Multidimensional fragmentomic profiling of cell-free DNA released from patient-derived organoids

Abstract Background Fragmentomics, the investigation of fragmentation patterns of cell-free DNA (cfDNA), has emerged as a promising strategy for the early detection of multiple cancers in the field of liquid biopsy. However, the clinical application of this approach has been hindered by a limited un...

Full description

Bibliographic Details
Main Authors: Jaeryuk Kim, Seung-Pyo Hong, Seyoon Lee, Woochan Lee, Dakyung Lee, Rokhyun Kim, Young Jun Park, Sungji Moon, Kyunghyuk Park, Bukyoung Cha, Jong-Il Kim
Format: Article
Language:English
Published: BMC 2023-10-01
Series:Human Genomics
Subjects:
Online Access:https://doi.org/10.1186/s40246-023-00533-0
_version_ 1797647273651339264
author Jaeryuk Kim
Seung-Pyo Hong
Seyoon Lee
Woochan Lee
Dakyung Lee
Rokhyun Kim
Young Jun Park
Sungji Moon
Kyunghyuk Park
Bukyoung Cha
Jong-Il Kim
author_facet Jaeryuk Kim
Seung-Pyo Hong
Seyoon Lee
Woochan Lee
Dakyung Lee
Rokhyun Kim
Young Jun Park
Sungji Moon
Kyunghyuk Park
Bukyoung Cha
Jong-Il Kim
author_sort Jaeryuk Kim
collection DOAJ
description Abstract Background Fragmentomics, the investigation of fragmentation patterns of cell-free DNA (cfDNA), has emerged as a promising strategy for the early detection of multiple cancers in the field of liquid biopsy. However, the clinical application of this approach has been hindered by a limited understanding of cfDNA biology. Furthermore, the prevalence of hematopoietic cell-derived cfDNA in plasma complicates the in vivo investigation of tissue-specific cfDNA other than that of hematopoietic origin. While conventional two-dimensional cell lines have contributed to research on cfDNA biology, their limited representation of in vivo tissue contexts underscores the need for more robust models. In this study, we propose three-dimensional organoids as a novel in vitro model for studying cfDNA biology, focusing on multifaceted fragmentomic analyses. Results We established nine patient-derived organoid lines from normal lung airway, normal gastric, and gastric cancer tissues. We then extracted cfDNA from the culture medium of these organoids in both proliferative and apoptotic states. Using whole-genome sequencing data from cfDNA, we analyzed various fragmentomic features, including fragment size, footprints, end motifs, and repeat types at the end. The distribution of cfDNA fragment sizes in organoids, especially in apoptosis samples, was similar to that found in plasma, implying occupancy by mononucleosomes. The footprints determined by sequencing depth exhibited distinct patterns depending on fragment sizes, reflecting occupancy by a variety of DNA-binding proteins. Notably, we discovered that short fragments (< 118 bp) were exclusively enriched in the proliferative state and exhibited distinct fragmentomic profiles, characterized by 3 bp palindromic end motifs and specific repeats. Conclusions In conclusion, our results highlight the utility of in vitro organoid models as a valuable tool for studying cfDNA biology and its associated fragmentation patterns. This, in turn, will pave the way for further enhancements in noninvasive cancer detection methodologies based on fragmentomics.
first_indexed 2024-03-11T15:13:55Z
format Article
id doaj.art-8b998c061fa34b3d9c74505a98d53d26
institution Directory Open Access Journal
issn 1479-7364
language English
last_indexed 2024-03-11T15:13:55Z
publishDate 2023-10-01
publisher BMC
record_format Article
series Human Genomics
spelling doaj.art-8b998c061fa34b3d9c74505a98d53d262023-10-29T12:30:24ZengBMCHuman Genomics1479-73642023-10-0117111510.1186/s40246-023-00533-0Multidimensional fragmentomic profiling of cell-free DNA released from patient-derived organoidsJaeryuk Kim0Seung-Pyo Hong1Seyoon Lee2Woochan Lee3Dakyung Lee4Rokhyun Kim5Young Jun Park6Sungji Moon7Kyunghyuk Park8Bukyoung Cha9Jong-Il Kim10Genomic Medicine Institute, Medical Research Center, Seoul National UniversityGenomic Medicine Institute, Medical Research Center, Seoul National UniversityGenomic Medicine Institute, Medical Research Center, Seoul National UniversityGenomic Medicine Institute, Medical Research Center, Seoul National UniversityGenomic Medicine Institute, Medical Research Center, Seoul National UniversityGenomic Medicine Institute, Medical Research Center, Seoul National UniversityGenomic Medicine Institute, Medical Research Center, Seoul National UniversityGenomic Medicine Institute, Medical Research Center, Seoul National UniversityGenomic Medicine Institute, Medical Research Center, Seoul National UniversityGenomic Medicine Institute, Medical Research Center, Seoul National UniversityGenomic Medicine Institute, Medical Research Center, Seoul National UniversityAbstract Background Fragmentomics, the investigation of fragmentation patterns of cell-free DNA (cfDNA), has emerged as a promising strategy for the early detection of multiple cancers in the field of liquid biopsy. However, the clinical application of this approach has been hindered by a limited understanding of cfDNA biology. Furthermore, the prevalence of hematopoietic cell-derived cfDNA in plasma complicates the in vivo investigation of tissue-specific cfDNA other than that of hematopoietic origin. While conventional two-dimensional cell lines have contributed to research on cfDNA biology, their limited representation of in vivo tissue contexts underscores the need for more robust models. In this study, we propose three-dimensional organoids as a novel in vitro model for studying cfDNA biology, focusing on multifaceted fragmentomic analyses. Results We established nine patient-derived organoid lines from normal lung airway, normal gastric, and gastric cancer tissues. We then extracted cfDNA from the culture medium of these organoids in both proliferative and apoptotic states. Using whole-genome sequencing data from cfDNA, we analyzed various fragmentomic features, including fragment size, footprints, end motifs, and repeat types at the end. The distribution of cfDNA fragment sizes in organoids, especially in apoptosis samples, was similar to that found in plasma, implying occupancy by mononucleosomes. The footprints determined by sequencing depth exhibited distinct patterns depending on fragment sizes, reflecting occupancy by a variety of DNA-binding proteins. Notably, we discovered that short fragments (< 118 bp) were exclusively enriched in the proliferative state and exhibited distinct fragmentomic profiles, characterized by 3 bp palindromic end motifs and specific repeats. Conclusions In conclusion, our results highlight the utility of in vitro organoid models as a valuable tool for studying cfDNA biology and its associated fragmentation patterns. This, in turn, will pave the way for further enhancements in noninvasive cancer detection methodologies based on fragmentomics.https://doi.org/10.1186/s40246-023-00533-0Cell-free DNA biologyIn vitro modelsFragmentomicsOrganoidsExtrachromosomal circular DNA
spellingShingle Jaeryuk Kim
Seung-Pyo Hong
Seyoon Lee
Woochan Lee
Dakyung Lee
Rokhyun Kim
Young Jun Park
Sungji Moon
Kyunghyuk Park
Bukyoung Cha
Jong-Il Kim
Multidimensional fragmentomic profiling of cell-free DNA released from patient-derived organoids
Human Genomics
Cell-free DNA biology
In vitro models
Fragmentomics
Organoids
Extrachromosomal circular DNA
title Multidimensional fragmentomic profiling of cell-free DNA released from patient-derived organoids
title_full Multidimensional fragmentomic profiling of cell-free DNA released from patient-derived organoids
title_fullStr Multidimensional fragmentomic profiling of cell-free DNA released from patient-derived organoids
title_full_unstemmed Multidimensional fragmentomic profiling of cell-free DNA released from patient-derived organoids
title_short Multidimensional fragmentomic profiling of cell-free DNA released from patient-derived organoids
title_sort multidimensional fragmentomic profiling of cell free dna released from patient derived organoids
topic Cell-free DNA biology
In vitro models
Fragmentomics
Organoids
Extrachromosomal circular DNA
url https://doi.org/10.1186/s40246-023-00533-0
work_keys_str_mv AT jaeryukkim multidimensionalfragmentomicprofilingofcellfreednareleasedfrompatientderivedorganoids
AT seungpyohong multidimensionalfragmentomicprofilingofcellfreednareleasedfrompatientderivedorganoids
AT seyoonlee multidimensionalfragmentomicprofilingofcellfreednareleasedfrompatientderivedorganoids
AT woochanlee multidimensionalfragmentomicprofilingofcellfreednareleasedfrompatientderivedorganoids
AT dakyunglee multidimensionalfragmentomicprofilingofcellfreednareleasedfrompatientderivedorganoids
AT rokhyunkim multidimensionalfragmentomicprofilingofcellfreednareleasedfrompatientderivedorganoids
AT youngjunpark multidimensionalfragmentomicprofilingofcellfreednareleasedfrompatientderivedorganoids
AT sungjimoon multidimensionalfragmentomicprofilingofcellfreednareleasedfrompatientderivedorganoids
AT kyunghyukpark multidimensionalfragmentomicprofilingofcellfreednareleasedfrompatientderivedorganoids
AT bukyoungcha multidimensionalfragmentomicprofilingofcellfreednareleasedfrompatientderivedorganoids
AT jongilkim multidimensionalfragmentomicprofilingofcellfreednareleasedfrompatientderivedorganoids