Skip to main content

Cell-free DNA levels associate with COPD exacerbations and mortality

Abstract

The question addressed by the study

Good biological indicators capable of predicting chronic obstructive pulmonary disease (COPD) phenotypes and clinical trajectories are lacking. Because nuclear and mitochondrial genomes are damaged and released by cigarette smoke exposure, plasma cell-free mitochondrial and nuclear DNA (cf-mtDNA and cf-nDNA) levels could potentially integrate disease physiology and clinical phenotypes in COPD. This study aimed to determine whether plasma cf-mtDNA and cf-nDNA levels are associated with COPD disease severity, exacerbations, and mortality risk.

Materials and methods

We quantified mtDNA and nDNA copy numbers in plasma from participants enrolled in the Evaluation of COPD Longitudinally to Identify Predictive Surrogate Endpoints (ECLIPSE, n = 2,702) study and determined associations with relevant clinical parameters.

Results

Of the 2,128 participants with COPD, 65% were male and the median age was 64 (interquartile range, 59–69) years. During the baseline visit, cf-mtDNA levels positively correlated with future exacerbation rates in subjects with mild/moderate and severe disease (Global Initiative for Obstructive Lung Disease [GOLD] I/II and III, respectively) or with high eosinophil count (≥ 300). cf-nDNA positively associated with an increased mortality risk (hazard ratio, 1.33 [95% confidence interval, 1.01–1.74] per each natural log of cf-nDNA copy number). Additional analysis revealed that individuals with low cf-mtDNA and high cf-nDNA abundance further increased the mortality risk (hazard ratio, 1.62 [95% confidence interval, 1.16–2.25] per each natural log of cf-nDNA copy number).

Answer to the question

Plasma cf-mtDNA and cf-nDNA, when integrated into quantitative clinical measurements, may aid in improving COPD severity and progression assessment.

Take-home message

In this COPD cohort, we found that elevated cf-nDNA predicts mortality while elevated cf-mtDNA predicts future exacerbations, with a hazard ratio of 1.3. Integration of both cf-mtDNA and cf-nDNA measures improved mortality predictions to 1.66 in our study.

Introduction

Chronic obstructive pulmonary disease (COPD) is a progressive lung disease characterized by alveolar destruction, airway remodeling, and inflammation [1, 2]. Environmental, behavioral, and genetic factors play pivotal roles in disease onset and progression. Cigarette smoke (CS) exposure is the most prominent risk factor for developing COPD [3].

CS exposure induces oxidative stress and mitochondrial dysfunction that can cause mitochondrial DNA (mtDNA) and nuclear (nDNA) damage and release into the cytosol or extracellular space [4,5,6,7]. Due to its evolution from bacterial origins, mislocalized mtDNA acts as a damage-associated molecular pattern that can promote inflammation by binding to DNA-sensing receptors [8, 9]. Indeed, cell-free mitochondrial (cf-mtDNA) and nuclear DNA (cf-nDNA) have been investigated as biomarkers of disease progression and mortality in critical illness [10] and idiopathic pulmonary fibrosis (IPF) [11, 12]. Cf-mtDNA may be mechanistically linked to pathology, as intratracheal mtDNA administration in mice triggered macrophage infiltration and proinflammatory cytokine production [13]. Mitochondrial dysfunction [5, 14] and cellular necrosis [7] are thought to contribute to COPD pathogenesis and are associated with the release of cf-mtDNA and cf-nDNA into the bloodstream and other compartments [15, 16]. Cf-mtDNA and cf-nDNA have been detected in the bronchoalveolar lavage [17, 18] and serum [6] of mice exposed to CS. Moreover, CS-induced release of DNA from neutrophils and macrophages contributed to the formation of extracellular traps. Decreasing these extracellular traps by intraperitoneal DNase-I reduced lung injury [19].

Despite the current understanding of COPD pathogenesis, there remains a lack of good biological indicators capable of predicting COPD disease phenotypes and clinical trajectories, including acute exacerbations [20,21,22]. The association of novel biomarkers with different COPD stages, phenotypes, and outcomes could provide insight into pathogenesis and the development of targeted therapeutics. The Rome Proposal from COPD panel experts highlighted the need to incorporate predictive biomarkers in assessing acute exacerbations in COPD [23]. A recent study reported that cf-mtDNA levels were higher in the plasma of 700 individuals with mild to moderate COPD in the SPIROMICS cohort [24]. However, these findings have not been validated in a larger, independent cohort. In this study, we tested the association of plasma cf-mtDNA and cf-nDNA levels with clinical measures of COPD severity, rate of exacerbations, and mortality in the Evaluation of COPD Longitudinally to Identify Predictive Surrogate Endpoints (ECLIPSE) cohort.

Methods

Study participants

We analyzed prospectively collected plasma samples from participants enrolled in the ECLIPSE cohort, a longitudinal multicenter study of non-smokers and current or former smokers with and without COPD. The study design and distribution of disease have been previously described [25, 26]. Details of the study design and clinical parameters collected are summarized in the online Supplementary Information.

Automated cell-free DNA (cf-DNA) isolation from plasma

Total cf-DNA was isolated from plasma using a previously validated automated, high throughput methodology [27]. A detailed description of cf-DNA isolation and quantitation from plasma are provided in the online Supplementary Information.

Quantitative polymerase chain reaction (qPCR) and digital polymerase chain reaction (dPCR)

Cf-mtDNA and cf-nDNA levels were measured simultaneously by TaqMan-based duplex qPCR as previously described [27]. The assay quantified mitochondrial-encoded human NADH:ubiquinone oxidoreductase core subunit 1 (ND1) and nuclear-encoded human beta-2-microglobulin (B2M). Serial dilutions of pooled human placenta DNA were used as a standard curve as we previously described [28]. Assay sequences (Supplementary Table S1) are provided in the online Supplementary Information.

mtDNA and nDNA copy numbers (copies/µL) of the standard curve were determined by dPCR as previously described [29] and used to calculate copy numbers for the experimental samples by linear regression. Using the QuantStudio 3D Digital PCR System and associated reagents (Thermo Fisher, Waltham, Massachusetts, USA), mtDNA and nDNA copy numbers were measured separately using singleplex ND1 and B2M assays. All reactions were performed in duplicate (two chips). Details of both qPCR and dPCR methods are provided in the Supplementary Information.

Statistical analysis

All analyses regarding the ECLIPSE cohort were performed using Stata version 16.0 (StataCorp). Continuous variables were presented as the median (interquartile range [IQR]) in three study subject groups and tested using ANOVA. We used natural logarithmic transformation of cf-mtDNA and cf-nDNA to optimally transform the data to a normal distribution for statistical comparison. In Figs. 13, cf-DNA levels were transformed back to nominal data (copies/µL) for ease of interpretation. Categorical variables were presented as frequencies and tested using Fisher’s exact test.

We applied linear regression analysis to calculate the unadjusted and adjusted (for sex, body mass index, and age) standardized regression (correlation) coefficient of continuous outcomes with cf-mtDNA and cf-nDNA levels (with natural log transformation). For binary outcomes, we used logistic regression analysis. We noted that the rate of exacerbation was positively skewed (mean = 1.2, SD = 1.4, P < 0.001 against a normal distribution). Therefore, we used the generalized linear model with gamma family distribution with a log link and robust variance estimation to analyze the association between cf-mtDNA and cf-nDNA levels and the rate of exacerbations. This model is appropriate for non-negative data with heteroscedasticity. Time to mortality was calculated using the Cox proportional regression analysis. Hazard ratio (95% confidence interval [CI]) was reported for high cf-mtDNA and cf-nDNA using a median level in all COPD patients. The assumption of Cox analysis was assessed and verified. We did not adjust the P values for multiple hypothesis testing. We did not adjust for missing outcomes because the effects were negligible in most cases.

Results

Characteristics of the tested cohort

The ECLIPSE cohort subgroups and their clinical characteristics are shown in Supplementary Figure S1 and Table 1, respectively. From the 3,186 samples provided, 484 were excluded due to insufficient plasma volume. The remaining 2,702 subjects were separated into 243 non-smokers, 331 current or former smokers without airflow obstruction, or 2,128 current or former smokers with COPD.

The ECLIPSE participants were recruited from United States of America, Canada, and Europe. The subjects were primarily white (97–98%) (Table 1), preventing the detection of socio-racial contributions. Compared to non-smokers and smokers, individuals with COPD were predominantly older males with a lower body mass index (BMI), which was adjusted for in subsequent analyses. Each participant with COPD was assigned a Global Initiative for Obstructive Lung Disease (GOLD) stage based on the severity of their airway limitation [30]. One subject was GOLD stage I (0.1%, mild disease), 943 individuals were stage II (44%, moderate disease), 893 were stage III (42%, severe disease), and 290 were stage IV (14%, very severe disease) (Table 1, Supplementary Figure S1). GOLD stages I and II were grouped for statistical analysis. At baseline, the median FEV1% of the COPD group was 47% (interquartile range [IQR], 36–61%) (Table 1). As expected, packs per year, FEV1% [31, 32], FVC% [31, 32], FEV1/FVC [31, 32], eosinophil blood counts [33], St. George’s Respiratory Questionnaire (SGRQ) total score [31], and emphysema were significantly different in the COPD group compared to both control groups (P < 0.001).

Table 1 Baseline characteristics and cell-free DNA levels of study participants

cf-mtDNA and cf-nDNA copy number in plasma

To determine if either cf-mtDNA or cf-nDNA levels in the plasma were significantly different between subgroups, cf-DNA was isolated in a blinded fashion from plasma samples collected during the baseline visit. Then, cf-mtDNA and cf-nDNA levels were determined by quantitative PCR followed by interpolation to a standard curve. Digital PCR (dPCR) was used to determine the copies/µL of the standard curve. Copies/µL of the experimental samples were interpolated from the standard curve by linear regression calculations. After adjusting for age, sex, and BMI, cf-mtDNA levels were significantly higher in the plasma of participants with COPD and smokers than non-smokers (P < 0.001, Fig. 1A). There was no difference in cf-mtDNA copy levels between COPD subjects and smokers. The median cf-mtDNA level was 1,042 copies/µL (IQR, 262-3,266 copies/µL) for COPD participants, 1,381 copies/µL (IQR, 563-2,933 copies/µL) for smokers, and 367 copies/µL (IQR, 91 − 1,299 copies/µL) for non-smokers (Table 1).

Fig. 1
figure 1

cf-mtDNA levels are higher in participants with COPD. Baseline (A) cf-mtDNA and (B) cf-nDNA levels were assessed in participants with COPD (n = 2,128), smokers (n = 331), and non-smokers (n = 243). The results are presented as median, with the box indicating Q1 and Q3 quartiles and the whiskers indicating the range. To format the y-axis, we used the natural logarithmic scale of data and then transformed it back to nominal data for ease of interpretation. Statistical analysis was performed using ANOVA with Bonferroni post hoc pairwise comparison. ***P value < 0.001. cf-mtDNA = cell-free mitochondrial DNA; cf-nDNA = cell-free nuclear DNA; COPD = chronic obstructive pulmonary disease

cf-nDNA copies were significantly higher in subjects with COPD or non-smokers than smokers (P < 0.001, Fig. 1B). In contrast, there was no difference in median cf-nDNA copy levels between individuals with COPD and non-smokers. The median cf-nDNA level was 0.54 copies/µL (IQR, 0.31–0.92 copies/µL) for COPD participants, 0.34 copies/µL (IQR, 0.20–0.61 copies/µL) for smokers, and 0.49 copies/µL (IQR, 0.29–0.80 copies/µL) for non-smokers (Table 1).

Associations between cf-mtDNA and cf-nDNA copies in plasma and clinical variables

To determine if COPD severity was associated with higher cf-DNA levels in this cohort, the participants were stratified by GOLD stage. The median cf-mtDNA level for mild/moderate (GOLD I/II) COPD cases was 1,220 copies/µL (IQR, 334-3,639), 911 copies/µL (IQR, 219-3,151) for severe (GOLD III), and 901 copies/µL (IQR, 244-2,689) for very severe (GOLD IV) airway obstruction (Table 2). cf-mtDNA levels were significantly increased in GOLD I/II cases compared to GOLD III (P < 0.05, Fig. 2A). There was no statistically significant difference in cf-mtDNA levels between GOLD I/II and GOLD IV groups or between GOLD III and GOLD IV groups. The median cf-nDNA level for GOLD I/II cases was 0.5 copies/µL (IQR, 0.3–0.8), 0.5 copies/µL (IQR, 0.3–0.9) for GOLD III, and 0.7 copies/µL (IQR, 0.4–1.3) for GOLD IV (Table 2). In contrast, cf-nDNA levels were significantly increased in samples collected from GOLD IV cases compared to either GOLD I/II or III cases (P values < 0.001, Fig. 2B). There was no difference between the levels of cf-nDNA in GOLD I/II and III groups.

Table 2 Baseline cell-free DNA Levels by GOLD Stage
Fig. 2
figure 2

cf-mtDNA levels are higher in participants classified as GOLD stage I/II while cf-nDNA levels are higher in participants classified as GOLD stage IV. Baseline (A) cf-mtDNA and (B) cf-nDNA levels were assessed in participants with COPD who were classified as GOLD stage I/II (n = 944), III (n = 893), or IV (n = 290). The results are presented as median, with the box indicating Q1 and Q3 quartiles and the whiskers indicating range. To format the y-axis, we used the natural logarithmic scale of data and then transformed it back to nominal data for ease of interpretation. Statistical analysis was performed using ANOVA with Bonferroni post hoc pairwise comparison. *P value < 0.05, ***P value < 0.001. cf-mtDNA = cell-free mitochondrial DNA; cf-nDNA = cell-free nuclear DNA; COPD = chronic obstructive pulmonary disease; GOLD = Global Initiative for Obstructive Lung Disease

We then sought to determine if either cf-mtDNA or cf-nDNA levels were associated with other clinical variables in the COPD group. The correlations (both unadjusted and adjusted) are presented in Table 3. The adjustment for age, sex, and BMI did not influence the associations for either DNA measure. cf-mtDNA levels were not associated with FEV1%, FEV1/FVC, the body mass index, airflow obstruction, dyspnea, and exercise (BODE) index, or SGRQ total score (Table 3). However, cf-mtDNA levels were positively correlated with FVC% (r = 0.05, P = 0.026), the Borg exertional dyspnea scale (r = 0.18, P < 0.001), and the number of prior exacerbations that occurred the year before the first study visit (r = 0.05, P = 0.031) and negatively correlated with GOLD stage (r = -0.06, P = 0.009) and 6-minute walk distance (6MWD) (r = -0.04, P = 0.035). cf-nDNA levels were negatively correlated with FEV1% (r = -0.13, P < 0.001), FVC% (r = -0.10, P < 0.001), FEV1/FVC (r = -0.10, P < 0.001), and the 6MWD (r = -0.13, P < 0.001). cf-nDNA levels were positively correlated with GOLD stage (r = 0.13, P < 0.001), the BODE index (r = 0.15, P < 0.001), SGRQ total score (r = 0.12, P < 0.001), Borg scale (r = 0.07, P = 0.029), and the number of prior exacerbations (r = 0.06, P = 0.005). Neither cf-mtDNA nor cf-nDNA levels were associated with the presence of emphysema.

Table 3 Correlation (P value) between baseline DNA measures and clinical parameters in participants with COPD

Associations between cf-mtDNA copies, rate of future exacerbations, and eosinophil count

COPD exacerbations were defined as a clinical episode of worsening pulmonary symptoms requiring systemic corticosteroids, antibiotics, or both [34]. The average rate of exacerbations over the three-year study period was provided in the clinical data. After adjusting for age, sex, and BMI, regression analysis showed that higher levels of cf-mtDNA were associated with an increased rate of exacerbations for the 3-year study (Fig. 3A). The ratio of the mean from the gamma regression was 1.03, indicating a 3% increase in exacerbations per each natural log increase of mtDNA (P = 0.021). A similar result arose from unadjusted data (ratio of the mean, 1.03, P = 0.030, data not shown). An association with exacerbation rate was not observed for cf-nDNA levels (Fig. 3B).

Fig. 3
figure 3

Higher cf-mtDNA levels at baseline are associated with an increased rate of yearly exacerbations while cf-nDNA levels are associated with an increased risk of mortality in participants with COPD. Baseline (A) cf-mtDNA and (B) cf-nDNA levels were evaluated for their association with the average number of exacerbations per year during the 3-year study period in all participants with COPD (n = 2,106). The results are presented as adjusted mean with 95% confidence intervals. Statistical analysis was performed using gamma regression after adjusting for age, sex, and body mass index. In (A) and (B), we used the natural logarithmic scale of data and then transformed back the x-axis to nominal data for ease of interpretation. Baseline (C) cf-mtDNA and (D) cf-nDNA levels were evaluated for their association with the risk of mortality in all participants with COPD (n = 2,126). Statistical analysis was performed using Cox proportional hazard regression. cf-mtDNA = cell-free mitochondrial DNA; cf-nDNA = cell-free nuclear DNA; COPD = chronic obstructive pulmonary disease; HR = hazard ratio

We next examined the association between each cf-DNA measure and the rate of exacerbations by GOLD stage (Table 4). Participants classified as GOLD I/II or GOLD III with higher cf-mtDNA levels experienced more exacerbations in the time between visits (ratio of the mean, 1.06, P = 0.011 for GOLD I/II and ratio of the mean, 1.05, P value = 0.012 for GOLD III). However, this association was not present in participants classified as GOLD IV. No associations were observed for cf-nDNA levels.

Table 4 Effects of DNA measures on the rate of exacerbations by GOLD stage

Using established thresholds [35], 1,640 COPD participants had normal eosinophil levels (< 300 cells/µL) and 430 had elevated levels (≥ 300 cells/µL) at the baseline visit. COPD participants with higher levels of cf-mtDNA and a high eosinophil count experienced an increased rate of exacerbations (ratio of the mean, 1.09, P = 0.007) (Table 5). This trend was not observed in COPD participants with a low eosinophil count. No association was observed for cf-nDNA levels.

Table 5 Effects of DNA measures on the rate of exacerbations by eosinophil count

Association between cf-nDNA copies and mortality

All-cause mortality was documented up to day 1,060 of the study. The cause of death was not further investigated [34]. In adjusted analyses, we observed no association between cf-mtDNA levels and mortality risk (Fig. 3C). In contrast, there was a significant association between higher cf-nDNA levels and decreased survival (hazard ratio [HR], 1.33, 95% confidence interval [CI], 1.01–1.74, P = 0.042) (Fig. 3D), indicating that there was 33% increase in mortality for each natural log increase of cf-nDNA.

Combined analysis of relative cf-mtDNA and cf-nDNA copies enhances survival prediction

Because cf-mtDNA and cf-nDNA demonstrated a reciprocal relationship regarding COPD severity (Table 2; Fig. 2) and are associated with distinct clinical parameters (exacerbation rate and mortality, respectively), we wanted to determine whether the combination of these measures would improve the prediction of survival. We established quartile assignments for cf-mtDNA and cf-nDNA levels for all participant samples and calculated the frequency of being in the highest quartile for each GOLD stage (Fig. 4A). The frequency of samples being in the top quartile for cf-nDNA increased as disease stage increased (P < 0.001), while the frequency for cf-mtDNA decreased (P = 0.040). Thus, having high cf-mtDNA and low cf-nDNA was a common feature of GOLD stage I/II, whereas having low cf-mtDNA and high cf-nDNA was a common feature of GOLD stage IV. Using low cf-mtDNA and low cf-nDNA as a neutral reference group, we calculated the survival HR for the other combinations (Fig. 4B). The HR for the high cf-mtDNA and low cf-nDNA group was 0.5 (95% CI, 0.29–0.84, overall P < 0.001), indicating that participants with this characteristic had twice the survival rate compared to the neutral reference group. Notably, the low cf-mtDNA and high cf-nDNA group showed worse outcomes, with a HR of 1.62 (95% CI, 1.16–2.25, overall P < 0.001).

Fig. 4
figure 4

Combined measures of low cf-mtDNA and high cf-nDNA levels are associated with increased mortality risk. (A) Samples across the COPD population were binned into quartiles. The frequency of the top quartile of cf-mtDNA and cf-nDNA was determined across GOLD stages. Statistical analysis was performed using test for linear trend. (B) Low and high quartiles of cf-mtDNA and cf-nDNA were combined, demonstrating an increase in the strength of the association with mortality. cf-mtDNA = cell-free mitochondrial DNA; cf-nDNA = cell-free nuclear DNA; GOLD = Global Initiative for Obstructive Lung Disease; HR = hazard ratio

Discussion

Quantifiable indicators that associate with COPD pathogenesis are needed to classify endotypes, predict outcomes, and assess treatment responsiveness. The incorporation of new biomarkers for the prediction of acute exacerbations was highlighted in the Rome Proposal by a panel of COPD experts [23]. The ability to assess the trajectories of COPD endotypes will involve recognizing associated biomarkers. Several attempts to categorize patients based on clinical and radiologic parameters have been made; however, there remains a missing link between such parameters and underlying disease mechanisms [36, 37].

In a subset of the SPIROMICS cohort, plasma cf-mtDNA was elevated in patients with mild to moderate COPD compared to severe COPD [24]. Initially, mtDNA was associated with exacerbations and clinical outcomes (6MWD, SGRQ total score, or COPD Assessment Test score) but was no longer significant after adjusting for loss of follow-up data. In the same cohort, higher urine mtDNA was associated with increased symptoms and worse exercise tolerance [31]. In a separate, larger cohort, our study validates the finding that cf-mtDNA was higher in the plasma of patients with mild to moderate COPD. Moreover, our study showed that elevated cf-mtDNA was positively associated with FVC%, the Borg exertional dyspnea scale, and number of prior exacerbations and negatively associated with GOLD stage and 6MWD. Furthermore, cf-mtDNA predicted future exacerbations, particularly in GOLD I-III participants or those with high eosinophil counts, suggesting its potential utility as a stratifying marker of at-risk subgroups for exacerbations. Cf-mtDNA has proinflammatory implications [13] and therefore may promote chronic inflammatory responses and exacerbations in COPD.

Neither study involving the SPIROMICS cohort reported cf-nDNA levels. In the present study, we showed that cf-nDNA was negatively associated with FEV1%, FVC%, FEV1/FVC, and 6MWD and positively associated with GOLD stage, BODE index, SGRQ total score, the Borg scale, number of prior exacerbations, and an increased risk for mortality. While cf-mtDNA is predictive of exacerbations, we note that it is not associated with increased mortality risk and the cf-mtDNA data seemingly separate exacerbations from survival. Integrating both cf-mtDNA and cf-nDNA measures improved mortality predictions in our study. Specifically, participants with low cf-mtDNA and high cf-nDNA levels showed worse outcomes among patients with COPD. As such, multi-factor assessments are likely necessary to refine risk prediction. The distinct predictive nature of integrated cf-mtDNA and cf-nDNA measurements should be investigated in additional COPD patient cohorts stratified by endotypes, such as early age-onset COPD, those with rapid decline in lung function, or frequent exacerbations.

cf-nDNA is passively released during cell death processes such as apoptosis or necrosis [15, 38]. In contrast, cf-mtDNA may result from active release mechanisms, including microvesicles [6, 39]. Cardiomyocytes release mitochondria-containing extracellular vesicles (EV) in response to stimulation with isoproterenol [40]. Therefore, some types of EV release may represent a compensatory mechanism in response to acute cellular damage that overwhelms the normal degradation pathways. Such damage leads to the redirection of endomembrane lysosomal trafficking to secretory pathways, where resident macrophages digest the contents of EVs. We suggest that the high levels of plasma cf-mtDNA in COPD may represent a compensatory response to smoke exposure, whereby systemic cellular damage triggers microparticle release. This cellular adaptation could prevent cell death, characterized by low cf-nDNA release in the plasma, as observed in this study. Consistent with this idea, the high cf-mtDNA and low cf-nDNA group had an HR of 0.5 relative to the reference group, suggesting a significant benefit from the potentially compensatory state. In contrast, low cf-mtDNA would indicate poor adaptation to stress with high cf-nDNA levels indicating increased cell death. The low cf-mtDNA/high cf-nDNA group had an HR of 1.62, which poses a significantly higher lethality than other groups. We acknowledge that this is a hypothetical scenario and may not represent the response of all cell types or cell-free DNA production mechanisms.

In summary, higher cf-mtDNA levels were associated with future exacerbations, while higher cf-nDNA levels were associated with increased mortality risk in participants with COPD. Future studies should evaluate changes in both cell-free genomes in the context of active exacerbations or specific treatments.

Data availability

The clinical data used for this study can be accessed through dbGaP Study Accession: phs001252.v1.p1. While the data are not publicly available, interested parties can apply to the pertinent data providers for data access requests.

References

  1. Mathers CD, Loncar D. Projections of global mortality and burden of Disease from 2002 to 2030. PLoS Med. 2006;3:e442.

    Article  PubMed  PubMed Central  Google Scholar 

  2. World Health Organization, Updated who projections of mortality and causes of. death 2016-2060 [Internet]. Updated who projections of mortality and causes of death 2016–2060 2018 [cited 2021 Jun 28].Available from: https://www.who.int/healthinfo/global_burden_disease/projections_method.pdf?ua=1.

  3. 2020 GLOBAL Global Initiative for chronic obstructive lung disease, strategy for prevention, diagnosis and management of COPD [Internet]. 2020 [cited 2021 Jun 29].Available from: https://goldcopd.org/gold-reports/.

  4. Cloonan SM, Choi AMK. Mitochondria in lung disease. J Clin Invest. 2016;126:809–20.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Mizumura K, Cloonan SM, Nakahira K, Bhashyam AR, Cervo M, Kitada T, Glass K, Owen CA, Mahmood A, Washko GR, Hashimoto S, Ryter SW, Choi AMK. Mitophagy-dependent necroptosis contributes to the pathogenesis of COPD. J Clin Invest. 2014;124:3987–4003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Giordano L, Gregory AD, Pérez Verdaguer M, Ware SA, Harvey H, DeVallance E, Brzoska T, Sundd P, Zhang Y, Sciurba FC, Shapiro SD, Kaufman BA. Extracellular release of mitochondrial DNA: triggered by cigarette smoke and detected in COPD. Cells 2022; 11.

  7. van der Toorn M, Slebos D-J, de Bruin HG, Leuvenink HG, Bakker SJL, Gans ROB, Koëter GH, van Oosterhout AJM, Kauffman HF. Cigarette smoke-induced blockade of the mitochondrial respiratory chain switches lung epithelial cell apoptosis into necrosis. Am J Physiol Lung Cell Mol Physiol. 2007;292:L1211–8.

    Article  PubMed  Google Scholar 

  8. Schumacker PT, Gillespie MN, Nakahira K, Choi AMK, Crouser ED, Piantadosi CA, Bhattacharya J. Mitochondria in lung biology and pathology: more than just a powerhouse. Am J Physiol Lung Cell Mol Physiol. 2014;306:L962–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Benmerzoug S, Ryffel B, Togbe D, Quesniaux VFJ. Self-DNA sensing in lung inflammatory diseases. Trends Immunol. 2019;40:719–34.

    Article  CAS  PubMed  Google Scholar 

  10. Harrington JS, Huh J-W, Schenck EJ, Nakahira K, Siempos II, Choi AMK. Circulating mitochondrial DNA as predictor of mortality in critically ill patients: a systematic review of clinical studies. Chest. 2019;156:1120–36.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Ryu C, Sun H, Gulati M, Herazo-Maya JD, Chen Y, Osafo-Addo A, Brandsdorfer C, Winkler J, Blaul C, Faunce J, Pan H, Woolard T, Tzouvelekis A, Antin-Ozerkis DE, Puchalski JT, Slade M, Gonzalez AL, Bogenhagen DF, Kirillov V, Feghali-Bostwick C, Gibson K, Lindell K, Herzog RI, Dela Cruz CS, Mehal W, Kaminski N, Herzog EL, Trujillo G. Extracellular mitochondrial DNA is generated by fibroblasts and predicts death in Idiopathic Pulmonary Fibrosis. Am J Respir Crit Care Med. 2017;196:1571–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Bueno M, Zank D, Buendia-Roldán I, Fiedler K, Mays BG, Alvarez D, Sembrat J, Kimball B, Bullock JK, Martin JL, Nouraie M, Kaufman BA, Rojas M, Pardo A, Selman M, Mora AL. PINK1 attenuates mtDNA release in alveolar epithelial cells and TLR9 mediated profibrotic responses. PLoS ONE. 2019;14:e0218003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Gu X, Wu G, Yao Y, Zeng J, Shi D, Lv T, Luo L, Song Y. Intratracheal administration of mitochondrial DNA directly provokes lung inflammation through the TLR9-p38 MAPK pathway. Free Radic Biol Med. 2015;83:149–58.

    Article  CAS  PubMed  Google Scholar 

  14. Cloonan SM, Glass K, Laucho-Contreras ME, Bhashyam AR, Cervo M, Pabón MA, Konrad C, Polverino F, Siempos II, Perez E, Mizumura K, Ghosh MC, Parameswaran H, Williams NC, Rooney KT, Chen Z-H, Goldklang MP, Yuan G-C, Moore SC, Demeo DL, Rouault TA, D’Armiento JM, Schon EA, Manfredi G, Quackenbush J, Mahmood A, Silverman EK, Owen CA, Choi AMK. Mitochondrial iron chelation ameliorates cigarette smoke-induced Bronchitis and Emphysema in mice. Nat Med. 2016;22:163–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. De Gaetano A, Solodka K, Zanini G, Selleri V, Mattioli AV, Nasi M, Pinti M. Molecular mechanisms of mtDNA-Mediated inflammation. Cells. 2021. 10.

  16. Kustanovich A, Schwartz R, Peretz T, Grinshpun A. Life and death of circulating cell-free DNA. Cancer Biol Ther. 2019;20:1057–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Szczesny B, Marcatti M, Ahmad A, Montalbano M, Brunyánszki A, Bibli S-I, Papapetropoulos A, Szabo C. Mitochondrial DNA damage and subsequent activation of Z-DNA binding protein 1 links oxidative stress to inflammation in epithelial cells. Sci Rep. 2018;8:914.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Nascimento M, Gombault A, Lacerda-Queiroz N, Panek C, Savigny F, Sbeity M, Bourinet M, Le Bert M, Riteau N, Ryffel B, Quesniaux VFJ, Couillin I. Self-DNA release and STING-dependent sensing drives inflammation to cigarette smoke in mice. Sci Rep. 2019;9:14848.

    Article  PubMed  PubMed Central  Google Scholar 

  19. King PT, Sharma R, O’Sullivan KM, Callaghan J, Dousha L, Thomas B, Ruwanpura S, Lim S, Farmer MW, Jennings BR, Finsterbusch M, Brooks G, Selemidis S, Anderson GP, Holdsworth SR, Bardin PG. Deoxyribonuclease 1 reduces pathogenic effects of cigarette smoke exposure in the lung. Sci Rep. 2017;7:12128.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Han MK, Agusti A, Calverley PM, Celli BR, Criner G, Curtis JL, Fabbri LM, Goldin JG, Jones PW, Macnee W, Make BJ, Rabe KF, Rennard SI, Sciurba FC, Silverman EK, Vestbo J, Washko GR, Wouters EFM, Martinez FJ. Chronic Obstructive Pulmonary Disease phenotypes: the future of COPD. Am J Respir Crit Care Med. 2010;182:598–604.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Stockley RA, Halpin DMG, Celli BR, Singh D. Chronic Obstructive Pulmonary Disease biomarkers and their interpretation. Am J Respir Crit Care Med. 2019;199:1195–204.

    Article  CAS  PubMed  Google Scholar 

  22. Han MK, Postma D, Mannino DM, Giardino ND, Buist S, Curtis JL, Martinez FJ. Gender and Chronic Obstructive Pulmonary Disease: why it matters. Am J Respir Crit Care Med. 2007;176:1179–84.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Celli BR, Fabbri LM, Aaron SD, Agusti A, Brook R, Criner GJ, Franssen FME, Humbert M, Hurst JR, O’Donnell D, Pantoni L, Papi A, Rodriguez-Roisin R, Sethi S, Torres A, Vogelmeier CF, Wedzicha JA. An updated definition and severity classification of Chronic Obstructive Pulmonary Disease exacerbations: the rome proposal. Am J Respir Crit Care Med. 2021;204:1251–8.

    Article  PubMed  Google Scholar 

  24. Zhang WZ, Hoffman KL, Schiffer KT, Oromendia C, Rice MC, Barjaktarevic I, Peters SP, Putcha N, Bowler RP, Wells JM, Couper DJ, Labaki WW, Curtis JL, Han MK, Paine R, Woodruff PG, Criner GJ, Hansel NN, Diaz I, Ballman KV, Nakahira K, Choi ME, Martinez FJ, Choi AMK, Cloonan SM. Association of plasma mitochondrial DNA with COPD severity and progression in the SPIROMICS cohort. Respir Res. 2021;22:126.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Vestbo J, Anderson W, Coxson HO, Crim C, Dawber F, Edwards L, Hagan G, Knobil K, Lomas DA, MacNee W, Silverman EK, Tal-Singer R. ECLIPSE investigators. Evaluation of COPD longitudinally to identify predictive surrogate end-points (ECLIPSE). Eur Respir J. 2008;31:869–73.

    Article  CAS  PubMed  Google Scholar 

  26. Agusti A, Calverley PMA, Celli B, Coxson HO, Edwards LD, Lomas DA, MacNee W, Miller BE, Rennard S, Silverman EK, Tal-Singer R, Wouters E, Yates JC, Vestbo J. Evaluation of COPD longitudinally to identify predictive surrogate endpoints (ECLIPSE) investigators. Characterisation of COPD heterogeneity in the ECLIPSE cohort. Respir Res. 2010;11:122.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Ware SA, Desai N, Lopez M, Leach D, Zhang Y, Giordano L, Nouraie M, Picard M, Kaufman BA. An automated, high-throughput methodology optimized for quantitative cell-free mitochondrial and nuclear DNA isolation from plasma. J Biol Chem. 2020;295:15677–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Trumpff C, Marsland AL, Basualto-Alarcón C, Martin JL, Carroll JE, Sturm G, Vincent AE, Mosharov EV, Gu Z, Kaufman BA, Picard M. Acute psychological stress increases serum circulating cell-free mitochondrial DNA. Psychoneuroendocrinology. 2019;106:268–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Belmonte FR, Martin JL, Frescura K, Damas J, Pereira F, Tarnopolsky MA, Kaufman BA. Digital PCR methods improve detection sensitivity and measurement precision of low abundance mtDNA deletions. Sci Rep. 2016;6:25186.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Rabe KF, Hurd S, Anzueto A, Barnes PJ, Buist SA, Calverley P, Fukuchi Y, Jenkins C, Rodriguez-Roisin R, van Weel C, Zielinski J, Global Initiative for Chronic Obstructive Lung Disease. Global strategy for the diagnosis, management, and prevention of Chronic Obstructive Pulmonary Disease: GOLD executive summary. Am J Respir Crit Care Med. 2007;176:532–55.

    Article  PubMed  Google Scholar 

  31. Zhang WZ, Rice MC, Hoffman KL, Oromendia C, Barjaktarevic IZ, Wells JM, Hastie AT, Labaki WW, Cooper CB, Comellas AP, Criner GJ, Krishnan JA, Paine R, Hansel NN, Bowler RP, Barr RG, Peters SP, Woodruff PG, Curtis JL, Han MK, Ballman KV, Martinez FJ, Choi AM, Nakahira K, Cloonan SM, Choi ME, SPIROMICS Investigators. Association of urine mitochondrial DNA with clinical measures of COPD in the SPIROMICS cohort. JCI Insight 2020; 5.

  32. Vestbo J, Hurd SS, Agustí AG, Jones PW, Vogelmeier C, Anzueto A, Barnes PJ, Fabbri LM, Martinez FJ, Nishimura M, Stockley RA, Sin DD, Rodriguez-Roisin R. Global strategy for the diagnosis, management, and prevention of Chronic Obstructive Pulmonary Disease: GOLD executive summary. Am J Respir Crit Care Med. 2013;187:347–65.

    Article  CAS  PubMed  Google Scholar 

  33. Tashkin DP, Wechsler ME. Role of eosinophils in airway inflammation of Chronic Obstructive Pulmonary Disease. Int J Chron Obstruct Pulmon Dis. 2018;13:335–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Celli BR, Locantore N, Yates J, Tal-Singer R, Miller BE, Bakke P, Calverley P, Coxson H, Crim C, Edwards LD, Lomas DA, Duvoix A, MacNee W, Rennard S, Silverman E, Vestbo J, Wouters E, Agustí A, ECLIPSE Investigators. Inflammatory biomarkers improve clinical prediction of mortality in Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med. 2012;185:1065–72.

    Article  CAS  PubMed  Google Scholar 

  35. Jabarkhil A, Moberg M, Janner J, Petersen MN, Jensen CB, Henrik Äangquist L, Vestbo J, Jess T, Porsbjerg C. Elevated blood eosinophils in acute COPD exacerbations: better short- and long-term prognosis. Eur Clin Respir J. 2020;7:1757274.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Burgel PR, Paillasseur JL, Caillaud D, Tillie-Leblond I, Chanez P, Escamilla R, Court-Fortune I, Perez T, Carré P, Roche N. Initiatives BPCO Scientific Committee. Clinical COPD phenotypes: a novel approach using principal component and cluster analyses. Eur Respir J. 2010;36:531–9.

    Article  PubMed  Google Scholar 

  37. Lange P, Celli B, Agustí A, Boje Jensen G, Divo M, Faner R, Guerra S, Marott JL, Martinez FD, Martinez-Camblor P, Meek P, Owen CA, Petersen H, Pinto-Plata V, Schnohr P, Sood A, Soriano JB, Tesfaigzi Y, Vestbo J. Lung-function trajectories leading to Chronic Obstructive Pulmonary Disease. N Engl J Med. 2015;373:111–22.

    Article  CAS  PubMed  Google Scholar 

  38. McIlroy DJ, Bigland M, White AE, Hardy BM, Lott N, Smith DW, Balogh ZJ. Cell necrosis-independent sustained mitochondrial and nuclear DNA release following trauma Surgery. J Trauma Acute Care Surg. 2015;78:282–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Trumpff C, Michelson J, Lagranha CJ, Taleon V, Karan KR, Sturm G, Lindqvist D, Fernström J, Moser D, Kaufman BA, Picard M. Stress and circulating cell-free mitochondrial DNA: a systematic review of human studies, physiological considerations, and technical recommendations. Mitochondrion. 2021;59:225–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Nicolás-Ávila JA, Lechuga-Vieco AV, Esteban-Martínez L, Sánchez-Díaz M, Díaz-García E, Santiago DJ, Rubio-Ponce A, Li JL, Balachander A, Quintana JA, Martínez-de-Mena R, Castejón-Vega B, Pun-García A, Través PG, Bonzón-Kulichenko E, García-Marqués F, Cussó L, A-González N, González-Guerra A, Roche-Molina M, Martin-Salamanca S, Crainiciuc G, Guzmán G, Larrazabal J, Herrero-Galán E, Alegre-Cebollada J, Lemke G, Rothlin CV, Jimenez-Borreguero LJ, Reyes G, et al. A network of macrophages supports mitochondrial homeostasis in the heart. Cell. 2020;183:94–109e23.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

A special thanks to the patients and scientists who participated in the ECLIPSE study.

Funding

Funding for this study was from GlaxoSmithKline (GSK) under a sponsored research agreement. GSK has not modified the results in any way.

Author information

Authors and Affiliations

Authors

Contributions

BAK and WLR contributed to the conception of the study. BAK, SMN, WLR, and SB designed the study. WLR and SB contributed to acquiring samples and clinical parameters. SAW performed DNA isolation and qPCR of the ECLIPSE samples. LG and KMR performed cigarette smoke extract experiments. SMN performed the statistical analysis. FS and YZ provided early preclinical samples to promote the study and consulted on manuscript development. CRK, SAW, SMN, LG, and BAK wrote the manuscript. CRK, SMN, and FS provided clinical oversight.

Corresponding authors

Correspondence to S. Mehdi Nouraie or Brett A. Kaufman.

Ethics declarations

Ethics approval and consent to participate

The human biological samples were sourced ethically in accordance with the Declaration of Helsinki, and their research use was in accord with the terms of the informed consent under IRB/EC-approved protocols in the original studies. Current work was University of Pittsburgh IRB exempt and the data met GDPR-requirements.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

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

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1:

Supplementary Methods

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

Ware, S.A., Kliment, C.R., Giordano, L. et al. Cell-free DNA levels associate with COPD exacerbations and mortality. Respir Res 25, 42 (2024). https://0-doi-org.brum.beds.ac.uk/10.1186/s12931-023-02658-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/s12931-023-02658-1