Abstract
This review examines the role of circulating cell-free DNA (cfDNA) as potential drivers of inflammation and their potential application as mechanistic biomarkers in Inflammatory Bowel Diseases (IBD). These DNA fragments contain significant information about their origins, the underlying host pathology leading to their release, and possess properties that can fuel the inflammatory process. Recent advances in sequencing and analytical approaches have made the translation of cfDNA into clinical practice a promising prospect. We focus on the functional relevance of cfDNA in the inflammatory process and discuss its potential for future assessments of IBD activity and identification of therapeutic options.
Keywords: IBD, UC, CD, cfDNA, mitochondria, liquid biopsy
Introduction
Inflammatory bowel diseases (IBD), Ulcerative colitis (UC) and Crohn’s disease (CD) are chronic immune-mediated conditions with complex pathogenic mechanisms that drive the abnormal process of gut mucosal inflammation. Genetic susceptibility (>300 genetic loci), environmental influences (such as diet, drugs, and smoking), altered microbiome, and dysregulated immune and gut barrier functions are key contributory factors to the development of IBD. With recent scientific advances, the quest for precision medicine to predict outcomes and select the right treatment has provided much promise but realistic clinical translation opportunities have not been forthcoming1. Rapid progress in identifying new potential drug targets has shifted the challenge of drug discovery towards identifying dominant inflammatory mechanisms within the heterogeneous nature of IBD to facilitate decision-making in selecting the right drug for the right patient. In this context, increasingly deeper multi-omic data in very large patient groups combined with powerful computational approaches will help, but the question remains as to whether there are entirely underexplored areas in inflammation biology that have not been fully appreciated.
Our review focuses on the roles of circulating cell-free DNA (cfDNA) as potential drivers and mechanistic biomarkers and their development as ‘liquid biopsies’ in IBD. We recently showed that circulating mitochondrial DNA could be found in active IBD2, an interesting observation in line with other conditions where liberated host DNA triggers inflammation, in a spectrum of conditions ranging from systemic inflammatory syndrome (SIRS) to chronic immune-mediated diseases, such as systemic lupus erythematosus (SLE). Furthermore, DNA complexed with proteins can be pathogenic drivers of immune-mediated diseases, such as psoriasis3,4. Recently, therapeutic developments of nucleic acid-based therapies using oligonucleotides to gene targets, such as SMAD7, to mimic the effect of bacterial DNA or to influence specific splicing of anti-inflammatory microRNA miR-124 by small-molecule ABX464 have highlighted the translational importance of such approaches in IBD (Table 1)5,6. In other disease areas, mRNA vaccines for COVID-19 and the use of circulating tumour DNA in oncology are in clinical use7. However, DNA pathways and translational technologies are virtually unexplored areas in IBD. Here, we present a comprehensive appraisal of circulating nucleic acids (with a particular focus on DNA) and their potential roles in IBD with a focus on translational concepts and opportunities.
Box 1. Key milestones in the development of cfDNA technology.
1948 Mandel and Metais described the presence of DNA in blood
1966 Elevated circulating DNA in SLE with anti-DNA antibody formation
1973 Presence of circulating tumour DNA (ctDNA) in cancer patients – ‘Liquid biopsy’
1990 Launch of the human genome project
1997 Discovery of circulating cell-free foetal DNA in maternal plasma
2001 First draft of human genome published
2004 First metagenomics study
2008 First successes of prenatal diagnosis using foetal cfDNA in maternal plasma
2010 High circulating mitochondrial DNA in non-infective Systemic Inflammatory Response Syndrome (SIRS).
2018 High circulating mitochondrial DNA in active IBD, a chronic inflammatory condition.
2020 The Galleri blood test to screen for ~50 cancer ctDNA in 165 000 patients in England.
Table 1. Nucleic acid-based therapies reaching human trials in IBD.
Name | Disease | Mechanism | Target Development Progress | Reference |
---|---|---|---|---|
Cobitolimod | Ulcerative colitis | DNA-based oligonucleotide activating TLR-9 receptor leading to upregulation of anti-inflammatory IL10+ macrophages and regulatory T cells and suppresses Th17 cells |
Phase 2b completed in 2020. Phase 3 trials underway |
5,6 |
Mongersen | Crohn’s disease | Oral Smad7 antisense oligonucleotide | Phase 3 completed in 2020. No efficacy vs placebo in active Crohn’s disease. |
8 |
ABX-464 | Ulcerative colitis | Selectively upregulates miR-124 in immune cells | Phase 2b completed. Phase 3 trials are in planning phase. |
9,10 |
Alicaforsen | Crohn’s disease Ulcerative colitis |
Antisense oligonucleotide inhibiting ICAM-1 | Phase 3 is completed. No efficacy in Crohn’s. Limited efficacy in ulcerative proctitis |
11,12 |
Discovery of circulating DNA – Origins
DNA, the blueprint of all biological information, is contained in specialised compartments - in the nucleus or intracellular mitochondria, separate from the rest of the eukaryotic cell. This partitions major DNA functions and cellular processes, including gene expression, genome replication, and the repair of damaged DNA. Multiple biological machineries exist to tightly regulate and maintain a finely tuned state of cellular and tissue homeostasis, where cells can replicate while safely transferring their DNA.
cfDNA is present in the circulating plasma, urine, and other bodily fluids in humans13. It comprises double-stranded DNA fragments that are overwhelmingly short, <200 base pairs (bp), and are normally present at low concentrations14. The circulation of cfDNA in blood was first described in 1948, shortly after World War II, by Mandel and Metais15 using a perchloric acid precipitation method16. It is interesting to note that this was only a few years after DNA was first demonstrated as the material of inheritance and preceded Watson and Crick’s seminal paper on the double-helical structure of DNA17. This discovery received little attention during that era, and it was not until 1966 that it was found that cfDNA levels were elevated in systemic lupus erythematosus, a disease characterised by the formation of anti-dsDNA antibodies18. Interest in cfDNA began to increase when it was shown to be elevated in cancer patients in 1973, with cancer patients having approximately 10 times more cfDNA than healthy controls19. It has only been in the last 10–20 years that improved technology has led to an explosion of interest in cfDNA, particularly in the field of oncology. More recently, there has been emerging evidence that cfDNA may play an important role in regulating immune responses in health and various disease states (Table 2).
Table 2. Summary of clinical studies of cell-free DNA and behaviour in various disease states.
Disease State | cfDNA subtype | Sample Size | Effects | References |
---|---|---|---|---|
Trauma | mtDNA | 86 patients 40 controls |
mtDNA was associated with risk of post-trauma SIRS |
20 |
Trauma | mtDNA | 15 patients 12 controls |
mtDNA activates PMNs through TLR9 | 21 |
Trauma | mtDNA, ncDNA | 104 patients 42 controls |
No association between mtDNA and injury severity. ncDNA correlates but does not directly contribute to immune response. |
22 |
Systemic Lupus Erythematosus |
Total cfDNA, mtDNA, ncDNA |
43 patients 50 controls |
Total cfDNA higher in SLE Relative mtDNA higher in SLE. Higher fragmentation in SLE. |
23 |
Systemic Lupus Erythematosus |
Total cfDNA | 52 patients 25 controls |
cfDNA correlates with CRP cfDNA negatively correlates with anti-dsDNA, ANA |
24 |
Systemic Lupus Erythematosus |
Total cfDNA | 58 patients 259 controls |
cfDNA higher in SLE cfDNA higher in active disease |
25 |
Rheumatoid Arthritis | mtDNA, ncDNA | 74 patients 63 controls |
ncDNA higher in RhA mtDNA no association in RhA |
26 |
Rheumatoid Arthritis | mtDNA, ncDNA | 32 patients, longitudinal sampling at 3 and 6 months, 0 controls |
ncDNA and mtDNA decreased significantly following treatment |
27 |
Rheumatoid Arthritis | Total cfDNA | 30 patients 21 controls |
cfDNA higher in RhA. | 28 |
Sepsis | Total cfDNA | 27 sepsis 7 trauma |
cfDNA higher in sepsis vs trauma | 29 |
Sepsis | Total cfDNA | 80 patients 14 controls |
cfDNA higher in non-survivors | 30 |
Sepsis | Total cfDNA | 255 patients 0 controls |
cfDNA higher in non-survivors | 31 |
Sepsis | mtDNA | 20 sepsis 40 post-surgery 20 controls |
mtDNA higher in sepsis vs post-surgical inflammation |
32 |
COVID-19 | mtDNA | 97 patients 0 controls |
mtDNA higher in non-survivors | 33 |
Major depression | mtDNA | 109 major depressive disorder 28 bipolar disorder 17 schizophrenia 29 controls |
mtDNA decrease in depression and bipolar disorders |
34 |
cfDNA – cell-free DNA; mtDNA – mitochondrial cell-free DNA; ncDNA – nuclear cell-free DNA; SIRS – systemic inflammatory response syndrome; PMNs – polymorphonuclear neutrophils; TLR9 – toll-like receptor-9; SLE – systemic lupus erythematosus; CRP – C-reactive protein; RhA – rheumatoid arthritis
cfDNA in blood plasma consists of a mixture of fragmented DNA molecules released from various tissues within the body. They are primarily nucleosome-associated fragments that are wrapped around histone proteins, such as spools rather than free strands of DNA35. Each cfDNA fragment bears the molecular signatures of its cell of origin, such as DNA methylation status. The cellular/tissue origins of cfDNA have been studied using several approaches, such as nucleosome positioning35,36 and methylation analysis37,38.
cfDNA mostly originates from haematopoietic cell death37,39–41. In health, Moss et al. estimated that cfDNA is derived from 55% white blood cells, 30% erythrocyte progenitors, and 10% vascular endothelial cells, accounting for approximately 95% of cfDNA37. Of the remainder of cfDNA from non-haematopoietic tissue, the liver is estimated to account for a further 1–1.3% of the total cfDNA, which is consistent with estimates of daily liver cell turnover via apoptosis38,42. Notably, the absence of cfDNA signals from other tissues in the body with a high turnover rate, such as intestinal epithelial cells, lung, kidney, and skin, may reflect alternative clearance routes.
In disease states such as sepsis and cirrhosis, the elevation in cfDNA is mostly derived from blood cells37,38. Owing to tight nucleosome-wrapping of cfDNA and generally low quantities in circulation, it is unclear if they impart a direct inflammatory impact. However, circulating mitochondrial DNA exists in short fragments that have a strong immunostimulatory effect and from this perspective, mitochondrial DNA has received much recent attention in inflammatory diseases43.
Circulating cfDNA as an inflammatory trigger – ‘Matzinger’s Danger Model’
In 2000, Polly Matzinger proposed the ‘Danger Model’, where our immune system is primarily geared towards recognising and responding to danger irrespective of its origins. Hence, the immune system can be rapidly mobilised to respond to pathogen-associated molecular patterns (PAMPs) when host defences are compromised (e.g. in septicaemia) and danger-associated molecular patterns (DAMPs) are released during significant tissue damage. Although the former is apposite in IBD, given its close juxtaposition with gut microbiota, endogenously released DAMPs may also be important44. In both settings, activation of pattern recognition receptors (PRRs) by PAMPs or DAMPs leads to inflammation45.
Nucleic acids, whether released from our cells or a foreign source (e.g. bacteria, viruses, and vaccines), encapsulate Matzinger’s dictum, in that our immune system has evolved to develop built-in ‘alarm systems’ to respond to intrusion and calibrate an appropriate response. Following significant tissue damage in major trauma, Hauser et al. showed that mitochondrial DAMPs, such as formylated peptides and mitochondrial DNA, are released into the circulation with a direct functional effect of activating the immune system, leading to systemic inflammatory response syndrome (SIRS)21. SIRS is a serious condition characterised by systemic inflammation (fever, tachycardia, and breathlessness), multi-organ dysfunction and failure associated with high morbidity and mortality. In patients with severe trauma, mitochondrial cfDNA was thousands of folds higher than in healthy controls and persisted for 24 hours after injury. The authors further showed that the strong immune response was mediated by the activation of TLR9 by mitochondrial cfDNA in neutrophils.
We have shown that elevated mitochondrial cfDNA can be detected in the blood of patients with active and extensive IBD2. In fulminant cases of IBD, patients can present with evidence of systemic inflammation (such as SIRS) without evidence of bacterial infection. In our study, a subset of patients with severe IBD showed significantly decreased mitochondrial cfDNA levels after the removal of the inflamed colon2. Overall, circulating mitochondrial cfDNA has been detected in many inflammatory conditions and, most recently, is associated with disease severity in COVID-1933.
Circulating cell-free mitochondrial versus genomic DNA
Mitochondria descended from ɑ-proteobacteria in the ancestry of eukaryotic cells 1.5 billion years ago46, and consequently, mitochondrial cfDNA possesses unmethylated CpG motifs similar to bacterial DNA47. CpG motifs are short fragments of DNA that have cytosine followed by guanine residues, and they have particular significance in stimulating immune responses, notably (but not exclusively) via the Toll-like Receptor 9 (TLR9) signalling pathway48. Mitochondrial cfDNA is shorter (40–60 bp), more fragmented, has a higher GC content, and is not bound to nucleosomes compared to nuclear cfDNA49.
On the other hand, genomic DNA generally circulates with a size profile in the region of 20–200 bp with a peak of approximately 166 bp, corresponding to the size of a nucleosome. Other peaks were noted at ~320 bp and ~1000 bp, corresponding to dinucleosomes and chromatin, respectively50.
The innate immune system has developed a tiered system of nucleic acid sensors to recognise and respond to mislocalised or foreign DNA51, which is strategically located in cell compartments free of resident DNA (e.g. cytoplasm) to detect tissue injury and infection51. There are three main DNA sensors: TLR952, cyclic GMP-AMP synthase (cGAS), and Absent in Melanoma 2 (AIM2). TLR9 detects DNA rich in unmethylated CpG dinucleotides, which are rare in mammalian genomes, but are common in microbial and mitochondrial DNA. cGAS binds double-stranded DNA leading to activation of STING (stimulator of interferon genes) and resulting in type I interferon release, and the activation of NF-κB pathway to drive the secretion of inflammatory cytokines, such as tumour necrosis factor (TNF) and interleukin-6 (IL-6)53. AIM2 signalling leads to inflammasome activity and the cleaving of pro–IL-1β and pro–IL-18 to enable the secretion of the active forms of these inflammatory cytokines and also drives an inflammatory program of cell lysis (called pyroptosis54). A diverse range of noxious stimuli by damaging the mitochondria can result in mitochondrial DNA release that triggers NLRP3 inflammasome activation, thus acting as a nexus for the cellular ‘alarm system’55. TLR9, cGAS and AIM2 have different nucleic acid-sensing capacities based on CpG methylation, strand, and fragment length; however, it is notable that these receptors are located within the cellular cytosol (or endosomes in the case of TLR9) and are expressed mainly in immune cells. Hence, the entry mechanisms and types of immune cells involved are important for circulating and extracellular DNA.
While naked cfDNA is easily degraded, oxidation induces structural changes, which make the DNA more resistant to degradation and, therefore, available for uptake56. This is particularly relevant for mitochondrial DNA, where high reactive oxygen species generation leads to DNA oxidation and thereby significantly confers its inflammatory potential to NLRP3-inflammasome activation. Other entry mechanisms exist; for example, a process mediated by the antimicrobial peptide LL37 that can bind to both genomic and mitochondrial DNA. When bound to LL37, cfDNA condenses and forms a stable complex that is resistant to nuclease degradation57. DNA-LL37 complexes can then be transported via lipid rafts into the endosomal component of plasmacytoid dendritic cells, for example, where they can activate TLR9 and subsequently induce a type 1 interferon response58. This process can be pathogenic and is implicated in a range of diseases, including type 1 diabetes59, atherosclerosis57,60, psoriasis4, and SLE61.
Circulating cfDNA release and clearance
Figure 1 summarises the current understanding of the life cycle of cfDNA. cfDNA is released during cell death and/or active secretion. Cell death processes include apoptosis, necrosis, mitotic catastrophe, autophagy, necroptosis and pyroptosis, whereas active secretory processes include active secretion of cfDNA, packaging and circulation within exosomes and release in processes such as NETosis62. In addition, cfDNA can arise from exogenous sources, such as bacterial and viral DNA, during infection. In healthy states, the majority of cfDNA is packaged into nucleosomes and released as a by-product during apoptosis. Conversely, in disease states, uncontrolled cell death from necrosis can occur leading to the release of long fragments of 10,000 bp63,64. Therefore, the balance between apoptosis and necrosis can alter the cfDNA fragment size profile and indicate the underlying pathological processes. In severe trauma, nuclear cfDNA, but not mitochondrial cfDNA, correlates with early host inflammatory response22. Conversely, there is a higher mitochondrial/nuclear cfDNA ratio in patients with sickle cell disease than in healthy controls65.
Figure 1. Lifecycle of circulating cell-free DNA.
The half-life of cfDNA is short and is generally estimated to range from a few minutes to 1–2 hours66–69. Different clearance processes are likely to account for this variability. For example, studies examining the clearance of foetal DNA from maternal plasma show a biphasic process consisting of a rapid phase with a half-life of 10–60 mins and a slow phase with a half-life of 13 hours70. In cancer, cfDNA has an estimated half-life of 1–2 hours71,72. Several factors are known to affect cfDNA clearance, including the formation of molecular complexes which reduce degradation, epigenetic regulation, levels of nucleases, and transport mechanisms within the liver, spleen, and kidneys that remove cfDNA. These factors are not fully understood. However, emerging evidence suggests that these processes are not random73.
Within the circulation, cfDNA is rapidly degraded by nucleases, such as DNA fragmentation factor subunit beta (DFFB), intracellular deoxyribonuclease 1 like 3 (DNASE1L3), and deoxyribonuclease 1 (DNASE1). Using nuclease-deficient mouse models, the Lo group74,75 showed that cfDNA fragments were first generated intracellularly using DFFB, DNASE1L3 and other nucleases. Subsequently, fragmentation occurred in the extracellular space via DNASE1 and DNASE1L3. The 10 bp periodicity seen in the size profile of cfDNA originates from the cutting of DNA within an intact nucleosomal structure. DNASE1 levels are elevated in the context of trauma and may play an immunoregulatory role by clearing high cfDNA levels76. It must be noted that the degradation of cfDNA by nucleases only plays a partial role in cfDNA elimination77 and it is postulated that there are other unknown mechanisms by which cfDNA is cleared.
From circulation, cfDNA is eliminated via the liver, spleen, and kidneys. Most cfDNA (71–85%) is cleared by the liver69. Kupffer cells in the liver and macrophages in the spleen are responsible for trapping and clearing DNA and nucleosomes78. The kidneys play a smaller role. The presence of plasma-derived cfDNA in urine suggests some clearance; however, patients with chronic renal failure do not have higher levels of cfDNA66.
Circulating DNA in IBD
Although there are increasing data in health and oncology, our understanding of cfDNA in inflammatory diseases, pertinently in IBD, is still at an early stage. In health, circulating cfDNA is derived mainly from the apoptosis of haematopoietic lineage cells with minimal contributions from other tissues. As discussed earlier, the short half-life suggests a tightly maintained system of degradation and filtration that limits their potential pathogenic effects77.
Two other factors have been demonstrated to be involved in inflammatory disease, such as IBD. First, the concentration levels of circulating cfDNA were significantly higher during active systemic inflammation. Second, the underlying conditions may contribute to the dysregulation of cfDNA release (i.e. higher non-apoptotic cell death, release from the inflamed gut), pro-inflammatory qualitative changes in cfDNA (shorter fragment sizes, higher oxidised mitochondrial DNA in the setting of impaired cellular mitophagy), and impaired/overwhelmed clearance mechanisms. Traditional views of cfDNA as an epiphenomenon have been challenged by rapid advances in sequencing technologies that are more affordable and require much lower DNA concentrations, the simultaneous expansion of disease-related datasets, and the development of simple methods to stabilise cfDNA during blood sampling. Here we present key conceptual developments that will be of translational relevance in IBD (Figure 2).
Figure 2. Factors affecting the release of cfDNA in IBD and translational opportunities.
Circulating DNA as mechanistic biomarkers of IBD
In active IBD, both total circulating cfDNA and mitochondrial cfDNA levels are elevated2,79. In our study, patients with acute severe IBD have significantly higher levels of mitochondrial DNA; this correlation with more severe illness is in line with other diseases such as COVID-19 and trauma. However, unlike advances in oncology, where the clinical utility of cfDNA is geared toward the detection of cancer, crude plasma cfDNA concentration may not be useful for predicting the level of systemic or mucosal inflammation in IBD. Rather its use may be more relevant as a biomarker to identify dominant mechanisms (mechanistic biomarkers) in a heterogeneous complex disease, such as IBD, and potentially stratify patients to relevant therapeutic interventions.
In SLE, higher neutrophil release of the oxidised and more pro-inflammatory form of mitochondrial DNA is linked to defective mitophagy, a process that is also highly relevant in IBD. In this context, the role of mitochondrial cfDNA is more defined, given the increasing lines of evidence to implicate mitochondrial dysfunction in IBD80. Two Phase 2b randomised controlled trials on the use of oral mitochondrial antioxidant therapy in adults and children with UC (Marvel and MiniMarvel ClinicalTrials.gov Identifier: NCT0427674081) will incorporate the quantification of plasma mitochondrial cfDNA into the study design. A further study involving 1500 IBD patients will measure cfDNA and assess its correlation with mucosal healing in IBD (MUSIC study; ClinicalTrials.gov Identifier NCT0476096482,83).
Finding new therapeutics from the role of cfDNA in inflammation
As discussed, nucleic acid sensors play a clear role, given that immune cells can take up cfDNA fragments with consequential effects. Earlier work has shown that this effect is not didactic, but more complex and dependent on features such as specific DNA sequence, fragment size, and structural modification, for example, oxidation of mitochondrial DNA. The earliest work was based on the study of CpG oligonucleotides (ODNs), in which three different classes of ODNs were characterised by their structure and pro-inflammatory effects. Class A ODNs are multimeric structures containing central unmethylated CpG motifs that accumulate in early endosomes, where they induce IFNα production by plasmacytoid dendritic cells (pDC) and can trigger B cell proliferation. In contrast, class B ODNs are monomeric, contain one or more unmethylated CpG motifs, and accumulate in late endosomes, where they induce pDC maturation via TLR9 signalling. Class C ODNs combine the features of class A and class B ODNs, accumulate in both early and late endosomes, and induce activation and maturation of pDCs84.
The triggering of PRR activation by host and bacterial DNA can have either pro- or anti-inflammatory effects depending on the tissue- and disease context. In a mouse model of colitis, bacterial CpG worsens the disease via TLR9 activation85,86. On the other hand, several studies have also shown the potential anti-inflammatory effect of TLR9-activation. Rachmilewitz et al. described synthetic Class B ODNs with multiple TCG repeats containing unmethylated CpG motifs that suppressed TNFα and IL1β generation in ex vivo cultured colonoscopy biopsies from UC patients87. This study also showed that the anti-inflammatory effect of certain unmethylated CpG ODNs depends on the exact sequence and that methylation of the CpG repeat abolished the anti-inflammatory potential. In 2017, Liu et al. described the immunosuppressive effects of a TLR-antagonistic CpG ODN by inhibiting innate immune signalling via TLR3 and 7/8 in a TLR9-independent manner88. The CpG ODN cobitolimod contains a single unmethylated CpG motif and has been shown to induce anti-inflammatory responses via TLR95.
In addition to synthetic CpG-ODNs, the anti-inflammatory potential of cfDNA has also been described; however, the exact mechanisms underlying this effect have not yet been elucidated. In this context, two studies showed that prophylactic administration of cfDNA from colitic into healthy mice was protective against DSS-induced colitis89,90. A recent study by Constantinovits et al. found that injecting healthy mice with colitis-derived cfDNA before the induction of DSS-colitis changed the expression of several autophagy and inflammatory cytokines in a clinically favourable manner at the gene and protein levels91.
Other potential therapeutic pathways will come from the better characterisation of cfDNA and its specific effects on inflammation. This includes how blood cfDNA is released, removed from circulation and/or its downstream effects on inflammatory pathways. Notably, these studies can be run in parallel with mechanistic studies to accelerate drug development timelines. One example is our current study on the efficacy of mitochondrial antioxidant therapy in UC, where one postulated mechanism of action is by reducing the release of mitochondrial cfDNA from gut mitochondrial damage92. In IBD, one of the challenges in the development of therapeutic antisense oligonucleotides (ASO) that inhibit or modulate gene targets via RNA is the abundant presence of nucleases in plasma and tissue that rapidly degrade ASOs93.
Development of the next generation highly specific biomarker of gut damage in IBD
A second development relates to the ability to leverage new sequencing technologies and analytical methods to identify the tissue origin of cfDNA. More recently, Esfahani et al. showed that the origin of cfDNA can be inferred from fragmentation profiles of cfDNA94. Circulating cfDNA molecules are primarily nucleosome-associated fragments; they represent distinctive chromatin configurations of the nuclear genome of cells from which they originate. Specifically, genomic regions densely associated with nucleosomal complexes are generally protected against the action of various endonucleases, whereas open chromatin is not. Hence, specific chromatin fragmentation features are potentially useful for the classification of tissue of origin by cfDNA profiling, which includes fragmentation profiles of lower-depth sequencing coverage and disruption of nucleosome positioning near transcription start sites in groups of genes relevant to a particular tissue site.
Such advances can be directly exploited to find very sensitive biomarkers of gut damage in IBD with realistic clinical applications: to track mucosal healing in IBD, to screen and detect pre-IBD in high-risk groups (i.e. those with a strong family history) and even to identify precancerous changes during IBD surveillance. From a theoretical perspective, this is likely to be superior to current clinical biomarkers such as calprotectin which relies on protein release into the gut from neutrophils or C-reactive protein produced by hepatocytes during systemic inflammation. If these technologies are proven effective, they could be compared against the current gold standard of colonoscopy, potentially decreasing the need for invasive assessment and improving upon the current non-invasive blood and stool tests used in clinics.
Using the Tabula Sapiens transcriptomic cell atlas and Human Protein Atlas RNA consensus dataset, Vorperian et al. generated cell-type signature scores, which allow the inference of cell types that contribute to cell-free RNA for a variety of diseases95. Here, they showed that loss of cell-free RNA (cfRNA)-specific signatures from tubular cells and neurons from the kidneys and brain are associated with chronic kidney disease and Alzheimer’s disease, respectively, whereas increased hepatocyte-specific cfRNA signatures are linked to hepatic steatosis96. Another study by the same group showed that cfRNA signatures could predict the development of preeclampsia during pregnancy97. This exciting progress has provided a conceptual platform for cfDNA in IBD, although much work remains to be done.
Box 2. cfDNA in IBD–Current questions and potential translational impact.
1. Does active IBD result in higher cfDNA release with functional relevance to the development of chronic inflammation?
2. Can advances in technologies allow very sensitive detection of gut inflammation and prioritisation of inflammatory mechanisms using liquid biopsy approach?
Conclusion
The understanding of the role of cfDNA in inflammation, in parallel with the development of technologies to accurately detect their presence, origins, and relationships with underlying inflammatory mechanisms, is likely to represent significant translational advances in the next 10 years. The use of nucleic acid-based therapies and technologies is already in the clinical mainstream – in vaccines, in ‘liquid biopsies’ in oncology, and prenatal diagnostics. In IBD, this approach is a new major step toward realising the potential of Precision Medicine. Incorporating knowledge from cellular pathophysiology, which often forms the basis of disease, into a liquid biopsy would more loosely match the resolution afforded by invasive procedures that can enable stratification and the discovery of a potentially new class of treatments in IBD.
Box 3. Definitions.
Nucleic acids – A class of macromolecules found in all cells and viruses composed of nucleotides that encode information and the two major classes are DNA and RNA.
DNA – Deoxyribonucleic acid. Double-stranded helical structure composed of one of the four nucleotide bases (adenine, thymine, cytosine, guanine) first described by Watson and Crick, allowing for stability of genomic information and ability for repair and replication with high accuracy.
RNA – Ribonucleic acid. In contrast to DNA, RNA is single stranded, making it more prone to mutation and degradation. RNA has no thymine (T) but instead is replaced by uracil (U) as one of its four component nucleotides. DNA is transcribed into messenger RNA which is exported from the nucleus leading to subsequent protein expression.
Nucleosome -A nucleosome is the basic structural unit of DNA packaging in eukaryotes. The structure of a nucleosome consists of a segment of DNA wound around eight histone proteins and resembles thread wrapped around a spool.
Cell-free DNA – All non-encapsulated DNA fragments found in blood plasma.
Liquid Biopsies – The genomic analyses of cell-free DNA that circulates in the blood can be used in prenatal testing, oncology, and monitoring organ transplant recipients.
Mitochondria – Organelle found in large numbers in most cells where biochemical processes of respiration and energy production occur.
Mitochondrial DNA – 16.6 kilobases in length encoding for 13 proteins (ATP synthase, cytochrome c oxidase, cytochrome b, NADH dehydrogenase). 2 ribosomal RNAs (12S and 16S), 22 transfer RNAs – all of which involved in the oxidative phosphorylation process. Its genome is rich in unmethylated CpG-like bacteria. CpGs in human genomic DNA, conversely, are usually methylated as a means of controlling gene expression.
Damage-associated molecular patterns (DAMPs) – Host-derived endogenous signals that are released during tissue damage and cellular stress in many human inflammatory conditions.
Pathogen-associated molecular patterns (PAMPs) – Molecular signatures conserved by bacteria but not present in the host.
Pattern recognition receptors (PRRs) – Germline encoded host sensors which help detect molecular signatures that lead to an inflammatory response. These molecular signatures can be classified as either PAMPs or DAMPs.
Nucleic acid sensors -Toll-like receptor 9 (TLR9), cyclic GMP-AMP synthase (cGAS) and Absent in Melanoma 2 (AIM2)
The peer reviewers who approve this article are:
Mairi McLean, Division of Molecular & Clinical Medicine, School of Medicine, University of Dundee
Craig Mowat, Department of Gastroenterology, Ninewells Hospital, Dundee, Scotland, UK
Funding Statement
The Leona M. and Harry B. Helmsley Charitable Trust (SC, GTH) and Crohn’s and Colitis UK (LF).
References
- 1. Verstockt B, Parkes M, Lee JC: How do we predict a patient’s disease course and whether they will respond to specific treatments? Gastroenterology. 2022; 162(5): 1383–1395. 10.1053/j.gastro.2021.12.245 [DOI] [PubMed] [Google Scholar]
- 2. Boyapati RK, Dorward DA, Tamborska A, et al. : Mitochondrial DNA Is a Pro-Inflammatory Damage-Associated Molecular Pattern Released During Active IBD. Inflamm Bowel Dis. 2018; 24(10): 2113–22. 10.1093/ibd/izy095 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Lande R, Gregorio J, Facchinetti V, et al. : Plasmacytoid dendritic cells sense self-DNA coupled with antimicrobial peptide. Nature. 2007; 449(7162): 564–9. 10.1038/nature06116 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 4. Liang H, Yan Y, Wu J, et al. : Topical nanoparticles interfering with the DNA-LL37 complex to alleviate psoriatic inflammation in mice and monkeys. Sci Adv. 2020; 6(31): eabb5274. 10.1126/sciadv.abb5274 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Schmitt H, Ulmschneider J, Billmeier U, et al. : The TLR9 Agonist Cobitolimod Induces IL10-Producing Wound Healing Macrophages and Regulatory T Cells in Ulcerative Colitis. J Crohns Colitis. 2020; 14(4): 508–24. 10.1093/ecco-jcc/jjz170 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Atreya R, Peyrin-Biroulet L, Klymenko A, et al. : Cobitolimod for moderate-to-severe, left-sided ulcerative colitis (CONDUCT): a phase 2b randomised, double-blind, placebo-controlled, dose-ranging induction trial. Lancet Gastroenterol Hepatol. 2020; 5(12): 1063–75. 10.1016/S2468-1253(20)30301-0 [DOI] [PubMed] [Google Scholar]
- 7. Otandault A, Anker P, Al Amir Dache Z, et al. : Recent advances in circulating nucleic acids in oncology. Ann Oncol. 2019; 30(3): 374–84. 10.1093/annonc/mdz031 [DOI] [PubMed] [Google Scholar]
- 8. Sands BE, Feagan BG, Sandborn WJ, et al. : Mongersen (GED-0301) for Active Crohn’s Disease: Results of a Phase 3 Study. Am J Gastroenterol. 2020; 115(5): 738–745. 10.14309/ajg.0000000000000493 [DOI] [PubMed] [Google Scholar]
- 9. Tazi J, Begon-Pescia C, Campos N, et al. : Specific and selective induction of miR-124 in immune cells by the quinoline ABX464: a transformative therapy for inflammatory diseases. Drug Discov Today. 2021; 26(4): 1030–1039. 10.1016/j.drudis.2020.12.019 [DOI] [PubMed] [Google Scholar]
- 10. Steens JM, Scherrer D, Gineste P, et al. : Safety, Pharmacokinetics, and Antiviral Activity of a Novel HIV Antiviral, ABX464, in Treatment-Naive HIV-Infected Subjects in a Phase 2 Randomized, Controlled Study. Antimicrob Agents Chemother. 2017; 61(7): e00545–17. 10.1128/AAC.00545-17 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Yacyshyn B, Chey WY, Wedel MK, et al. : A Randomized, Double-Masked, Placebo-Controlled Study of Alicaforsen, an Antisense Inhibitor of Intercellular Adhesion Molecule 1, for the Treatment of Subjects With Active Crohn’s Disease. Clin Gastroenterol Hepatol. 2007; 5(2): 215–20. 10.1016/j.cgh.2006.11.001 [DOI] [PubMed] [Google Scholar]
- 12. Vegter S, Tolley K, Wilson Waterworth T, et al. : Meta-analysis using individual patient data: efficacy and durability of topical alicaforsen for the treatment of active ulcerative colitis. Aliment Pharmacol Ther. 2013; 38(3): 284–93. 10.1111/apt.12369 [DOI] [PubMed] [Google Scholar]
- 13. Chan AKC, Chiu RWK, Lo YMD, et al. : Cell-free nucleic acids in plasma, serum and urine: a new tool in molecular diagnosis. Ann Clin Biochem. 2003; 40(Pt 2): 122–30. 10.1258/000456303763046030 [DOI] [PubMed] [Google Scholar]
- 14. Fleischhacker M, Schmidt B: Circulating nucleic acids (CNAs) and cancer--a survey. Biochim Biophys Acta. 2007; 1775(1): 181–232. 10.1016/j.bbcan.2006.10.001 [DOI] [PubMed] [Google Scholar]
- 15. Mandel P, Métais P: Les acides nucléiques du plasma sanguin chez l’homme. C R Acad Sci Paris 142. 1948; 142: 241–3. 10.1373/clinchem.2005.051003 [DOI] [PubMed] [Google Scholar]
- 16. Anker P, Stroun M: Circulating DNA in Plasma or Serum. Fetal Cells Fetal DNA Matern Blood. 2001; 1–7. [Google Scholar]
- 17. Thierry AR, El Messaoudi S, Gahan PB, et al. : Origins, structures, and functions of circulating DNA in oncology. Cancer Metastasis Rev. 2016; 35(3): 347–76. 10.1007/s10555-016-9629-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Tan EM, Schur PH, Carr RI, et al. : Deoxybonucleic acid (DNA) and antibodies to DNA in the serum of patients with systemic lupus erythematosus. J Clin Invest. 1966; 45(11): 1732–40. 10.1172/JCI105479 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Leon SA, Shapiro B, Sklaroff DM, et al. : Free DNA in the serum of cancer patients and the effect of therapy. Cancer Res. 1977; 37(3): 646–50. [PubMed] [Google Scholar]
- 20. Gu X, Yao Y, Wu G, et al. : The Plasma Mitochondrial DNA Is an Independent Predictor for Post-Traumatic Systemic Inflammatory Response Syndrome. PLoS One. 2013; 8(8): e72834. 10.1371/journal.pone.0072834 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 21. Zhang Q, Raoof M, Chen Y, et al. : Circulating Mitochondrial DAMPs Cause Inflammatory Responses to Injury. Nature. 2010; 464(7285): 104–7. 10.1038/nature08780 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 22. Stortz JA, Hawkins RB, Holden DC, et al. : Cell-free nuclear, but not mitochondrial, DNA concentrations correlate with the early host inflammatory response after severe trauma. Sci Rep. 2019; 9(1): 13648. 10.1038/s41598-019-50044-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Truszewska A, Wirkowska A, Gala K, et al. : Cell-free DNA profiling in patients with lupus nephritis. Lupus. 2020; 29(13): 1759–1772. 10.1177/0961203320957717 [DOI] [PubMed] [Google Scholar]
- 24. Hendy OM, Motalib TA, El Shafie MA, et al. : Circulating cell free DNA as a predictor of systemic lupus erythematosus severity and monitoring of therapy. Egypt J Med Hum Genet. 2016; 17(1): 79–85. 10.1016/j.ejmhg.2015.07.001 [DOI] [Google Scholar]
- 25. Xu Y, Song Y, Chang J, et al. : High levels of circulating cell-free DNA are a biomarker of active SLE. Eur J Clin Invest. 2018; 48(11): e13015. 10.1111/eci.13015 [DOI] [PubMed] [Google Scholar]
- 26. Rykova E, Sizikov A, Roggenbuck D, et al. : Circulating DNA in rheumatoid arthritis: pathological changes and association with clinically used serological markers. Arthritis Res Ther. 2017; 19(1): 85. 10.1186/s13075-017-1295-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Lauková L, Konečná B, Vlková B, et al. : Anti-cytokine therapy and plasma DNA in patients with rheumatoid arthritis. Rheumatol Int. 2018; 38(8): 1449–54. 10.1007/s00296-018-4055-8 [DOI] [PubMed] [Google Scholar]
- 28. Hashimoto T, Yoshida K, Hashimoto N, et al. : Circulating cell free DNA: a marker to predict the therapeutic response for biological DMARDs in rheumatoid arthritis. Int J Rheum Dis. 2017; 20(6): 722–30. 10.1111/1756-185X.12959 [DOI] [PubMed] [Google Scholar]
- 29. Clementi A, Virzì GM, Brocca A, et al. : The Role of Cell-Free Plasma DNA in Critically Ill Patients with Sepsis. Blood Purif. 2016; 41(1–3): 34–40. 10.1159/000440975 [DOI] [PubMed] [Google Scholar]
- 30. Dwivedi DJ, Toltl LJ, Swystun LL, et al. : Prognostic utility and characterization of cell-free DNA in patients with severe sepsis. Crit Care. 2012; 16(4): R151. 10.1186/cc11466 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Saukkonen K, Lakkisto P, Pettilä V, et al. : Cell-Free Plasma DNA as a Predictor of Outcome in Severe Sepsis and Septic Shock. Clin Chem. 2008; 54(6): 1000–7. 10.1373/clinchem.2007.101030 [DOI] [PubMed] [Google Scholar]
- 32. Schneck E, Edinger F, Hecker M, et al. : Blood Levels of Free-Circulating Mitochondrial DNA in Septic Shock and Postsurgical Systemic Inflammation and Its Influence on Coagulation: A Secondary Analysis of a Prospective Observational Study. J Clin Med. 2020; 9(7): 2056. 10.3390/jcm9072056 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Scozzi D, Cano M, Ma L, et al. : Circulating mitochondrial DNA is an early indicator of severe illness and mortality from COVID-19. JCI Insight. 2021; 6(4): e143299. 10.1172/jci.insight.143299 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Kageyama Y, Kasahara T, Kato M, et al. : The relationship between circulating mitochondrial DNA and inflammatory cytokines in patients with major depression. J Affect Disord. 2018; 233: 15–20. 10.1016/j.jad.2017.06.001 [DOI] [PubMed] [Google Scholar]
- 35. Snyder MW, Kircher M, Hill AJ, et al. : Cell-free DNA comprises an in vivo nucleosome footprint that informs its tissues-of-origin. Cell. 2016; 164(1–2): 57–68. 10.1016/j.cell.2015.11.050 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 36. Ulz P, Thallinger GG, Auer M, et al. : Inferring expressed genes by whole-genome sequencing of plasma DNA. Nat Genet. 2016; 48(10): 1273–8. 10.1038/ng.3648 [DOI] [PubMed] [Google Scholar]
- 37. Moss J, Magenheim J, Neiman D, et al. : Comprehensive human cell-type methylation atlas reveals origins of circulating cell-free DNA in health and disease. Nat Commun. 2018; 9(1): 5068. 10.1038/s41467-018-07466-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Liu X, Ren J, Luo N, et al. : Comprehensive DNA methylation analysis of tissue of origin of plasma cell-free DNA by methylated CpG tandem amplification and sequencing (MCTA-Seq). Clin Epigenetics. 2019; 11(1): 93. 10.1186/s13148-019-0689-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Lam WKJ, Gai W, Sun K, et al. : DNA of Erythroid Origin Is Present in Human Plasma and Informs the Types of Anemia. Clin Chem. 2017; 63(10): 1614–23. 10.1373/clinchem.2017.272401 [DOI] [PubMed] [Google Scholar]
- 40. Lui YY, Chik KW, Chiu RW, et al. : Predominant Hematopoietic Origin of Cell-free DNA in Plasma and Serum after Sex-mismatched Bone Marrow Transplantation. Clin Chem. 2002; 48(3): 421–7. 10.1093/clinchem/48.3.421 [DOI] [PubMed] [Google Scholar]
- 41. Zheng YWL, Chan KCA, Sun H, et al. : Nonhematopoietically derived DNA is shorter than hematopoietically derived DNA in plasma: a transplantation model. Clin Chem. 2012; 58(3): 549–58. 10.1373/clinchem.2011.169318 [DOI] [PubMed] [Google Scholar]
- 42. Lehmann-Werman R, Magenheim J, Moss J, et al. : Monitoring liver damage using hepatocyte-specific methylation markers in cell-free circulating DNA. JCI Insight. 2018; 3(12): e120687. 10.1172/jci.insight.120687 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Boyapati RK, Tamborska A, Dorward DA, et al. : Advances in the understanding of mitochondrial DNA as a pathogenic factor in inflammatory diseases [version 1; peer review: 3 approved]. F1000Res. 2017; 6: 169. 10.12688/f1000research.10397.1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Boyapati RK, Rossi AG, Satsangi J, et al. : Gut mucosal DAMPs in IBD: from mechanisms to therapeutic implications. Mucosal Immunol. 2016; 9(3): 567–82. 10.1038/mi.2016.14 [DOI] [PubMed] [Google Scholar]
- 45. Kotas ME, Medzhitov R: Homeostasis, inflammation, and disease susceptibility. Cell. 2015; 160(5): 816–27. 10.1016/j.cell.2015.02.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Gray MW: Mitochondrial Evolution. Cold Spring Harb Perspect Biol. 2012; 4(9): a011403. 10.1101/cshperspect.a011403 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Meyer A, Laverny G, Bernardi L, et al. : Mitochondria: An Organelle of Bacterial Origin Controlling Inflammation. Front Immunol. 2018; 9: 536. 10.3389/fimmu.2018.00536 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 48. Krieg AM: CpG motifs in bacterial DNA and their immune effects. Annu Rev Immunol. 2002; 20: 709–60. 10.1146/annurev.immunol.20.100301.064842 [DOI] [PubMed] [Google Scholar]
- 49. West AP, Shadel GS: Mitochondrial DNA in innate immune responses and inflammatory pathology. Nat Rev Immunol. 2017; 17(6): 363–75. 10.1038/nri.2017.21 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 50. Sanchez C, Roch B, Mazard T, et al. : Circulating nuclear DNA structural features, origins, and complete size profile revealed by fragmentomics. JCI Insight. 2021; 6(7): e144561. 10.1172/jci.insight.144561 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Emming S, Schroder K: Tiered DNA sensors for escalating responses. Science. 2019; 365(6460): 1375–6. 10.1126/science.aay2701 [DOI] [PubMed] [Google Scholar]
- 52. Hemmi H, Takeuchi O, Kawai T, et al. : A Toll-like receptor recognizes bacterial DNA. Nature. 2000; 408(6813): 740–5. 10.1038/35047123 [DOI] [PubMed] [Google Scholar]
- 53. Kato K, Omura H, Ishitani R, et al. : Cyclic GMP-AMP as an Endogenous Second Messenger in Innate Immune Signaling by Cytosolic DNA. Annu Rev Biochem. 2017; 86: 541–66. 10.1146/annurev-biochem-061516-044813 [DOI] [PubMed] [Google Scholar]
- 54. Boucher D, Monteleone M, Coll RC, et al. : Caspase-1 self-cleavage is an intrinsic mechanism to terminate inflammasome activity. J Exp Med. 2018; 215(3): 827–40. 10.1084/jem.20172222 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Zhong Z, Liang S, Sanchez-Lopez E, et al. : New mitochondrial DNA synthesis enables NLRP3 inflammasome activation. Nature. 2018; 560(7717): 198–203. 10.1038/s41586-018-0372-z [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 56. Gehrke N, Mertens C, Zillinger T, et al. : Oxidative damage of DNA confers resistance to cytosolic nuclease TREX1 degradation and potentiates STING-dependent immune sensing. Immunity. 2013; 39(3): 482–95. 10.1016/j.immuni.2013.08.004 [DOI] [PubMed] [Google Scholar]
- 57. Zhang Z, Meng P, Han Y, et al. : Mitochondrial DNA-LL-37 Complex Promotes Atherosclerosis by Escaping from Autophagic Recognition. Immunity. 2015; 43(6): 1137–47. 10.1016/j.immuni.2015.10.018 [DOI] [PubMed] [Google Scholar]
- 58. Sandgren S, Wittrup A, Cheng F, et al. : The human antimicrobial peptide LL-37 transfers extracellular DNA plasmid to the nuclear compartment of mammalian cells via lipid rafts and proteoglycan-dependent endocytosis. J Biol Chem. 2004; 279(17): 17951–6. 10.1074/jbc.M311440200 [DOI] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 59. Badal D, Dayal D, Singh G, et al. : Role of DNA-LL37 complexes in the activation of plasmacytoid dendritic cells and monocytes in subjects with type 1 diabetes. Sci Rep. 2020; 10(1): 8896. 10.1038/s41598-020-65851-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Döring Y, Manthey HD, Drechsler M, et al. : Auto-antigenic protein-DNA complexes stimulate plasmacytoid dendritic cells to promote atherosclerosis. Circulation. 2012; 125(13): 1673–83. 10.1161/CIRCULATIONAHA.111.046755 [DOI] [PubMed] [Google Scholar]
- 61. Lande R, Ganguly D, Facchinetti V, et al. : Neutrophils Activate Plasmacytoid Dendritic Cells by Releasing Self-DNA-Peptide Complexes in Systemic Lupus Erythematosus. Sci Transl Med. 2011; 3(73): 73ra19. 10.1126/scitranslmed.3001180 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 62. Aucamp J, Bronkhorst AJ, Badenhorst CPS, et al. : The diverse origins of circulating cell-free DNA in the human body: a critical re-evaluation of the literature. Biol Rev Camb Philos Soc. 2018; 93(3): 1649–83. 10.1111/brv.12413 [DOI] [PubMed] [Google Scholar]
- 63. Schwarzenbach H, Pantel K: Circulating DNA as biomarker in breast cancer. Breast Cancer Res. 2015; 17(1): 136. 10.1186/s13058-015-0645-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Mouliere F, Robert B, Arnau Peyrotte E, et al. : High fragmentation characterizes tumour-derived circulating DNA. PLoS One. 2011; 6(9): e23418. 10.1371/journal.pone.0023418 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Tumburu L, Ghosh-Choudhary S, Seifuddin FT, et al. : Circulating mitochondrial DNA is a proinflammatory DAMP in sickle cell disease. Blood. 2021; 137(22): 3116–26. 10.1182/blood.2020009063 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Celec P, Vlková B, Lauková L, et al. : Cell-free DNA: the role in pathophysiology and as a biomarker in kidney diseases. Expert Rev Mol Med. 2018; 20: e1. 10.1017/erm.2017.12 [DOI] [PubMed] [Google Scholar]
- 67. Thierry AR, Mouliere F, Gongora C, et al. : Origin and quantification of circulating DNA in mice with human colorectal cancer xenografts. Nucleic Acids Res. 2010; 38(18): 6159–75. 10.1093/nar/gkq421 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. García Moreira V, de la Cera Martínez T, Gago González E, et al. : Increase in and clearance of cell-free plasma DNA in hemodialysis quantified by real-time PCR. Clin Chem Lab Med. 2006; 44(12): 1410–5. 10.1515/CCLM.2006.252 [DOI] [PubMed] [Google Scholar]
- 69. Gauthier VJ, Tyler LN, Mannik M: Blood clearance kinetics and liver uptake of mononucleosomes in mice. J Immunol. 1996; 156(3): 1151–6. 10.4049/jimmunol.156.3.1151 [DOI] [PubMed] [Google Scholar]
- 70. Yu SCY, Lee SWY, Jiang P, et al. : High-resolution profiling of fetal DNA clearance from maternal plasma by massively parallel sequencing. Clin Chem. 2013; 59(8): 1228–37. 10.1373/clinchem.2013.203679 [DOI] [PubMed] [Google Scholar]
- 71. Muhanna N, Di Grappa MA, Chan HHL, et al. : Cell-Free DNA Kinetics in a Pre-Clinical Model of Head and Neck Cancer. Sci Rep. 2017; 7(1): 16723. 10.1038/s41598-017-17079-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Diehl F, Schmidt K, Choti MA, et al. : Circulating mutant DNA to assess tumor dynamics. Nat Med. 2008; 14(9): 985–90. 10.1038/nm.1789 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Ivanov M, Baranova A, Butler T, et al. : Non-random fragmentation patterns in circulating cell-free DNA reflect epigenetic regulation. BMC Genomics. 2015; 16 Suppl 13(Suppl 13): S1. 10.1186/1471-2164-16-S13-S1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Han DSC, Ni M, Chan RWY, et al. : The Biology of Cell-free DNA Fragmentation and the Roles of DNASE1, DNASE1L3, and DFFB. Am J Hum Genet. 2020; 106(2): 202–14. 10.1016/j.ajhg.2020.01.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Serpas L, Chan RWY, Jiang P, et al. : Dnase1l3 deletion causes aberrations in length and end-motif frequencies in plasma DNA. Proc Natl Acad Sci U S A. 2019; 116(2): 641–9. 10.1073/pnas.1815031116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Meng W, Paunel-Görgülü A, Flohé S, et al. : Deoxyribonuclease is a potential counter regulator of aberrant neutrophil extracellular traps formation after major trauma. Mediators Inflamm. 2012; 2012: 149560. 10.1155/2012/149560 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Lo YM, Zhang J, Leung TN, et al. : Rapid clearance of fetal DNA from maternal plasma. Am J Hum Genet. 1999; 64(1): 218–24. 10.1086/302205 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Du Clos TW, Volzer MA, Hahn FF, et al. : Chromatin clearance in C57Bl/10 mice: interaction with heparan sulphate proteoglycans and receptors on Kupffer cells. Clin Exp Immunol. 1999; 117(2): 403–11. 10.1046/j.1365-2249.1999.00976.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Vrablicova Z, Tomova K, Tothova L, et al. : Nuclear and Mitochondrial Circulating Cell-Free DNA Is Increased in Patients With Inflammatory Bowel Disease in Clinical Remission. Front Med (Lausanne). 2020; 7: 593316. 10.3389/fmed.2020.593316 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Ho GT, Theiss AL: Mitochondria and Inflammatory Bowel Diseases: Toward a Stratified Therapeutic Intervention. Annu Rev Physiol. 2022; 84: 435–459. 10.1146/annurev-physiol-060821-083306 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. University of Edinburgh: Mitochondrial Anti-oxidant Therapy to Resolve Inflammation in Ulcerative Colitis (MARVEL): A Randomised Placebo-controlled Trial on Oral MitoQ in Moderate UC. clinicaltrials.gov; Report No.: NCT04276740, 2021; [cited 2022 May 5]. Reference Source [Google Scholar]
- 82. Chuah S, Hall R, Ward E, et al. : Mitochondrial DAMPs as mechanistic biomarkers of mucosal inflammation in Crohn’s disease: Study protocol for prospective longitudinal cohort study in Scotland. medRxiv. 2022; [cited 2022 May 9]. 10.1101/2022.03.21.22270313 [DOI] [Google Scholar]
- 83. University of Edinburgh: Mitochondrial DAMPs as Mechanistic Biomarkers of Mucosal Inflammation in Crohn’s Disease and Ulcerative Colitis (MUSIC). clinicaltrials.gov; Report No.: NCT04760964, 2021; [cited 2022 May 5]. Reference Source [Google Scholar]
- 84. Marshall JD, Fearon K, Abbate C, et al. : Identification of a novel CpG DNA class and motif that optimally stimulate B cell and plasmacytoid dendritic cell functions. J Leukoc Biol. 2003; 73(6): 781–92. 10.1189/jlb.1202630 [DOI] [PubMed] [Google Scholar]
- 85. Obermeier F, Dunger N, Strauch UG, et al. : CpG Motifs of Bacterial DNA Essentially Contribute to the Perpetuation of Chronic Intestinal Inflammation. Gastroenterology. 2005; 129(3): 913–27. 10.1053/j.gastro.2005.06.061 [DOI] [PubMed] [Google Scholar]
- 86. O’Hara JR, Feener TD, Fischer CD, et al. : Campylobacter jejuni disrupts protective Toll-like receptor 9 signaling in colonic epithelial cells and increases the severity of dextran sulfate sodium-induced colitis in mice. Infect Immun. 2012; 80(4): 1563–71. 10.1128/IAI.06066-11 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Rachmilewitz D, Karmeli F, Shteingart S, et al. : Immunostimulatory oligonucleotides inhibit colonic proinflammatory cytokine production in ulcerative colitis. Inflamm Bowel Dis. 2006; 12(5): 339–45. [DOI] [PubMed] [Google Scholar]
- 88. Liu W, Yang X, Wang N, et al. : Multiple Immunosuppressive Effects of CpG-c41 on Intracellular TLR-Mediated Inflammation. Mediators Inflamm. 2017; 2017: 6541729. 10.1155/2017/6541729 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Sipos F, Műzes G, Fűri I, et al. : Intravenous administration of a single-dose free-circulating DNA of colitic origin improves severe murine DSS-colitis. Pathol Oncol Res. 2014; 20(4): 867–77. 10.1007/s12253-014-9766-x [DOI] [PubMed] [Google Scholar]
- 90. Műzes G, Sipos F, Fűri I, et al. : Preconditioning with intravenous colitic cell-free DNA prevents DSS-colitis by altering TLR9-associated gene expression profile. Dig Dis Sci. 2014; 59(12): 2935–46. 10.1007/s10620-014-3325-x [DOI] [PubMed] [Google Scholar]
- 91. Constantinovits M, Sipos F, Kiss AL, et al. : Preconditioning with cell-free DNA prevents DSS-colitis by promoting cell protective autophagy. J Investig Med. 2020; 68(5): 992–1001. 10.1136/jim-2020-001296 [DOI] [PubMed] [Google Scholar]
- 92. Gwyer Findlay E, Sutton G, Ho GT: The MARVEL trial: a phase 2b randomised placebo-controlled trial of oral MitoQ in moderate ulcerative colitis. Immunother Adv. 2021; 1(1): ltaa002. 10.1093/immadv/ltaa002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Eder PS, DeVine RJ, Dagle JM, et al. : Substrate specificity and kinetics of degradation of antisense oligonucleotides by a 3’ exonuclease in plasma. Antisense Res Dev. 1991; 1(2): 141–51. 10.1089/ard.1991.1.141 [DOI] [PubMed] [Google Scholar]
- 94. Esfahani MS, Hamilton EG, Mehrmohamadi M, et al. : Inferring gene expression from cell-free DNA fragmentation profiles. Nat Biotechnol. 2022; 40(4): 585–597. 10.1038/s41587-022-01222-4 [DOI] [PMC free article] [PubMed] [Google Scholar]; Faculty Opinions Recommendation
- 95. Tabula Sapiens. [cited 2022 Apr 29]. Reference Source [Google Scholar]
- 96. Vorperian SK, Moufarrej MN, , Tabula Sapiens Consortium, et al. : Cell types of origin of the cell-free transcriptome. Nat Biotechnol. 2022; 40(6): 855–861. 10.1038/s41587-021-01188-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Moufarrej MN, Vorperian SK, Wong RJ, et al. : Early prediction of preeclampsia in pregnancy with cell-free RNA. Nature. 2022; 602(7898): 689–694. 10.1038/s41586-022-04410-z [DOI] [PMC free article] [PubMed] [Google Scholar]