Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Fam Cancer ; 2024 May 14.
Article in English | MEDLINE | ID: mdl-38743206

ABSTRACT

TP53 pathogenic variants cause Li-Fraumeni syndrome (LFS), with some variants causing an attenuated phenotype. Herein, we describe the clinical phenotype and genetic characteristics of carriers of NM_000546.6 (TP53): c.541C > T, (p.Arg181Cys) treated at Hadassah Medical Center. We retrospectively examined our genetic databases to identify all carriers of TP53 p.Arg181Cys. We reached out to carriers and their relatives and collected clinical and demographic data, lifestyle factors, carcinogenic exposures as well as additional blood samples for genetic testing and whole exome sequencing. Between 2005 and 2022 a total of 2875 cancer patients underwent genetic testing using genetic panels, whole exome sequencing or targeted TP53 assays. A total of 30 cancer patients, all of Arab-Muslim descent, were found to be carriers of TP53 p.Arg181Cys, the majority from Jerusalem and Hebron, two of which were homozygous for the variant. Carriers were from 24 distinct families of them, 15 families (62.5%) met updated Chompret criteria for LFS. Median age of diagnosis was 35 years-old (range 1-69) with cancers characteristic of LFS (16 Breast cancer; 6 primary CNS tumors; 3 sarcomas) including 4 children with choroid plexus carcinoma, medulloblastoma, or glioblastoma. A total of 21 healthy carriers of TP53 p.Arg181Cys were identified at a median age of 39 years-old (range 2-54)-19 relatives and 2 additional pediatric non-cancer patients, in which the finding was incidental. We report a shared haplotype of 350kb among carriers, limited co-morbidities and low BMI in both cancer patients and healthy carriers. There were no demographic factors or carcinogenic exposures unique to carriers who developed malignancy. Upon exome analysis no other known pathogenic variants in cancer predisposing genes were identified. TP53 p.Arg181Cys is a founder pathogenic variant predominant to the Arab-Muslim population in Jerusalem and Hebron, causing attenuated-LFS. We suggest strict surveillance in established carriers and encourage referral to genetic testing for all cancer patients of Arab-Muslim descent in this region with LFS-associated malignancies as well as family members of established carriers.

2.
PLoS One ; 18(11): e0285646, 2023.
Article in English | MEDLINE | ID: mdl-38015964

ABSTRACT

BACKGROUND: Radiotherapy has an important role in the treatment of brain metastases but carries risk of short and/or long-term toxicity, termed radiation-induced brain injury (RBI). As the diagnosis of RBI is crucial for correct patient management, there is an unmet need for reliable biomarkers for RBI. The aim of this proof-of concept study is to determine the utility of brain-derived circulating free DNA (BncfDNA), identified by specific methylation patterns for neurons, astrocytes, and oligodendrocytes, as biomarkers brain injury induced by radiotherapy. METHODS: Twenty-four patients with brain metastases were monitored clinically and radiologically before, during and after brain radiotherapy, and blood for BncfDNA analysis (98 samples) was concurrently collected. Sixteen patients were treated with whole brain radiotherapy and eight patients with stereotactic radiosurgery. RESULTS: During follow-up nine RBI events were detected, and all correlated with significant increase in BncfDNA levels compared to baseline. Additionally, resolution of RBI correlated with a decrease in BncfDNA. Changes in BncfDNA were independent of tumor response. CONCLUSIONS: Elevated BncfDNA levels reflects brain cell injury incurred by radiotherapy. further research is needed to establish BncfDNA as a novel plasma-based biomarker for brain injury induced by radiotherapy.


Subject(s)
Brain Injuries , Brain Neoplasms , Radiation Injuries , Radiosurgery , Humans , Pilot Projects , Brain , Brain Neoplasms/secondary , Brain Injuries/etiology , Brain Injuries/surgery , Radiation Injuries/etiology
3.
Cancers (Basel) ; 15(16)2023 Aug 18.
Article in English | MEDLINE | ID: mdl-37627196

ABSTRACT

Sarcoma classification is challenging and can lead to treatment delays. Previous studies used DNA aberrations and machine-learning classifiers based on methylation profiles for diagnosis. We aimed to classify sarcomas by analyzing methylation signatures obtained from low-coverage whole-genome sequencing, which also identifies copy-number alterations. DNA was extracted from 23 suspected sarcoma samples and sequenced on an Oxford Nanopore sequencer. The methylation-based classifier, applied in the nanoDx pipeline, was customized using a reference set based on processed Illumina-based methylation data. Classification analysis utilized the Random Forest algorithm and t-distributed stochastic neighbor embedding, while copy-number alterations were detected using a designated R package. Out of the 23 samples encompassing a restricted range of sarcoma types, 20 were successfully sequenced, but two did not contain tumor tissue, according to the pathologist. Among the 18 tumor samples, 14 were classified as reported in the pathology results. Four classifications were discordant with the pathological report, with one compatible and three showing discrepancies. Improving tissue handling, DNA extraction methods, and detecting point mutations and translocations could enhance accuracy. We envision that rapid, accurate, point-of-care sarcoma classification using nanopore sequencing could be achieved through additional validation in a diverse tumor cohort and the integration of methylation-based classification and other DNA aberrations.

4.
Int J Cancer ; 152(7): 1444-1451, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36468189

ABSTRACT

The standard treatment approach for stage II/III rectal cancer is neoadjuvant chemoradiation therapy (nCRT) followed by surgery. In recent years, new treatment approaches have led to higher rates of complete tumor eradication combined with organ-preservation strategies. However, better tools are still needed to personalize therapy for the individual patient. In this prospective observational study, we analyzed colon-derived cell-free (cf)DNA (c-cfDNA) using a tissue-specific DNA methylation signature, and its association with therapy outcomes. Analyzing plasma samples (n = 303) collected during nCRT from 37 patients with locally advanced rectal cancer (LARC), we identified colon-specific methylation markers that discriminated healthy individuals from patients with untreated LARC (area under the curve, 0.81; 95% confidence interval, 0.70-0.92; P < .0001). Baseline c-cfDNA predicted tumor response, with increased levels linked to larger residual cancer. c-cfDNA measured after the first week of therapy identified patients with maximal response and complete cancer eradication, who had significantly lower c-cfDNA compared with those who had residual disease (8.6 vs 57.7 average copies/ml, respectively; P = .013). Increased c-cfDNA after 1 week of therapy was also associated with disease recurrence. Methylation-based liquid biopsy can predict nCRT outcomes and facilitate patient selection for escalation and de-escalation strategies.


Subject(s)
Cell-Free Nucleic Acids , Rectal Neoplasms , Humans , Cell-Free Nucleic Acids/genetics , Neoplasm Recurrence, Local , Chemoradiotherapy , Rectal Neoplasms/genetics , Rectal Neoplasms/therapy , Rectal Neoplasms/pathology , Rectum/pathology , Neoadjuvant Therapy , Treatment Outcome , Retrospective Studies
5.
Nat Biotechnol ; 41(2): 212-221, 2023 02.
Article in English | MEDLINE | ID: mdl-36076083

ABSTRACT

The analysis of cell-free DNA (cfDNA) in plasma provides information on pathological processes in the body. Blood cfDNA is in the form of nucleosomes, which maintain their tissue- and cancer-specific epigenetic state. We developed a single-molecule multiparametric assay to comprehensively profile the epigenetics of plasma-isolated nucleosomes (EPINUC), DNA methylation and cancer-specific protein biomarkers. Our system allows for high-resolution detection of six active and repressive histone modifications and their ratios and combinatorial patterns on millions of individual nucleosomes by single-molecule imaging. In addition, our system provides sensitive and quantitative data on plasma proteins, including detection of non-secreted tumor-specific proteins, such as mutant p53. EPINUC analysis of a cohort of 63 colorectal cancer, 10 pancreatic cancer and 33 healthy plasma samples detected cancer with high accuracy and sensitivity, even at early stages. Finally, combining EPINUC with direct single-molecule DNA sequencing revealed the tissue of origin of colorectal, pancreatic, lung and breast tumors. EPINUC provides multilayered information of potential clinical relevance from limited (<1 ml) liquid biopsy material.


Subject(s)
Cell-Free Nucleic Acids , Neoplasms , Nucleosomes , Humans , Biomarkers, Tumor , Cell-Free Nucleic Acids/metabolism , DNA Methylation/genetics , Epigenesis, Genetic/genetics , Neoplasm Proteins/genetics , Neoplasms/diagnosis , Neoplasms/genetics , Nucleosomes/genetics , Nucleosomes/metabolism , Single Molecule Imaging
6.
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.
Nat Biotechnol ; 39(5): 586-598, 2021 05.
Article in English | MEDLINE | ID: mdl-33432199

ABSTRACT

Cell-free DNA (cfDNA) in human plasma provides access to molecular information about the pathological processes in the organs or tumors from which it originates. These DNA fragments are derived from fragmented chromatin in dying cells and retain some of the cell-of-origin histone modifications. In this study, we applied chromatin immunoprecipitation of cell-free nucleosomes carrying active chromatin modifications followed by sequencing (cfChIP-seq) to 268 human samples. In healthy donors, we identified bone marrow megakaryocytes, but not erythroblasts, as major contributors to the cfDNA pool. In patients with a range of liver diseases, we showed that we can identify pathology-related changes in hepatocyte transcriptional programs. In patients with metastatic colorectal carcinoma, we detected clinically relevant and patient-specific information, including transcriptionally active human epidermal growth factor receptor 2 (HER2) amplifications. Altogether, cfChIP-seq, using low sequencing depth, provides systemic and genome-wide information and can inform diagnosis and facilitate interrogation of physiological and pathological processes using blood samples.


Subject(s)
Chromatin Immunoprecipitation , Colorectal Neoplasms/genetics , Enhancer Elements, Genetic/genetics , Promoter Regions, Genetic/genetics , Cell-Free System , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Neoplasm Metastasis , Nucleosomes/genetics , Sequence Analysis, DNA/methods
10.
FASEB J ; 34(12): 15701-15717, 2020 12.
Article in English | MEDLINE | ID: mdl-33136328

ABSTRACT

While the involvement of protease-activated receptors (PARs) in the physiological regulation of human placenta development, as in tumor biology, is recognized, the molecular pathway is unknown. We evaluated the impact of PAR1 and PAR2 function in cytotrophoblast (CTB) proliferation and invasion in a system of extravillous trophoblast (EVT) organ culture and in human cell-lines. Activation of PAR1 - and PAR2 -induced EVT invasion and proliferation, while the shRNA silencing of low-density lipoprotein receptor-related protein 5/6 (LRP5/6) inhibited these processes. PAR1 and PAR2 effectively induce ß-catenin stabilization in a manner similar to that shown for the canonical ß-catenin stabilization pathway yet independent of Wnts. Immunoprecipitation analyses and protein-protein docking demonstrated the co-association between either PAR1 or PAR2 with LRP5/6 forming an axis of PAR-LRP5/6-Axin. Noticeably, in PAR1 -PAR2 heterodimers a dominant role is assigned to PAR2 over PAR1 as shown by inhibition of PAR1 -induced ß-catenin levels, and Dvl nuclear localization. This inhibition takes place either by shRNA silenced hPar2 or in the presence of a TrPAR2 devoid its cytoplasmic tail. Indeed, TrPAR2 cannot form the PAR1 -PAR2 complex, obstructing thereby the flow of signals downstream. Elucidation of the mechanism of PAR-induced invasion contributes to therapeutic options highlighting key partners in the process.


Subject(s)
Low Density Lipoprotein Receptor-Related Protein-5/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Placenta/metabolism , Receptor, PAR-1/metabolism , Receptor, PAR-2/metabolism , Cell Line , Cells, Cultured , Cytoplasm/metabolism , Female , HEK293 Cells , Humans , Placentation/physiology , Pregnancy , RNA, Small Interfering/metabolism , Signal Transduction/physiology , Trophoblasts/metabolism , beta Catenin/metabolism
12.
Sci Rep ; 9(1): 18795, 2019 12 11.
Article in English | MEDLINE | ID: mdl-31827209

ABSTRACT

ERBB2 amplification is a prognostic marker for aggressive tumors and a predictive marker for prolonged survival following treatment with HER2 inhibitors. We attempt to sub-group HER2+ tumors based on amplicon structures and co-amplified genes. We examined five HER2+ cell lines, three HER2+ xenographs and 57 HER2+ tumor tissues. ERBB2 amplification was analyzed using digital droplet PCR and low coverage whole genome sequencing. In some HER2+ tumors PPM1D, that encodes WIP1, is co-amplified. Cell lines were treated with HER2 and WIP1 inhibitors. We find that inverted duplication is the amplicon structure in the majority of HER2+ tumors. In patients suffering from an early stage disease the ERBB2 amplicon is composed of a single segment while in patients suffering from advanced cancer the amplicon is composed of several different segments. We find robust WIP1 inhibition in some HER2+ PPM1D amplified cell lines. Sub-grouping HER2+ tumors using low coverage whole genome sequencing identifies inverted duplications as the main amplicon structure and based on the number of segments, differentiates between local and advanced tumors. In addition, we found that we could determine if a tumor is a recurrent tumor or second primary tumor and identify co-amplified oncogenes that may serve as targets for therapy.


Subject(s)
Gene Amplification , Neoplasms/classification , Receptor, ErbB-2/genetics , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Disease Progression , Enzyme Inhibitors/pharmacology , Female , Genes, erbB-2 , Humans , Male , Middle Aged , Neoplasms/genetics , Polymerase Chain Reaction , Protein Phosphatase 2C/antagonists & inhibitors , Protein Phosphatase 2C/genetics , Whole Genome Sequencing , Young Adult
13.
Nat Commun ; 9(1): 5068, 2018 11 29.
Article in English | MEDLINE | ID: mdl-30498206

ABSTRACT

Methylation patterns of circulating cell-free DNA (cfDNA) contain rich information about recent cell death events in the body. Here, we present an approach for unbiased determination of the tissue origins of cfDNA, using a reference methylation atlas of 25 human tissues and cell types. The method is validated using in silico simulations as well as in vitro mixes of DNA from different tissue sources at known proportions. We show that plasma cfDNA of healthy donors originates from white blood cells (55%), erythrocyte progenitors (30%), vascular endothelial cells (10%) and hepatocytes (1%). Deconvolution of cfDNA from patients reveals tissue contributions that agree with clinical findings in sepsis, islet transplantation, cancer of the colon, lung, breast and prostate, and cancer of unknown primary. We propose a procedure which can be easily adapted to study the cellular contributors to cfDNA in many settings, opening a broad window into healthy and pathologic human tissue dynamics.


Subject(s)
Cell-Free Nucleic Acids/genetics , Algorithms , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Cells, Cultured , Colonic Neoplasms/genetics , CpG Islands/genetics , DNA Methylation/genetics , Endothelial Cells/metabolism , Erythrocytes/metabolism , Hepatocytes/metabolism , Humans , Leukocytes/metabolism , Lung Neoplasms/genetics , Promoter Regions, Genetic/genetics , Sepsis/genetics
14.
Mol Clin Oncol ; 8(5): 683-685, 2018 May.
Article in English | MEDLINE | ID: mdl-29725535

ABSTRACT

Tumor genome sequencing is important for increasing our understanding of the development of cancer, which may be affected by different therapies. In the present study, genomic evolution was investigated in a patient with stage IV pancreatic cancer bearing a germline breast cancer 2 (BRCA2) mutation. The patient received cisplatin, a DNA cross-linking agent, which led to a long-lasting complete response. Eventually the patient developed brain metastasis, suggesting the acquisition of resistance to cisplatin. He subsequently underwent brain lesion resection, radiofrequency ablation and chemotherapy, again resulting in long-lasting response. Samples of blood, pancreatic tumor tissue and brain metastases were collected and the extracted DNA was sequenced. The pancreatic and brain lesions, when compared with the blood samples, exhibited mutations in the BRCA1 and checkpoint kinase 2 genes, in addition to the germline BRCA2 mutation. The brain lesion, when compared with the primary tumor, harbored no additional mutations or copy-number variations. These findings suggest that the isolated relapse in the brain was due to pharmacological sanctuary rather than genomic alterations. It may be suggested that the presence of defects in the homologous recombination repair pathways are associated with a good prognosis and clinical sensitivity to agents that damage the DNA in pancreatic cancer.

15.
Cancer Biol Ther ; 19(8): 645-648, 2018 08 03.
Article in English | MEDLINE | ID: mdl-29565707

ABSTRACT

The growing use of genomic testing presents new treatment options but also new dilemmas. We describe here a heavily-pretreated metastatic triple negative breast cancer patient who failed to respond to conventional treatment. Genomic analyses were performed that discovered several targetable alterations (e.g. FGFR1, CDK6, INSR) and created a clinical challenge - which target to target first? Our solution to this relatively common scenario was using ex-vivo organ culture (EVOC) system to prioritize treatment directed toward the best molecular target. EVOC enabled the trial of several potent targeted agents (Everolimus, Linsitinib, Palbociclib, AZD4547) and allowed semi-quantitative measurement of tumor response. The best response was to FGFR inhibitor, AZD4547. Consequently, the most accessible FGFR inhibiting agents (Pazopanib, then Nintedanib) were administered and some response was achieved. This report provides a potential rationale for utilizing EVOC system to predict tumor response to targeted therapy when multiple targets are proposed.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor , Organ Culture Techniques , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Drug Screening Assays, Antitumor/methods , Female , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Immunohistochemistry , Mice , Middle Aged , Molecular Targeted Therapy , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
16.
Cancer Metastasis Rev ; 37(1): 197, 2018 03.
Article in English | MEDLINE | ID: mdl-29260351

ABSTRACT

The original version of this article unfortunately contained a mistake. The family name of Beatrice Uziely was mistakenly spelled as Uzieky. The correct name is now presented above.

17.
Cancer Metastasis Rev ; 37(1): 147-157, 2018 03.
Article in English | MEDLINE | ID: mdl-29222765

ABSTRACT

G protein-coupled receptors (GPCRs) comprise the main signal-transmitting components in the cell membrane. Over the past several years, biochemical and structural analyses have immensely enhanced our knowledge of GPCR involvement in health and disease states. The present review focuses on GPCRs that are cancer drivers, involved in tumor growth and development. Our aim is to highlight the involvement of stabilized ß-catenin molecular machinery with a specific array of GPCRs. We discuss recent advances in understanding the molecular path leading to ß-catenin nuclear localization and transcriptional activity and their implications for future cancer therapy research.


Subject(s)
Neoplasms/metabolism , Receptors, G-Protein-Coupled/metabolism , beta Catenin/metabolism , Animals , Endothelins/metabolism , Hippo Signaling Pathway , Humans , Neoplasms/genetics , Protein Binding , Protein Interaction Domains and Motifs , Protein Serine-Threonine Kinases/metabolism , Protein Stability , Protein Transport , Receptor, Parathyroid Hormone, Type 1/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Receptors, Prostaglandin/metabolism , Signal Transduction , Transcription, Genetic , Wnt Signaling Pathway
18.
Oncotarget ; 8(24): 38650-38667, 2017 Jun 13.
Article in English | MEDLINE | ID: mdl-28418856

ABSTRACT

Protease-activated receptor-2 (PAR2) plays a central role in cancer; however, the molecular machinery of PAR2-instigated tumors remains to be elucidated. We show that PAR2 is a potent inducer of ß-catenin stabilization, a core process in cancer biology, leading to its transcriptional activity. Novel association of low-density lipoprotein-related protein 6 (LRP6), a known coreceptor of Frizzleds (Fz), with PAR2 takes place following PAR2 activation. The association between PAR2 and LRP6 was demonstrated employing co-immunoprecipitation, bioluminescence resonance energy transfer (BRET), and confocal microscopy analysis. The association was further supported by ZDOCK protein-protein server. PAR2-LRP6 interaction promotes rapid phosphorylation of LRP6, which results in the recruitment of Axin. Confocal microscopy of PAR2-driven mammary gland tumors in vivo, as well as in vitro confirms the association between PAR2 and LRP6. Indeed, shRNA silencing of LRP6 potently inhibits PAR2-induced ß-catenin stabilization, demonstrating its critical role in the induced path. We have previously shown a novel link between protease-activated receptor-1 (PAR1) and ß-catenin stabilization, both in a transgenic (tg) mouse model with overexpression of human PAR1 (hPar1) in the mammary glands, and in cancer epithelial cell lines. Unlike in PAR1-Gα13 axis, both Gα12 and Gα13 are equally involved in PAR2-induced ß-catenin stabilization. Disheveled (DVL) is translocated to the cell nucleus through the DVL-PDZ domain. Collectively, our data demonstrate a novel PAR2-LRP6-Axin interaction as a key axis of PAR2-induced ß-catenin stabilization in cancer. This newly described axis enhances our understanding of cancer biology, and opens new avenues for future development of anti-cancer therapies.


Subject(s)
Biomarkers, Tumor/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Receptors, G-Protein-Coupled/metabolism , beta Catenin/chemistry , Amino Acid Sequence , Apoptosis , Axin Protein/genetics , Axin Protein/metabolism , Biomarkers, Tumor/genetics , Cell Proliferation , Humans , Low Density Lipoprotein Receptor-Related Protein-6/antagonists & inhibitors , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Neoplasms/genetics , Phosphorylation , Protein Conformation , RNA, Small Interfering/genetics , Receptor, PAR-2 , Receptors, G-Protein-Coupled/genetics , Sequence Homology , Signal Transduction , Tumor Cells, Cultured , Wnt Proteins/genetics , Wnt Proteins/metabolism , beta Catenin/genetics , beta Catenin/metabolism
19.
Int J Mol Sci ; 17(8)2016 Aug 12.
Article in English | MEDLINE | ID: mdl-27529230

ABSTRACT

Despite the fact that G protein-coupled receptors (GPCRs) are the largest signal-conveying receptor family and mediate many physiological processes, their role in tumor biology is underappreciated. Numerous lines of evidence now associate GPCRs and their downstream signaling targets in cancer growth and development. Indeed, GPCRs control many features of tumorigenesis, including immune cell-mediated functions, proliferation, invasion and survival at the secondary site. Technological advances have further substantiated GPCR modifications in human tumors. Among these are point mutations, gene overexpression, GPCR silencing by promoter methylation and the number of gene copies. At this point, it is imperative to elucidate specific signaling pathways of "cancer driver" GPCRs. Emerging data on GPCR biology point to functional selectivity and "biased agonism"; hence, there is a diminishing enthusiasm for the concept of "one drug per GPCR target" and increasing interest in the identification of several drug options. Therefore, determining the appropriate context-dependent conformation of a functional GPCR as well as the contribution of GPCR alterations to cancer development remain significant challenges for the discovery of dominant cancer genes and the development of targeted therapeutics.


Subject(s)
Neoplasms/metabolism , Animals , Humans , Neoplasms/pathology , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology
20.
Proc Natl Acad Sci U S A ; 113(13): E1826-34, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26976580

ABSTRACT

Minimally invasive detection of cell death could prove an invaluable resource in many physiologic and pathologic situations. Cell-free circulating DNA (cfDNA) released from dying cells is emerging as a diagnostic tool for monitoring cancer dynamics and graft failure. However, existing methods rely on differences in DNA sequences in source tissues, so that cell death cannot be identified in tissues with a normal genome. We developed a method of detecting tissue-specific cell death in humans based on tissue-specific methylation patterns in cfDNA. We interrogated tissue-specific methylome databases to identify cell type-specific DNA methylation signatures and developed a method to detect these signatures in mixed DNA samples. We isolated cfDNA from plasma or serum of donors, treated the cfDNA with bisulfite, PCR-amplified the cfDNA, and sequenced it to quantify cfDNA carrying the methylation markers of the cell type of interest. Pancreatic ß-cell DNA was identified in the circulation of patients with recently diagnosed type-1 diabetes and islet-graft recipients; oligodendrocyte DNA was identified in patients with relapsing multiple sclerosis; neuronal/glial DNA was identified in patients after traumatic brain injury or cardiac arrest; and exocrine pancreas DNA was identified in patients with pancreatic cancer or pancreatitis. This proof-of-concept study demonstrates that the tissue origins of cfDNA and thus the rate of death of specific cell types can be determined in humans. The approach can be adapted to identify cfDNA derived from any cell type in the body, offering a minimally invasive window for diagnosing and monitoring a broad spectrum of human pathologies as well as providing a better understanding of normal tissue dynamics.


Subject(s)
DNA Methylation , DNA/blood , Insulin-Secreting Cells/pathology , Oligodendroglia/pathology , Adolescent , Adult , Aged , Brain Ischemia/genetics , Brain Ischemia/pathology , Case-Control Studies , Cell Death , Child , Child, Preschool , DNA/metabolism , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Female , Genetic Markers , Humans , Male , Middle Aged , Multiple Sclerosis, Relapsing-Remitting/genetics , Multiple Sclerosis, Relapsing-Remitting/pathology , Organ Specificity , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatitis, Chronic/genetics , Pancreatitis, Chronic/pathology , Promoter Regions, Genetic , Sensitivity and Specificity , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...