Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 134(2)2024 Jan 16.
Article in English | MEDLINE | ID: mdl-37971879

ABSTRACT

BACKGROUND: Accurate detection of graft-versus-host disease (GVHD) is a major challenge in the management of patients undergoing hematopoietic stem cell transplantation (HCT). Here, we demonstrated the use of circulating cell-free DNA (cfDNA) for detection of tissue turnover and chronic GVHD (cGVHD) in specific organs. METHODS: We established a cocktail of tissue-specific DNA methylation markers and used it to determine the concentration of cfDNA molecules derived from the liver, skin, lungs, colon, and specific immune cells in 101 patients undergoing HCT. RESULTS: Patients with active cGVHD showed elevated concentrations of cfDNA, as well as tissue-specific methylation markers that agreed with clinical scores. Strikingly, transplanted patients with no clinical symptoms had abnormally high levels of tissue-specific markers, suggesting hidden tissue turnover even in the absence of evident clinical pathology. An integrative model taking into account total cfDNA concentration, monocyte/macrophage cfDNA levels and alanine transaminase was able to correctly identify GVHD with a specificity of 86% and precision of 89% (AUC of 0.8). CONCLUSION: cfDNA markers can be used for the detection of cGVHD, opening a window into underlying tissue dynamics in patients that receive allogeneic stem cell transplants. FUNDING: This work was supported by grants from the Ernest and Bonnie Beutler Research Program of Excellence in Genomic Medicine, The Israel Science Foundation, the Waldholtz/Pakula family, the Robert M. and Marilyn Sternberg Family Charitable Foundation and the Helmsley Charitable Trust (to YD).


Subject(s)
Bronchiolitis Obliterans Syndrome , Cell-Free Nucleic Acids , Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Humans , DNA Methylation , Cell-Free Nucleic Acids/genetics , Graft vs Host Disease/diagnosis , Biomarkers , Genetic Markers , Chronic Disease
2.
Gut ; 73(4): 639-648, 2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38123998

ABSTRACT

OBJECTIVE: Pancreatic ductal adenocarcinoma (PDAC) is commonly diagnosed at an advanced stage. Liquid biopsy approaches may facilitate detection of early stage PDAC when curative treatments can be employed. DESIGN: To assess circulating marker discrimination in training, testing and validation patient cohorts (total n=426 patients), plasma markers were measured among PDAC cases and patients with chronic pancreatitis, colorectal cancer (CRC), and healthy controls. Using CA19-9 as an anchor marker, measurements were made of two protein markers (TIMP1, LRG1) and cell-free DNA (cfDNA) pancreas-specific methylation at 9 loci encompassing 61 CpG sites. RESULTS: Comparative methylome analysis identified nine loci that were differentially methylated in exocrine pancreas DNA. In the training set (n=124 patients), cfDNA methylation markers distinguished PDAC from healthy and CRC controls. In the testing set of 86 early stage PDAC and 86 matched healthy controls, CA19-9 had an area under the receiver operating characteristic curve (AUC) of 0.88 (95% CI 0.83 to 0.94), which was increased by adding TIMP1 (AUC 0.92; 95% CI 0.88 to 0.96; p=0.06), LRG1 (AUC 0.92; 95% CI 0.88 to 0.96; p=0.02) or exocrine pancreas-specific cfDNA methylation markers at nine loci (AUC 0.92; 95% CI 0.88 to 0.96; p=0.02). In the validation set of 40 early stage PDAC and 40 matched healthy controls, a combined panel including CA19-9, TIMP1 and a 9-loci cfDNA methylation panel had greater discrimination (AUC 0.86, 95% CI 0.77 to 0.95) than CA19-9 alone (AUC 0.82; 95% CI 0.72 to 0.92). CONCLUSION: A combined panel of circulating markers including proteins and methylated cfDNA increased discrimination compared with CA19-9 alone for early stage PDAC.


Subject(s)
Adenocarcinoma , Carcinoma, Pancreatic Ductal , Cell-Free Nucleic Acids , Pancreatic Neoplasms , Humans , CA-19-9 Antigen , Biomarkers, Tumor , Cell-Free Nucleic Acids/metabolism , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Carcinoma, Pancreatic Ductal/diagnosis , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Pancreas/pathology , Adenocarcinoma/diagnosis , Adenocarcinoma/genetics , Adenocarcinoma/pathology , DNA Methylation
3.
Med ; 4(4): 263-281.e4, 2023 04 14.
Article in English | MEDLINE | ID: mdl-37060900

ABSTRACT

BACKGROUND: Vascular endothelial cells (VECs) are an essential component of each tissue, contribute to multiple pathologies, and are targeted by important drugs. Yet, there is a shortage of biomarkers to assess VEC turnover. METHODS: To develop DNA methylation-based liquid biopsies for VECs, we determined the methylome of VECs isolated from freshly dissociated human tissues. FINDINGS: A comparison with a human cell-type methylome atlas yielded thousands of loci that are uniquely unmethylated in VECs. These sites are typically gene enhancers, often residing adjacent to VEC-specific genes. We also identified hundreds of genomic loci that are differentially methylated in organotypic VECs, indicating that VECs feeding specific organs are distinct cell types with a stable epigenetic identity. We established universal and lung-specific VEC markers and evaluated their presence in circulating cell-free DNA (cfDNA). Nearly 2.5% of cfDNA in the plasma of healthy individuals originates from VECs. Sepsis, graft versus host disease, and cardiac catheterization are associated with elevated levels of VEC-derived cfDNA, indicative of vascular damage. Lung-specific VEC cfDNA is selectively elevated in patients with chronic obstructive pulmonary disease (COPD) or lung cancer, revealing tissue-specific vascular turnover. CONCLUSIONS: VEC cfDNA biomarkers inform vascular dynamics in health and disease, potentially contributing to early diagnosis and monitoring of pathologies, and assessment of drug activity. FUNDING: This work was supported by the Beutler Research Program, Helmsley Charitable Trust, JDRF, Grail and the DON Foundation (to Y.D.). Y.D holds the Walter & Greta Stiel Chair in heart studies. B.G., R.S., J.M., D.N., T.K., and Y.D. filed patents on cfDNA analysis.


Subject(s)
Cell-Free Nucleic Acids , Epigenome , Humans , Endothelium, Vascular , Endothelial Cells/metabolism , Biomarkers/metabolism , Liquid Biopsy
4.
Elife ; 112022 06 14.
Article in English | MEDLINE | ID: mdl-35699419

ABSTRACT

Schizophrenia is a common, severe, and debilitating psychiatric disorder. Despite extensive research there is as yet no biological marker that can aid in its diagnosis and course prediction. This precludes early detection and intervention. Imaging studies suggest brain volume loss around the onset and over the first few years of schizophrenia, and apoptosis has been proposed as the underlying mechanism. Cell-free DNA (cfDNA) fragments are released into the bloodstream following cell death. Tissue-specific methylation patterns allow the identification of the tissue origins of cfDNA. We developed a cocktail of brain-specific DNA methylation markers, and used it to assess the presence of brain-derived cfDNA in the plasma of patients with a first psychotic episode. We detected significantly elevated neuron- (p=0.0013), astrocyte- (p=0.0016), oligodendrocyte- (p=0.0129), and whole brain-derived (p=0.0012) cfDNA in the plasma of patients during their first psychotic episode (n=29), compared with healthy controls (n=31). Increased cfDNA levels were not correlated with psychotropic medications use. Area under the curve (AUC) was 0.77, with 65% sensitivity at 90% specificity in patients with a psychotic episode. Potential interpretations of these findings include increased brain cell death, disruption of the blood-brain barrier, or a defect in clearance of material from dying brain cells. Brain-specific cfDNA methylation markers can potentially assist early detection and monitoring of schizophrenia and thus allow early intervention and adequate therapy.


Subject(s)
Cell-Free Nucleic Acids , Psychotic Disorders , Biomarkers, Tumor/genetics , Brain , Cell-Free Nucleic Acids/genetics , DNA Methylation , Genetic Markers , Humans , Psychotic Disorders/genetics
5.
Med ; 3(7): 468-480.e5, 2022 07 08.
Article in English | MEDLINE | ID: mdl-35716665

ABSTRACT

BACKGROUND: Much remains unknown regarding the response of the immune system to severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) vaccination. METHODS: We employed circulating cell-free DNA (cfDNA) to assess the turnover of specific immune cell types following administration of the Pfizer/BioNTech vaccine. FINDINGS: The levels of B cell cfDNA after the primary dose correlated with development of neutralizing antibodies and memory B cells after the booster, revealing a link between early B cell turnover-potentially reflecting affinity maturation-and later development of effective humoral response. We also observed co-elevation of B cell, T cell, and monocyte cfDNA after the booster, underscoring the involvement of innate immune cell turnover in the development of humoral and cellular adaptive immunity. Actual cell counts remained largely stable following vaccination, other than a previously demonstrated temporary reduction in neutrophil and lymphocyte counts. CONCLUSIONS: Immune cfDNA dynamics reveal the crucial role of the primary SARS-CoV-2 vaccine in shaping responses of the immune system following the booster vaccine. FUNDING: This work was supported by a generous gift from Shlomo Kramer. Supported by grants from Human Islet Research Network (HIRN UC4DK116274 and UC4DK104216 to R.S. and Y.D.), Ernest and Bonnie Beutler Research Program of Excellence in Genomic Medicine, The Alex U Soyka Pancreatic Cancer Fund, The Israel Science Foundation, the Waldholtz/Pakula family, the Robert M. and Marilyn Sternberg Family Charitable Foundation, the Helmsley Charitable Trust, Grail, and the DON Foundation (to Y.D.). Y.D. holds the Walter and Greta Stiel Chair and Research Grant in Heart Studies. I.F.-F. received a fellowship from the Glassman Hebrew University Diabetes Center.


Subject(s)
BNT162 Vaccine , COVID-19 , Cell-Free Nucleic Acids , SARS-CoV-2 , Adult , Aged , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/immunology , Antibodies, Viral/genetics , Antibodies, Viral/immunology , BNT162 Vaccine/administration & dosage , COVID-19/immunology , COVID-19/prevention & control , Cell-Free Nucleic Acids/genetics , Cell-Free Nucleic Acids/immunology , Female , Humans , Immunization, Secondary , Male , Memory B Cells/immunology , Memory B Cells/metabolism , Middle Aged , SARS-CoV-2/immunology , Young Adult
6.
Eur Respir J ; 60(5)2022 11.
Article in English | MEDLINE | ID: mdl-35450968

ABSTRACT

BACKGROUND: Circulating biomarkers for lung damage are lacking. Lung epithelium-specific DNA methylation patterns can potentially report the presence of lung-derived cell-free DNA (cfDNA) in blood, as an indication of lung cell death. METHODS: We sorted human lung alveolar and bronchial epithelial cells from surgical specimens, and obtained their methylomes using whole-genome bisulfite sequencing. We developed a PCR sequencing assay determining the methylation status of 17 loci with lung-specific methylation patterns, and used it to assess lung-derived cfDNA in the plasma of healthy volunteers and patients with lung disease. RESULTS: Loci that are uniquely unmethylated in alveolar or bronchial epithelial cells are enriched for enhancers controlling lung-specific genes. Methylation markers extracted from these methylomes revealed that normal lung cell turnover probably releases cfDNA into the air spaces, rather than to blood. People with advanced lung cancer show a massive elevation of lung cfDNA concentration in blood. Among individuals undergoing bronchoscopy, lung-derived cfDNA is observed in the plasma of those later diagnosed with lung cancer, and to a lesser extent in those diagnosed with other lung diseases. Lung cfDNA is also elevated in patients with acute exacerbation of COPD compared with patients with stable disease, and is associated with future exacerbation and mortality in these patients. CONCLUSIONS: Universal cfDNA methylation markers of normal lung epithelium allow for mutation-independent, sensitive and specific detection of lung-derived cfDNA, reporting on ongoing lung injury. Such markers can find broad utility in the study of normal and pathologic human lung dynamics.


Subject(s)
Cell-Free Nucleic Acids , Lung Neoplasms , Humans , DNA Methylation , Cell-Free Nucleic Acids/genetics , Liquid Biopsy , Biomarkers , Epithelium , Lung , Lung Neoplasms/genetics , Biomarkers, Tumor/genetics
7.
JCI Insight ; 7(2)2022 01 25.
Article in English | MEDLINE | ID: mdl-35076021

ABSTRACT

Cancer inflicts damage to surrounding normal tissues, which can culminate in fatal organ failure. Here, we demonstrate that cell death in organs affected by cancer can be detected by tissue-specific methylation patterns of circulating cell-free DNA (cfDNA). We detected elevated levels of hepatocyte-derived cfDNA in the plasma of patients with liver metastases originating from different primary tumors, compared with cancer patients without liver metastases. In addition, patients with localized pancreatic or colon cancer showed elevated hepatocyte cfDNA, suggesting liver damage inflicted by micrometastatic disease, by primary pancreatic tumor pressing the bile duct, or by a systemic response to the primary tumor. We also identified elevated neuron-, oligodendrocyte-, and astrocyte-derived cfDNA in a subpopulation of patients with brain metastases compared with cancer patients without brain metastasis. Cell type-specific cfDNA methylation markers enabled the identification of collateral tissue damage in cancer, revealing the presence of metastases in specific locations and potentially assisting in early cancer detection.


Subject(s)
Brain Neoplasms , Cell-Free Nucleic Acids , DNA Methylation , Liquid Biopsy/methods , Liver Neoplasms , Neoplasm Metastasis , Pancreatic Neoplasms , Biomarkers, Tumor/analysis , Biomarkers, Tumor/blood , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/secondary , Cell-Free Nucleic Acids/analysis , Cell-Free Nucleic Acids/blood , Early Detection of Cancer/methods , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/secondary , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Pancreatic Neoplasms/complications , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology
8.
Elife ; 102021 11 29.
Article in English | MEDLINE | ID: mdl-34842142

ABSTRACT

Blood cell counts often fail to report on immune processes occurring in remote tissues. Here, we use immune cell type-specific methylation patterns in circulating cell-free DNA (cfDNA) for studying human immune cell dynamics. We characterized cfDNA released from specific immune cell types in healthy individuals (N = 242), cross sectionally and longitudinally. Immune cfDNA levels had no individual steady state as opposed to blood cell counts, suggesting that cfDNA concentration reflects adjustment of cell survival to maintain homeostatic cell numbers. We also observed selective elevation of immune-derived cfDNA upon perturbations of immune homeostasis. Following influenza vaccination (N = 92), B-cell-derived cfDNA levels increased prior to elevated B-cell counts and predicted efficacy of antibody production. Patients with eosinophilic esophagitis (N = 21) and B-cell lymphoma (N = 27) showed selective elevation of eosinophil and B-cell cfDNA, respectively, which were undetectable by cell counts in blood. Immune-derived cfDNA provides a novel biomarker for monitoring immune responses to physiological and pathological processes that are not accessible using conventional methods.


Subject(s)
Biomarkers, Tumor/metabolism , Cell-Free Nucleic Acids/metabolism , DNA Methylation , Immunity , Adult , Aged , Female , Humans , Male , Middle Aged , Young Adult
9.
JCI Insight ; 5(14)2020 07 23.
Article in English | MEDLINE | ID: mdl-32573495

ABSTRACT

It has been proposed that unmethylated insulin promoter fragments in plasma derive exclusively from ß cells, reflect their recent demise, and can be used to assess ß cell damage in type 1 diabetes. Herein we describe an ultrasensitive assay for detection of a ß cell-specific DNA methylation signature, by simultaneous assessment of 6 DNA methylation markers, that identifies ß cell DNA in mixtures containing as little as 0.03% ß cell DNA (less than 1 ß cell genome equivalent). Based on this assay, plasma from nondiabetic individuals (N = 218, aged 4-78 years) contained on average only 1 ß cell genome equivalent/mL. As expected, cell-free DNA (cfDNA) from ß cells was significantly elevated in islet transplant recipients shortly after transplantation. We also detected ß cell cfDNA in a patient with KATP congenital hyperinsulinism, in which substantial ß cell turnover is thought to occur. Strikingly, in contrast to previous reports, we observed no elevation of ß cell-derived cfDNA in autoantibody-positive subjects at risk for type 1 diabetes (N = 32), individuals with recent-onset type 1 diabetes (<4 months, N = 92), or those with long-standing disease (>4 months, N = 38). We discuss the utility of sensitive ß cell cfDNA analysis and potential explanations for the lack of a ß cell cfDNA signal in type 1 diabetes.


Subject(s)
Cell-Free Nucleic Acids/blood , DNA Methylation/genetics , Diabetes Mellitus, Type 1/blood , Insulin-Secreting Cells/metabolism , Adolescent , Adult , Aged , Biomarkers/blood , Child , Child, Preschool , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Female , Humans , Insulin/genetics , Insulin/metabolism , Insulin-Secreting Cells/pathology , Male , Middle Aged , Young Adult
10.
JCI Insight ; 3(12)2018 06 21.
Article in English | MEDLINE | ID: mdl-29925683

ABSTRACT

Liver damage is typically inferred from serum measurements of cytoplasmic liver enzymes. DNA molecules released from dying hepatocytes are an alternative biomarker, unexplored so far, potentially allowing for quantitative assessment of liver cell death. Here we describe a method for detecting acute hepatocyte death, based on quantification of circulating, cell-free DNA (cfDNA) fragments carrying hepatocyte-specific methylation patterns. We identified 3 genomic loci that are unmethylated specifically in hepatocytes, and used bisulfite conversion, PCR, and massively parallel sequencing to quantify the concentration of hepatocyte-derived DNA in mixed samples. Healthy donors had, on average, 30 hepatocyte genomes/ml plasma, reflective of basal cell turnover in the liver. We identified elevations of hepatocyte cfDNA in patients shortly after liver transplantation, during acute rejection of an established liver transplant, and also in healthy individuals after partial hepatectomy. Furthermore, patients with sepsis had high levels of hepatocyte cfDNA, which correlated with levels of liver enzymes aspartate aminotransferase (AST) and alanine aminotransferase (ALT). Duchenne muscular dystrophy patients, in which elevated AST and ALT derive from damaged muscle rather than liver, did not have elevated hepatocyte cfDNA. We conclude that measurements of hepatocyte-derived cfDNA can provide specific and sensitive information on hepatocyte death, for monitoring human liver dynamics, disease, and toxicity.


Subject(s)
Biomarkers/blood , Cell-Free Nucleic Acids/blood , Hepatocytes/metabolism , Liver Diseases/blood , Liver Diseases/diagnosis , Liver/metabolism , Alanine Transaminase/analysis , Aspartate Aminotransferases/analysis , Blood Proteins/genetics , Cell Death , DNA Methylation , Glycoproteins/genetics , Hepatectomy , High-Throughput Nucleotide Sequencing , Humans , Liver/enzymology , Liver Transplantation , Proteinase Inhibitory Proteins, Secretory/genetics , Receptor, IGF Type 2/genetics , Sensitivity and Specificity
11.
Proc Natl Acad Sci U S A ; 114(51): 13525-13530, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29203669

ABSTRACT

DNA methylation at promoters is an important determinant of gene expression. Earlier studies suggested that the insulin gene promoter is uniquely unmethylated in insulin-expressing pancreatic ß-cells, providing a classic example of this paradigm. Here we show that islet cells expressing insulin, glucagon, or somatostatin share a lack of methylation at the promoters of the insulin and glucagon genes. This is achieved by rapid demethylation of the insulin and glucagon gene promoters during differentiation of Neurogenin3+ embryonic endocrine progenitors, regardless of the specific endocrine cell-type chosen. Similar methylation dynamics were observed in transgenic mice containing a human insulin promoter fragment, pointing to the responsible cis element. Whole-methylome comparison of human α- and ß-cells revealed generality of the findings: genes active in one cell type and silent in the other tend to share demethylated promoters, while methylation differences between α- and ß-cells are concentrated in enhancers. These findings suggest an epigenetic basis for the observed plastic identity of islet cell types, and have implications for ß-cell reprogramming in diabetes and diagnosis of ß-cell death using methylation patterns of circulating DNA.


Subject(s)
DNA Methylation , Enhancer Elements, Genetic , Glucagon-Secreting Cells/metabolism , Insulin-Secreting Cells/metabolism , Promoter Regions, Genetic , Animals , Cell Differentiation , Cell Line , Cells, Cultured , Epigenesis, Genetic , Glucagon-Secreting Cells/cytology , Humans , Insulin-Secreting Cells/cytology , Mice , Mice, Inbred ICR
SELECTION OF CITATIONS
SEARCH DETAIL
...