DNA methylation alterations in muscle of critically ill patients

Lisa Van Dyck, Fabian Güiza, Inge Derese, Lies Pauwels, Michaël P Casaer, Greet Hermans, Pieter J Wouters, Greet Van den Berghe, Ilse Vanhorebeek, Lisa Van Dyck, Fabian Güiza, Inge Derese, Lies Pauwels, Michaël P Casaer, Greet Hermans, Pieter J Wouters, Greet Van den Berghe, Ilse Vanhorebeek

Abstract

Background: Intensive care unit (ICU)-acquired weakness can persist beyond ICU stay and has been associated with long-term functional impairment of ICU survivors. Recently, DNA methylation alterations were found in the blood of ICU patients, partially explaining long-term developmental impairment of critically ill children. As illness-induced aberrant DNA methylation theoretically could also be involved in long-term weakness, we investigated whether the DNA methylation signature in muscle of adult critically ill patients differs from that in muscle of healthy controls.

Methods: Genome-wide methylation was determined (Infinium® HumanMethylationEPIC BeadChips) in DNA extracted from skeletal muscle biopsies that had been collected on Day 8 ± 1 in ICU from 172 EPaNIC-trial patients [66% male sex, median age 62.7 years, median body mass index (BMI) 25.9 kg/m2 ] and 20 matched healthy controls (70% male sex, median age 58.0 years, median BMI 24.4 kg/m2 ). Methylation status of individual cytosine-phosphate-guanine (CpG) sites of patients and controls was compared with F-tests, using the Benjamini-Hochberg false discovery rate to correct for multiple comparisons. Differential methylation of DNA regions was assessed with bump hunting, with 1000 permutations assessing uncertainty, expressed as family-wise error rate. Gene expression was investigated for 10 representative affected genes.

Results: In DNA from ICU patients, 565 CpG sites, associated with 400 unique genes, were differentially methylated as compared with controls (average difference 3.2 ± 0.1% ranging up to 16.9%, P < 0.00005). Many of the associated genes appeared highly relevant for muscle structure and function/weakness, including genes involved in myogenesis, muscle regeneration, nerve/muscle membrane excitability, muscle denervation/re-innervation, axon guidance/myelination/degeneration/regeneration, synapse function, ion channelling with especially calcium signalling, metabolism (glucose, protein, and fat), insulin signalling, neuroendocrine hormone regulation, mitochondrial function, autophagy, apoptosis, oxidative stress, Wnt signalling, transcription regulation, muscle fat infiltration during regeneration, and fibrosis. In patients as compared with controls, we also identified two hypomethylated regions, spanning 18 and 3 CpG sites in the promoters of the HIC1 and NADK2 genes, respectively (average differences 5.8 ± 0.01% and 12.1 ± 0.04%, family-wise error rate <0.05). HIC1 and NADK2 play important roles in muscle regeneration and postsynaptic acetylcholine receptors and in mitochondrial processes, respectively. Nine of 10 investigated genes containing DNA methylation alterations were differentially expressed in patients as compared with controls (P ≤ 0.03).

Conclusions: Critically ill patients present with a different DNA methylation signature in skeletal muscle as compared with healthy controls, which in theory could provide a biological basis for long-term persistence of weakness in ICU survivors.

Trial registration: ClinicalTrials.gov: NCT00512122, registered on 31 July 2007.

Keywords: Critical illness; DNA methylation; Epigenetics; Intensive care unit-acquired muscle weakness; Muscle.

Conflict of interest statement

None declared.

All authors certify that they comply with the ethical guidelines for authorship and publishing in the Journal of Cachexia, Sarcopenia and Muscle. Institutional review board approval was obtained (ML4190). The study was performed in accordance with the 1964 Declaration of Helsinki and its later amendments. Written informed consent was acquired from all patients or their next of kin.

© 2022 The Authors. Journal of Cachexia, Sarcopenia and Muscle published by John Wiley & Sons Ltd on behalf of Society on Sarcopenia, Cachexia and Wasting Disorders.

Figures

Figure 1
Figure 1
Consort diagram. Data file‐related problem refers to unrepairable damage to one of the IDAT data files, which became unreadable. DNA, deoxyribonucleic acid; EPaNIC, Early Parenteral Nutrition Completing Enteral Nutrition in Adult Critically Ill Patients; ICU, intensive care unit; RCT, randomized controlled trial.
Figure 2
Figure 2
Location of differentially methylated cytosine–phosphate–guanine (CpG) sites. (A) Distribution over the autosomes, (B) association with known gene transcripts, and (C) relative position to CpG islands are shown for the 565 CpG sites that were significantly differentially methylated in critically ill patients as compared with healthy controls. Gene transcript regions are divided into the promotor region, where proteins bind to initiate transcription of the gene; the 5′ untranslated region (5′‐UTR) region, which regulates translation; the gene body, which actually encodes the protein; the 3′ untranslated region (3′‐UTR) region, which is a regulatory unit; and the intergenic region, which is not related to a known gene transcript. In relation to CpG islands, the regions are divided into islands, which are regions with a high frequency of CpG sites; shores, which immediately flank the islands (within 2000 bp); shelves, which flank the shores (within 4000 bp from a CpG island); and open‐sea regions, which are not related to a CpG island. The height of the bars indicates the total number of CpG sites. Red and blue bars indicate the number of CpG sites that were respectively hypermethylated and hypomethylated in patients as compared with controls. In relation to gene transcripts, the 78 CpG sites that were associated with more than one gene section type are included for each of these associations.
Figure 3
Figure 3
Visualisation of the differentially methylated regions in patients vs. controls. Regions that are differentially methylated in muscle of critically ill patients as compared with healthy controls are visualised. The red line on the chromosome ideogram (top line of each panel) indicates the position of the differentially methylated region (DMR) relative to the chromosome. The light blue highlighted area shows the DMR. In blue and red, average cytosine–phosphate–guanine (CpG) methylation (beta‐value) and its confidence interval are shown for healthy controls and patients, respectively. At the bottom of each panel, the location of CpG sites evaluated by the microarray is indicated. Purple areas show locations of exons of known gene transcripts in this region. (A) The first DMR, which is located on chromosome 17 and spans 636 nucleotides containing 18 CpG sites. All 18 CpG sites were hypomethylated in patients as compared with controls. This region spans the hypermethylated in cancer 1 (HIC1) gene. (B) The second DMR, which is located on chromosome 5, spans 34 nucleotides, and contains three CpG sites that were hypomethylated in patients as compared with controls. This region spans the NAD kinase 2 (NADK2) gene.
Figure 4
Figure 4
Gene expression. mRNA expression of selected genes affected by DNA methylation alterations is shown for controls (n = 19) and patients (n = 162). Results are expressed relatively to the median of the healthy controls, with cancer susceptibility candidate gene 3 (CASC3) used as housekeeping gene. Boxes represent medians and inter‐quartile ranges, and whiskers are drawn to the furthest point within 1.5 × inter‐quartile range from the box. P‐values for comparison of expression between patients and controls are shown for each gene.

References

    1. Zimmerman JE, Kramer AA, Knaus WA. Changes in hospital mortality for United States intensive care unit admissions from 1988 to 2012. Crit Care 2013;17:R81.
    1. Desai SV, Law TJ, Needham DM. Long‐term complications of critical care. Crit Care Med 2011;39:371–379.
    1. Herridge MS, Tansey CM, Matte A, Tomlinson G, Diaz‐Granados N, Cooper A, et al. Functional disability 5 years after acute respiratory distress syndrome. N Engl J Med 2011;364:1293–1304.
    1. Kress JP, Hall JB. ICU‐acquired weakness and recovery from critical illness. N Engl J Med 2014;370:26–1635.
    1. De Jonghe B, Sharshar T, Lefaucheur JP, Authier FJ, Durand‐Zaleski I, Boussarsar M, et al. Paresis acquired in the intensive care unit: a prospective multicenter study. JAMA 2002;288:2859–2867.
    1. Vanhorebeek I, Latronico N, Van den Berghe G. ICU‐acquired weakness. Intensive Care Med 2020;46:637–653.
    1. Hermans G, Van Aerde N, Meersseman P, Van Mechelen H, Debaveye Y, Wilmer A, et al. Five‐year mortality and morbidity impact of prolonged versus brief ICU stay: a propensity score matched cohort study. Thorax 2019;74:1037–1045.
    1. Van Aerde N, Meersseman P, Debaveye Y, Wilmer A, Gunst J, Casaer MP, et al. Five‐year impact of ICU‐acquired neuromuscular complications: a prospective, observational study. Intensive Care Med 2020;46:1184–1193.
    1. Friedrich O, Reid MB, Van den Berghe G, Vanhorebeek I, Hermans G, Rich MM, et al. The sick and the weak: neuropathies/myopathies in the critically ill. Physiol Rev 2015;95:1025–1109.
    1. Dos Santos C, Hussain SN, Mathur S, Picard M, Herridge M, Correa J, et al. Mechanisms of chronic muscle wasting and dysfunction after an intensive care unit stay. A pilot study. Am J Respir Crit Care Med 2016;194:821–830.
    1. Walsh CJ, Batt J, Herridge MS, Mathur S, Bader GD, Hu P, et al. Transcriptomic analysis reveals abnormal muscle repair and remodeling in survivors of critical illness with sustained weakness. Sci Rep 2016;6:29334.
    1. Duarte JD. Epigenetics primer: why the clinician should care about epigenetics. Pharmacotherapy 2013;33:1362–1368.
    1. Waterland RA, Michels KB. Epigenetic epidemiology of the developmental origins hypothesis. Annu Rev Nutr 2007;27:363–388.
    1. Gut P, Verdin E. The nexus of chromatin regulation and intermediary metabolism. Nature 2013;502:489–498.
    1. Sharples AP, Stewart CE, Seaborne RA. Does skeletal muscle have an ‘epi’‐memory? The role of epigenetics in nutritional programming, metabolic disease, aging and exercise. Aging Cell 2016;15:603–616.
    1. Zykovich A, Hubbard A, Flynn JM, Tarnopolsky M, Fraga MF, Kerksick C, et al. Genome‐wide DNA methylation changes with age in disease‐free human skeletal muscle. Aging Cell 2014;13:360–366.
    1. Liu L, Cheung TH, Charville GW, Hurgo BM, Leavitt T, Shih J, et al. Chromatin modifications as determinants of muscle stem cell quiescence and chronological aging. Cell Rep 2013;4:189–204.
    1. Palacios D, Puri PL. The epigenetic network regulating muscle development and regeneration. J Cell Physiol 2006;207:1–11.
    1. Moresi V, Marroncelli N, Coletti D, Adamo S. Regulation of skeletal muscle development and homeostasis by gene imprinting, histone acetylation and microRNA. Biochim Biophys Acta 2015;1849:309–316.
    1. Sui X, Zhu J, Zhou J, Wang X, Li D, Han W, et al. Epigenetic modifications as regulatory elements of autophagy in cancer. Cancer Lett 2015;360:106–113.
    1. Hermans G, Casaer MP, Clerckx B, Güiza F, Vanhullebusch T, Derde S, et al. Effect of tolerating macronutrient deficit on the development of intensive‐care unit acquired weakness: a subanalysis of the EPaNIC trial. Lancet Respir Med 2013;1:621–629.
    1. Shih CC, Hii HP, Tsao CM, Chen SJ, Ka SM, Liao MH, et al. Therapeutic effects of procainamide on endotoxin‐induced rhabdomyolysis in rats. PLoS ONE 2016;11:e0150319.
    1. Güiza F, Vanhorebeek I, Verstraete S, Verlinden I, Derese I, Ingels C, et al. Effect of early parenteral nutrition during paediatric critical illness on DNA methylation as a potential mediator of impaired neurocognitive development: a pre‐planned secondary analysis of the PEPaNIC international randomised controlled trial. Lancet Respir Med 2020;8:288–303.
    1. Verlinden I, Güiza F, Derese I, Wouters PJ, Joosten K, Verbruggen SC, et al. Time course of altered DNA methylation evoked by critical illness and by early administration of parenteral nutrition in the paediatric ICU. Clin Epigenetics 2020;12:155.
    1. Binnie A, Walsh CJ, Hu P, Dwivedi DJ, Fox‐Robichaud A, Liaw PC, et al. Epigenetic profiling in severe sepsis: a pilot study of DNA methylation profiles in critical illness. Crit Care Med 2020;48:142–150.
    1. Casaer MP, Mesotten D, Hermans G, Wouters PJ, Schetz M, Meyfroidt G, et al. Early versus late parenteral nutrition in critically ill adults. N Engl J Med 2011;365:506–517.
    1. Casaer MP, Hermans G, Wilmer A, Van den Berghe G. Impact of early parenteral nutrition completing enteral nutrition in adult critically ill patients (EPaNIC trial): a study protocol and statistical analysis plan for a randomized controlled trial. Trials 2011;12:21.
    1. Aryee MJ, Jaffe AE, Corrada‐Bravo H, Ladd‐Acosta C, Feinberg AP, Hansen KD, et al. Minfi: a flexible and comprehensive Bioconductor package for the analysis of Infinium DNA methylation microarrays. Bioinformatics 2014;30:1363–1369.
    1. Fortin JP, Triche TJ Jr, Hansen KD. Preprocessing, normalization and integration of the Illumina HumanMethylationEPIC array with minfi. Bioinformatics 2017;33:558–560.
    1. Touleimat N, Tost J. Complete pipeline for Infinium® Human Methylation 450K BeadChip data processing using subset quantile normalization for accurate DNA methylation estimation. Epigenomics 2012;4:325–341.
    1. Johnson WE, Li C, Rabinovic A. Adjusting batch effects in microarray expression data using empirical Bayes methods. Biostatistics 2007;8:118–127.
    1. Leek JT, Johnson WE, Parker HS, Jaffe AE, Storey JD. The sva package for removing batch effects and other unwanted variation in high‐throughput experiments. Bioinformatics 2012;28:882–883.
    1. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Statist Soc B 1995;57:289–300.
    1. Liao Y, Wang J, Jaehnig EJ, Shi Z, Zhang B. WebGestalt 2019: gene set analysis toolkit with revamped UIs and APIs. Nucleic Acids Res 2019;47:W199–W205.
    1. Jaffe AE, Murakami P, Lee H, Leek JT, Fallin MD, Feinberg AP, et al. Bump hunting to identify differentially methylated regions in epigenetic epidemiology studies. Int J Epidemiol 2012;41:200–209.
    1. Scott RW, Arostegui M, Schweitzer R, Rossi FMV, Underhill TM. Hic1 Defines quiescent mesenchymal progenitor subpopulations with distinct functions and fates in skeletal muscle regeneration. Cell Stem Cell 2019;25:797, e9–813.
    1. Ohashi K, Kawai S, Murata K. Identification and characterization of a human mitochondrial NAD kinase. Nat Commun 2012;3:1248.
    1. Houten SM, Denis S, Te Brinke H, Jongejan A, van Kampen AH, Bradley EJ, et al. Mitochondrial NADP(H) deficiency due to a mutation in NADK2 causes dienoyl‐CoA reductase deficiency with hyperlysinemia. Hum Mol Genet 2014;23:5009–5016.
    1. Jacobsen SC, Brons C, Bork‐Jensen J, Ribel‐Madsen R, Yang B, Lara E, et al. Effects of short‐term high‐fat overfeeding on genome‐wide DNA methylation in the skeletal muscle of healthy young men. Diabetologia 2012;55:3341–3349.
    1. Nitert MD, Dayeh T, Volkov P, Elgzyri T, Hall E, Nilsson E, et al. Impact of an exercise intervention on DNA methylation in skeletal muscle from first‐degree relatives of patients with type 2 diabetes. Diabetes 2012;61:3322–3332.
    1. Fredriksson K, Tjäder I, Keller P, Petrovic N, Ahlman B, Schéele C, et al. Dysregulation of mitochondrial dynamics and the muscle transcriptome in ICU patients suffering from sepsis induced multiple organ failure. PLoS ONE 2008;3:e3686.
    1. Llano‐Diez M, Fury W, Okamoto H, Bai Y, Gromada J, Larsson L. RNA‐sequencing reveals altered skeletal muscle contraction, E3 ligases, autophagy, apoptosis, and chaperone expression in patients with critical illness myopathy. Skelet Muscle 2019;9:9.
    1. Pollina EA, Brunet A. Epigenetic regulation of aging stem cells. Oncogene 2011;30:3105–3126.
    1. Rocheteau P, Chatre L, Briand D, Mebarki M, Jouvion G, Bardon J, et al. Sepsis induces long‐term metabolic and mitochondrial muscle stem cell dysfunction amenable by mesenchymal stem cell therapy. Nat Commun 2015;6:10145.
    1. Bloch SA, Lee JY, Syburra T, Rosendahl U, Griffiths MJ, Kemp PR, et al. Increased expression of GDF‐15 may mediate ICU‐acquired weakness by down‐regulating muscle microRNAs. Thorax 2015;70:219–228.
    1. Garros RF, Paul R, Connolly M, Lewis A, Garfield BE, Natanek SA, et al. MicroRNA‐542 promotes mitochondrial dysfunction and SMAD activity and is elevated in intensive care unit‐acquired weakness. Am J Respir Crit Care Med 2017;196:1422–1433.
    1. Poddar S, Kesharwani D, Datta M. Interplay between the miRNome and the epigenetic machinery: implications in health and disease. J Cell Physiol 2017;232:2938–2945.
    1. Tikiyani V, Li L, Sharma P, Liu H, Hu Z, Babu K. Wnt secretion is regulated by the tetraspan protein HIC‐1 through its interaction with Neurabin/NAB‐1. Cell Rep 2018;25:1856, e6–1871.
    1. Lim U, Song M‐A. Dietary and lifestyle factors of DNA methylation. Methods Mol Biol 2012;863:359–376.
    1. Ferrante LE, Pisani MA, Murphy TE, Gahbauer EA, Leo‐Summers LS, Gill TM. Functional trajectories among older persons before and after critical illness. JAMA Intern Med 2015;175:523–529.
    1. Begue G, Raue U, Jemiolo B, Trappa S. DNA methylation assessment from human slow‐ and fast‐twitch skeletal muscle fibers. J Appl Physiol (1985) 2017;122:952–967.
    1. von Haehling S, Morley JE, Coats AJS, Anker SD. Ethical guidelines for publishing in the Journal of Cachexia, Sarcopenia and Muscle: update 2021. J Cachexia Sarcopenia Muscle 2021;12:2259–2261.

Source: PubMed

3
Sottoscrivi