitochondrial heteroplasmy using next generation sequencing: a note of caution. Mitochondrion. 2019;46:302–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Salas A, Schönherr S, Bandelt HJ, Gómez-Carballa A, Weissensteiner H. Extraordinary claims require extraordinary evidence in asserted mtDNA biparental inheritance. Forensic Sci Int Genet. 2020;47: 102274.

    Article  CAS  PubMed  Google Scholar 

  25. Corces MR, Trevino AE, Hamilton EG, Greenside PG, Sinnott-Armstrong NA, Vesuna S, Satpathy AT, Rubin AJ, Montine KS, Wu B, et al. An improved ATAC-seq protocol reduces background and enables interrogation of frozen tissues. Nat Methods. 2017;14:959–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Laricchia KM, Lake NJ, Watts NA, Shand M, Haessly A, Gauthier L, Benjamin D, Banks E, Soto J, Garimella K, et al. Mitochondrial DNA variation across 56,434 individuals in gnomAD. Genome Res. 2022;32:569–82.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Legati A, Zanetti N, Nasca A, Peron C, Lamperti C, Lamantea E, Ghezzi D. Current and New Next-Generation Sequencing Approaches to Study Mitochondrial DNA. J Mol Diagn. 2021;23:732–41.

    Article  CAS  PubMed  Google Scholar 

  28. Keraite I, Becker P, Canevazzi D, Frias-Lopez C, Dabad M, Tonda-Hernandez R, Paramonov I, Ingham MJ, Brun-Heath I, Leno J, et al. A method for multiplexed full-length single-molecule sequencing of the human mitochondrial genome. Nat Commun. 2022;13:5902.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Walsh DJ, Bernard DJ, Pangilinan F, Esposito M, Harold D, Parle-McDermott A, Brody LC. Mito-SiPE is a sequence-independent and PCR-free mtDNA enrichment method for accurate ultra-deep mitochondrial sequencing. Commun Biol. 2022;5:1269.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Liang D, Lin Y, Qiao F, Li H, Wang Y, Zhang J, Liu A, Ji X, Ma D, Jiang T, et al. Perinatal outcomes following cell-free DNA screening in >32 000 women: clinical follow-up data from a single tertiary center. Prenat Diagn. 2018;38:755–64.

    Article  CAS  PubMed  Google Scholar 

  31. Wei W, Tuna S, Keogh MJ, Smith KR, Aitman TJ, Beales PL, Bennett DL, Gale DP, Bitner-Glindzicz MAK, Black GC, et al: Germline selection shapes human mitochondrial DNA diversity. Science 2019, 364.

  32. Steffann J, Monnot S, Magen M, Assouline Z, Gigarel N, Ville Y, Salomon L, Bessiere B, Martinovic J, Rotig A, et al. A retrospective study on the efficacy of prenatal diagnosis for pregnancies at risk of mitochondrial DNA disorders. Genet Med. 2020. https://doi.org/10.1038/s41436-020-01043-3.

    Article  PubMed  Google Scholar 

  33. Bacman SR, Kauppila JHK, Pereira CV, Nissanka N, Miranda M, Pinto M, Williams SL, Larsson NG, Stewart JB, Moraes CT. MitoTALEN reduces mutant mtDNA load and restores tRNA(Ala) levels in a mouse model of heteroplasmic mtDNA mutation. Nat Med. 2018;24:1696–700.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Zaidi AA, Wilton PR, Su MS, Paul IM, Arbeithuber B, Anthony K, Nekrutenko A, Nielsen R, Makova KD. Bottleneck and selection in the germline and maternal age influence transmission of mitochondrial DNA in human pedigrees. Proc Natl Acad Sci U S A. 2019;116:25172–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Chen X, Liang D, Guo J, Zhang J, Sun H, Zhang X, Jin J, Dai Y, Bao Q, Qian X, et al. DdCBE-mediated mitochondrial base editing in human 3PN embryos. Cell Discov. 2022;8:8.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Parikh S, Goldstein A, Koenig MK, Scaglia F, Enns GM, Saneto R, Anselm I, Cohen BH, Falk MJ, Greene C, et al. Diagnosis and management of mitochondrial disease: a consensus statement from the Mitochondrial Medicine Society. Genet Med. 2015;17:689–701.

    Article  CAS  PubMed  Google Scholar 

  37. Yuan Y, Ju YS, Kim Y, Li J, Wang Y, Yoon CJ, Yang Y, Martincorena I, Creighton CJ, Weinstein JN, et al. Comprehensive molecular characterization of mitochondrial genomes in human cancers. Nat Genet. 2020;52:342–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Guo Y, Li CI, Sheng Q, Winther JF, Cai Q, Boice JD, Shyr Y. Very low-level heteroplasmy mtDNA variations are inherited in humans. J Genet Genomics. 2013;40:607–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Ivanov PL, Wadhams MJ, Roby RK, Holland MM, Weedn VW, Parsons TJ. Mitochondrial DNA sequence heteroplasmy in the Grand Duke of Russia Georgij Romanov establishes the authenticity of the remains of Tsar Nicholas II. Nat Genet. 1996;12:417–20.

    Article  CAS  PubMed  Google Scholar 

  40. Calabrese C, Pyle A, Griffin H, Coxhead J, Hussain R, Braund PS, Li L, Burgess A, Munroe PB, Little L, et al. Heteroplasmic mitochondrial DNA variants in cardiovascular diseases. PLoS Genet. 2022;18: e1010068.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Just RS, Irwin JA, Parson W. Mitochondrial DNA heteroplasmy in the emerging field of massively parallel sequencing. Forensic Sci Int Genet. 2015;18:131–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Chen R, Aldred MA, Xu W, Zein J, Bazeley P, Comhair SAA, Meyers DA, Bleecker ER, Liu C, Erzurum SC, et al. Comparison of whole genome sequencing and targeted sequencing for mitochondrial DNA. Mitochondrion. 2021;58:303–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Hazkani-Covo E, Zeller RM, Martin W. Molecular poltergeists: mitochondrial DNA copies (numts) in sequenced nuclear genomes. PLoS Genet. 2010;6: e1000834.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Calabrese FM, Simone D, Attimonelli M. Primates and mouse NumtS in the UCSC Genome Browser. BMC Bioinformatics. 2012;13(Suppl 4):S15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Blanchard JL, Schmidt GW. Mitochondrial DNA migration events in yeast and humans: integration by a common end-joining mechanism and alternative perspectives on nucleotide substitution patterns. Mol Biol Evol. 1996;13:537–48.

    Article  CAS  PubMed  Google Scholar 

  46. Yuan JD, Shi JX, Meng GX, An LG, Hu GX. Nuclear pseudogenes of mitochondrial DNA as a variable part of the human genome. Cell Res. 1999;9:281–90.

    Article  CAS  PubMed  Google Scholar 

  47. Wei W, Schon KR, Elgar G, Orioli A, Tanguy M, Giess A, Tischkowitz M, Caulfield MJ, Chinnery PF. Nuclear-embedded mitochondrial DNA sequences in 66,083 human genomes. Nature. 2022;611:105–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Yao YG, Kong QP, Salas A, Bandelt HJ. Pseudomitochondrial genome haunts disease studies. J Med Genet. 2008;45:769–72.

    Article  CAS  PubMed  Google Scholar 

  49. Wallace DC, Stugard C, Murdock D, Schurr T, Brown MD. Ancient mtDNA sequences in the human nuclear genome: a potential source of errors in identifying pathogenic mutations. Proc Natl Acad Sci U S A. 1997;94:14900–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Wang D, Xiang H, Ning C, Liu H, Liu JF, Zhao X. Mitochondrial DNA enrichment reduced NUMT contamination in porcine NGS analyses. Brief Bioinform. 2020;21:1368–77.

    Article  CAS  PubMed  Google Scholar 

  51. Calvignac S, Konecny L, Malard F, Douady CJ. Preventing the pollution of mitochondrial datasets with nuclear mitochondrial paralogs (numts). Mitochondrion. 2011;11:246–54.

    Article  CAS  PubMed  Google Scholar 

  52. Goios A, Prieto L, Amorim A, Pereira L. Specificity of mtDNA-directed PCR-influence of NUclear MTDNA insertion (NUMT) contamination in routine samples and techniques. Int J Legal Med. 2008;122:341–5.

    Article  PubMed  Google Scholar 

  53. Luo S, Valencia CA, Zhang J, Lee NC, Slone J, Gui B, Wang X, Li Z, Dell S, Brown J, et al. Biparental Inheritance of Mitochondrial DNA in Humans. Proc Natl Acad Sci U S A. 2018;115:13039–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Lutz-Bonengel S, Parson W. No further evidence for paternal leakage of mitochondrial DNA in humans yet. Proc Natl Acad Sci U S A. 2019;116:1821–2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Mavraki E, Labrum R, Sergeant K, Alston CL, Woodward C, Smith C, Knowles CVY, Patel Y, Hodsdon P, Baines JP, et al. Genetic testing for mitochondrial disease: the United Kingdom best practice guidelines. Eur J Hum Genet. 2023;31:148–63.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by the National Key Research and Development Program of China 2022YFC2703400 (Z. X.), 2022YFC2702705 (J. Z.), the National Natural Science Foundation of China 32371546 (B. S.), 31871445 (J. Z.), and the Jiangsu Province Capability Improvement Project through Science, Technology and Education Jiangsu Provincial Medical Key Discipline ZDXK202211 (Z. X.).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: DL, JZ, BS; Formal Analysis: DL, LZ, YZ, MH; Investigation: LZ, YZ; Project Administration: ZX; Resources: YL, HL, PH; Writing: DL, JZ, BS, ZX.

Corresponding authors

Correspondence to Jun Zhang, Bin Shen or Zhengfeng Xu.

Ethics declarations

Ethics approval and consent to participate

This study was ethically approved by the Medical Ethics Committee of Nanjing Maternal and Child Health Care Hospital, reference number 2021KY-130. All participants signed the informed consent, in accordance with the principles of Declaration of Helsinki.

Consent for publication

All authors give their consent to publish this manuscript.

Competing interests

The authors declare that they have 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

Additional file 1: Table S1.

Primer information.

Additional file 2: Table S2.

Information of the sequencing reads of the 20 PBMC samples generated by Pime-Seq.

Additional file 3: Table S3.

Information of the sequencing reads of the 3 urine samples generated by Pime-Seq.

Additional file 4: Table S4.

All variants called by Pime-Seq in 3 technical repeats on 3 human PBMC samples.

Additional file 5: Table S5.

Variants called by Pime-Seq and lrPCR-NGS in 45 samples.

Additional file 6: Table S6.

A total of 146 discordant variants detected in 45 samples by Pime-Seq and lrPCR-NGS.

Additional file 7: Table S7.

A total of 453 heteroplasmic variants called in 192 prospective samples.

Additional file 8: Figure S1.

Examining the nuclear DNA and mtDNA post mtDNA enrichment to yield an optimal enrichment condition.

Additional file 9: Figure S2.

Pyrosequencing result for m.9540T > C in two negative control samples.

Additional file 10: Figure S3.

Pime-Seq results and Sanger sequencing verification in the 4 previously diagnosed PMD patients.

Additional file 11: Figure S4.

Pime-Seq results and Sanger sequencing verification of the 4 pathogenic mtDNA variants identified from the 192 apparently healthy pregnant women.

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

Liang, D., Zhu, L., Zhu, Y. et al. A PCR-independent approach for mtDNA enrichment and next-generation sequencing: comprehensive evaluation and clinical application. J Transl Med 22, 386 (2024). https://doi.org/10.1186/s12967-024-05213-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12967-024-05213-8

Keywords