Next Article in Journal
Effects of Omega-3 and Antioxidant Cocktail Supplement on Prolonged Bed Rest: Results from Serum Proteome and Sphingolipids Analysis
Next Article in Special Issue
HIV and Schistosoma Co-Exposure Leads to Exacerbated Pulmonary Endothelial Remodeling and Dysfunction Associated with Altered Cytokine Landscape
Previous Article in Journal
Tissue-Based Proteomic Profiling in Patients with Hyperplasia and Endometrial Cancer
Previous Article in Special Issue
Methodological Progress of Stereology in Cardiac Research and Its Application to Normal and Pathological Heart Development
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Brief Report

Association of Clonal Hematopoiesis of Indeterminate Potential with Inflammatory Gene Expression in Patients with COPD

by
Stefan Kuhnert
1,
Siavash Mansouri
2,
Michael A. Rieger
3,4,
Rajkumar Savai
1,2,4,5,
Edibe Avci
2,
Gabriela Díaz-Piña
2,
Manju Padmasekar
2,
Mario Looso
2,
Stefan Hadzic
1,
Till Acker
6,
Stephan Klatt
7,
Jochen Wilhelm
1,5,
Ingrid Fleming
7,
Natascha Sommer
1,
Norbert Weissmann
1,5,
Claus Vogelmeier
8,
Robert Bals
9,
Andreas Zeiher
10,
Stefanie Dimmeler
11,
Werner Seeger
1,2,5 and
Soni S. Pullamsetti
1,2,5,*
add Show full author list remove Hide full author list
1
University of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Justus Liebig University, 35392 Giessen, Germany
2
Max Planck Institute for Heart and Lung Research, DZL, CPI, 61231 Bad Nauheim, Germany
3
Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, CPI, Goethe University, 60596 Frankfurt am Main, Germany
4
Frankfurt Cancer Institute (FCI), CPI, Goethe University, 60596 Frankfurt am Main, Germany
5
Institute for Lung Health (ILH), Justus Liebig University, 35392 Giessen, Germany
6
Institute for Neuropathology, CPI, Justus Liebig University, 35392 Giessen, Germany
7
Institute of Vascular Signalling, Department of Molecular Medicine, CPI, Goethe University, 60596 Frankfurt am Main, Germany
8
Department of Medicine, Pulmonary and Critical Care Medicine, Philipps University of Marburg, DZL, 35043 Marburg, Germany
9
Department of Internal Medicine V-Pulmonology, Allergology and Critical Care Medicine, Saarland University, 66421 Homburg, Germany
10
Department of Medicine, Cardiology, CPI, Goethe University Hospital, 60596 Frankfurt am Main, Germany
11
Institute for Cardiovascular Regeneration, CPI, Goethe University, 60596 Frankfurt am Main, Germany
*
Author to whom correspondence should be addressed.
Cells 2022, 11(13), 2121; https://doi.org/10.3390/cells11132121
Submission received: 11 May 2022 / Revised: 23 June 2022 / Accepted: 24 June 2022 / Published: 5 July 2022

Abstract

:
Chronic obstructive pulmonary disease (COPD) is a disease with an inflammatory phenotype with increasing prevalence in the elderly. Expanded population of mutant blood cells carrying somatic mutations is termed clonal hematopoiesis of indeterminate potential (CHIP). The association between CHIP and COPD and its relevant effects on DNA methylation in aging are mainly unknown. Analyzing the deep-targeted amplicon sequencing from 125 COPD patients, we found enhanced incidence of CHIP mutations (~20%) with a predominance of DNMT3A CHIP-mediated hypomethylation of Phospholipase D Family Member 5 (PLD5), which in turn is positively correlated with increased levels of glycerol phosphocholine, pro-inflammatory cytokines, and deteriorating lung function.

1. Introduction

Accumulation of somatic mutations throughout human life is a clearly defined phenomenon becoming more pronounced by aging. Hematopoietic stem cells (HSCs) with extensive self-renewal capacity are one of the most vulnerable cells to various somatic mutation with 70 mutations per gene for one HSC by age of 70 [1]. Different studies have shown that expansion of mutated HSCs, known as “Clonal hematopoiesis (CH)” is highly increased by aging [2]. CHIP has been described previously to be a driver of altered inflammatory response in chronic diseases, e.g., cardiovascular disease [3]. Age and smoking are the two strongest risk factors of chronic obstructive pulmonary disease (COPD), which contribute to the aberrant inflammatory phenotype of the disease [4]. In a very recent study, CHIP was shown to be associated with disease development and severity of COPD from four different cohorts. Miller et al. identified the increased risk of COPD related to CHIP independently of age, cigarette smoke exposure, and inherited polygenic risk score [5]. In other aspects, the association between CHIP and COPD and more importantly, its relevant effects on DNA methylation as one of the key players in aging are mainly unknown. In this study, we aimed (I) to investigate the prevalence of CHIP in COPD patients, (II) to investigate CHIP-mediated molecular consequences by DNA methylation, metabolomics, and cytokine analyses, (III) to correlate these with lung function parameters, and (IV) to evaluate the causality in macrophages derived from human peripheral blood mononuclear cells (PBMCs) and human precision cut lung slides (PCLS).

2. Materials and Methods

2.1. Study Population

Data were obtained from the baseline examination (Visit 1) of the German multicenter COPD cohort COSYCONET (German COPD and Systemic Consequences–Comorbidities Network). COPD was defined according to the GOLD criteria after performing standardized post-bronchodilator spirometry. COSYCONET is a German multicenter prospective observational trial, which recruited 2741 patients aged 40 years and older with diagnosis of COPD between 2010 and 2013 in 31 study centers. The study protocol has been previously described in detail [6] and in the Supplementary Materials.

2.2. DNA Methylation

DNA was isolated from the EDTA blood samples obtained from COSYCONET. The Infinium Human-Methylation EPIC BeadChip (850k) (WG-317, Illumina, San Diego, CA, USA) was used to determine the DNA methylation status following the producer’s guidelines. Infinium human methylation EPIC array analysis was performed by using RnBeads2.0, Max Planck Institute for Heart and Lung Research, release 2.0. https://rnbeads.org (accessed on 5 July 2019) [7] and ADMIRE, Max Planck Institute for Heart and Lung Research, release 2.0 https://github.molgen.mpg.de/loosolab/admire (accessed on 5 July 2019) [8]. More information regarding DNA isolation and methylation analysis has been provided in the Supplementary Materials.

2.3. Sample Preparation for NGS and High-Throughput Sequencing

Samples of 125 COPD patients were assessed for the presence of CHIP by the Illumina TruSeq Custom Amplicon Low Input assay. The panel includes 594 amplicons in 56 genes commonly mutated in CHIP and myeloid malignancies (Supplementary Table S1) [9]. The pooled libraries were sequenced on a NextSeq 500 sequencer (Illumina) using the NextSeq 500/550 Mid Output, version 2 kit (300 cycles) according to the manufacturer’s instructions. The procedure has been described in detail in the Supplementary Materials.

2.4. Metabolomics and Cytokine Analysis

Oxylipins and choline pathway metabolites were extracted from plasma by adding 3× the volume of ethyl acetate and 4× the volume of methanol, respectively. Either the upper layer (oxylipins) or supernatant (cholines) was analyzed on an Agilent 1290 Infinity LC system coupled to a Sciex QTrap 5500 mass spectrometer in negative (oxylipins) or positive (cholines) ionization modes. TNF-α and IL-6 were analyzed using ELISA kits. The detailed protocol for plasma collection and metabolome analysis has been explained in the Supplementary Materials.

2.5. Cell Culture and Gene Expression Analysis by qPCR

Generation of human macrophages from PBMCs, RNA isolation, complementary DNA synthesis, and quantitative PCR were performed as previously described [10]. The brief information has been provided in the Supplementary Materials.

2.6. Human COPD Precision Cut Lung Slides (PCLS)

PCLS were cut 400 µm thick using a vibratome (Microm HM650V, Thermo Fisher Scientific Inc., Waltham, MA, USA) and cultured in growth media. PCLS were then treated with conditional media of a DNMT3A-deficient monocyte cell line (THP1) for 48 h. The tissue lysates were taken for protein expression studies. Detailed protocol has been explained in the Supplementary Materials.

2.7. Western Blotting

The tissues were lysed in RIPA lysis buffer containing protease and phosphatase inhibitors. Subsequent step have been described in detail in the supplementary file.

2.8. Statistics

All analyses were carried out with the statistics software R v3.4 https://www.R-project.org (accessed on 5 July 2019). Lung function data were analyzed with general linear models using the log-transformed response values, and p values were adjusted by confounding factors including age, sex, smoking status, and pack years.

3. Results and Discussion

In this study, we investigated the prevalence of CHIP in a COPD patient cohort (COSYCONET) (Table 1).
Deep targeted amplicon sequencing in the patients showed an enhanced incidence of CHIP mutations compared with age-matched controls (~20%), with the striking predominance of somatic mutations in the gene DNMT3A (18 out of 25) (Figure 1A). The same profile was detected in early-stage COPD (Gold 0). In-depth analysis of the mutations and in silico analysis via Swiss Model https://swissmodel.expasy.org/ (accessed on 5 July 2019) identified 6 mutations (from 20) in DNMT3A which significantly alter its structure, suggesting reduced or even loss of methyl-transferase activity (Supplementary Figure S1, In silico analysis of DNMT3A and Supplementary Table S4). Interestingly, DNA methylation analysis demonstrated massive hypomethylation in DNMT3A-CHIP COPD versus Non-CHIP COPD patients, in up to 95% of top differentially methylated CpG islands, and 80% of differentially methylated CpG sites and tilings (Figure 1B).
As a possible indicator of CHIP mutations [3], we evaluated differences in proinflammatory cytokine formation and lung function between CHIP-positive and CHIP-negative early COPD patients. In early-stage COPD patients, significant higher levels of circulating inflammatory mediator IL6 and slight increase of TNFα were indeed noted in the CHIP mutation carriers, concomitant with increased functional residual capacity and decreased PaCO2 levels, indicating ventilation/perfusion mismatch and forced ventilatory drive (Figure 1C,D). The numbers of CHIP-positive early COPD patients were too low to link these findings to the smoking history of these patients.
To prove causality, DNMT3A was silenced in non-stimulated macrophages derived from human PBMCs, a significant resulting in an upregulation of pro-inflammatory cytokines, especially IL6 and slightly TNFα (Figure 1E). Human PCLS cultured with conditional media of DNMT3A deficient-THP1 confirmed a link between DNMT3A dysfunction and enhancement of pro-inflammatory (IL-1ß and TNFα) responses (Figure 1F–H). Nevertheless, the DNA methylation status of inflammatory cytokines was not altered (Supplementary Table S5), raising the intriguing possibility of additional molecular players driving the pro-inflammatory gene expression. The top hypomethylated tiling of genes includes Transmembrane Protein 232 (TMEM232), Glycophorin A (GYPA), and Follistatin Like 4 (FSTL4), which were previously shown to be associated with COPD and inflammation [11] (Figure 2A).
Notably, one of the top hypomethylated gene families that could be associated with both inflammation and COPD is Phospholipase D Family Member 5 (PLD5). We observed that in DNMT3A-CHIP COPD patients, PLD5 is hypomethylated (Figure 2B) and positively correlated with PaCO2 levels of DNMT3A-CHIP COPD, a phenomenon that was not observed in non-CHIP COPD patients (Figure 2C,D). Importantly, PLD5 methylation levels negatively correlated with inflammatory cytokines in CHIP-positive COPD patients, but not in CHIP-negative patients (Figure 2E,F), confirming that PLD5-mediated metabolic pathway modulates systemic oxidative stress and inflammation [12].
Phospholipase D isoforms are lipid-signaling enzymes that preferentially hydrolyze phosphatidylcholine (PC), thus generating phosphatidic acid and choline. Phosphatidic acid is further metabolized to either diacylglycerol (DAG) by phosphatidate phosphohydrolase (lipid phosphate phosphohydrolase) or lysophosphatidic acid (LPA) by phospholipase A1/A2 (PLA). PLAs are the rate-limiting enzyme for the downstream synthesis of prostaglandins and leukotrienes that are the main mediators of inflammation [12]. Importantly, previous studies confirmed the involvement of PLAs in the COPD-associated inflammation [13] although there are some gaps related to the regulation of specific enzymes in this pathway. To explore whether DNMT3A-CHIP mutations may explain these underlying molecular mechanisms, we performed a metabolome analysis of arachidonic acid-related metabolites as well as choline-containing metabolites in human plasma obtained from DNMT3A-CHIP COPD and Non-CHIP COPD patients. Notably, higher levels of glycerol phosphocholine were observed in DNMT3A-CHIP COPD versus Non-CHIP COPD patients (Figure 2G,H), which are negatively correlated with PLD5 methylation levels.
We conclude that somatic mutations in hematopoietic cells, specifically in the most commonly mutated CHIP driver gene DNMT3A, are increased in COPD and may be associated with metabolic and inflammatory sequelae with the functional outcome (Figure 2I). Underlying mechanisms such as smoking or the lung disease itself may enhance the CHIP frequency in the bone marrow stem cell niche, which then—as a vicious feedback loop—may amplify the pulmonary pathogenetic sequelae. Long-term observations on the impact of CHIP on the course of COPD are mandatory.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/cells11132121/s1, Figure S1: In silico analysis of DNMT3A; Table S1: List of genes analyzed by the custom TruSeq targeted sequencing panel; Table S2: qPCR primer list; Table S3: List of primary antibodies; Table S4: DNMT3A SNPs; Table S5: Methylation level of inflammatory markers. Supporting information can be found in supplementary materials [14,15,16,17,18].

Author Contributions

R.S., S.D., W.S., S.S.P. designed the experiments and supervised the study. S.K. (Stefan Kuhnert), S.M., M.A.R., S.H., G.D.-P., M.P., M.L., N.S., N.W., R.S., C.V., R.B., A.Z., S.D., E.A., T.A., S.K. (Stephan Klatt), I.F., J.W. collected data, interpreted the results, and revised them for important intellectual content. S.K. (Stefan Kuhnert), S.M., W.S., S.S.P. wrote the manuscript. All authors reviewed and edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Max Planck Society, the Cardio-Pulmonary Institute (CPI), the German Center for Lung Research (DZL), and the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation)—Project number 268555672—SFB 1213 (Project A01, A05 to SSP, A07 to NW and Project A10* to RS) and European Research Council (ERC) Consolidator Grant (#866051 to SSP).

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Jaiswal, S.; Ebert, B.L. Clonal hematopoiesis in human aging and disease. Science 2019, 366, eaan4673. [Google Scholar] [CrossRef] [PubMed]
  2. Jaiswal, S.; Libby, P. Clonal haematopoiesis: Connecting ageing and inflammation in cardiovascular disease. Nat. Rev. Cardiol. 2020, 17, 137–144. [Google Scholar] [CrossRef] [PubMed]
  3. Jaiswal, S.; Natarajan, P.; Silver, A.J.; Gibson, C.J.; Bick, A.G.; Shvartz, E.; McConkey, M.; Gupta, N.; Gabriel, S.; Ardissino, D.; et al. Clonal Hematopoiesis and Risk of Atherosclerotic Cardiovascular Disease. N. Engl. J. Med. 2017, 377, 111–121. [Google Scholar] [CrossRef] [PubMed]
  4. Caramori, G.; Adcock, I.M.; Di Stefano, A.; Chung, K.F. Cytokine inhibition in the treatment of COPD. Int. J. Chron. Obstruct. Pulmon. Dis. 2014, 9, 397–412. [Google Scholar] [PubMed] [Green Version]
  5. Miller, P.G.; Qiao, D.; Rojas-Quintero, J.; Honigberg, M.C.; Sperling, A.S.; Gibson, C.J.; Bick, A.G.; Niroula, A.; McConkey, M.E.; Sandoval, B.; et al. Association of clonal hematopoiesis with chronic obstructive pulmonary disease. Blood 2022, 139, 357–368. [Google Scholar] [CrossRef] [PubMed]
  6. Quanjer, P.H.; Stanojevic, S.; Cole, T.J.; Baur, X.; Hall, G.L.; Culver, B.H.; Enright, P.L.; Hankinson, J.L.; Ip, M.S.M.; Zheng, J.; et al. Multi-ethnic reference values for spirometry for the 3–95-yr age range: The global lung function 2012 equations. Eur. Respir. J. 2012, 40, 1324–1343. [Google Scholar] [CrossRef] [PubMed]
  7. Müller, F.; Scherer, M.; Assenov, Y.; Lutsik, P.; Walter, J.; Lengauer, T.; Bock, C. RnBeads 2.0: Comprehensive analysis of DNA methylation data. Genome Biol. 2019, 20, 55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Preussner, J.; Bayer, J.; Kuenne, C.; Looso, M. ADMIRE: Analysis and visualization of differential methylation in genomic re-gions using the Infinium HumanMethylation450 Assay. Epigenetics Chromatin 2015, 8, 51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Dorsheimer, L.; Assmus, B.; Rasper, T.; Ortmann, C.A.; Ecke, A.; Abou-El-Ardat, K.; Schmid, T.; Brüne, B.; Wagner, S.; Serve, H.; et al. Association of Mutations Contributing to Clonal Hematopoiesis With Prognosis in Chronic Ischemic Heart Failure. JAMA Cardiol. 2019, 4, 25–33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Pullamsetti, S.S.; Kojonazarov, B.; Storn, S.; Gall, H.; Salazar, Y.; Wolf, J.; Weigert, A.; El-Nikhely, N.; Ghofrani, H.A.; Krombach, G.A.; et al. Lung cancer–associated pulmonary hypertension: Role of microenvironmental inflammation based on tumor cell–immune cell cross-talk. Sci. Transl. Med. 2017, 9, eaai9048. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Celarain, N.; Tomas-Roig, J. Aberrant DNA methylation profile exacerbates inflammation and neurodegeneration in multi-ple sclerosis patients. J. Neuroinflamm. 2020, 17, 21. [Google Scholar] [CrossRef] [PubMed]
  12. Colombo, S.; Martín-Sierra, C.; Melo, T.; Laranjeira, P.; Paiva, A.; Domingues, P.; Domingues, M.R. Modulation of the inflammatory response of immune cells in human peripheral blood by oxidized arachidonoyl aminophospholipids. Arch. Biochem. Biophys. 2018, 660, 64–71. [Google Scholar] [CrossRef] [PubMed]
  13. Pniewska, E.; Pawliczak, R. The Involvement of Phospholipases A2 in Asthma and Chronic Obstructive Pulmonary Disease. Mediat. Inflamm. 2013, 2013, 793505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Tombor, L.S.; John, D.; Glaser, S.F.; Luxán, G.; Forte, E.; Furtado, M.; Rosenthal, N.; Baumgarten, N.; Schulz, M.H.; Wittig, J.; et al. Single cell sequencing reveals endothelial plasticity with transient mesenchymal activation after myocardial infarction. Nat. Commun. 2021, 12, 681. [Google Scholar] [CrossRef]
  15. Mas-Peiro, S.; Hoffmann, J.; Fichtlscherer, S.; Dorsheimer, L.; Rieger, M.A.; Dimmeler, S.; Vasa-Nicotera, M.; Zeiher, A.M. Clonal haematopoiesis in patients with degenerative aortic valve stenosis undergoing transcatheter aortic valve implantation. Eur. Heart J. 2020, 41, 933–939. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Ortmann, C.A.; Dorsheimer, L.; Abou-El-Ardat, K.; Hoffrichter, J.; Assmus, B.; Bonig, H.; Scholz, A.; Pfeifer, H.; Martin, H.; Schmid, T.; et al. Functional Dominance of CHIP-Mutated Hematopoietic Stem Cells in Patients Undergoing Autologous Transplantation. Cell Rep. 2019, 27, 2022–2028. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Sarode, P.; Zheng, X.; Giotopoulou, G.A.; Weigert, A.; Kuenne, C.; Günther, S.; Friedrich, A.; Gattenlöhner, S.; Stiewe, T.; Brüne, B.; et al. Reprogramming of tumor-associated macrophages by targeting β-catenin/FOSL2/ARID5A signaling: A potential treatment of lung cancer. Sci. Adv. 2020, 6, 6105. [Google Scholar] [CrossRef] [PubMed]
  18. Zheng, X.; Weigert, A.; Reu, S.; Guenther, S.; Mansouri, S.; Bassaly, B.; Gattenlöhner, S.; Grimminger, F.; Pullamsetti, S.; Seeger, W.; et al. Spatial Density and Distribution of Tumor-Associated Macrophages Predict Survival in Non-Small Cell Lung Carcinoma. Cancer Res. 2020, 80, 4414–4425. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Clonal hematopoiesis of indeterminate potential (CHIP) and mutations in DNMT3A gene is prevalent in COPD patients. (A) The % of CHIP mutation carriers and the proportion of CHIP-associated mutations (in 56 analyzed genes) in patients with COPD is given. (B) Distribution of hypomethylated (Hypo) and hypermethylated (Hyper) CpG sites in DNMT3A-CHIP and Non-CHIP patients as well as the depiction of the methylation status of top-ranked CpGs and tiling sites in DNMT3A-CHIP COPD and non-CHIP COPD patients. (C) Lung function parameters FRCpleth, Exa and PaCO2 in 26 early-stage patients with CHIP mutation (CHIP; n = 6) or without CHIP mutation (Non-CHIP; n = 20), observed over 1.5 years, are given. V1—first visit upon entry into the COSYCONET cohort; V3—third visit after 18 ± 1 months. (D) Plasma IL6 and TNFα levels in CHIP (n = 6 for IL6 and TNFα) and Non-CHIP patients (n = 19 for IL6 and TNFα) with early-stage COPD. p values are adjusted by confounding factors including age, sex, smoking status, and pack years. (E) mRNA expression of DNMT3A, TNFα, and IL6 in PBMC-derived monocytes, that were subsequently differentiated into macrophages and are transfected with siControl (non-silencing control siRNA) and siDNMT3A for 48 h (n = 3 independently performed experiments) to mimic the hypomethylation status observed by DNMT3A SNPs in COPD, 2-tailed Student’s t test. (F) Schematic presentation of ex vivo cultured human PCLS with conditional media of DNMT3A deficient monocyte cell line (THP1), that was transfected with siControl (non-silencing control siRNA) and siDNMT3A for 48 h. (G,H) Representative western blotting images of IL1β and TNFα followed by densitometric quantification of relative expression IL1β and TNFα with GAPDH as the loading control in PCLS (n = 5), 2-tailed Student’s t-test. p values are adjusted by confounding factors including age, sex, smoking status, and pack years. PaCO2—Partial pressure of carbon dioxide in mmHg. FRCpleth—functional residual capacity measured by body plethysmography. Exa—exacerbations. *: p < 0.05, **: p < 0.01, ***: p < 0.001.
Figure 1. Clonal hematopoiesis of indeterminate potential (CHIP) and mutations in DNMT3A gene is prevalent in COPD patients. (A) The % of CHIP mutation carriers and the proportion of CHIP-associated mutations (in 56 analyzed genes) in patients with COPD is given. (B) Distribution of hypomethylated (Hypo) and hypermethylated (Hyper) CpG sites in DNMT3A-CHIP and Non-CHIP patients as well as the depiction of the methylation status of top-ranked CpGs and tiling sites in DNMT3A-CHIP COPD and non-CHIP COPD patients. (C) Lung function parameters FRCpleth, Exa and PaCO2 in 26 early-stage patients with CHIP mutation (CHIP; n = 6) or without CHIP mutation (Non-CHIP; n = 20), observed over 1.5 years, are given. V1—first visit upon entry into the COSYCONET cohort; V3—third visit after 18 ± 1 months. (D) Plasma IL6 and TNFα levels in CHIP (n = 6 for IL6 and TNFα) and Non-CHIP patients (n = 19 for IL6 and TNFα) with early-stage COPD. p values are adjusted by confounding factors including age, sex, smoking status, and pack years. (E) mRNA expression of DNMT3A, TNFα, and IL6 in PBMC-derived monocytes, that were subsequently differentiated into macrophages and are transfected with siControl (non-silencing control siRNA) and siDNMT3A for 48 h (n = 3 independently performed experiments) to mimic the hypomethylation status observed by DNMT3A SNPs in COPD, 2-tailed Student’s t test. (F) Schematic presentation of ex vivo cultured human PCLS with conditional media of DNMT3A deficient monocyte cell line (THP1), that was transfected with siControl (non-silencing control siRNA) and siDNMT3A for 48 h. (G,H) Representative western blotting images of IL1β and TNFα followed by densitometric quantification of relative expression IL1β and TNFα with GAPDH as the loading control in PCLS (n = 5), 2-tailed Student’s t-test. p values are adjusted by confounding factors including age, sex, smoking status, and pack years. PaCO2—Partial pressure of carbon dioxide in mmHg. FRCpleth—functional residual capacity measured by body plethysmography. Exa—exacerbations. *: p < 0.05, **: p < 0.01, ***: p < 0.001.
Cells 11 02121 g001
Figure 2. PLD5 gene methylation association with pro-inflammatory signaling and impaired lung function in early COPD. (A) Characteristics of top 15 differentially methylated tiling sites between DNMT3A-CHIP and Non-CHIP patients. (B) Methylation level of tiling-associated PDL5 between DNMT3A-CHIP and Non-CHIP patients, ***: p < 0.001, 2-tailed Student’s t test. (C,D) Pearson correlation analysis between PLD5 methylation and PaCO2 level in DNMT3A-CHIP (N = 11) and Non-CHIP (N = 10) patients. (E) Pearson correlation analysis between PLD5 methylation and plasma IL6 concentration in DNMT3A-CHIP (N = 11) and Non-CHIP (N = 10) patients. (F) Pearson correlation analysis between PLD5 methylation and plasma TNF concentration in DNMT3A-CHIP (N = 11) and Non-CHIP (N = 10) patients. (G) Volcano plot of metabolome analysis of serum in DNMT3A-CHIP (N = 18) and Non-CHIP (N = 22) patients. **: p < 0.01, 2-tailed Student’s t test (H) Pearson correlation analysis between PLD5 methylation and serum glycerolphosphocholine concentration in DNMT3A-CHIP (N = 11). (I) Schematic diagram displaying the link between DNMT3A dysfunction and enhancement of metabolic and pro-inflammatory responses in COPD.
Figure 2. PLD5 gene methylation association with pro-inflammatory signaling and impaired lung function in early COPD. (A) Characteristics of top 15 differentially methylated tiling sites between DNMT3A-CHIP and Non-CHIP patients. (B) Methylation level of tiling-associated PDL5 between DNMT3A-CHIP and Non-CHIP patients, ***: p < 0.001, 2-tailed Student’s t test. (C,D) Pearson correlation analysis between PLD5 methylation and PaCO2 level in DNMT3A-CHIP (N = 11) and Non-CHIP (N = 10) patients. (E) Pearson correlation analysis between PLD5 methylation and plasma IL6 concentration in DNMT3A-CHIP (N = 11) and Non-CHIP (N = 10) patients. (F) Pearson correlation analysis between PLD5 methylation and plasma TNF concentration in DNMT3A-CHIP (N = 11) and Non-CHIP (N = 10) patients. (G) Volcano plot of metabolome analysis of serum in DNMT3A-CHIP (N = 18) and Non-CHIP (N = 22) patients. **: p < 0.01, 2-tailed Student’s t test (H) Pearson correlation analysis between PLD5 methylation and serum glycerolphosphocholine concentration in DNMT3A-CHIP (N = 11). (I) Schematic diagram displaying the link between DNMT3A dysfunction and enhancement of metabolic and pro-inflammatory responses in COPD.
Cells 11 02121 g002
Table 1. Detailed characteristics of CHIP in COPD patient cohort (COSYCONET).
Table 1. Detailed characteristics of CHIP in COPD patient cohort (COSYCONET).
SubsetGOLD 0 PatientsGOLD I–III PatientsGOLD III/IV Patients
CHIPAllNegativePositiveAllNegativePositiveAllNegativePositive
Number26206907119362610
SexMale42%30%83%68%69%63%61%58%70%
Female58%70%17%32%31%37%39%42%30%
Age60 (5)59 (4.7)62 (5.6)62 (5.5)62 (5.8)62 (4.4)62 (5.7)62 (6.1)62 (4.5)
BMI27 (4.8)26 (5.4)28 (2.1)27 (5.8)27 (6.2)26 (4.1)26 (4.7)27 (5.2)25 (3.2)
SmokingPackyears37 (19)34 (17)46 (26)42 (31)41 (31)46 (29)48 (29)48 (28)50 (33)
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kuhnert, S.; Mansouri, S.; Rieger, M.A.; Savai, R.; Avci, E.; Díaz-Piña, G.; Padmasekar, M.; Looso, M.; Hadzic, S.; Acker, T.; et al. Association of Clonal Hematopoiesis of Indeterminate Potential with Inflammatory Gene Expression in Patients with COPD. Cells 2022, 11, 2121. https://doi.org/10.3390/cells11132121

AMA Style

Kuhnert S, Mansouri S, Rieger MA, Savai R, Avci E, Díaz-Piña G, Padmasekar M, Looso M, Hadzic S, Acker T, et al. Association of Clonal Hematopoiesis of Indeterminate Potential with Inflammatory Gene Expression in Patients with COPD. Cells. 2022; 11(13):2121. https://doi.org/10.3390/cells11132121

Chicago/Turabian Style

Kuhnert, Stefan, Siavash Mansouri, Michael A. Rieger, Rajkumar Savai, Edibe Avci, Gabriela Díaz-Piña, Manju Padmasekar, Mario Looso, Stefan Hadzic, Till Acker, and et al. 2022. "Association of Clonal Hematopoiesis of Indeterminate Potential with Inflammatory Gene Expression in Patients with COPD" Cells 11, no. 13: 2121. https://doi.org/10.3390/cells11132121

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop