Abstract
Mitochondrial DNA copy number (mtDNA-CN) is a biomarker of mitochondrial function and levels of mtDNA-CN have been reproducibly associated with overall mortality and a number of age-related diseases, including cardiovascular disease, chronic kidney disease, and cancer. Recent advancements in techniques for estimating mtDNA-CN, in particular the use of DNA microarrays and next-generation sequencing data, have led to the comprehensive assessment of mtDNA-CN across these and other diseases and traits. The importance of mtDNA-CN measures to disease and these advancing technologies suggest the potential for mtDNA-CN to be a useful biomarker in the clinic. While the exact mechanism(s) underlying the association of mtDNA-CN with disease remain to be elucidated, we review the existing literature which supports roles for inflammatory dynamics, immune function and alterations to cell signaling as consequences of variation in mtDNA-CN. We propose that future studies should focus on characterizing longitudinal, cell-type and cross-tissue profiles of mtDNA-CN as well as improving methods for measuring mtDNA-CN which will expand the potential for its use as a clinical biomarker.
Keywords: mitochondrial DNA, clinical biomarker, mtDNA, complex disease
Introduction
The mitochondrion is a double membraned organelle that produces over 90% of the cell’s chemical energy via respiration. Mitochondria have essential roles in the production of adenosine triphosphate (ATP), in the regulation of bioenergetic processing, in multiple homeostatic, apoptotic and signaling pathways, (Wallace, 1992) and in the biosynthesis of macromolecules, such as nucleotides, lipids, heme and iron-sulfur clusters (Vakifahmetoglu-Norberg et al., 2017).
Mitochondrial dysfunction, generally characterized as a loss of efficiency in oxidative phosphorylation, is a hallmark of aging and a variety of chronic diseases (Nicolson, 2014). Mitochondrial dysfunction results in inefficient cellular energy production and in increased levels of reactive oxygen species (ROS) which may damage lipids, proteins and nucleic acids (Guyatt et al., 2017). Mitochondrial dysfunction also affects the expression of nuclear genes involved in metabolism, growth, differentiation, and apoptosis (Pello et al., 2008). All these changes may explain the contribution of mitochondrial dysfunction to chronic and complex human diseases.
A major limitation to the routine evaluation of mitochondrial dysfunction in clinical practice is the lack of reliable measures of mitochondrial dysfunction available for clinical use. Mitochondrial DNA copy number (mtDNA-CN) is a promising biomarker of mitochondrial dysfunction that has the potential to become widely available in clinical practice. Other measures of mitochondrial dysfunction, including cell culture-based methods such as the Seahorse XF Cell Mito Stress Test (Agilent, USA), are optimized in vitro, do not make use of pre-existing datasets and cannot be scaled-up for widespread use. In this review, we describe the rationale for using mtDNA-CN as a biomarker of mitochondrial dysfunction, the issues associated with its measurement and interpretation in clinical and population studies, and the evidence linking variation in mtDNA-CN to human health disorders, including cardiovascular disease (Ashar et al., 2017; Dai et al., 2012), chronic kidney disease (Tin et al., 2016), cancer (Taylor and Turnbull, 2005), and all-cause mortality (Ashar et al., 2015; Mengel-From et al., 2014). mtDNA-CN is also modified in patients with primary mitochondrial disease, often due to mutations in genes involved in mtDNA integrity (Grady et al., 2018). Increases in mtDNA-CN in primary mitochondrial diseases may represent a compensatory response to attenuate the effect of the primary mtDNA mutation (Bianco et al., 2018). The identity and mechanisms behind mtDNA-CN variation in primary mitochondrial diseases are not discussed in this review.
The mitochondrial genome
The mitochondrial genome (mtDNA) is a circular, intron-free, double-stranded, haploid DNA strand 16.5 kb in length that encodes 37 genes (Raffoul et al., 2012). The organization of the mammalian mitochondrial genome is highly conserved (Clayton, 1992). The human mtDNA encodes 13 essential genes involved in the oxidative phosphorylation system, while the remaining genes play essential roles in assembling amino acids into functional proteins.
The growth, movement, division and fusion of mitochondria is highly regulated (Yamada et al., 2016). Division of mitochondria is important for proper organelle functioning, distribution and turnover (Yamada et al., 2016). The number of mitochondria in each cell changes due to fission and fusion of mitochondria, two processes that play a key role in maintaining mitochondrial function. Since the total number of mitochondria in each cell is difficult to determine and does not fully reflect functional capacity, we must rely on proxy measures such as mtDNA-CN.
Replication of mtDNA is regulated by the D-loop, (Nicholls and Minczuk, 2014) a non-coding region within mtDNA, and occurs independently from nuclear DNA replication, although many factors associated with mtDNA replication are encoded by nuclear DNA (Bogenhagen and Clayton, 1977). In contrast to the nuclear genome, which contains only two copies per cell, the mitochondrial genome is present in multiple copies per cell (from 100 to 10,000), depending on cell type (Wai et al., 2010). High ATP requirement cells such as heart and skeletal muscle cells have as high as ~7,000 mtDNA copies per cell, while low energy requirement cells such as spleen and liver cells have as low as ~100 copies per cell (Chabi et al., 2003; Kelly et al., 2012; Miller et al., 2003).
mtDNA-CN as a biomarker of mitochondrial function
Levels of mtDNA-CN are directly correlated with energy reserves, oxidative stress, and changes in mitochondrial membrane potential (Guha and Avadhani, 2013). The relationship of mtDNA-CN to mitochondrial function was evidenced in knockdown cell models of reduced mtDNA-CN, which resulted in reduced expression of vital complex proteins, altered cellular morphology, and lower respiratory enzyme activity (Jeng et al., 2008). In these models, mitochondrial function was rescued after restoring mtDNA-CN to wild-type levels. mtDNA-CN levels are dependent on the maintenance of mitochondrial genome stability via proper mitochondrial translation, as evidenced by reduction of mtDNA-CN due to dysfunction in the synthesis of mitochondrial ribosomal protein components (Zhang and Singh, 2014). Further, although mitochondrial fission is coordinated with DNA replication, it does not seem to influence mtDNA-CN levels (Ishihara et al., 2009). In contrast, fusion is critical for mtDNA-CN maintenance (Chen et al., 2010; Y. Chen et al., 2011; Silva Ramos et al., 2019). Taken together, these observations highlight that reduced mtDNA-CN serves as a useful biomarker of mitochondrial dysfunction that can be measured in clinical and population studies.
Measurement of mtDNA-CN
mtDNA-CN levels can be readily measured in extracted DNA from peripheral blood or other tissues, although in practice most measurements are performed in peripheral blood due to ease of accessibility. Measurements of mtDNA-CN in blood have been shown to be relevant to a variety of chronic diseases (Ashar et al., 2017, 2015; Chen et al., 2014; Tin et al., 2016; Yiyi Zhang et al., 2017). However, when available, mtDNA-CN should be measured directly in the tissue of interest.
Until recently, quantitative PCR (qPCR) has been widely considered the gold standard for measuring mtDNA-CN. The standard protocol consists of a multiplexed Taqman assay in which the number of copies of a mitochondrial gene is compared to the number of copies of a nuclear gene to achieve a relative measure of mtDNA-CN (Ashar et al., 2015). Numerous target genes have been used successfully including, but not limited to, MT-ND1, MT-ND4, MT-CYB, and MT-TL1 from the mitochondrial genome, and B2M, RPLPO, ACTB, and RPPH1 for the nuclear genome (Ashar et al., 2015; Bai and Wong, 2005; Dimmock et al., 2010; Gu et al., 2013; Jędrak et al., 2017; Kamfar et al., 2016; Knez et al., 2016). Recently, digital PCR (dPCR) has emerged as an additional method for calculating mtDNA-CN particularly due to its ability to quantify absolute copy number by determining the ratio of the number of positive mitochondrial probe copies to the number of nuclear probe copies (Li et al., 2018; Memon et al., 2017; O’Hara et al., 2019; Ye et al., 2017). mtDNA-CN levels can also be measured from pre-existing genotyping microarray, whole exome, and whole genome sequence data (Table 1) (Ding et al., 2015; MitoPipeline, 2018; Qian et al., 2017).
Table 1.
mtDNA-CN estimation methods
| Platform | qPCR | digitalPCR | Microarray | Sequencing |
|---|---|---|---|---|
| mtDNA-CN Measurement | Ct mitochondrial probe - Ct nuclear probe | Ratio of number of positive mitochondrial probe copies/uL to the number of nuclear probe copies/uL | Median mitochondrial probe intensity adjusted for PCs generated from nuclear probes | (Mitochondrial DNA coverage divided by autosomal coverage)* 2 |
| Absolute measure of mtDNA-CN | No | Yes | No | No |
| Number of mtDNA loci interrogated | 1–2 | 1–2 | ~25–150 | Whole Mitochondrial Genome |
| Cost | $ | $$ | $$$$ | $$$$$ |
| Detect mtDNA deletions | Yes, major deletions with additional probe | Yes, major deletions with additional probe | No | Yes |
| Detect Single Base-Pair Heteroplasmy | No | No | No | Yes |
| Haplogroup determination | No | No | No | Yes |
PCs: Principal Components; Ct: Cycle threshold; qPCR: Quantitative PCR; mtDNA: Mitochondrial DNA; mtDNA-CN: Mitochondrial DNA Copy Number.
Further, the DNA extraction method affects the accuracy of mtDNA-CN measurements. Specifically, organic solvent extraction (phenol-chloroform-isoamyl alcohol) provides a more consistent and accurate mtDNA-CN estimation compared to silica-based methods, (Guo et al., 2009) likely because column kit parameters are typically optimized for the isolation of DNA fragments ≥ 50 Kb leading to inadequate isolation of the smaller mtDNA genome (Nacheva et al., 2017). One solution to reduce biases introduced by DNA extraction may be to use direct measurements that avoid traditional DNA extraction, such as measuring mtDNA-CN from direct cell lysis, an approach that outperforms both organic solvent extraction and a column-based method (Longchamps et al., 2020). This method involves pelleting cells and removing the supernatant followed by agitation and heating in solution to disrupt the pellet and subsequent centrifugation to pellet out any insoluble inhibitors (Longchamps et al., 2020). Further, direct cell lysis provides a more cost-effective approach and is faster than traditional DNA extraction, although its use limits the utility of the sample for downstream applications.
While qPCR, dPCR, genotyping microarray, and sequence data reliably quantify mtDNA-CN, there are two major challenges to the widespread use of these measurements in clinical practice. First, other than dPCR, these methods do not produce absolute measures of mtDNA-CN, but relative measures with respect to the number of nuclear DNA copies in the sample. Second, these measurements are affected by batch effects which must be adjusted for prior to data interpretation. Because mtDNA-CN values are often relative values and there is between-batch variability, mtDNA-CN values cannot be directly compared across studies or when assays are performed over time in the same individuals. In addition, since the number of mitochondria varies by cell type, mtDNA-CN levels will depend on the cell composition of the sample. For peripheral blood, mtDNA-CN are positively correlated with platelet count and inversely correlated with white blood cell (WBC) count (Ashar et al., 2015; Hurtado-Roca et al., 2016; Knez et al., 2016; Tin et al., 2016). As a consequence, mtDNA-CN measurements need to be standardized by sex, age, platelet count and WBC count.
In addition to tissue-specificity, average mtDNA-CN levels in blood decrease with age after approximately age 50 and are on average higher in women relative to men (Ashar et al., 2015; Mengel-From et al., 2014). Although little is known about the precise mechanisms that lead to variation in mtDNA-CN, genetic and environmental contributions are hypothesized to interact to determine the number of mitochondria in cells. In a genome-wide association study (GWAS) of 10,442 Chinese women, two loci were associated with mtDNA-CN levels, one within the 3’ UTR of mitochondrial transcription factor A (TFAM) and the other within intron 1 of cyclin dependent kinase 6 (CDK6) (Cai et al., 2015). In addition to genetic factors, various chemicals and drugs are known to affect mtDNA-CN levels (Young, 2017).
Detecting large heteroplasmic deletions and single base-pair heteroplasmies
Mitochondrial heteroplasmy reflects the coexistence of multiple mitochondrial haplotypes in a single organism. The technical ease of multiplexing in qPCR and digital PCR has led to the development of several techniques which enable the simultaneous detection of large heteroplasmic deletions and mtDNA-CN quantification in a single assay (Bai and Wong, 2005; Phillips et al., 2014; Rygiel et al., 2015). Similarly, mtDNA deletion detection methods for next generation sequence data have been packaged into several useful bioinformatics pipelines (Bosworth et al., 2017; Goudenège et al., 2019). Low frequency single base-pair heteroplasmies have traditionally been difficult to detect due to the coverage necessary to eliminate the contribution of sequencing error. However, with sufficient coverage several pipelines such as mitoCaller and mtDNA-Server exist which enable users to detect heteroplasmies contributing to as little as 1% of mtDNA (Ding et al., 2015; Weissensteiner et al., 2016).
Molecular consequences of variation in mtDNA-CN
In vitro reductions in mtDNA-CN are associated with downregulation of mitochondrial transcription and decreased levels of proteins involved in oxidative phosphorylation, specifically ND1, CYTB and COX-I (Jeng et al., 2008). Furthermore, natural variation in mtDNA-CN in cancer tissues is highly correlated with expression levels of genes involved in cellular respiration and mitochondrial function (Reznik et al., 2016). The reduction in respiratory capacity induced by reduced mtDNA-CN levels may have important pathogenic consequences.
Several theories involving immune dysfunction, energy demands, inflammation and altered cell signaling may explain the mechanisms linking mitochondrial dysfunction in whole blood to chronic disease (Figure 1). Mitochondrial-mediated changes in oxidative capacity may lead to chronic inflammation via macrophage polarization. Macrophages can change sub-types based on the surrounding microenvironment, with TNF-α and IFN-γ signaling pro-inflammatory M1 macrophages and TGF-β and IL-10 signaling anti-inflammatory M2 macrophages (Martinez et al., 2008). Interestingly, M1 macrophages generate ATP through glycolysis while M2 macrophages use oxidative phosphorylation (Ravi et al., 2014; Rodríguez-Prados et al., 2010; Vats et al., 2006). In the presence of reduced mtDNA-CN, mitochondrial dysfunction may be the consequence of an insufficient number of mtDNA molecules to encode for proteins of the cellular respiration pathway. As a result, macrophages may no longer be able to switch to their anti-inflammatory M2 subtype, leading to a reduction in anti-inflammatory mediators that would assist in tissue repair and the resolution of inflammation.
Figure 1.

Summary of proposed relationship between mtDNA-CN and disease.
Lending further support to the macrophage hypothesis, many of the diseases associated with mtDNA-CN involve immune dysfunction. Cardiovascular disease (CVD), and more specifically the atherosclerotic processes which leads to CVD, involves a chronic inflammatory processes from initiation through progression and eventual thrombotic events (Libby et al., 2002). Similar to CVD, a chronic pro-inflammatory state directly contributes to morbidity and mortality in chronic kidney disease and liver disease (Czaja, 2014; Silverstein, 2009). Furthermore, immune dysfunction and microglial activation have been shown to be signature landmarks of neurodegenerative disorders such as Alzheimer’s disease and Parkinson’s disease (Amor et al., 2010; Morgan et al., 2012; Rodriguez and Kern, 2011).
Mitochondrial dysfunction may also affect nuclear gene expression and methylation patterns (Friis et al., 2014; Picard et al., 2014; Srinivasan et al., 2016). Mitochondrial retrograde signaling (signaling from the mitochondrion to the nucleus) affects nuclear gene expression through disruption of the mitochondrial membrane potential which in turn leads to global changes in nuclear gene expression (Guantes et al., 2015; Guha et al., 2016; Muir et al., 2016) via chromatin activation, regulation of transcriptional activity, alternative splicing and regulation of protein synthesis (Guantes et al., 2015). The impact of mtDNA-CN levels seem to be particularly strong for alternative splicing, modulating both the abundance and type of mRNAs through direct impact on RNA Pol II kinetics (Guantes et al., 2015). In the presence of a reduced number of mitochondria, the cell enters a state of energy crisis and experiences changes in nuclear gene expression that may increase the risk of chronic disease, cancer and/or premature aging (Guantes et al., 2015).
Finally, reduced mtDNA-CN levels has been linked to increased oxidative stress via increased production of ROS. In CVD, mtDNA-CN was associated with the activation of proatherogenic genes, atherogenesis, plaque instability, thrombosis via LDL oxidation, and altered electrophysiology (Aggarwal and Makielski, 2013; Berliner and Heinecke, 1996; Erusalimsky, 2009; Jain et al., 2013; Liu et al., 2010; Lo et al., 2005; Sánchez-Santos et al., 2018; Yu and Bennett, 2014). Mitochondrial-mediated oxidative stress and resulting hepatocyte apoptosis along with impaired fatty acid oxidation have long been implicated in liver disease (Cichoż-Lach and Michalak, 2014; Sookoian et al., 2010). Furthermore, mitochondrial-mediated oxidative stress is also involved in aging-related diseases and neurodegeneration, such as promoting the production of amyloid-β peptide (Aβ) in Alzheimer’s disease (AD) (Gandhi and Abramov, 2012; Kim et al., 2015). In fact, mitochondrial dysfunction has been established as a hallmark of the aging process (Tidwell et al., 2017) and lifespan studies in C.elegans suggest that mitochondrial-nuclear crosstalk is a major determinant of lifespan (Cristina et al., 2009).
mtDNA Copy Number and the Risk of Chronic Disease
Changes in mtDNA-CN precede disease onset, implicating mtDNA regulation in the development of a variety of chronic diseases (Table 2).
Table 2.
Studies reporting association with mtDNA-CN and risk of chronic disease
| Study | Setting | Study Design | Sample Size | Tissue | Method for mtdNA estimation | Endpoint | Results |
|---|---|---|---|---|---|---|---|
| Cardiovascular | |||||||
| Ashar et al (2017)(Asharet al., 2017) | Prospective | Population-based | 21,870(11,153 from ARIC, 4,830 from CHS, and 5,887 from MESA) | Buffy coat, peripheral leukocytes | ARIC/MESA: Array-based; CHS: qPCR | Coronary heart disease (CHD) | ↓ Decreased mtDNA-CN is associated with CHD(OR=1.29, 95% Cl, 1.24–1.33) |
| Chen et al (2014)(Chen et al., 2014) | Retrospective | Case-control | 378 CHD cases, 378 controls | Peripheral leukocytes | qPCR | ↓ Decreased mtDNA-CN is associated with CHD (OR=2.38, 95% Cl, 1.33–4.69) | |
| Ashar et al (2017)(Asharet al., 2017) | Prospective | Population-based | 21,870(11,153 from ARIC, 4,830 from CHS, and 5,887 from MESA) | Buffy coat, peripheral leukocytes | ARIC/MESA: Array-based; CHS: qPCR | Stroke | ↓ Decreased mtDNA-CN is associated with Stroke (OR= 1.11, 95% Cl, 1.06–1.16) |
| Zhang et al (2017)(Yiyi Zhang et al., 2017) | Prospective | Population-based | 11,093 participants | Buffy coat | Array-based | Sudden cardiac death (SCD) | ↓ Decreased mtDNA-CN is associated with SCD (HR: 2.24, 95% Cl, 1.58–3.19; P-trend <0.001) |
| Kidney | |||||||
| Tin et al (2016)(Tinetal., 2016) | Prospective | Population-based | 9,058 participants | Buffy coat | Array-based | Chronic kidney disease (CKD) | ↓ Decreased mtDNA-CN is associated with CKD (HR=0.65, 95% Cl, 0.56 to 0.75; PO.001) |
| Zhang et al (2017)(Yuheng Zhang et al., 2017) | Retrospective | Case-control | 109ESRD cases, 112 controls | Peripheral leukocytes | qPCR | End stage renal disease (ESRD) | 1 Decreased mtDNA-CN is associated with ESRD (2.85 ±3.01 versus 5.94 ± 5.63; P<0.001) |
| Zhang etal (2017)(Yuheng Zhang et al., 2017) | Retrospective | Case-control | 56 ESRD cases, 58 controls | Plasma | qPCR | ↑ Increased mtDNA-CN is associated with ESRD (6.97 ±1.16 versus 5.50 ± 0.93, P<0.001) | |
| Liver Disease | |||||||
| Sookoian et al (2010)(Sookoian etal., 2010) | Retrospective | Case-control | 63 NAFLD cases, 11 controls | Liver biopsy | qPCR | Non-alcoholic fatty liver disease (NAFLD) | ↓ Decreased mtDNA-CN is associated with NAFLD (PO.01) |
| Pirola et al (2015)(Pirola et al., 2015) | Retrospective | Case-control | 67 NAFLD cases, 23 controls | Liver biopsy | qPCR | ↓ Patients with NAFLD have a significantly lower liver mtDNA copy number (62.5 ±41.0), in comparison with controls (103.5 ±88.0) | |
| Neurodegenerative Disorders | |||||||
| Pyle et al (2016)(Pyle et al., 2016) | Retrospective | Case-control | 363 peripheral blood patient samples (262 controls), 151 substantia nigra pars compacta (SNpc) patient samples (33 controls) | Peripheral leukocytes, substantia nigra pars compacta | qPCR | Parkinson’s Disease (PD) | ↓ Decreased mtDNA-CN is associated with PD (Blood: P=2.86×10–4; Substantia nigra: P=4.Oxio-3) |
| Wei et al (2017)(W. Wei et al., 2017) | Retrospective | Case-control | 282 cases, 351 controls | Cerebellum, cerebral cortex | Exome sequencing | Alzheimer’s Disease (AD) | ↓ Decreased mtDNA-CN is associated with AD(P=2.85×10–7) |
| Wei et al (2017)(W. Wei et al.,2017) | Retrospective | Case-control | 181 cases, 351 controls | Cerebellum, cerebral cortex | Exome sequencing | Creutzfeldt-Jakob Disease (CJD) | ↓ Decreased mtDNA-CN is associated with CJD(P=3.34><10–7) |
| Cancer Risk | |||||||
| Xing et al (2008)(Xing et al., 2008) | Retrospective | Case-control | 260 cases, 281 controls | Peripheral leukocytes | qPCR | Kidney Cancer | ↓ Decreased mtDNA-CN is associated with Kidney Cancer (OR = |
| 1.53, 95% Cl, 1.07 to 2.19) | |||||||
| Thyagarajan et al (2012)(Thyagaraj anetal., 2012) | Retrospective | Case-control | 422 cases, 874 controls | Peripheral blood | qPCR | Colorectal Cancer | ↓⁄↑ Compared to 2nd quartile, OR (95% Cl) for subjects in the lowest and highest quartiles of relative mtDNA copy numbers were 1.81 (1.13–2.89) and 3.40 (2.15–5.36), respectively (Pcurvilinearity<0.0001) |
| Lynch etal (2011)(Lynchet al., 2011) | Retrospective | Case-control | 203 cases, 656 controls | Whole blood | qPCR | Pancreatic Cancer | ↑ Increased mtDNA-CN is associated with pancreatic cancer (OR=1.14, 95% Cl, 1.06–1.23) |
OR: Odds Ratio; CI: Confidence Interval; HR: Hazard Ratio; qPCR: Quantitative PCR; mtDNA: Mitochondrial DNA; mtDNA-CN: Mitochondrial DNA Copy Number; ARIC: Atherosclerosis Risk in Communities Study; MESA: Multi-Ethnic Study of Atherosclerosis; CHS: Cardiovascular Health Study; CHD: Coronary heart disease; SCD: Sudden Cardiac Death; CKD: Chronic Kidney Disease; ESRD: End stage renal disease; NAFLD: Non-alcoholic fatty liver disease; PD: Parkinson’s Disease; AD: Alzheimer’s Disease; CJD: Creutzfeldt-Jakob Disease.
Cardiovascular disease
In a large pooled analysis of three major cohorts, mtDNA-CN measured from buffy coat was inversely associated with prevalent and incident CVD, coronary heart disease (CHD), stroke, and sudden cardiac death (SCD), independent of traditional risk factors (3665, 2460, 1583, and 361 incident events for CVD, CHD, stroke, and SCD, respectively) (Ashar et al., 2017; Yiyi Zhang et al., 2017). In these cohorts, adding mtDNA-CN to the ACC/AHA risk evaluation criteria for primary prevention significantly improved risk reclassification for CVD (Ashar et al., 2017), and it improved the sensitivity and specificity for the ACC/AHA recommendations on initiating statin therapy for primary prevention of atherosclerotic cardiovascular disease (continuous net reclassification index, 0.194; 95% CI, 0.130–0.258; P< 0.001) (Ashar et al., 2017). In addition, mtDNA-CN measured from leukocytes was associated with the presence of coronary atherosclerotic plaque in a small case-control study (378 CHD cases, 378 controls) (Chen et al., 2014) and mtDNA-CN from peripheral blood was inversely associated with an angiographic severity score in coronary atherosclerosis in cross-sectional analysis (Liu et al., 2017; Neeland et al., 2012).
Kidney Disease
In the Atherosclerosis Risk in Communities (ARIC) cohort, higher mtDNA-CN measured from buffy coat was associated with a reduced risk of incident chronic kidney disease (CKD) after adjusting for traditional CKD risk factors, such as prevalent diabetes, hypertension, and C-reactive protein levels (Tin et al., 2016). Others have found similar associations with kidney disease, including a recent study which identified decreased copy number measured from peripheral blood in patients with end stage renal disease (ESRD) (Yuheng Zhang et al., 2017). Interestingly, mtDNA-CN measured from serum was increased in patients with ESRD and in the presence of markers of renal injury (Eirin et al., 2016; Yuheng Zhang et al., 2017). Further, decreased mtDNA is observed in urinary supernatant compared to intra-renal samples which suggests different mechanisms are at play in these tissues (P. Z. Wei et al., 2017). One possible explanation for this observation may be that cell-free mtDNA measured from serum or urine could reflect higher rates of apoptosis rather than mitochondrial dysfunction, thus leading to elevated mtDNA-CN associations with disease. Further, in a large independent CKD cohort, decreased mtDNA-CN was significantly associated with infection-related deaths (Fazzini et al., 2019) which lends additional support to recent findings linking HIV disease severity with decreased mtDNA-CN (Sun et al., 2019).
Liver Disease
mtDNA-CN has been shown to be associated with several classes of liver disease, including non-alcoholic fatty liver disease (NAFLD). In NALFD, mtDNA-CN is reduced in liver tissue of patients as compared to controls (Pirola et al., 2015; Sookoian et al., 2010). Additionally, the mtDNA/nDNA ratio has been found to be inversely correlated with insulin resistance, serum fasting glucose and plasma fasting insulin (Sookoian et al., 2010).
Neurodegenerative Disorders
Cerebellar neuron dysfunction has been associated with impaired mitochondrial structure and bioenergetic function (Bartesaghi et al., 2010). Patients with polymerase gamma encephalopathy display dopaminergic substanta nigral neurons with significantly lower mtDNA-CN and an overall loss of dopaminergic neurons in the pars compacta region (Tzoulis et al., 2013). The link to neurodegeneration appears independent of rare mitochondrial DNA depletion syndromes as independent analysis found significantly lower mtDNA-CN in substantia nigra pars compacta tissue and peripheral blood samples in patients with Parkinson’s disease (PD) (Pyle et al., 2016). Interestingly, the reduction of copy number appears to be brain region specific as no reduction in mtDNA-CN was observed in the frontal cortex (Pyle et al., 2016). Although recent work did not replicate the previous findings for PD, reduced mtDNA-CN was observed in AD and Creutzfeldt-Jakob Disease (W. Wei et al., 2017).
Cancer
A number of case-control studies which measured mtDNA-CN from blood between cancer patients and normal healthy controls have identified associations between mtDNA-CN levels and increased risk of cancer. Specifically, one such study found an association between decreased mtDNA-CN in peripheral leukocytes and renal cell carcinoma (Xing et al., 2008). Further, pancreatic cancer risk correlates with increased mtDNA-CN levels in whole blood (Lynch et al., 2011). Additionally, colorectal cancer risk has been shown to be associated with both high levels and low levels of mtDNA-CN in peripheral blood (Thyagarajan et al., 2012). Interestingly, increased mtDNA-CN in colorectal cancer correlates with the presence of the common 4977 mitochondrial deletion which spans five tRNA genes and seven protein-coding genes (T. Chen et al., 2011).
Differences in mtDNA Copy Number between Cancer and Normal Tissue
A hallmark characteristic of cancer is the presence of defective mitochondria in cancer cells (Reznik et al., 2016), including mutations, deletions and copy number variation of mtDNA. In addition to studies which have assessed the contribution of mtDNA-CN to the risk of cancer, mtDNA-CN differences have also been found between cancer and normal tissue for a variety of cancer types (Table 3). Specifically, depletion of mtDNA relative to matched normal tissue is particularly common in bladder, breast, and kidney cancers (Reznik et al., 2016). In addition, there is an association between the incidence of several somatic alterations, such as IDH1 mutations in gliomas, and mtDNA content (Reznik et al., 2016). Further, in some cancer types, mtDNA content is correlated with the expression of respiratory genes and negatively correlated with the expression of immune response and cell cycle genes (Reznik et al., 2016). These observations suggest that mtDNA-CN measurement may be useful as a clinical marker of tumor identification, progression, and treatment.
Table 3.
Studies reporting differences in mtDNA-CN between cancer and normal tissue
| Study | Setting | Study Design | Sample Size | Tissue | Method for mtdNA estimation | Endpoint | Results |
|---|---|---|---|---|---|---|---|
| Reznik et al (2016)(Rezni ketal., 2016) | Retrospective | Tumor-normal | 45 matched samples | Bladder tissue | Whole exome/ whole genome | Bladder | ↓ Decreased mtDNA-CN in bladder tumor tissue (BH-corrected Mann-Whitney p-value <0.05) |
| Reznik et al (2016)(Rezni ketal., 2016) | Retrospective | Tumor-normal | 81 matched samples | Breast tissue | Whole exome/ whole genome | Breast | ↓ Decreased mtDNA-CN in breast tumor tissue (BH-corrected Mann-Whitney p-value <0.05) |
| Yu et al (2007)(Yu et al., 2007) | Retrospective | Tumor-normal | 59 matched samples | qPCR | ↓ Decreased mtDNA-CN in breast tumor tissue (P=0.001) | ||
| Reznik et al (2016)(Rezni ketal., 2016) | Retrospective | Tumor-normal | Clear-cell: 249 matched samples; Papillary: 55 matched samples | Kidney tissue | Whole exome/ whole genome | Kidney | ↓ Decreased mtDNA-CN in kidney tumor tissue (BH-corrected Mann-Whitney p-value <0.05) |
| Yuan et al (2020)(Yuan etal.,2020) | Retrospective | Tumor-normal | 27 matched samples | Whole genome | ↓ Decreased mtDNA-CN in renal clear-cell carcinoma tissue (two-sided Wilcoxon signed-rank p-value <0.001) | ||
| Reznik et al (2016)(Rezni ketal., 2016) | Retrospective | Tumor-normal | 25 matched samples | Esophegeal tissue | Whole exome/ whole genome | Esophageal | ↓ Decreased mtDNA-CN in esophageal tumor tissue (BH-corrected Mann-Whitney p-value <0.05) |
| Reznik et al (2016)(Rezni ketal., 2016) | Retrospective | Tumor-normal | 92 matched samples | Squamous cell tissue | Whole exome/ whole genome | Head/neck squamous cell | ↓ Decreased mtDNA-CN in squamous cell tumor tissue (BH-corrected Mann-Whitney p-value <0.05) |
| Reznik et al (2016)(Rezni ketal.,2016) | Retrospective | Tumor-normal | 83 matched samples | Liver tissue | Whole exome/ whole genome | Liver | ↓ Decreased mtDNA-CN in liver tumor tissue (BH-corrected Mann-Whitney p-value <0.05) |
| Yuan et al (2020)(Yuan etal.,2020) | Retrospective | Tumor-normal | 29 matched samples | Whole genome | ↓ Decreased mtDNA-CN in liver tumor tissue (two-sided Wilcoxon signed-rank p-value <0.01) | ||
| Yuan et al (2020)(Yuan etal.,2020) | Retrospective | Tumor-normal | 25 matched samples | Myeloid tissue | Whole genome | Myeloproliferative neoplasm | ↓ Decreased mtDNA-CN in tumor tissue (two-sided Wilcoxon signed-rank p-value O.001) |
| Reznik et al (2016)(Rezni ketal.,2016) | Retrospective | Tumor-normal | 137 matched samples | Lung tissue | Whole exome/ whole genome | Lung | ↓ Decreased mtDNA-CN in lung adenocarcinoma tumor tissue (BH-corrected Mann-Whitney p-value <0.05) |
| Yuan et al (2020)(Yuan etal.,2020) | Retrospective | Tumor-normal | 25 matched samples | Whole genome | ↓ Incresed mtDNA-CN in lung squamous cell carcinoma tissue (two-sided Wilcoxon signed-rank p-value <0.01) | ||
| Savagner et al (2001)(Savag ner et al., 2001) | Retrospective | Tumor-normal | 22 matched samples | Thyroid tissue | Southern blot analysis of rRNA | Thyroid | ↓ Increased mtDNA-CN in thyroid tumor tissue (PO.001) |
| Mizumachi et al (2008)(Mizu machi et al., 2008) | Retrospective | Tumor-normal | 9 cell lines | Prostate cancer cell line (LNCaP) | qPCR | Prostate | ↓ mtDNA was increased in 7 of 9 prostate cancer lines compared to adjacent cells |
| Yuan et al (2020)(Yuan etal.,2020) | Retrospective | Tumor-normal | 81 matched samples | Lymph tissue | Whole genome | Chronic Lymphocytic Leukemia | ↓ Increased mtDNA-CN in turmor tissue (two-sided Wilcoxon signed-rank p-value <0.001) |
| Yuan et al (2020)(Yuan etal.,2020) | Retrospective | Tumor-normal | 111 matched samples | Pancreatic tissue | Whole genome | Pancreatic adenocarcinoma | ↓ Increased mtDNA-CN in pancreatic tissue (two-sided Wilcoxon signed-rank p-value <0.001) |
qPCR: Quantitative PCR; mtDNA: Mitochondrial DNA; mtDNA-CN: Mitochondrial DNA Copy Number; BH: Benjamini-Hochberg; rRNA: ribosomal RNA.
Mutations of the mitochondrial genome have been associated with the formation, growth and metastasis of tumor cells in multiple types of cancer (Ju et al., 2014) (Table 3). In the most comprehensive comparison of mtDNA content across 15 cancer types with normal adjacent tissue (Reznik et al., 2016), six cancer types displayed decreased mtDNA-CN (bladder, breast, esophageal, head/neck squamous cell, kidney, and liver), one cancer type showed increased mtDNA-CN (lung adenocarcinoma), and six cancer types displayed no difference (colorectal, pancreatic, prostate, gastric, thyroid, and uterine) (Reznik et al., 2016). Other studies have confirmed decreased mtDNA-CN in breast cancer (Yu et al., 2007). Further, recent molecular characterization of mitochondrial genomes in human cancers showed increased mtDNA-CN in chronic lymphocytic leukemia, lung squamous cell carcinoma and pancreatic adenocarcinoma, and decreased mtDNA-CN in kidney clear cell carcinoma, liver cancer and myeloproliferative neoplasm (Yuan et al., 2020). Thyroid and prostate cancer cells have been found by other groups to be enriched for mitochondrial content, with prostate cancer showing an increase of up to 78% (Mizumachi et al., 2008; Savagner et al., 2001). This increase has been suggested to be the consequence of cell compensation for defective oxidative phosphorylation, leading to lower ATP production and increased risk for cancer (Hertweck and Dasgupta, 2017; Mizumachi et al., 2008).
Across five cancer types, statistically significant associations have been identified between mtDNA-CN and patient survival and across three cancer types higher mtDNA content has been associated with increased survival (Reznik et al., 2016). Interestingly, it has been suggested that tumor cells may increase mtDNA-CN as a self-protective mechanism to prevent apoptosis, given that reduced mtDNA-CN increases ROS levels in tumor cells, tumor cell sensitivity to chemotherapeutic drugs, and the rate of apoptosis. Thus, mtDNA-CN variation may be a possible therapeutic target for treatment of tumors in a clinical setting (Mei et al., 2015). However, it is plausible that mitochondrial biogenesis is upregulated in cancers as a compensatory mechanism only (Gasparre et al., 2011) and therefore further research is necessary to elucidate the precise mechanism underlying the association of mtDNA-CN and cancer etiology/progression.
Conclusion
In understanding the etiology of complex diseases, it has become clear that the complete knowledge of nuclear DNA sequence is not enough to fully predict complex disease risk. As discussed here, an emerging body of evidence supports roles for mtDNA in the complex underpinnings of a variety of diseases, including a number of cancers and aging-related disorders. A common link in these studies include anti-inflammatory pathways. These mechanisms will be further elucidated as our ability to measure mtDNA-CN from sequencing and microarray technologies expands. As studies increase in power and functional assessment of mechanisms underlying the effect of mtDNA on mitochondrial function and gene expression improve, our understanding of variation in mtDNA-CN as cause or consequence of disease development will rapidly improve.
mtDNA-CN is an especially attractive biomarker because its measurement in blood is both non-invasive and relatively cost-friendly to obtain. The proposed utility of mtDNA-CN as a biomarker for disease has been suggested by the observation that mtDNA content can differentiate healthy controls from patients with cancer and other diseases (Memon et al., 2017). In addition, mtDNA-CN has been shown to be relevant for risk reclassification for cardiovascular disease (Ashar et al., 2017). Currently, these applications are limited by several analytical factors affecting the accurate and reproducible quantification of mtDNA-CN, as discussed in this review. The recent confirmation that human mtDNA is methylated adds yet another level of complexity to the crosstalk between the nucleus and mitochondrion and its control (Ghosh et al., 2014). We close by suggesting that improved detection techniques for mtDNA-CN as well as greater understanding of the mechanisms underlying individual, cell-type, and tissue-specific variation in mtDNA-CN are essential to determining the direct pathological, therapeutic and/or clinical relevance of this relatively cost-effective and easily measured biomarker.
Highlights.
mtDNA-CN, a proxy for mitochondrial function, is associated with a number of age-related diseases and cancers
mtDNA-CN can be measured using a variety of technologies, including pre-existing microarray and sequencing data
Mitochondrial dysfunction may lead to alterations to gene expression and subsequent disease outcomes via a number of relevant pathways
mtDNA-CN shows promise as a potential biomarker for chronic disease
Acknowledgements
We wish to thank Stephanie Y Yang, ThuyVy Duong, and Adrienne Tin for their thorough contributions and editing of this review. This work was supported by the US National Institutes of Health [grant numbers: R01HL131573, R01HL14469].
Role of the Funding Sources
The funding sources had no involvement in the design, collection, analysis, interpretation of data, writing of the report nor decision to submit the article for publication.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Competing Interests Statement
Declarations of interest: none other than funding sources noted in acknowledgements above.
Search Strategy and Selection Criteria
Data for this Review were identified by searches in PubMed and Google Scholar and references from relevant articles. Only articles published in English between 1950 and 2020 in high impact journals with rigorous sample sizes and matched controls were included.
References
- Aggarwal NT, Makielski JC, 2013. Redox control of cardiac excitability. Antioxid. Redox Signal 18, 432–468. 10.1089/ars.2011.4234 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Amor S, Puentes F, Baker D, van der Valk P, 2010. Inflammation in neurodegenerative diseases. Immunology 129, 154–169. 10.1111/j.1365-2567.2009.03225.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ashar FN, Moes A, Moore AZ, Grove ML, Chaves PHM, Coresh J, Newman AB, Matteini AM, Bandeen-Roche K, Boerwinkle E, Walston JD, Arking DE, 2015. Association of mitochondrial DNA levels with frailty and all-cause mortality. J. Mol. Med. Berl. Ger 93, 177–186. 10.1007/s00109-014-1233-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ashar FN, Zhang Y, Longchamps RJ, Lane J, Moes A, Grove ML, Mychaleckyj JC, Taylor KD, Coresh J, Rotter JI, Boerwinkle E, Pankratz N, Guallar E, Arking DE, 2017. Association of Mitochondrial DNA Copy Number With Cardiovascular Disease. JAMA Cardiol. 2, 1247–1255. 10.1001/jamacardio.2017.3683 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bai R-K, Wong L-JC, 2005. Simultaneous Detection and Quantification of Mitochondrial DNA Deletion(s), Depletion, and Over-Replication in Patients with Mitochondrial Disease. J. Mol. Diagn 7, 613–622. 10.1016/S1525-1578(10)60595-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bartesaghi S, Betts-Henderson J, Cain K, Dinsdale D, Zhou X, Karlsson A, Salomoni P, Nicotera P, 2010. Loss of thymidine kinase 2 alters neuronal bioenergetics and leads to neurodegeneration. Hum. Mol. Genet 19, 1669–1677. 10.1093/hmg/ddq043 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berliner JA, Heinecke JW, 1996. The role of oxidized lipoproteins in atherogenesis. Free Radic. Biol. Med 20, 707–727. [DOI] [PubMed] [Google Scholar]
- Bianco A, Valletti A, Longo G, Bisceglia L, Montoya J, Emperador S, Guerriero S, Petruzzella V, 2018. Mitochondrial DNA copy number in affected and unaffected LHON mutation carriers. BMC Res. Notes 11 10.1186/s13104-018-4025-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bogenhagen D, Clayton DA, 1977. Mouse L cell mitochondrial DNA molecules are selected randomly for replication throughout the cell cycle. Cell 11, 719–727. [DOI] [PubMed] [Google Scholar]
- Bosworth CM, Grandhi S, Gould MP, LaFramboise T, 2017. Detection and quantification of mitochondrial DNA deletions from next-generation sequence data. BMC Bioinformatics 18, 407 10.1186/s12859-017-1821-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cai N, Li Y, Chang S, Liang J, Lin C, Zhang X, Liang L, Hu J, Chan W, Kendler KS, Malinauskas T, Huang G-J, Li Q, Mott R, Flint J, 2015. Genetic Control over mtDNA and Its Relationship to Major Depressive Disorder. Curr. Biol 25, 3170–3177. 10.1016/j.cub.2015.10.065 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chabi B, Mousson de Camaret B, Duborjal H, Issartel J-P, Stepien G, 2003. Quantification of mitochondrial DNA deletion, depletion, and overreplication: application to diagnosis. Clin. Chem 49, 1309–1317. [DOI] [PubMed] [Google Scholar]
- Chen H, Vermulst M, Wang YE, Chomyn A, Prolla TA, McCaffery JM, Chan DC, 2010. Mitochondrial fusion is required for mtDNA stability in skeletal muscle and tolerance of mtDNA mutations. Cell 141, 280–289. 10.1016/j.cell.2010.02.026 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen S, Xie, Xuejian, Wang Y., Gao Y., Xie, Xiaoli, Yang J., Ye J., 2014. Association between leukocyte mitochondrial DNA content and risk of coronary heart disease: A case-control study. Atherosclerosis 237, 220–226. 10.1016/j.atherosclerosis.2014.08.051 [DOI] [PubMed] [Google Scholar]
- Chen T, He J, Shen L, Fang H, Nie H, Jin T, Wei X, Xin Y, Jiang Y, Li H, Chen G, Lu J, Bai Y, 2011. The mitochondrial DNA 4,977-bp deletion and its implication in copy number alteration in colorectal cancer. BMC Med. Genet 12, 8 10.1186/1471-2350-12-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen Y, Liu Y, Dorn GW, 2011. Mitochondrial fusion is essential for organelle function and cardiac homeostasis. Circ. Res 109, 1327–1331. 10.1161/CIRCRESAHA.111.258723 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cichoż-Lach H, Michalak A, 2014. Oxidative stress as a crucial factor in liver diseases. World J. Gastroenterol WJG 20, 8082–8091. 10.3748/wjg.v20.i25.8082 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Clayton DA, 1992. Transcription and replication of animal mitochondrial DNAs. Int. Rev. Cytol 141, 217–232. [DOI] [PubMed] [Google Scholar]
- Cristina D, Cary M, Lunceford A, Clarke C, Kenyon C, 2009. A regulated response to impaired respiration slows behavioral rates and increases lifespan in Caenorhabditis elegans. PLoS Genet. 5, e1000450 10.1371/journal.pgen.1000450 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Czaja AJ, 2014. Hepatic inflammation and progressive liver fibrosis in chronic liver disease. World J. Gastroenterol WJG 20, 2515–2532. 10.3748/wjg.v20.i10.2515 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dai D-F, Rabinovitch PS, Ungvari Z, 2012. Mitochondria and cardiovascular aging. Circ. Res 110, 1109–1124. 10.1161/CIRCRESAHA.111.246140 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dimmock D, Tang L-Y, Schmitt ES, Wong L-JC, 2010. Quantitative Evaluation of the Mitochondrial DNA Depletion Syndrome. Clin. Chem 56, 1119–1127. 10.1373/clinchem.2009.141549 [DOI] [PubMed] [Google Scholar]
- Ding J, Sidore C, Butler TJ, Wing MK, Qian Y, Meirelles O, Busonero F, Tsoi LC, Maschio A, Angius A, Kang HM, Nagaraja R, Cucca F, Abecasis GR, Schlessinger D, 2015. Assessing Mitochondrial DNA Variation and Copy Number in Lymphocytes of ~2,000 Sardinians Using Tailored Sequencing Analysis Tools. PLOS Genet. 11, e1005306 10.1371/journal.pgen.1005306 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eirin A, Saad A, Tang H, Herrmann SM, Woollard JR, Lerman A, Textor SC, Lerman LO, 2016. Urinary Mitochondrial DNA Copy Number Identifies Chronic Renal Injury in Hypertensive PatientsNovelty and Significance. Hypertension 68, 401–410. 10.1161/HYPERTENSIONAHA.116.07849 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Erusalimsky JD, 2009. Vascular endothelial senescence: from mechanisms to pathophysiology. J. Appl. Physiol Bethesda Md 1985 106, 326–332. 10.1152/japplphysiol.91353.2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fazzini F, Lamina C, Fendt L, Schultheiss UT, Kotsis F, Hicks AA, Meiselbach H, Weissensteiner H, Forer L, Krane V, Eckardt K-U, Köttgen A, Kronenberg F, GCKD Investigators, 2019. Mitochondrial DNA copy number is associated with mortality and infections in a large cohort of patients with chronic kidney disease. Kidney Int. 96, 480–488. 10.1016/j.kint.2019.04.021 [DOI] [PubMed] [Google Scholar]
- Friis RMN, Glaves JP, Huan T, Li L, Sykes BD, Schultz MC, 2014. Rewiring AMPK and mitochondrial retrograde signaling for metabolic control of aging and histone acetylation in respiratory-defective cells. Cell Rep. 7, 565–574. 10.1016/j.celrep.2014.03.029 [DOI] [PubMed] [Google Scholar]
- Gandhi S, Abramov AY, 2012. Mechanism of Oxidative Stress in Neurodegeneration. Oxid. Med. Cell. Longev. 2012 10.1155/2012/428010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gasparre G, Romeo G, Rugolo M, Porcelli AM, 2011. Learning from oncocytic tumors: Why choose inefficient mitochondria? Biochim. Biophys. Acta 1807, 633–642. 10.1016/j.bbabio.2010.08.006 [DOI] [PubMed] [Google Scholar]
- Ghosh S, Sengupta S, Scaria V, 2014. Comparative analysis of human mitochondrial methylomes shows distinct patterns of epigenetic regulation in mitochondria. Mitochondrion 18, 58–62. 10.1016/j.mito.2014.07.007 [DOI] [PubMed] [Google Scholar]
- Goudenège D, Bris C, Hoffmann V, Desquiret-Dumas V, Jardel C, Rucheton B, Bannwarth S, Paquis-Flucklinger V, Lebre AS, Colin E, Amati-Bonneau P, Bonneau D, Reynier P, Lenaers G, Procaccio V, 2019. eKLIPse: a sensitive tool for the detection and quantification of mitochondrial DNA deletions from next-generation sequencing data. Genet. Med. Off. J. Am. Coll. Med. Genet 21, 1407–1416. 10.1038/s41436-018-0350-8 [DOI] [PubMed] [Google Scholar]
- Grady JP, Pickett SJ, Ng YS, Alston CL, Blakely EL, Hardy SA, Feeney CL, Bright AA, Schaefer AM, Gorman GS, McNally RJ, Taylor RW, Turnbull DM, McFarland R, 2018. mtDNA heteroplasmy level and copy number indicate disease burden in m.3243A>G mitochondrial disease. EMBO Mol. Med 10, e8262 10.15252/emmm.201708262 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gu F, Chauhan V, Kaur K, Brown WT, LaFauci G, Wegiel J, Chauhan A, 2013. Alterations in mitochondrial DNA copy number and the activities of electron transport chain complexes and pyruvate dehydrogenase in the frontal cortex from subjects with autism. Transl. Psychiatry 3, e299 10.1038/tp.2013.68 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guantes R, Rastrojo A, Neves R, Lima A, Aguado B, Iborra FJ, 2015. Global variability in gene expression and alternative splicing is modulated by mitochondrial content. Genome Res. 25, 633–644. 10.1101/gr.178426.114 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guha M, Avadhani NG, 2013. Mitochondrial retrograde signaling at the crossroads of tumor bioenergetics, genetics and epigenetics. Mitochondrion 13, 577–591. 10.1016/j.mito.2013.08.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guha M, Srinivasan S, Guja K, Mejia E, Garcia-Diaz M, Johnson FB, Ruthel G, Kaufman BA, Rappaport EF, Glineburg MR, Fang J-K, Szanto AK, Nakagawa H, Basha J, Kundu T, Avadhani NG, 2016. HnRNPA2 is a novel histone acetyltransferase that mediates mitochondrial stress-induced nuclear gene expression. Cell Discov. 2, 16045 10.1038/celldisc.2016.45 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guo W, Jiang L, Bhasin S, Khan SM, Swerdlow RH, 2009. DNA Extraction Procedures Meaningfully Influence qPCR-Based mtDNA Copy Number Determination. Mitochondrion 9, 261–265. 10.1016/j.mito.2009.03.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guyatt AL, Burrows K, Guthrie PAI, Ring S, McArdle W, Day INM, Ascione R, Lawlor DA, Gaunt TR, Rodriguez S, 2017. Cardiometabolic phenotypes and mitochondrial DNA copy number in two cohorts of UK women. Mitochondrion. 10.1016/j.mito.2017.08.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hertweck KL, Dasgupta S, 2017. The Landscape of mtDNA Modifications in Cancer: A Tale of Two Cities. Front. Oncol 7, 262 10.3389/fonc.2017.00262 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hurtado-Roca Y, Ledesma M, Gonzalez-Lazaro M, Moreno-Loshuertos R, Fernandez-Silva P, Enriquez JA, Laclaustra M, 2016. Adjusting MtDNA Quantification in Whole Blood for Peripheral Blood Platelet and Leukocyte Counts. PLOS ONE 11, e0163770 10.1371/journal.pone.0163770 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ishihara N, Nomura M, Jofuku A, Kato H, Suzuki SO, Masuda K, Otera H, Nakanishi Y, Nonaka I, Goto Y-I, Taguchi N, Morinaga H, Maeda M, Takayanagi R, Yokota S, Mihara K, 2009. Mitochondrial fission factor Drp1 is essential for embryonic development and synapse formation in mice. Nat. Cell Biol 11, 958–966. 10.1038/ncb1907 [DOI] [PubMed] [Google Scholar]
- Jain M, Rivera S, Monclus EA, Synenki L, Zirk A, Eisenbart J, Feghali-Bostwick C, Mutlu GM, Budinger GRS, Chandel NS, 2013. Mitochondrial reactive oxygen species regulate transforming growth factor-β signaling. J. Biol. Chem 288, 770–777. 10.1074/jbc.M112.431973 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jędrak P, Krygier M, Tońska K, Drozd M, Kaliszewska M, Bartnik E, Sołtan W, Sitek EJ, Stanisławska-Sachadyn A, Limon J, Sławek J, Węgrzyn G, Barańska S, 2017. Mitochondrial DNA levels in Huntington disease leukocytes and dermal fibroblasts. Metab. Brain Dis 32, 1237–1247. 10.1007/s11011-017-0026-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jeng J-Y, Yeh T-S, Lee J-W, Lin S-H, Fong T-H, Hsieh R-H, 2008. Maintenance of mitochondrial DNA copy number and expression are essential for preservation of mitochondrial function and cell growth. J. Cell. Biochem 103, 347–357. 10.1002/jcb.21625 [DOI] [PubMed] [Google Scholar]
- Ju YS, Alexandrov LB, Gerstung M, Martincorena I, Nik-Zainal S, Ramakrishna M, Davies HR, Papaemmanuil E, Gundem G, Shlien A, Bolli N, Behjati S, Tarpey PS, Nangalia J, Massie CE, Butler AP, Teague JW, Vassiliou GS, Green AR, Du M-Q, Unnikrishnan A, Pimanda JE, Teh BT, Munshi N, Greaves M, Vyas P, El-Naggar AK, Santarius T, Collins VP, Grundy R, Taylor JA, Hayes DN, Malkin D, ICGC Breast Cancer Group, ICGC Chronic Myeloid Disorders Group, ICGC Prostate Cancer Group, Foster CS, Warren AY, Whitaker HC, Brewer D, Eeles R, Cooper C, Neal D, Visakorpi T, Isaacs WB, Bova GS, Flanagan AM, Futreal PA, Lynch AG, Chinnery PF, McDermott U, Stratton MR, Campbell PJ, 2014. Origins and functional consequences of somatic mitochondrial DNA mutations in human cancer. eLife 3 10.7554/eLife.02935 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kamfar S, Alavian SM, Houshmand M, Yadegarazari R, Seifi Zarei B, Khalaj A, Shabab N, Saidijam M, 2016. Liver Mitochondrial DNA Copy Number and Deletion Levels May Contribute to Nonalcoholic Fatty Liver Disease Susceptibility. Hepat. Mon 16, e40774 10.5812/hepatmon.40774 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kelly RDW, Mahmud A, McKenzie M, Trounce IA, St John JC, 2012. 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 40, 10124–10138. 10.1093/nar/gks770 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim GH, Kim JE, Rhie SJ, Yoon S, 2015. The Role of Oxidative Stress in Neurodegenerative Diseases. Exp. Neurobiol 24, 325–340. 10.5607/en.2015.24.4.325 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Knez J, Winckelmans E, Plusquin M, Thijs L, Cauwenberghs N, Gu Y, Staessen JA, Nawrot TS, Kuznetsova T, 2016. Correlates of Peripheral Blood Mitochondrial DNA Content in a General Population. Am. J. Epidemiol 183, 138–146. 10.1093/aje/kwv175 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li B, Kaushik S, Kalinowski P, Kim B, Gershome C, Ching J, Poburko D, 2018. Droplet digital PCR shows the D-Loop to be an error prone locus for mitochondrial DNA copy number determination. Sci. Rep 8, 11392 10.1038/s41598-018-29621-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Libby P, Ridker PM, Maseri A, 2002. Inflammation and atherosclerosis. Circulation 105, 1135–1143. [DOI] [PubMed] [Google Scholar]
- Liu L-P, Cheng K, Ning M-A, Li H-H, Wang H-C, Li F, Chen S-Y, Qu F-L, Guo W-Y, 2017. Association between peripheral blood cells mitochondrial DNA content and severity of coronary heart disease. Atherosclerosis 261, 105–110. 10.1016/j.atherosclerosis.2017.02.013 [DOI] [PubMed] [Google Scholar]
- Liu M, Liu H, Dudley SC, 2010. Reactive oxygen species originating from mitochondria regulate the cardiac sodium channel. Circ. Res 107, 967–974. 10.1161/CIRCRESAHA.110.220673 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lo I-C, Shih J-M, Jiang MJ, 2005. Reactive oxygen species and ERK 1/2 mediate monocyte chemotactic protein-1-stimulated smooth muscle cell migration. J. Biomed. Sci 12, 377–388. 10.1007/s11373-005-1703-2 [DOI] [PubMed] [Google Scholar]
- Longchamps RJ, Castellani CA, Yang SY, Newcomb CE, Sumpter JA, Lane J, Grove ML, Guallar E, Pankratz N, Taylor KD, Rotter JI, Boerwinkle E, Arking DE, 2020. Evaluation of mitochondrial DNA copy number estimation techniques. PloS One 15, e0228166 10.1371/journal.pone.0228166 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lynch SM, Weinstein SJ, Virtamo J, Lan Q, Liu C-S, Cheng W-L, Rothman N, Albanes D, Stolzenberg-Solomon RZ, 2011. Mitochondrial DNA copy number and pancreatic cancer in the alpha-tocopherol beta-carotene cancer prevention study. Cancer Prev. Res. Phila. Pa 4, 1912–1919. 10.1158/1940-6207.CAPR-11-0002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martinez FO, Sica A, Mantovani A, Locati M, 2008. Macrophage activation and polarization. Front. Biosci. J. Virtual Libr 13, 453–461. [DOI] [PubMed] [Google Scholar]
- Mei H, Sun S, Bai Y, Chen Y, Chai R, Li H, 2015. Reduced mtDNA copy number increases the sensitivity of tumor cells to chemotherapeutic drugs. Cell Death Dis 6, e1710 10.1038/cddis.2015.78 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Memon AA, Zöller B, Hedelius A, Wang X, Stenman E, Sundquist J, Sundquist K, 2017. Quantification of mitochondrial DNA copy number in suspected cancer patients by a well optimized ddPCR method. Biomol. Detect. Quantif 13, 32–39. 10.1016/j.bdq.2017.08.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mengel-From J, Thinggaard M, Dalgård C, Kyvik KO, Christensen K, Christiansen L, 2014. Mitochondrial DNA copy number in peripheral blood cells declines with age and is associated with general health among elderly. Hum. Genet 133, 1149–1159. 10.1007/s00439-014-1458-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miller FJ, Rosenfeldt FL, Zhang C, Linnane AW, Nagley P, 2003. Precise determination of mitochondrial DNA copy number in human skeletal and cardiac muscle by a PCR-based assay: lack of change of copy number with age. Nucleic Acids Res 31, e61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- MitoPipeline G, 2018. MitoPipeline [WWW Document]. MitoPipeline URL http://genvisis.org/MitoPipeline/ (accessed 11.27.19).
- Mizumachi T, Muskhelishvili L, Naito A, Furusawa J, Fan C-Y, Siegel ER, Kadlubar FF, Kumar U, Higuchi M, 2008. Increased distributional variance of mitochondrial DNA content associated with prostate cancer cells as compared with normal prostate cells. The Prostate 68, 408–417. 10.1002/pros.20697 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morgan JT, Chana G, Abramson I, Semendeferi K, Courchesne E, Everall IP, 2012. Abnormal microglial–neuronal spatial organization in the dorsolateral prefrontal cortex in autism. Brain Res. 1456, 72–81. 10.1016/j.brainres.2012.03.036 [DOI] [PubMed] [Google Scholar]
- Muir R, Diot A, Poulton J, 2016. Mitochondrial content is central to nuclear gene expression: Profound implications for human health. BioEssays News Rev. Mol. Cell. Dev. Biol 38, 150–156. 10.1002/bies.201500105 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nacheva E, Mokretar K, Soenmez A, Pittman AM, Grace C, Valli R, Ejaz A, Vattathil S, Maserati E, Houlden H, Taanman J-W, Schapira AH, Proukakis C, 2017. DNA isolation protocol effects on nuclear DNA analysis by microarrays, droplet digital PCR, and whole genome sequencing, and on mitochondrial DNA copy number estimation. PLoS ONE 12 10.1371/journal.pone.0180467 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Neeland IJ, Patel RS, Eshtehardi P, Dhawan S, McDaniel MC, Rab ST, Vaccarino V, Zafari AM, Samady H, Quyyumi AA, 2012. Coronary angiographic scoring systems: An evaluation of their equivalence and validity. Am. Heart J 164, 547–552.e1. 10.1016/j.ahj.2012.07.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nicholls TJ, Minczuk M, 2014. In D-loop: 40 years of mitochondrial 7S DNA. Exp. Gerontol 56, 175–181. 10.1016/j.exger.2014.03.027 [DOI] [PubMed] [Google Scholar]
- Nicolson GL, 2014. Mitochondrial Dysfunction and Chronic Disease: Treatment With Natural Supplements. Integr. Med. Clin. J 13, 35–43. [PMC free article] [PubMed] [Google Scholar]
- O’Hara R, Tedone E, Ludlow A, Huang E, Arosio B, Mari D, Shay JW, 2019. Quantitative mitochondrial DNA copy number determination using droplet digital PCR with single-cell resolution. Genome Res. 10.1101/gr.250480.119 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pello R, Martín MA, Carelli V, Nijtmans LG, Achilli A, Pala M, Torroni A, Gómez-Durán A, Ruiz-Pesini E, Martinuzzi A, Smeitink JA, Arenas J, Ugalde C, 2008. Mitochondrial DNA background modulates the assembly kinetics of OXPHOS complexes in a cellular model of mitochondrial disease. Hum. Mol. Genet 17, 4001–4011. 10.1093/hmg/ddn303 [DOI] [PubMed] [Google Scholar]
- Phillips NR, Sprouse ML, Roby RK, 2014. Simultaneous quantification of mitochondrial DNA copy number and deletion ratio: a multiplex real-time PCR assay. Sci. Rep 4, 3887 10.1038/srep03887 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Picard M, Zhang J, Hancock S, Derbeneva O, Golhar R, Golik P, O’Hearn S, Levy S, Potluri P, Lvova M, Davila A, Lin CS, Perin JC, Rappaport EF, Hakonarson H, Trounce IA, Procaccio V, Wallace DC, 2014. Progressive increase in mtDNA 3243A>G heteroplasmy causes abrupt transcriptional reprogramming. Proc. Natl. Acad. Sci. U. S. A 111, E4033–4042. 10.1073/pnas.1414028111 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pirola CJ, Scian R, Gianotti TF, Dopazo H, Rohr C, Martino JS, Castaño GO, Sookoian S, 2015. Epigenetic Modifications in the Biology of Nonalcoholic Fatty Liver Disease: The Role of DNA Hydroxymethylation and TET Proteins. Medicine (Baltimore) 94, e1480 10.1097/MD.0000000000001480 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pyle A, Anugrha H, Kurzawa-Akanbi M, Yarnall A, Burn D, Hudson G, 2016. Reduced mitochondrial DNA copy number is a biomarker of Parkinson’s disease. Neurobiol. Aging 38, 216.e7–216.e10. 10.1016/j.neurobiolaging.2015.10.033 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qian Y, Butler TJ, Opsahl-Ong K, Giroux NS, Sidore C, Nagaraja R, Cucca F, Ferrucci L, Abecasis GR, Schlessinger D, Ding J, 2017. fastMitoCalc: an ultra-fast program to estimate mitochondrial DNA copy number from wholegenome sequences. Bioinformatics 33, 1399–1401. 10.1093/bioinformatics/btw835 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Raffoul JJ, Heydari AR, Hillman GG, 2012. DNA Repair and Cancer Therapy: Targeting APE1/Ref-1 Using Dietary Agents. J. Oncol 2012, 370481 10.1155/2012/370481 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ravi S, Mitchell T, Kramer P, Chacko B, Darley-Usmar VM, 2014. MITOCHONDRIA in Monocytes and Macrophages-Implications for Translational and Basic Research. Int. J. Biochem. Cell Biol 53, 202–207. 10.1016/j.biocel.2014.05.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reznik E, Miller ML, Şenbabaoğlu Y, Riaz N, Sarungbam J, Tickoo SK, Al-Ahmadie HA, Lee W, Seshan VE, Hakimi AA, Sander C, 2016. Mitochondrial DNA copy number variation across human cancers. eLife 5 10.7554/eLife.10769 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rodriguez JI, Kern JK, 2011. Evidence of microglial activation in autism and its possible role in brain underconnectivity. Neuron Glia Biol. 7, 205–213. 10.1017/S1740925X12000142 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rodríguez-Prados J-C, Través PG, Cuenca J, Rico D, Aragonés J, Martín-Sanz P, Cascante M, Boscá L, 2010. Substrate fate in activated macrophages: a comparison between innate, classic, and alternative activation. J. Immunol. Baltim. Md 1950 185, 605–614. 10.4049/jimmunol.0901698 [DOI] [PubMed] [Google Scholar]
- Rygiel KA, Grady JP, Taylor RW, Tuppen HAL, Turnbull DM, 2015. Triplex real-time PCR--an improved method to detect a wide spectrum of mitochondrial DNA deletions in single cells. Sci. Rep 5, 9906 10.1038/srep09906 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sánchez-Santos A, Martínez-Hernández MG, Contreras-Ramos A, Ortega-Camarillo C, Baiza-Gutman LA, 2018. Hyperglycemia-induced mouse trophoblast spreading is mediated by reactive oxygen species. Mol. Reprod. Dev 10.1002/mrd.22965 [DOI] [PubMed] [Google Scholar]
- Savagner F, Franc B, Guyetant S, Rodien P, Reynier P, Malthiery Y, 2001. Defective mitochondrial ATP synthesis in oxyphilic thyroid tumors. J. Clin. Endocrinol. Metab 86, 4920–4925. 10.1210/jcem.86.10.7894 [DOI] [PubMed] [Google Scholar]
- Silva Ramos E, Motori E, Brüser C, Kühl I, Yeroslaviz A, Ruzzenente B, Kauppila JHK, Busch JD, Hultenby K, Habermann BH, Jakobs S, Larsson N-G, Mourier A, 2019. Mitochondrial fusion is required for regulation of mitochondrial DNA replication. PLoS Genet. 15, e1008085 10.1371/journal.pgen.1008085 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Silverstein DM, 2009. Inflammation in chronic kidney disease: role in the progression of renal and cardiovascular disease. Pediatr. Nephrol 24, 1445–1452. 10.1007/s00467-008-1046-0 [DOI] [PubMed] [Google Scholar]
- Sookoian S, Rosselli MS, Gemma C, Burgueño AL, Fernández Gianotti T, Castaño GO, Pirola CJ, 2010. Epigenetic regulation of insulin resistance in nonalcoholic fatty liver disease: Impact of liver methylation of the peroxisome proliferator–activated receptor γ coactivator 1α promoter. Hepatology 52, 1992–2000. 10.1002/hep.23927 [DOI] [PubMed] [Google Scholar]
- Srinivasan S, Guha M, Dong DW, Whelan KA, Ruthel G, Uchikado Y, Natsugoe S, Nakagawa H, Avadhani NG, 2016. Disruption of cytochrome c oxidase function induces the Warburg effect and metabolic reprogramming. Oncogene 35, 1585–1595. 10.1038/onc.2015.227 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun J, Longchamps RJ, Piggott DA, Castellani CA, Sumpter JA, Brown TT, Mehta SH, Arking DE, Kirk GD, 2019. Association Between HIV Infection and Mitochondrial DNA Copy Number in Peripheral Blood: A Population-Based, Prospective Cohort Study. J. Infect. Dis 219, 1285–1293. 10.1093/infdis/jiy658 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Taylor RW, Turnbull DM, 2005. Mitochondrial DNA mutations in human disease. Nat. Rev. Genet 6, 389–402. 10.1038/nrg1606 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thyagarajan B, Wang R, Barcelo H, Koh W-P, Yuan J-M, 2012. Mitochondrial copy number is associated with colorectal cancer risk. Cancer Epidemiol. Biomark. Prev. Publ. Am. Assoc. Cancer Res. Cosponsored Am. Soc. Prev. Oncol 21, 1574–1581. 10.1158/1055-9965.EPI-12-0138-T [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tidwell TR, Søreide K, Hagland HR, 2017. Aging, Metabolism, and Cancer Development: from Peto’s Paradox to the Warburg Effect. Aging Dis. 8, 662–676. 10.14336/AD.2017.0713 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tin A, Grams ME, Ashar FN, Lane JA, Rosenberg AZ, Grove ML, Boerwinkle E, Selvin E, Coresh J, Pankratz N, Arking DE, 2016. Association between Mitochondrial DNA Copy Number in Peripheral Blood and Incident CKD in the Atherosclerosis Risk in Communities Study. J. Am. Soc. Nephrol JASN 27, 2467–2473. 10.1681/ASN.2015060661 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tzoulis C, Tran GT, Schwarzlmüller T, Specht K, Haugarvoll K, Balafkan N, Lilleng PK, Miletic H, Biermann M, Bindoff LA, 2013. Severe nigrostriatal degeneration without clinical parkinsonism in patients with polymerase gamma mutations. Brain 136, 2393–2404. 10.1093/brain/awt103 [DOI] [PubMed] [Google Scholar]
- Vakifahmetoglu-Norberg H, Ouchida AT, Norberg E, 2017. The role of mitochondria in metabolism and cell death. Biochem. Biophys. Res. Commun., Special Issue on Cell Death in Honor of Sten Orrenius 482, 426–431. 10.1016/j.bbrc.2016.11.088 [DOI] [PubMed] [Google Scholar]
- Vats D, Mukundan L, Odegaard JI, Zhang L, Smith KL, Morel CR, Wagner RA, Greaves DR, Murray PJ, Chawla A, 2006. Oxidative metabolism and PGC-1beta attenuate macrophage-mediated inflammation. Cell Metab. 4, 13–24. 10.1016/j.cmet.2006.05.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wai T, Ao A, Zhang X, Cyr D, Dufort D, Shoubridge EA, 2010. The role of mitochondrial DNA copy number in mammalian fertility. Biol. Reprod 83, 52–62. 10.1095/biolreprod.109.080887 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wallace DC, 1992. Diseases of the mitochondrial DNA. Annu. Rev. Biochem 61, 1175–1212. 10.1146/annurev.bi.61.070192.005523 [DOI] [PubMed] [Google Scholar]
- Wei PZ, Kwan BC-H, Chow KM, Cheng PM-S, Luk CC-W, Li PK-T, Szeto CC, 2017. Urinary mitochondrial DNA level is an indicator of intra-renal mitochondrial depletion and renal scarring in diabetic nephropathy. Nephrol. Dial. Transplant. Off. Publ. Eur. Dial. Transpl. Assoc. - Eur. Ren. Assoc 10.1093/ndt/gfx339 [DOI] [PubMed] [Google Scholar]
- Wei W, Keogh MJ, Wilson I, Coxhead J, Ryan S, Rollinson S, Griffin H, Kurzawa-Akanbi M, Santibanez-Koref M, Talbot K, Turner MR, McKenzie C-A, Troakes C, Attems J, Smith C, Al Sarraj S, Morris CM, Ansorge O, Pickering-Brown S, Ironside JW, Chinnery PF, 2017. Mitochondrial DNA point mutations and relative copy number in 1363 disease and control human brains. Acta Neuropathol. Commun 5, 13 10.1186/s40478-016-0404-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weissensteiner H, Forer L, Fuchsberger C, Schöpf B, Kloss-Brandstätter A, Specht G, Kronenberg F, Schönherr S, 2016. mtDNA-Server: next-generation sequencing data analysis of human mitochondrial DNA in the cloud. Nucleic Acids Res 44, W64–69. 10.1093/nar/gkw247 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xing J, Chen M, Wood CG, Lin J, Spitz MR, Ma J, Amos CI, Shields PG, Benowitz NL, Gu J, de Andrade M, Swan GE, Wu X, 2008. Mitochondrial DNA content: its genetic heritability and association with renal cell carcinoma. J. Natl. Cancer Inst 100, 1104–1112. 10.1093/jnci/djn213 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yamada T, Adachi Y, Iijima M, Sesaki H, 2016. Making a Division Apparatus on Mitochondria. Trends Biochem. Sci 41, 209–210. 10.1016/j.tibs.2016.02.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ye W, Tang X, Liu C, Wen C, Li W, Lyu J, 2017. Accurate quantitation of circulating cell-free mitochondrial DNA in plasma by droplet digital PCR. Anal. Bioanal. Chem 409, 2727–2735. 10.1007/s00216-017-0217-x [DOI] [PubMed] [Google Scholar]
- Young MJ, 2017. Off-Target Effects of Drugs that Disrupt Human Mitochondrial DNA Maintenance. Front. Mol. Biosci 4 10.3389/fmolb.2017.00074 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yu EPK, Bennett MR, 2014. Mitochondrial DNA damage and atherosclerosis. Trends Endocrinol. Metab TEM 25, 481–487. 10.1016/j.tem.2014.06.008 [DOI] [PubMed] [Google Scholar]
- Yu M, Zhou Y, Shi Y, Ning L, Yang Y, Wei X, Zhang N, Hao X, Niu R, 2007. Reduced mitochondrial DNA copy number is correlated with tumor progression and prognosis in Chinese breast cancer patients. IUBMB Life 59, 450–457. 10.1080/15216540701509955 [DOI] [PubMed] [Google Scholar]
- Yuan Y, Ju YS, Kim Y, Li J, Wang Y, Yoon CJ, Yang Y, Martincorena I, Creighton CJ, Weinstein JN, Xu Y, Han L, Kim H-L, Nakagawa H, Park K, Campbell PJ, Liang H, 2020. Comprehensive molecular characterization of mitochondrial genomes in human cancers. Nat. Genet 52, 342–352. 10.1038/s41588-019-0557-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang H, Singh KK, 2014. Global Genetic Determinants of Mitochondrial DNA Copy Number. PLoS ONE 9 10.1371/journal.pone.0105242 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang Yiyi, Guallar E, Ashar FN, Longchamps RJ, Castellani CA, Lane J, Grove ML, Coresh J, Sotoodehnia N, Ilkhanoff L, Boerwinkle E, Pankratz N, Arking DE, 2017. Association between mitochondrial DNA copy number and sudden cardiac death: findings from the Atherosclerosis Risk in Communities study (ARIC). Eur. Heart J 10.1093/eurheartj/ehx354 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang Yuheng, Zhao Y, Wen S, Yan R, Yang Q, Chen H, 2017. Associations of mitochondrial haplogroups and mitochondrial DNA copy numbers with end-stage renal disease in a Han population. Mitochondrial DNA Part A 28, 725–731. 10.1080/24701394.2016.1177038 [DOI] [PubMed] [Google Scholar]
