Origins, structures, and functions of circulating DNA in oncology

A R Thierry, S El Messaoudi, P B Gahan, P Anker, M Stroun, A R Thierry, S El Messaoudi, P B Gahan, P Anker, M Stroun

Abstract

While various clinical applications especially in oncology are now in progress such as diagnosis, prognosis, therapy monitoring, or patient follow-up, the determination of structural characteristics of cell-free circulating DNA (cirDNA) are still being researched. Nevertheless, some specific structures have been identified and cirDNA has been shown to be composed of many "kinds." This structural description goes hand-in-hand with the mechanisms of its origins such as apoptosis, necrosis, active release, phagocytosis, and exocytose. There are multiple structural forms of cirDNA depending upon the mechanism of release: particulate structures (exosomes, microparticles, apoptotic bodies) or macromolecular structures (nucleosomes, virtosomes/proteolipidonucleic acid complexes, DNA traps, links with serum proteins or to the cell-free membrane parts). In addition, cirDNA concerns both nuclear and/or mitochondrial DNA with both species exhibiting different structural characteristics that potentially reveal different forms of biological stability or diagnostic significance. This review focuses on the origins, structures and functional aspects that are paradoxically less well described in the literature while numerous reviews are directed to the clinical application of cirDNA. Differentiation of the various structures and better knowledge of the fate of cirDNA would considerably expand the diagnostic power of cirDNA analysis especially with regard to the patient follow-up enlarging the scope of personalized medicine. A better understanding of the subsequent fate of cirDNA would also help in deciphering its functional aspects such as their capacity for either genometastasis or their pro-inflammatory and immunological effects.

Keywords: Cancer; Cell-free circulating DNA; Functions; Origins; Structures.

Conflict of interest statement

The authors declare that they have are no conflicts of interest.

Figures

Fig. 1
Fig. 1
The first identification of extracellular nucleic acids in human blood compartment by Mandel and Metais in 1948 (adapted from Mandel and Métais [8])
Fig. 2
Fig. 2
Potential vesicular structures of circulating DNA (from Rykova et al. [48])
Fig. 3
Fig. 3
Distinct cellular origins of circulating DNA found in the blood of cancer patients
Fig. 4
Fig. 4
Circulating DNA levels correlate with tumor burden. a SW620 xenografted mouse model. Quantification by Q-PCR of cirDNA derived from malignant and non-malignant cells in the mouse model. Tumor weight is represented by the red curve (right axis). Concentration of cirDNA derived from mouse (normal) cells (mWT cirDNA) in control (not grafted) mice (mouse nos. 1–3) and in athymic nude mice (mouse nos. 4–11) xenografted with the SW620 colorectal human cells, determined using a primer set targeting a mouse KRAS second intron WT sequence. b SW620 xenografted mouse model. Concentration of cirDNA derived from human cells (hWT cirDNA) using a primer set targeting a human KRAS second intron WT sequence. c SW620 xenografted mouse model. Concentration of cirDNA derived from human cells (hKRASm cirDNA) using a primer set targeting a human KRAS second exon sequence that contains the G12V point mutation present in SW620. d Clinical mCRC plasma samples. Correlation between total cirDNA level and mutant cirDNA level in 4 KRAS mutant mCRC patients (adapted from Mouliere et al [68] and El Messaoudi  et al. [74]) (color figure online)
Fig. 5
Fig. 5
Strong interindividual heterogeneity of cirDNA mutation load values (mutant allele frequencies) (adapted from Mouliere et al. [68])
Fig. 6
Fig. 6
Correlation of circulating DNA parameters and overall survival. Overall survival analysis on a set of mCRC patients with KRAS or BRAF mutation. a Kaplan-Meier survival curve and log-rank test according to mA (mutant cirDNA concentration) determined by cirDNA analysis dichotomized around the median (3.06 ng/mL, n = 43). b Kaplan-Meier survival curve and log-rank test according to mA% (mutation load) dichotomized to the first tertile (4.14 %) determined by cirDNA analysis (n = 43). c Kaplan-Meier survival curve and log-rank test according to Ref A KRAS (total cirDNA concentration) dichotomized around the second tertile (107.0 ng/mL, n = 43). d Kaplan-Meier survival curve and log-rank test according to DNA integrity index (DII) determined by cirDNA analysis dichotomized around the second tertile (0.20, n = 43) (from El Messaoudi et al [74])
Fig. 7
Fig. 7
Neutrophil extracellular DNA traps. aKlebsiella pneumonia bacterium (pink) snared in a neutrophil extracellular trap (green) in a mouse lung. Credit: Papayannopoulos et al. (image by Volker Brinkman and Abdul Hakkim). b Immunostaining double-labeling of neutrophils releasing NETs isolated from tumor-bearing mice following 1 h activation with calcium ionophore (scale bar = 5 μm) (from Demers and Wagner 224)
Fig. 8
Fig. 8
Vizualization of circulating DNA by atomic force microscopy. Circulating DNA extracted from a metastatic colorectal cancer patient plasma sample vizualized by atomic force microscopy (AFM) and compared to SW620 genomic DNA extracted in the same conditions than circulating DNA (adapted from Mouliere et al. 69)
Fig. 9
Fig. 9
Malignant-derived circulating DNA is more fragmented than non-malignant derived circulating DNA. SW620 xenografted mouse model: a malignant (black bars) and non-malignant (hatched bars) cirDNA mean concentrations are expressed as ng/ml of plasma; the bar height is the sum of malignant and non-malignant cirDNA concentrations (estimated as the total ctDNA concentration). CirDNA was quantified by amplifying WT KRAS exon 2 sequences of increasing size: 60, 100, 150, 200, 250, 350, and 400 bp. b Fractional fragment size distribution of cirDNA amount from malignant and non-malignant cirDNA in xenografted mice and control cirDNA in non-xenografted mice. The cirDNA amount was arbitrarily estimated for the 60–100-, 100–150-, 150–400-, and >400-bp fragment size ranges. Clinical samples: c comparison of the cirDNA fragment size distribution of mutant (black bars) and WT (gray bars) cirDNA in four plasma samples from KRAS mutant mCRC patients. Mutant cirDNA was quantified by amplifying mutant KRAS exon 2 sequences of increasing size from 60 to 390 bp. Non-mutant cirDNA was quantified by amplifying WT KRAS exon 2 sequences of increasing size from 60 to 390 bp (adapted from Mouliere et al. [68, 72])
Fig. 10
Fig. 10
a Size distribution of malignant-derived circulating DNA. Ninety hepatocellular carcinoma patient plasma samples were examined by paired-end massively parallel sequencing. Different malignant-derived fractional concentrations were determined in HCC plasma samples (from <2 to >8 %). While size distribution peaks at 166 bp, it shifted progressively to the shorter size when malignant-derived cirDNA fraction increases (from Jiang et al. [107]). b Size distribution of circulating DNA in plasma sample from healthy individual as determined by next-generation sequencing from a single-stranded library preparation. Short fragments of 50–120 bp are considerably enriched as compared to conventional library preparation (from Snyder et al. [109])
Fig. 11
Fig. 11
CirDNA structure, origin, and function as regard to its analysis toward clinical application. Physicochemical analysis of cirDNA, i.e., analysis of its sequence, its quantity, and its structure brings qualitative and quantitative information useful for theragnostics, diagnostic, prognosis, and follow-up in the context of cancer management care. Analysis of the functions of cirDNA acting as an intercellular messenger, an immune activator, and a mediator in metastatic progression provides useful information for the follow-up of cancer patients by cirDNA analysis
Fig. 12
Fig. 12
Demonstration of transmission of inherited characters by hybridization in eggplants. Heterografts between S. melongena variety Long violet (mentor plant) and the variety White round (pupil plant) (from Stroun [16])
Fig. 13
Fig. 13
Representation of the main results showing the involvement of cirDNA released by T cells in the humoral immune response in vivo. Neutralizing activity of the serum of nude mice injected with DNA released by human T cells previously exposed to UV inactivated HSV or polio virus. Mice were injected with either 0.02 or 2 μg of human DNA. Control mice remained uninjected. After 5 days, the mice were killed and their serum was collected and frozen until tested. Numbers in ordinate represent highest dilutions of serum still presenting neutralizing activity (more than 3 units below virus control). Neutralization of HSV (hatched area); neutralization of polio (solid area). Human released T-DNA (α HSV) or T-DNA (α Polio) = DNA released by HSV or polio virus-exposed human T-lymph (from Anker et al. [184])
Fig. 14
Fig. 14
Schematic representation of the observing genometastasis. Plasma from colon cancer patients, in which a KRAS mutation had been detected, had been added to cultures of NIH/3T3 cells. Human KRAS DNA was detected soon afterward in these cells. Treated cells were injected, subsequently, into NOD-SCID mice and macroscopic tumors were generated. After sacrifice, human mutant KRAS sequences were detected in primary tumors, plasma, lungs, and livers of injected animals (from Garcia Olmo et al. [190])
Fig. 15
Fig. 15
Induction of double-strand DNA breaks and apoptosis by cirDNA into NIH/3T3 cells. Induction of γ H2AX and active caspase-3 by fragmented circulating DNA (DNAfs) and circulating chromatin (Cfs) derived from healthy volunteers (red bars) and cancer patients (gray bars). In vitro analysis of γ H2AX and active caspase-3: NIH/3T3 cells (10 × 104) were treated with DNAfs and Cfs (5 ng DNA each) for 6 h for detection of γ-H2AX (left) and for 24 h for detection of active caspase-3 (right) by immunofluorescence. For γ-H2AX (left-hand panel), 300 nuclei were counted and the percentage of nuclei showing positive foci were calculated and analyzed by chi-squared test. For active caspase-3 (right-hand panel), 200 cells were counted and the percentage of cells showing positive fluorescent signals were calculated and analyzed by chi-squared test (from Mittra et al. [195]) (color figure online)
Fig. 16
Fig. 16
Interaction between DNA and TLR9. Class III phosphatidylinositol 3-kinase (PI3K) facilitates the internalization of CpG oligodeoxynucleotides (ODNs) into endosomal vesicles that contain Toll-like receptor 9 (TLR9). The interaction between CpG DNA and TLR9 transduces an intracytoplasmic activation signal. The signal initiates with the recruitment of myeloid differentiation primary response gene 88 (MYD88) to the Toll–interleukin-1 receptor (TIR) domain of TLR9, followed by activation of the IRAK–TRAF6 complex. This leads to the activation of both the mitogen-activated protein kinase (MAPK: JNK1/2 and p38) and inhibitor of nuclear factor-kB (NF-kB) kinase (IKK) complexes, culminating in the upregulation of transcription factors, including NF-kB and activating protein 1 (AP1). ATF1, activating transcription factor 1; IRAK, IL-1 receptor-activated kinase; JNKK1, c-JUN N-terminal kinase (JNK) kinase 1; NIK, NF-kB-inducing kinase; TRAF6, tumor-necrosis factor receptor-associated factor 6 (from Klinman [215])
Fig. 17
Fig. 17
Involvement of mitochondrial cirDNA in the inflammatory process. Similar to the release of bacterial DNA following sepsis, the mitochondrial DNA released by severe trauma can also act through the Toll-like receptor 9 (TLR9) to activate neutrophils by activating p38 MAP kinase (MAPK). DAMPs damage-associated molecular patterns, PAMPs pathogen-associated molecular patterns. Adapted from © 2010 Nature Publishing Group Calfee, C. S. & Matthay, M. A. Clinical immunology: culprits with evolutionary ties. Nature 464, 41–42 (2010). All rights reserved
Fig. 18
Fig. 18
The metastasis suppression effect of RNase A or DNase I on Lewis lung carcinoma (LLC) metastatic mouse model. Typical histotopograms of lung lobes in the control and experimental groups treated with RNase A and DNase I. Hematoxylin and eosin staining. Arrows indicate large metastases. Bar corresponds to 5 mm (from Patutina et al. [228])
Fig. 19
Fig. 19
Real-time monitoring in the course of colorectal patients management care by cirDNA analysis. m mutant, WT wild type, MRD minimal residual disease, CDx companion diagnostics, CT chemotherapy, CRC colorectal cancer
Fig. 20
Fig. 20
Timeline of the main important discoveries about circulating DNA applications and functions

References

    1. Fleischhacker M, Schmidt B. Circulating nucleic acids (CNAs) and cancer--a survey. Biochimica et Biophysica Acta. 2007;1775(1):181–232.
    1. Gahan PB, Swaminathan R. Circulating nucleic acids in plasma and serum. Recent developments. Annals of the New York Academy of Sciences. 2008;1137:1–6.
    1. Miescher F. Ueber die chemische Zusammensetzung der Eiterzellen. Medicinisch-chemische Untersuchungen. 1871;4:441–460.
    1. Levene PA, Haller HL. On the configurational relationship of 3-Chlorobutyric and 3-Hydroxybutyric acids. Science (New York, N.Y.) 1929;69(1776):47.
    1. Beadle GW, Tatum EL. Genetic Control of Biochemical Reactions in Neurospora. Proceedings of the National Academy of Sciences of the United States of America. 1941;27(11):499–506.
    1. McClintock B. The Stability of Broken Ends of Chromosomes in Zea Mays. Genetics. 1941;26(2):234–282.
    1. Avery OT, Macleod CM, McCarty M. Studies on the chemical nature of the substance inducing transformation of pneumococcal types : induction of transformation by a Desoxyribonucleic acid fraction isolated from Pneumococcus TYPE III. The Journal of Experimental Medicine. 1944;79(2):137–158.
    1. Mandel P, Métais P. Les acides nucléiques du plasma sanguin chez l’Homme. Comptes Rendus des Seances de la Societe de Biologie et de ses Filiales. 1948;142:241–243.
    1. Chayen J, Norris KP. Cytoplasmic localization of nucleic acids in plant cells. Nature. 1953;171(4350):472–473.
    1. Swift H. The constancy of desoxyribose nucleic acid in plant nuclei. Proceedings of the National Academy of Sciences of the United States of America. 1950;36(11):643–654.
    1. Chargaff E. Chemical specificity of nucleic acids and mechanism of their enzymatic degradation. Experientia. 1950;6(6):201–209.
    1. Watson JD, Crick FH. The structure of DNA. Cold Spring Harbor Symposia on Quantitative Biology. 1953;18:123–131.
    1. Wilkins MHF, Stokes AR, Wilson HR. Molecular Structure of Nucleic Acids: Molecular Structure of Deoxypentose Nucleic Acids. Nature. 1953;171(4356):738–740.
    1. Sinsheimer RL. First steps toward a genetic chemistry. Science (New York, N.Y.) 1957;125(3258):1123–1128.
    1. Gartler SM. Cellular uptake of deoxyribonucleic acid by human tissue culture cells. Nature. 1959;184(Suppl 19):1505–1506.
    1. Stroun M. Modifications des caractères à la suite de greffes intervariétales chez le Solanum melongena. Comptes Rendues á l’Academie de Science Paris. 1962;255:361–363.
    1. Gahan PB, Chayen J, Silcox AA. Cytoplasmic localization of deoxyribonucleic acid in Allium cepa. Nature. 1962;195:1115–1116.
    1. Gahan PB, Chayen J. Cytoplasmic deoxyribonucleic acid. International Review of Cytology. 1965;18:223–247.
    1. Stroun M, Anker P. Nucleic acids spontaneously released by living frog auricles. The Biochemical Journal. 1972;128(3):100P–101P.
    1. Stroun M, Anker P, Maurice P, Gahan PB. Circulating nucleic acids in higher organisms. International Review of Cytology. 1977;51:1–48.
    1. Leon SA, Shapiro B, Sklaroff DM, Yaros MJ. Free DNA in the serum of cancer patients and the effect of therapy. Cancer Research. 1977;37(3):646–650.
    1. Stroun M, Anker P, Maurice P, Lyautey J, Lederrey C, Beljanski M. Neoplastic characteristics of the DNA found in the plasma of cancer patients. Oncology. 1989;46(5):318–322.
    1. Vasioukhin V, Anker P, Maurice P, Lyautey J, Lederrey C, Stroun M. Point mutations of the N-ras gene in the blood plasma DNA of patients with myelodysplastic syndrome or acute myelogenous leukaemia. British Journal of Haematology. 1994;86(4):774–779.
    1. Sorenson GD, Pribish DM, Valone FH, Memoli VA, Bzik DJ, Yao SL. Soluble normal and mutated DNA sequences from single-copy genes in human blood. Cancer Epidemiology, Biomarkers and Prevention. 1994;3(1):67–71.
    1. Lo YM, Corbetta N, Chamberlain PF, Rai V, Sargent IL, Redman CW, Wainscoat JS. Presence of fetal DNA in maternal plasma and serum. Lancet. 1997;350(9076):485–487.
    1. Diehl F, Li M, Dressman D, He Y, Shen D, Szabo S, Vogelstein B. Detection and quantification of mutations in the plasma of patients with colorectal tumors. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(45):16368–16373.
    1. Tan EM, Schur PH, Carr RI, Kunkel HG. Deoxybonucleic acid (DNA) and antibodies to DNA in the serum of patients with systemic lupus erythematosus. The Journal of Clinical Investigation. 1966;45(11):1732–1740.
    1. Yu SCY, Chan KCA, Zheng YWL, Jiang P, Liao GJW, Sun H, Lo YMD. Size-based molecular diagnostics using plasma DNA for noninvasive prenatal testing. Proceedings of the National Academy of Sciences of the United States of America. 2014;111(23):8583–8588.
    1. Mennuti MT, Chandrasekaran S, Khalek N, Dugoff L. Cell-free DNA screening and sex chromosome aneuploidies. Prenatal Diagnosis. 2015;35(10):980–985.
    1. Rieneck K, Clausen FB, Dziegiel MH. Noninvasive Antenatal Determination of Fetal Blood Group Using Next-Generation Sequencing. Cold Spring Harbor Perspectives in Medicine. 2015;6(1):a023093.
    1. Assou S, Aït-Ahmed O, El Messaoudi S, Thierry AR, Hamamah S. Non-invasive pre-implantation genetic diagnosis of X-linked disorders. Medical Hypotheses. 2014;83(4):506–508.
    1. Swarup V, Rajeswari MR. Circulating (cell-free) nucleic acids--a promising, non-invasive tool for early detection of several human diseases. FEBS Letters. 2007;581(5):795–799.
    1. Crowley E, Di Nicolantonio F, Loupakis F, Bardelli A. Liquid biopsy: monitoring cancer-genetics in the blood. Nature Reviews. Clinical Oncology. 2013;10(8):472–484.
    1. Heitzer E, Ulz P, Geigl JB. Circulating tumor DNA as a liquid biopsy for cancer. Clinical Chemistry. 2015;61(1):112–123.
    1. Thierry AR. Circulating DNA and cancer. AACR Education Book. 2015;2015(1):45.
    1. Harbeck RJ, Bardana EJ, Kohler PF, Carr RI. DNA:anti-DNA complexes: their detection in systemic lupus erythematosus sera. The Journal of Clinical Investigation. 1973;52(4):789–795.
    1. Compton LJ, Steinberg AD, Sano H. Nuclear DNA degradation in lymphocytes of patients with systemic lupus erythematosus. Journal of Immunology (Baltimore, Md.: 1950) 1984;133(1):213–216.
    1. Koffler D, Agnello V, Winchester R, Kunkel HG. The Occurrence of Single-Stranded DNA in the Serum of Patients with Systemic Lupus Erythematosus and Other Diseases. Journal of Clinical Investigation. 1973;52(1):198–204.
    1. Li JZ, Steinman CR. Plasma DNA in systemic lupus erythematosus. Characterization of cloned base sequences. Arthritis and Rheumatism. 1989;32(6):726–733.
    1. Raptis L, Menard HA. Quantitation and characterization of plasma DNA in normals and patients with systemic lupus erythematosus. The Journal of Clinical Investigation. 1980;66(6):1391–1399.
    1. Rumore PM, Steinman CR. Endogenous circulating DNA in systemic lupus erythematosus. Occurrence as multimeric complexes bound to histone. The Journal of Clinical Investigation. 1990;86(1):69–74.
    1. Steinman CR. Free DNA in serum and plasma from normal adults. Journal of Clinical Investigation. 1975;56(2):512–515.
    1. Tan EM. An immunologic precipitin system between soluble nucleoprotein and serum antibody in systemic lupus erythematosus. The Journal of Clinical Investigation. 1967;46(5):735–745.
    1. Volkova ZI, Solov’ev GI, Folomeeva OM. Isolation and characteristics of the DNA of donor and systemic lupus erythematosus patient blood plasma. Biulleten' Eksperimental'noĭ Biologii i Meditsiny. 1980;89(6):689–691.
    1. Emlen W, Mannik M. Effect of DNA size and strandedness on the in vivo clearance and organ localization of DNA. Clinical and Experimental Immunology. 1984;56(1):185–192.
    1. Stroun M, Anker P, Lyautey J, Lederrey C, Maurice PA. Isolation and characterization of DNA from the plasma of cancer patients. European Journal of Cancer & Clinical Oncology. 1987;23(6):707–712.
    1. Mouliere F, Thierry AR. The importance of examining the proportion of circulating DNA originating from tumor, microenvironment and normal cells in colorectal cancer patients. Expert Opinion on Biological Therapy. 2012;12(S1):S209–S215.
    1. Rykova EY, Morozkin ES, Ponomaryova AA, Loseva EM, Zaporozhchenko IA, Cherdyntseva NV, Laktionov PP. Cell-free and cell-bound circulating nucleic acid complexes: mechanisms of generation, concentration and content. Expert Opinion on Biological Therapy. 2012;12(Suppl 1):S141–153.
    1. Laktionov PP, Tamkovich SN, Rykova EY, Bryzgunova OE, Starikov AV, Kuznetsova NP, Vlassov VV. Cell-surface-bound nucleic acids: Free and cell-surface-bound nucleic acids in blood of healthy donors and breast cancer patients. Annals of the New York Academy of Sciences. 2004;1022:221–227.
    1. Mittra I, Nair NK, Mishra PK. Nucleic acids in circulation: are they harmful to the host? Journal of Biosciences. 2012;37(2):301–312.
    1. Gahan PB, Stroun M. The virtosome-a novel cytosolic informative entity and intercellular messenger. Cell Biochemistry and Function. 2010;28(7):529–538.
    1. Rogers JC, Boldt D, Kornfeld S, Skinner A, Valeri CR. Excretion of deoxyribonucleic acid by lymphocytes stimulated with phytohemagglutinin or antigen. Proceedings of the National Academy of Sciences of the United States of America. 1972;69(7):1685–1689.
    1. Anker P, Stroun M. Bacterial ribonucleic acid synthesis in frog organs after intraperitoneal injection of bacteria. Biochemical Journal. 1972;128(3):101P.
    1. Anker P, Stroun M. Bacterial ribonucleic acid in the frog brain after a bacterial peritoneal infection. Science (New York, N.Y.) 1972;178(4061):621–623.
    1. Anker P, Mulcahy H, Chen XQ, Stroun M. Detection of circulating tumour DNA in the blood (plasma/serum) of cancer patients. Cancer Metastasis Reviews. 1999;18(1):65–73.
    1. Stroun M, Anker P. Bacterial nucleic acid synthesis in plants following bacterial contact. Molecular and General Genetics MGG. 1971;113(1):92–98.
    1. Stroun M, Anker P. Transcription of Spontaneously Released Bacterial Deoxyribonucleic Acid in Frog Auricles. Journal of Bacteriology. 1973;114(1):114–120.
    1. Stroun M, Charles P, Anker P, Pelc SR. Metabolic DNA in heart and skeletal muscle and in the intestine of mice. Nature. 1967;216(5116):716–717.
    1. Stroun M, Anker P, Gahan P, Rossier A, Greppin H. Agrobacterium tumefaciens ribonucleic acid synthesis in tomato cells and crown gall induction. Journal of Bacteriology. 1971;106(2):634–639.
    1. Anker P, Stroun M, Maurice PA. Spontaneous release of DNA by human blood lymphocytes as shown in an in vitro system. Cancer Research. 1975;35(9):2375–2382.
    1. Stroun M, Lyautey J, Lederrey C, Olson-Sand A, Anker P. About the possible origin and mechanism of circulating DNA: Apoptosis and active DNA release. Clinica Chimica Acta. 2001;313(1–2):139–142.
    1. Gahan PB, Anker P, Stroun M. Metabolic DNA as the origin of spontaneously released DNA? Annals of the New York Academy of Sciences. 2008;1137:7–17.
    1. Abolhassani M, Tillotson J, Chiao J. Characterization of the release of DNA by a human leukemia-cell line HL-60. International Journal of Oncology. 1994;4(2):417–421.
    1. Meyer JS. Growth and cell kinetic measurements in human tumors. Pathology Annual. 1981;16(Pt 2):53–81.
    1. Ota DM, Drewinko B. Growth kinetics of human colorectal carcinoma. Cancer Research. 1985;45(5):2128–2131.
    1. Lo CF. Stochastic Gompertz model of tumour cell growth. Journal of Theoretical Biology. 2007;248(2):317–321.
    1. Bettegowda C, Sausen M, Leary RJ, Kinde I, Wang Y, Agrawal N, Diaz LA. Detection of Circulating Tumor DNA in Early- and Late-Stage Human Malignancies. Science Translational Medicine. 2014;6(224):224ra24–224ra24.
    1. Mouliere F, El Messaoudi S, Gongora C, Guedj A-S, Robert B, Del Rio M, Thierry AR. Circulating Cell-Free DNA from Colorectal Cancer Patients May Reveal High KRAS or BRAF Mutation Load. Translational Oncology. 2013;6(3):319–328.
    1. Mouliere F, El Messaoudi S, Pang D, Dritschilo A, Thierry AR. Multi-marker analysis of circulating cell-free DNA toward personalized medicine for colorectal cancer. Molecular Oncology. 2014;8(5):927–941.
    1. Thierry AR, Mouliere F, El Messaoudi S, Mollevi C, Lopez-Crapez E, Rolet F, Ychou M. Clinical validation of the detection of KRAS and BRAF mutations from circulating tumor DNA. Nature Medicine. 2014;20(4):430–435.
    1. Esposito A, Bardelli A, Criscitiello C, Colombo N, Gelao L, Fumagalli L, Curigliano G. Monitoring tumor-derived cell-free DNA in patients with solid tumors: Clinical perspectives and research opportunities. Cancer Treatment Reviews. 2014;40(5):648–655.
    1. Mouliere F, Robert B, Arnau Peyrotte E, Del Rio M, Ychou M, Molina F, Thierry AR. High fragmentation characterizes tumour-derived circulating DNA. PloS One. 2011;6(9):e23418.
    1. Thierry AR, Mouliere F, Gongora C, Ollier J, Robert B, Ychou M, Molina F. Origin and quantification of circulating DNA in mice with human colorectal cancer xenografts. Nucleic Acids Research. 2010;38(18):6159–6175.
    1. El Messaoudi S, Mouliere F, Du Manoir S, Bascoul-Mollevi C, Gillet B, Nouaille M, Thierry AR. Circulating DNA as a strong multi-marker prognostic tool for metastatic colorectal cancer patient management care. Clinical Cancer Research. 2016
    1. Murtaza M, Dawson S-J, Tsui DWY, Gale D, Forshew T, Piskorz AM, Rosenfeld N. Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature. 2013;497(7447):108–112.
    1. West NP, Dattani M, McShane P, Hutchins G, Grabsch J, Mueller W, Grabsch H. The proportion of tumour cells is an independent predictor for survival in colorectal cancer patients. British Journal of Cancer. 2010;102(10):1519–1523.
    1. Spindler K-LG, Pallisgaard N, Vogelius I, Jakobsen A. Quantitative cell-free DNA, KRAS, and BRAF mutations in plasma from patients with metastatic colorectal cancer during treatment with cetuximab and irinotecan. Clinical Cancer Research. 2012;18(4):1177–1185.
    1. Boone BA, Orlichenko L, Schapiro NE, Loughran P, Gianfrate GC, Ellis JT, Zeh HJ. The receptor for advanced glycation end products (RAGE) enhances autophagy and neutrophil extracellular traps in pancreatic cancer. Cancer Gene Therapy. 2015;22(6):326–334.
    1. Cortese R, Almendros I, Wang Y, Gozal D. Tumor circulating DNA profiling in xenografted mice exposed to intermittent hypoxia. Oncotarget. 2014;6(1):556–569.
    1. Sato K, Tsuchihara K, Fujii S, Sugiyama M, Goya T, Atomi Y, Esumi H. Autophagy is activated in colorectal cancer cells and contributes to the tolerance to nutrient deprivation. Cancer Research. 2007;67(20):9677–9684.
    1. Ho AS, Huang X, Cao H, Christman-Skieller C, Bennewith K, Le Q-T, Koong AC. Circulating miR-210 as a Novel Hypoxia Marker in Pancreatic Cancer. Translational Oncology. 2010;3(2):109–113.
    1. Yu M. Circulating cell-free mitochondrial DNA as a novel cancer biomarker: opportunities and challenges. Mitochondrial DNA. 2012;23(5):329–332.
    1. Ellinger J, Müller DC, Müller SC, Hauser S, Heukamp LC, von Ruecker A, Walgenbach-Brunagel G. Circulating mitochondrial DNA in serum: a universal diagnostic biomarker for patients with urological malignancies. Urologic Oncology. 2012;30(4):509–515.
    1. Kohler C, Radpour R, Barekati Z, Asadollahi R, Bitzer J, Wight E, Zhong XY. Levels of plasma circulating cell free nuclear and mitochondrial DNA as potential biomarkers for breast tumors. Molecular Cancer. 2009;8:105.
    1. Budnik LT, Kloth S, Baur X, Preisser AM, Schwarzenbach H. Circulating mitochondrial DNA as biomarker linking environmental chemical exposure to early preclinical lesions elevation of mtDNA in human serum after exposure to carcinogenic halo-alkane-based pesticides. PloS One. 2013;8(5):e64413.
    1. Ding W-X, Yin X-M. Mitophagy: mechanisms, pathophysiological roles, and analysis. Biological Chemistry. 2012;393(7):547–564.
    1. Chiu RWK, Chan LYS, Lam NYL, Tsui NBY, Ng EKO, Rainer TH, Lo YMD. Quantitative analysis of circulating mitochondrial DNA in plasma. Clinical Chemistry. 2003;49(5):719–726.
    1. Malik AN, Parsade CK, Ajaz S, Crosby-Nwaobi R, Gnudi L, Czajka A, Sivaprasad S. Altered circulating mitochondrial DNA and increased inflammation in patients with diabetic retinopathy. Diabetes Research and Clinical Practice. 2015;110(3):257–265.
    1. Liu J, Cai X, Xie L, Tang Y, Cheng J, Wang J, Gong J. Circulating Cell Free Mitochondrial DNA is a Biomarker in the Development of Coronary Heart Disease in the Patients with Type 2 Diabetes. Clinical Laboratory. 2015;61(7):661–667.
    1. Sudakov NP, Popkova TP, Katyshev AI, Goldberg OA, Nikiforov SB, Pushkarev BG, Konstantinov YM. Level of Blood Cell-Free Circulating Mitochondrial DNA as a Novel Biomarker of Acute Myocardial Ischemia. Biochemistry. Biokhimiia. 2015;80(10):1387–1392.
    1. Surmiak MP, Hubalewska-Mazgaj M, Wawrzycka-Adamczyk K, Szczeklik W, Musiał J, Sanak M. Circulating mitochondrial DNA in serum of patients with granulomatosis with polyangiitis. Clinical and Experimental Immunology. 2015;181(1):150–155.
    1. Zhang Q, Itagaki K, Hauser CJ. Mitochondrial DNA is released by shock and activates neutrophils via p38 map kinase. Shock (Augusta, Ga.) 2010;34(1):55–59.
    1. Nasi M, Cristani A, Pinti M, Lamberti I, Gibellini L, De Biasi S, Cossarizza A. Decreased Circulating mtDNA Levels in Professional Male Volleyball Players. International Journal of Sports Physiology and Performance. 2016;11(1):116–121.
    1. Shockett PE, Khanal J, Sitaula A, Oglesby C, Meachum WA, Castracane VD, Kraemer RR. Plasma cell free mitochondrial DNA declines in response to prolonged moderate aerobic exercise. Physiological Reports. 2016;4(1):e12672.
    1. Kandel ES. Mutations in circulating mitochondrial DNA: Cassandra of oral cancer? Oncotarget. 2012;3(7):664–665.
    1. Uzawa K, Baba T, Uchida F, Yamatoji M, Kasamatsu A, Sakamoto Y, Tanzawa H. Circulating tumor-derived mutant mitochondrial DNA: a predictive biomarker of clinical prognosis in human squamous cell carcinoma. Oncotarget. 2012;3(7):670–677.
    1. Huang C-Y, Chen Y-M, Wu C-H, Tsai C-M, Lee Y-C, Perng R-P, Whang-Peng J. Circulating free mitochondrial DNA concentration and its association with erlotinib treatment in patients with adenocarcinoma of the lung. Oncology Letters. 2014;7(6):2180–2184.
    1. Zhang Q, Raoof M, Chen Y, Sumi Y, Sursal T, Junger W, Hauser CJ. Circulating mitochondrial DAMPs cause inflammatory responses to injury. Nature. 2010;464(7285):104–107.
    1. Cao H, Ye H, Sun Z, Shen X, Song Z, Wu X, Yang J. Circulatory Mitochondrial DNA Is a Pro-Inflammatory Agent in Maintenance Hemodialysis Patients. PLoS ONE. 2014;9(12):e113179.
    1. Itagaki K, Kaczmarek E, Lee YT, Tang IT, Isal B, Adibnia Y, Hauser CJ. Mitochondrial DNA released by trauma induces neutrophil extracellular traps. PloS One. 2015;10(3):e0120549.
    1. Cools-Lartigue J, Spicer J, McDonald B, Gowing S, Chow S, Giannias B, Ferri L. Neutrophil extracellular traps sequester circulating tumor cells and promote metastasis. The Journal of Clinical Investigation. 2013
    1. Tohme S, Yazdani HO, Al-Khafaji AB, Chidi AP, Loughran P, Mowen K, Tsung A. Neutrophil Extracellular Traps Promote the Development and Progression of Liver Metastases after Surgical Stress. Cancer Research. 2016
    1. Yu M. Generation, function and diagnostic value of mitochondrial DNA copy number alterations in human cancers. Life Sciences. 2011;89(3–4):65–71.
    1. Yu M. Somatic mitochondrial DNA mutations in human cancers. Advances in Clinical Chemistry. 2012;57:99–138.
    1. Chatterjee A, Mambo E, Sidransky D. Mitochondrial DNA mutations in human cancer. Oncogene. 2006;25(34):4663–4674.
    1. Lu H, Busch J, Jung M, Rabenhorst S, Ralla B, Kilic E, Jung K. Diagnostic and prognostic potential of circulating cell-free genomic and mitochondrial DNA fragments in clear cell renal cell carcinoma patients. Clinica Chimica Acta. 2016;452:109–119.
    1. Jiang P, Chan CWM, Chan KCA, Cheng SH, Wong J, Wong VW-S, Lo YMD. Lengthening and shortening of plasma DNA in hepatocellular carcinoma patients. Proceedings of the National Academy of Sciences of the United States of America. 2015;112(11):E1317–1325.
    1. Jahr S, Hentze H, Englisch S, Hardt D, Fackelmayer FO, Hesch RD, Knippers R. DNA fragments in the blood plasma of cancer patients: quantitations and evidence for their origin from apoptotic and necrotic cells. Cancer Research. 2001;61(4):1659–1665.
    1. Snyder MW, Kircher M, Hill AJ, Daza RM, Shendure J. Cell-free DNA Comprises an In Vivo Nucleosome Footprint that Informs Its Tissues-Of-Origin. Cell. 2016;164(1–2):57–68.
    1. Lehmann-Werman R, Neiman D, Zemmour H, Moss J, Magenheim J, Vaknin-Dembinsky A, Dor Y. Identification of tissue-specific cell death using methylation patterns of circulating DNA. Proceedings of the National Academy of Sciences of the United States of America. 2016
    1. Nagata S, Nagase H, Kawane K, Mukae N, Fukuyama H. Degradation of chromosomal DNA during apoptosis. Cell Death and Differentiation. 2003;10(1):108–116.
    1. Holdenrieder S, Stieber P, Bodenmüller H, Busch M, Von Pawel J, Schalhorn A, Seidel D. Circulating Nucleosomes in Serum. Annals of the New York Academy of Sciences. 2001;945(1):93–102.
    1. Holdenrieder S, Stieber P. Clinical use of circulating nucleosomes. Critical Reviews in Clinical Laboratory Sciences. 2009;46(1):1–24.
    1. Weerasinghe P, Buja LM. Oncosis: an important non-apoptotic mode of cell death. Experimental and Molecular Pathology. 2012;93(3):302–308.
    1. Gezer U, Holdenrieder S. Post-translational histone modifications in circulating nucleosomes as new biomarkers in colorectal cancer. In Vivo (Athens, Greece) 2014;28(3):287–292.
    1. D’Souza-Schorey C, Clancy JW. Tumor-derived microvesicles: shedding light on novel microenvironment modulators and prospective cancer biomarkers. Genes & Development. 2012;26(12):1287–1299.
    1. Lee Y, El Andaloussi S, Wood MJA. Exosomes and microvesicles: extracellular vesicles for genetic information transfer and gene therapy. Human Molecular Genetics. 2012;21(R1):R125–134.
    1. Turturici G, Tinnirello R, Sconzo G, Geraci F. Extracellular membrane vesicles as a mechanism of cell-to-cell communication: advantages and disadvantages. American Journal of Physiology - Cellular Physiology. 2014;306(7):C621–633.
    1. De Jong OG, Van Balkom BWM, Schiffelers RM, Bouten CVC, Verhaar MC. Extracellular vesicles: potential roles in regenerative medicine. Frontiers in Immunology. 2014;5:608.
    1. Tetta C, Ghigo E, Silengo L, Deregibus MC, Camussi G. Extracellular vesicles as an emerging mechanism of cell-to-cell communication. Endocrine. 2013;44(1):11–19.
    1. Chevillet JR, Kang Q, Ruf IK, Briggs HA, Vojtech LN, Hughes SM, Tewari M. Quantitative and stoichiometric analysis of the microRNA content of exosomes. Proceedings of the National Academy of Sciences of the United States of America. 2014;111(41):14888–14893.
    1. Thakur BK, Zhang H, Becker A, Matei I, Huang Y, Costa-Silva B, Lyden D. Double-stranded DNA in exosomes: a novel biomarker in cancer detection. Cell Research. 2014;24(6):766–769.
    1. Chistiakov DA, Chekhonin VP. Extracellular vesicles shed by glioma cells: pathogenic role and clinical value. Tumour Biology. 2014;35(9):8425–8438.
    1. Lowe SW, Lin AW. Apoptosis in cancer. Carcinogenesis. 2000;21(3):485–495.
    1. Vlassov VV, Laktionov PP, Rykova EY. Extracellular nucleic acids. BioEssays. 2007;29(7):654–667.
    1. Rogers JC. Characterization of DNA excreted from phytohemagglutinin-stimulated lymphocytes. The Journal of Experimental Medicine. 1976;143(5):1249–1264.
    1. Boldt DH, MacDermott RP, Speckart SF, Nash GS. Excretion of DNA by purified human lymphocyte subpopulations. Journal of Immunology (Baltimore, Md.: 1950) 1977;118(4):1495–1498.
    1. Hoessli DC, Jones AP, Eisenstadt JM, Waksman BH. Studies on DNA release by cultured rat lymphoblasts. International Archives of Allergy and Applied Immunology. 1977;54(6):517–528.
    1. Olsen I, Harris G. Uptake and release of DNA by lymphoid tissue and cells. Immunology. 1974;27(6):973–987.
    1. Stroun, M., Anker, P., Gahan, P. B., & Henri, J. (1977). Spontaneous release of newly synthesized DNA from frog auricles. Archives des Sciences.
    1. Stroun M, Anker P, Beljanski M, Henri J, Lederrey C, Ojha M, Maurice PA. Presence of RNA in the nucleoprotein complex spontaneously released by human lymphocytes and frog auricles in culture. Cancer Research. 1978;38(10):3546–3554.
    1. Adams DH, Gahan PB. The DNA extruded by rat spleen cells in culture. The International Journal of Biochemistry. 1983;15(4):547–552.
    1. Adams DH, McIntosh AA. Studies on the cytosolic DNA of chick embryo fibroblasts and its uptake by recipient cultured cells. The International Journal of Biochemistry. 1985;17(10):1041–1051.
    1. McIntosh AA, Adams DH. Further studies on the extrusion of cytosol macromolecules by cultured chick embryo fibroblast cells. The International Journal of Biochemistry. 1985;17(2):147–153.
    1. Challen C, Adams DH. The assembly of the DNA complex present in chick embryo cell cytosol. The International Journal of Biochemistry. 1987;19(3):235–243.
    1. Ochatt, S., Gahan, P. (2005). Viable protoplasts release newly synthesised DNA. In: COST 843 action: “Quality enhancement of plant production throught tissue culture”, COST 851 Action “Gametic cells and molecular breeding for crops improvement” (p. 218–219).
    1. Adams DH, Diaz N, Gahan PB. In vitro stimulation by tumour cell media of [3H]-thymidine incorporation by mouse spleen lymphocytes. Cell Biochemistry and Function. 1997;15(2):119–126.
    1. Kim J, Bae J-S. Tumor-Associated Macrophages and Neutrophils in Tumor Microenvironment. Mediators of Inflammation. 2016;2016:6058147.
    1. Berger-Achituv S, Brinkmann V, Abed UA, Kühn LI, Ben-Ezra J, Elhasid R, Zychlinsky A. A proposed role for neutrophil extracellular traps in cancer immunoediting. Frontiers in Immunology. 2013;4:48.
    1. Mishalian I, Bayuh R, Levy L, Zolotarov L, Michaeli J, Fridlender ZG. Tumor-associated neutrophils (TAN) develop pro-tumorigenic properties during tumor progression. Cancer Immunology, Immunotherapy. 2013;62(11):1745–1756.
    1. Fridlender ZG, Albelda SM. Tumor Associated Neutrophils: Friend or Foe? Carcinogenesis. 2012
    1. Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS, Zychlinsky A. Neutrophil extracellular traps kill bacteria. Science (New York, N.Y.) 2004;303(5663):1532–1535.
    1. Branzk N, Papayannopoulos V. Molecular mechanisms regulating NETosis in infection and disease. Seminars in Immunopathology. 2013;35(4):513–530.
    1. Papayannopoulos V, Metzler KD, Hakkim A, Zychlinsky A. Neutrophil elastase and myeloperoxidase regulate the formation of neutrophil extracellular traps. The Journal of Cell Biology. 2010;191(3):677–691.
    1. Yipp BG, Kubes P. NETosis: how vital is it? Blood. 2013;122(16):2784–2794.
    1. Cooper PR, Palmer LJ, Chapple ILC. Neutrophil extracellular traps as a new paradigm in innate immunity: friend or foe? Periodontology 2000. 2013;63(1):165–197.
    1. Kaplan MJ, Radic M. Neutrophil extracellular traps (NETs): Double-edged swords of innate immunity. Journal of Immunology (Baltimore, Md. : 1950) 2012;189(6):2689–2695.
    1. Brinkmann V, Zychlinsky A. Neutrophil extracellular traps: Is immunity the second function of chromatin? The Journal of Cell Biology. 2012;198(5):773–783.
    1. Luo L, Zhang S, Wang Y, Rahman M, Syk I, Zhang E, Thorlacius H. Proinflammatory role of neutrophil extracellular traps in abdominal sepsis. American Journal of Physiology - Lung Cellular and Molecular Physiology. 2014;307(7):L586–596.
    1. Fuchs TA, Brill A, Duerschmied D, Schatzberg D, Monestier M, Myers DD, Wagner DD. Extracellular DNA traps promote thrombosis. Proceedings of the National Academy of Sciences. 2010;107(36):15880–15885.
    1. Beiter T, Fragasso A, Hudemann J, Schild M, Steinacker J, Mooren FC, Niess AM. Neutrophils release extracellular DNA traps in response to exercise. Journal of Applied Physiology (Bethesda, Md.: 1985) 2014;117(3):325–333.
    1. Davis BP, Rothenberg ME. Eosinophils and cancer. Cancer Immunology Research. 2014;2(1):1–8.
    1. Yousefi S, Simon D, Simon H-U. Eosinophil extracellular DNA traps: molecular mechanisms and potential roles in disease. Current Opinion in Immunology. 2012;24(6):736–739.
    1. Yousefi S, Gold JA, Andina N, Lee JJ, Kelly AM, Kozlowski E, Simon H-U. Catapult-like release of mitochondrial DNA by eosinophils contributes to antibacterial defense. Nature Medicine. 2008;14(9):949–953.
    1. Rakoff-Nahoum S. Why cancer and inflammation? The Yale Journal of Biology and Medicine. 2006;79(3–4):123–130.
    1. Chelobanov BP, Laktionov PP, Vlasov VV. Proteins involved in binding and cellular uptake of nucleic acids. Biochemistry. Biokhimiia. 2006;71(6):583–596.
    1. Rykova EY, Pautova LV, Yakubov LA, Karamyshev VN, Vlassov VV. Serum immunoglobulins interact with oligonucleotides. FEBS Letters. 1994;344(1):96–98.
    1. Bryzgunova OE, Tamkovich SN, Cherepanova AV, Yarmoshchuk SV, Permyakova VI, Anykeeva OY, Laktionov PP. Redistribution of Free- and Cell-Surface-Bound DNA in Blood of Benign and Malignant Prostate Tumor Patients. Acta Naturae. 2015;7(2):115–118.
    1. Morozkin ES, Laktionov PP, Rykova EY, Vlassov VV. Extracellular nucleic acids in cultures of long-term cultivated eukaryotic cells. Annals of the New York Academy of Sciences. 2004;1022:244–249.
    1. Schwarzenbach H, Hoon DSB, Pantel K. Cell-free nucleic acids as biomarkers in cancer patients. Nature reviews. Cancer. 2011;11(6):426–437.
    1. van der Vaart M, Pretorius PJ. Is the role of circulating DNA as a biomarker of cancer being prematurely overrated? Clinical Biochemistry. 2010;43(1–2):26–36.
    1. El Messaoudi S, Rolet F, Mouliere F, Thierry AR. Circulating cell free DNA: Preanalytical considerations. Clinica Chimica Acta. 2013
    1. El Messaoudi S, Thierry AR. Pre-analytical Requirements for Analyzing Nucleic Acids from Blood. In: Gahan PB, editor. Circulating Nucleic Acids in Early Diagnosis, Prognosis and Treatment Monitoring. Dordrecht: Springer Netherlands; 2015. pp. 45–69.
    1. Gormally E, Caboux E, Vineis P, Hainaut P. Circulating free DNA in plasma or serum as biomarker of carcinogenesis: practical aspects and biological significance. Mutation Research. 2007;635(2–3):105–117.
    1. Wang BG, Huang H-Y, Chen Y-C, Bristow RE, Kassauei K, Cheng C-C, Shih I-M. Increased plasma DNA integrity in cancer patients. Cancer Research. 2003;63(14):3966–3968.
    1. Pang D, Thierry AR, Dritschilo A. DNA studies using atomic force microscopy: capabilities for measurement of short DNA fragments. Frontiers in Molecular Biosciences. 2015;2:1.
    1. Lo YM, Zhang J, Leung TN, Lau TK, Chang AM, Hjelm NM. Rapid clearance of fetal DNA from maternal plasma. American Journal of Human Genetics. 1999;64(1):218–224.
    1. Yu SCY, Lee SWY, Jiang P, Leung TY, Chan KCA, Chiu RWK, Lo YMD. High-resolution profiling of fetal DNA clearance from maternal plasma by massively parallel sequencing. Clinical Chemistry. 2013;59(8):1228–1237.
    1. Diehl F, Schmidt K, Choti MA, Romans K, Goodman S, Li M, Diaz LA. Circulating mutant DNA to assess tumor dynamics. Nature Medicine. 2008;14(9):985–990.
    1. Stroun M, Anker P. Prehistory of the notion of circulating nucleic acids in plasma/serum (CNAPS): birth of a hypothesis. Annals of the New York Academy of Sciences. 2006;1075:10–20.
    1. Stroun, M. (2011). Reflections on a Life of CNAPS: From Circulating DNA to the Virtosome. In P. B. Gahan (Ed.), Circulating Nucleic Acids in Plasma and Serum (pp. 15–20). Springer Netherlands. Retrieved from
    1. Anker P, Stroun M. Circulating nucleic acids and evolution. Expert Opinion on Biological Therapy. 2012;12(Suppl 1):S113–117.
    1. Darwin C. Animals and Plants under domestication. Vol II. 2. New York: Appleton & Company; 1876.
    1. Liu Y. Historical and modern genetics of plant graft hybridization. Advances in Genetics. 2006;56:101–129.
    1. Liu Y. A new perspective on Darwin’s Pangenesis. Biological Reviews of the Cambridge Philosophical Society. 2008;83(2):141–149.
    1. Stroun J, Stroun-Guttieres L, Rossi J, Stroun M. Alteration of the color of the feathers in white Leghorn chickens by repeated injections of the blood of guinea hens. Observations on 5 generations. Comptes Rendus Hebdomadaires des Séances de l'Académie des Sciences. 1962;255:781–783.
    1. Stroun, M., Mathon, C. C., & Stroun, J. (1963). Alteration of hereditary traits in Solanum melongena induced by grafts with Solanum nigrum. In: Proceedings of the 11th International Congress of Genetics (la Haye) (Vol. 1, p. 218).
    1. Gahan PB. Messenger DNA in higher plants. Cell Biochemistry and Function. 2003;21(3):207–209.
    1. Stroun M, Anker P. In vitro synthesis of DNA spontaneously released by bacteria or frog auricles. Biochimie. 1972;54(11):1443–1452.
    1. Thierry AR, Dritschilo A. Intracellular availability of unmodified, phosphorothioated and liposomally encapsulated oligodeoxynucleotides for antisense activity. Nucleic Acids Research. 1992;20(21):5691–5698.
    1. Anker P, Lyautey J, Lefort F, Lederrey C, Stroun M. Transformation de cellules NIH/3 T3 et cellules SW 480 porteuses d’une mutation K-ras. Comptes Rendus de l'Academie des Sciences. Serie III, Sciences de la Vie. 1994;317(10):869–874.
    1. Anker P, Jachertz D, Stroun M, Brögger R, Lederrey C, Maurice PA. [Transfer of genetic information from T to B human lymphocytes during an immune response to herpes simplex virus] Comptes Rendus Des Séances De l’Académie Des Sciences. Série D, Sciences Naturelles. 1979;289(2):217–220.
    1. Anker P, Jachertz D, Stroun M, Brögger R, Lederrey C, Henri J, Maurice PA. The role of extracellular DNA in the transfer of information from T to B human lymphocytes in the course of an immune response. Journal of Immunogenetics. 1980;7(6):475–481.
    1. Anker P, Jachertz D, Maurice PA, Stroun M. Nude mice injected with DNA released by antigen stimulated human T lymphocytes produce specific antibodies expressing human characteristics. Cell Biochemistry and Function. 1984;2(1):33–37.
    1. Anker P, Stroun M, Maurice PA. Spontaneous extracellular synthesis of DNA released by human blood lymphocytes. Cancer Research. 1976;36(8):2832–2839.
    1. Anker P, Stroun M, Maurice P. Characteristics of nucleic acids excreted by non-stimulated normal human lymphocytes. Schweizerische Medizinische Wochenschrift. 1977;107(41):1457.
    1. Anker P, Stroun M. Immunological aspects of circulating DNA. Annals of the New York Academy of Sciences. 2006;1075:34–39.
    1. Jachertz, D., Stroun, M., Anker, P., Brogger, R., Lederrey, C., & Maurice, P. A. (1979). Transfer of genetic information from T to B lymphocytes in the course of an immune response. Biomedicine/[publiée Pour l’A.A.I.C.I.G.], 31(6), 153–154.
    1. Bendich A, Wilczok T, Borenfreund E. Circulating DNA as a possible factor in oncogenesis. Science (New York, N.Y.) 1965;148(3668):374–376.
    1. García-Olmo DC, Domínguez C, García-Arranz M, Anker P, Stroun M, García-Verdugo JM, García-Olmo D. Cell-free nucleic acids circulating in the plasma of colorectal cancer patients induce the oncogenic transformation of susceptible cultured cells. Cancer Research. 2010;70(2):560–567.
    1. García-Olmo DC, García-Olmo D. Biological role of cell-free nucleic acids in cancer: the theory of genometastasis. Critical Reviews in Oncogenesis. 2013;18(1–2):153–161.
    1. García-Olmo DC, Picazo MG, García-Olmo D. Transformation of non-tumor host cells during tumor progression: theories and evidence. Expert Opinion on Biological Therapy. 2012;12(Suppl 1):S199–207.
    1. Peters DL, Pretorius PJ. Continuous adaptation through genetic communication - a putative role for cell-free DNA. Expert Opinion on Biological Therapy. 2012;12(Suppl 1):S127–132.
    1. Hariton-Gazal E, Rosenbluh J, Graessmann A, Gilon C, Loyter A. Direct translocation of histone molecules across cell membranes. Journal of Cell Science. 2003;116(Pt 22):4577–4586.
    1. Mittra I, Khare NK, Raghuram GV, Chaubal R, Khambatti F, Gupta D, Dutt A. Circulating nucleic acids damage DNA of healthy cells by integrating into their genomes. Journal of Biosciences. 2015;40(1):91–111.
    1. Mittra I. Circulating nucleic acids: a new class of physiological mobile genetic elements. F1000Research. 2015;4:924.
    1. Gahan PB, Anker P, Stroun M, Jacob K. DNA-Induced Chromosome Damage in Vicia Faba. Caryologia. 1969;22(4):307–310.
    1. Gahan PB, Perry IJ, Stroun M, Anker P. Effect of Exogenous DNA on Acid Deoxyribonuclease Activity in Intact Roots of Vicia faba L. Annals of Botany. 1974;38(3):701–704.
    1. Klieve AV, Yokoyama MT, Forster RJ, Ouwerkerk D, Bain PA, Mawhinney EL. Naturally occurring DNA transfer system associated with membrane vesicles in cellulolytic Ruminococcus spp. of ruminal origin. Applied and Environmental Microbiology. 2005;71(8):4248–4253.
    1. de la Cruz F, Davies J. Horizontal gene transfer and the origin of species: lessons from bacteria. Trends in Microbiology. 2000;8(3):128–133.
    1. Moriguchi K, Yamamoto S, Tanaka K, Kurata N, Suzuki K. Trans-kingdom horizontal DNA transfer from bacteria to yeast is highly plastic due to natural polymorphisms in auxiliary nonessential recipient genes. PloS One. 2013;8(9):e74590.
    1. Heinemann JA, Sprague GF. Bacterial conjugative plasmids mobilize DNA transfer between bacteria and yeast. Nature. 1989;340(6230):205–209.
    1. Syvanen M. Cross-species gene transfer; implications for a new theory of evolution. Journal of Theoretical Biology. 1985;112(2):333–343.
    1. Jabr, F. (n.d.). Genes that jump species: does this shake the tree of life? — Ferris Jabr — Aeon Essays. Aeon. Retrieved March 16, 2016, from .
    1. Stroun M, Anker P, Gahan P. B and Sheikh K. (1971). Bacterial infection due to natural release of nucleic acids from bacteria in plant cells. Informative Molecules in Biological Systems, 187–192.
    1. Anker P, Stroun M, Laroche J. Bacterial RNA synthesis in frog auricles after intraperitoneal injection of bacteria. Experientia. 1972;28(4):488–489.
    1. Pisetsky DS. The origin and properties of extracellular DNA: from PAMP to DAMP. Clinical Immunology (Orlando, Fla.) 2012;144(1):32–40.
    1. Kawasaki T, Kawai T, Akira S. Recognition of nucleic acids by pattern-recognition receptors and its relevance in autoimmunity. Immunological Reviews. 2011;243(1):61–73.
    1. Napirei M, Karsunky H, Zevnik B, Stephan H, Mannherz HG, Möröy T. Features of systemic lupus erythematosus in Dnase1-deficient mice. Nature Genetics. 2000;25(2):177–181.
    1. Jacob M, Napirei M, Ricken A, Dixkens C, Mannherz HG. Histopathology of lupus-like nephritis in Dnase1-deficient mice in comparison to NZB/W F1 mice. Lupus. 2002;11(8):514–527.
    1. Martinez-Valle F, Balada E, Ordi-Ros J, Bujan-Rivas S, Sellas-Fernandez A, Vilardell-Tarres M. DNase 1 activity in patients with systemic lupus erythematosus: relationship with epidemiological, clinical, immunological and therapeutical features. Lupus. 2009;18(5):418–423.
    1. Chitrabamrung S, Rubin RL, Tan EM. Serum deoxyribonuclease I and clinical activity in systemic lupus erythematosus. Rheumatology International. 1981;1(2):55–60.
    1. Kawai T, Akira S. The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nature Immunology. 2010;11(5):373–384.
    1. Pisetsky DS, Ullal AJ. The blood nucleome in the pathogenesis of SLE. Autoimmunity Reviews. 2010;10(1):35–37.
    1. Klinman DM. Immunotherapeutic uses of CpG oligodeoxynucleotides. Nature Reviews Immunology. 2004;4(4):249–259.
    1. Margulis L. Symbiotic theory of the origin of eukaryotic organelles; criteria for proof. Symposia of the Society for Experimental Biology. 1975;29:21–38.
    1. McCarthy CG, Wenceslau CF, Goulopoulou S, Ogbi S, Baban B, Sullivan JC, Webb RC. Circulating mitochondrial DNA and Toll-like receptor 9 are associated with vascular dysfunction in spontaneously hypertensive rats. Cardiovascular Research. 2015;107(1):119–130.
    1. Lewis EJ, Schwartz MM. Pathology of lupus nephritis. Lupus. 2005;14(1):31–38.
    1. Scharfe-Nugent A, Corr SC, Carpenter SB, Keogh L, Doyle B, Martin C, O’Neill LAJ. TLR9 provokes inflammation in response to fetal DNA: mechanism for fetal loss in preterm birth and preeclampsia. Journal of Immunology (Baltimore, Md.: 1950) 2012;188(11):5706–5712.
    1. Cotter AM, Martin CM, O’leary JJ, Daly SF. Increased fetal DNA in the maternal circulation in early pregnancy is associated with an increased risk of preeclampsia. American Journal of Obstetrics and Gynecology. 2004;191(2):515–520.
    1. Seval MM, Karabulut HG, Tükün A, Koç A. Cell free fetal DNA in the plasma of pregnant women with preeclampsia. Clinical and Experimental Obstetrics & Gynecology. 2015;42(6):787–791.
    1. Wartha F, Beiter K, Normark S, Henriques-Normark B. Neutrophil extracellular traps: casting the NET over pathogenesis. Current Opinion in Microbiology. 2007;10(1):52–56.
    1. Zawrotniak M, Rapala-Kozik M. Neutrophil extracellular traps (NETs) - formation and implications. Acta Biochimica Polonica. 2013;60(3):277–284.
    1. Demers M, Wagner DD. Neutrophil extracellular traps: A new link to cancer-associated thrombosis and potential implications for tumor progression. Oncoimmunology. 2013;2(2):e22946.
    1. Spicer JD, McDonald B, Cools-Lartigue JJ, Chow SC, Giannias B, Kubes P, Ferri LE. Neutrophils promote liver metastasis via Mac-1-mediated interactions with circulating tumor cells. Cancer Research. 2012;72(16):3919–3927.
    1. McDonald B, Spicer J, Giannais B, Fallavollita L, Brodt P, Ferri LE. Systemic inflammation increases cancer cell adhesion to hepatic sinusoids by neutrophil mediated mechanisms. International Journal of Cancer. 2009;125(6):1298–1305.
    1. Hawes MC, Wen F, Elquza E. Extracellular DNA: A Bridge to Cancer. Cancer Research. 2015;75(20):4260–4264.
    1. Patutina O, Mironova N, Ryabchikova E, Popova N, Nikolin V, Kaledin V, Zenkova M. Inhibition of metastasis development by daily administration of ultralow doses of RNase A and DNase I. Biochimie. 2011;93(4):689–696.
    1. Patutina OA, Mironova NL, Ryabchikova EI, Popova NA, Nikolin VP, Kaledin VI, Zenkova MA. Tumoricidal Activity of RNase A and DNase I. Acta Naturae. 2010;2(1):88–94.
    1. Trejo-Becerril C, Pérez-Cárdenas E, Taja-Chayeb L, Anker P, Herrera-Goepfert R, Medina-Velázquez LA, Dueñas-González A. Cancer progression mediated by horizontal gene transfer in an in vivo model. PloS One. 2012;7(12):e52754.
    1. Lee J, Sohn JW, Zhang Y, Leong KW, Pisetsky D, Sullenger BA. Nucleic acid-binding polymers as anti-inflammatory agents. Proceedings of the National Academy of Sciences of the United States of America. 2011;108(34):14055–14060.
    1. Holl EK, Shumansky KL, Pitoc G, Ramsburg E, Sullenger BA. Nucleic acid scavenging polymers inhibit extracellular DNA-mediated innate immune activation without inhibiting anti-viral responses. PloS One. 2013;8(7):e69413.
    1. Stroun, M. (2012). “Mon cher collègue je ne serai pas recteur”: Une aventure dans le monde de l’Université et de la recherche scientifique suisse. Editions L’Harmattan.
    1. Dawson S-J, Tsui DWY, Murtaza M, Biggs H, Rueda OM, Chin S-F, Rosenfeld N. Analysis of Circulating Tumor DNA to Monitor Metastatic Breast Cancer. New England Journal of Medicine. 2013;368(13):1199–1209.
    1. Misale S, Yaeger R, Hobor S, Scala E, Janakiraman M, Liska D, Bardelli A. Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature. 2012;486(7404):532–536.
    1. Garcia-Murillas I, Schiavon G, Weigelt B, Ng C, Hrebien S, Cutts RJ, Turner NC. Mutation tracking in circulating tumor DNA predicts relapse in early breast cancer. Science Translational Medicine. 2015;7(302):302ra133.
    1. Rago C, Huso DL, Diehl F, Karim B, Liu G, Papadopoulos N, Diaz LA. Serial assessment of human tumor burdens in mice by the analysis of circulating DNA. Cancer Research. 2007;67(19):9364–9370.
    1. Diaz LA, Jr, Williams RT, Wu J, Kinde I, Hecht JR, Berlin J, Vogelstein B. The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers. Nature. 2012;486(7404):537–540.

Source: PubMed

3
Tilaa