Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters

Publication year range
1.
Nature ; 610(7930): 190-198, 2022 10.
Article in English | MEDLINE | ID: mdl-36131018

ABSTRACT

Although melanoma is notorious for its high degree of heterogeneity and plasticity1,2, the origin and magnitude of cell-state diversity remains poorly understood. Equally, it is unclear whether growth and metastatic dissemination are supported by overlapping or distinct melanoma subpopulations. Here, by combining mouse genetics, single-cell and spatial transcriptomics, lineage tracing and quantitative modelling, we provide evidence of a hierarchical model of tumour growth that mirrors the cellular and molecular logic underlying the cell-fate specification and differentiation of the embryonic neural crest. We show that tumorigenic competence is associated with a spatially localized perivascular niche, a phenotype acquired through an intercellular communication pathway established by endothelial cells. Consistent with a model in which only a fraction of cells are fated to fuel growth, temporal single-cell tracing of a population of melanoma cells with a mesenchymal-like state revealed that these cells do not contribute to primary tumour growth but, instead, constitute a pool of metastatic initiating cells that switch cell identity while disseminating to secondary organs. Our data provide a spatially and temporally resolved map of the diversity and trajectories of melanoma cell states and suggest that the ability to support growth and metastasis are limited to distinct pools of cells. The observation that these phenotypic competencies can be dynamically acquired after exposure to specific niche signals warrant the development of therapeutic strategies that interfere with the cancer cell reprogramming activity of such microenvironmental cues.


Subject(s)
Cell Proliferation , Melanoma , Neoplasm Metastasis , Animals , Cell Communication , Cell Differentiation , Cell Lineage , Cell Tracking , Cellular Reprogramming , Endothelial Cells , Melanoma/genetics , Melanoma/pathology , Mesoderm/pathology , Mice , Neoplasm Metastasis/pathology , Neural Crest/embryology , Phenotype , Single-Cell Analysis , Transcriptome , Tumor Microenvironment
2.
Nat Methods ; 20(9): 1355-1367, 2023 09.
Article in English | MEDLINE | ID: mdl-37443338

ABSTRACT

Joint profiling of chromatin accessibility and gene expression in individual cells provides an opportunity to decipher enhancer-driven gene regulatory networks (GRNs). Here we present a method for the inference of enhancer-driven GRNs, called SCENIC+. SCENIC+ predicts genomic enhancers along with candidate upstream transcription factors (TFs) and links these enhancers to candidate target genes. To improve both recall and precision of TF identification, we curated and clustered a motif collection with more than 30,000 motifs. We benchmarked SCENIC+ on diverse datasets from different species, including human peripheral blood mononuclear cells, ENCODE cell lines, melanoma cell states and Drosophila retinal development. Next, we exploit SCENIC+ predictions to study conserved TFs, enhancers and GRNs between human and mouse cell types in the cerebral cortex. Finally, we use SCENIC+ to study the dynamics of gene regulation along differentiation trajectories and the effect of TF perturbations on cell state. SCENIC+ is available at scenicplus.readthedocs.io .


Subject(s)
Gene Regulatory Networks , Multiomics , Animals , Humans , Mice , Leukocytes, Mononuclear , Gene Expression Regulation , Chromatin/genetics , Drosophila/genetics , Enhancer Elements, Genetic
3.
EMBO J ; 40(10): e106214, 2021 05 17.
Article in English | MEDLINE | ID: mdl-33932034

ABSTRACT

BNIP3 is a mitophagy receptor with context-dependent roles in cancer, but whether and how it modulates melanoma growth in vivo remains unknown. Here, we found that elevated BNIP3 levels correlated with poorer melanoma patient's survival and depletion of BNIP3 in B16-F10 melanoma cells compromised tumor growth in vivo. BNIP3 depletion halted mitophagy and enforced a PHD2-mediated downregulation of HIF-1α and its glycolytic program both in vitro and in vivo. Mechanistically, we found that BNIP3-deprived melanoma cells displayed increased intracellular iron levels caused by heightened NCOA4-mediated ferritinophagy, which fostered PHD2-mediated HIF-1α destabilization. These effects were not phenocopied by ATG5 or NIX silencing. Restoring HIF-1α levels in BNIP3-depleted melanoma cells rescued their metabolic phenotype and tumor growth in vivo, but did not affect NCOA4 turnover, underscoring that these BNIP3 effects are not secondary to HIF-1α. These results unravel an unexpected role of BNIP3 as upstream regulator of the pro-tumorigenic HIF-1α glycolytic program in melanoma cells.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Melanoma/metabolism , Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Cell Line, Tumor , Computational Biology , Female , Gas Chromatography-Mass Spectrometry , Humans , Immunoblotting , Immunohistochemistry , Magnetic Resonance Spectroscopy , Mice , Mice, Inbred C57BL , Signal Transduction/genetics , Signal Transduction/physiology
4.
Genome Res ; 31(6): 1082-1096, 2021 06.
Article in English | MEDLINE | ID: mdl-33832990

ABSTRACT

Genomic sequence variation within enhancers and promoters can have a significant impact on the cellular state and phenotype. However, sifting through the millions of candidate variants in a personal genome or a cancer genome, to identify those that impact cis-regulatory function, remains a major challenge. Interpretation of noncoding genome variation benefits from explainable artificial intelligence to predict and interpret the impact of a mutation on gene regulation. Here we generate phased whole genomes with matched chromatin accessibility, histone modifications, and gene expression for 10 melanoma cell lines. We find that training a specialized deep learning model, called DeepMEL2, on melanoma chromatin accessibility data can capture the various regulatory programs of the melanocytic and mesenchymal-like melanoma cell states. This model outperforms motif-based variant scoring, as well as more generic deep learning models. We detect hundreds to thousands of allele-specific chromatin accessibility variants (ASCAVs) in each melanoma genome, of which 15%-20% can be explained by gains or losses of transcription factor binding sites. A considerable fraction of ASCAVs are caused by changes in AP-1 binding, as confirmed by matched ChIP-seq data to identify allele-specific binding of JUN and FOSL1. Finally, by augmenting the DeepMEL2 model with ChIP-seq data for GABPA, the TERT promoter mutation, as well as additional ETS motif gains, can be identified with high confidence. In conclusion, we present a new integrative genomics approach and a deep learning model to identify and interpret functional enhancer mutations with allelic imbalance of chromatin accessibility and gene expression.


Subject(s)
Chromatin , Deep Learning , Alleles , Artificial Intelligence , Chromatin/genetics , Promoter Regions, Genetic
5.
Genome Res ; 30(12): 1815-1834, 2020 12.
Article in English | MEDLINE | ID: mdl-32732264

ABSTRACT

Deciphering the genomic regulatory code of enhancers is a key challenge in biology because this code underlies cellular identity. A better understanding of how enhancers work will improve the interpretation of noncoding genome variation and empower the generation of cell type-specific drivers for gene therapy. Here, we explore the combination of deep learning and cross-species chromatin accessibility profiling to build explainable enhancer models. We apply this strategy to decipher the enhancer code in melanoma, a relevant case study owing to the presence of distinct melanoma cell states. We trained and validated a deep learning model, called DeepMEL, using chromatin accessibility data of 26 melanoma samples across six different species. We show the accuracy of DeepMEL predictions on the CAGI5 challenge, where it significantly outperforms existing models on the melanoma enhancer of IRF4 Next, we exploit DeepMEL to analyze enhancer architectures and identify accurate transcription factor binding sites for the core regulatory complexes in the two different melanoma states, with distinct roles for each transcription factor, in terms of nucleosome displacement or enhancer activation. Finally, DeepMEL identifies orthologous enhancers across distantly related species, where sequence alignment fails, and the model highlights specific nucleotide substitutions that underlie enhancer turnover. DeepMEL can be used from the Kipoi database to predict and optimize candidate enhancers and to prioritize enhancer mutations. In addition, our computational strategy can be applied to other cancer or normal cell types.


Subject(s)
Computational Biology/methods , Melanoma/genetics , Zebrafish/genetics , Animals , Deep Learning , Dogs , Enhancer Elements, Genetic , Gene Expression Regulation, Neoplastic , Horses , Humans , Mice , Swine
7.
J Hepatol ; 76(5): 1001-1012, 2022 05.
Article in English | MEDLINE | ID: mdl-34942286

ABSTRACT

BACKGROUND & AIMS: Obesity-associated inflammation is a key player in the pathogenesis of non-alcoholic fatty liver disease (NAFLD). However, the role of macrophage scavenger receptor 1 (MSR1, CD204) remains incompletely understood. METHODS: A total of 170 NAFLD liver biopsies were processed for transcriptomic analysis and correlated with clinicopathological features. Msr1-/- and wild-type mice were subjected to a 16-week high-fat and high-cholesterol diet. Mice and ex vivo human liver slices were treated with a monoclonal antibody against MSR1. Genetic susceptibility was assessed using genome-wide association study data from 1,483 patients with NAFLD and 430,101 participants of the UK Biobank. RESULTS: MSR1 expression was associated with the occurrence of hepatic lipid-laden foamy macrophages and correlated with the degree of steatosis and steatohepatitis in patients with NAFLD. Mice lacking Msr1 were protected against diet-induced metabolic disorder, showing fewer hepatic foamy macrophages, less hepatic inflammation, improved dyslipidaemia and glucose tolerance, and altered hepatic lipid metabolism. Upon induction by saturated fatty acids, MSR1 induced a pro-inflammatory response via the JNK signalling pathway. In vitro blockade of the receptor prevented the accumulation of lipids in primary macrophages which inhibited the switch towards a pro-inflammatory phenotype and the release of cytokines such as TNF-ɑ. Targeting MSR1 using monoclonal antibody therapy in an obesity-associated NAFLD mouse model and human liver slices resulted in the prevention of foamy macrophage formation and inflammation. Moreover, we identified that rs41505344, a polymorphism in the upstream transcriptional region of MSR1, was associated with altered serum triglycerides and aspartate aminotransferase levels in a cohort of over 400,000 patients. CONCLUSIONS: Taken together, our data suggest that MSR1 plays a critical role in lipid-induced inflammation and could thus be a potential therapeutic target for the treatment of NAFLD. LAY SUMMARY: Non-alcoholic fatty liver disease (NAFLD) is a chronic disease primarily caused by excessive consumption of fat and sugar combined with a lack of exercise or a sedentary lifestyle. Herein, we show that the macrophage scavenger receptor MSR1, an innate immune receptor, mediates lipid uptake and accumulation in Kupffer cells, resulting in liver inflammation and thereby promoting the progression of NAFLD in humans and mice.


Subject(s)
Non-alcoholic Fatty Liver Disease , Animals , Antibodies, Monoclonal , Diet, High-Fat/adverse effects , Genome-Wide Association Study , Humans , Inflammation/metabolism , Lipids , Liver/pathology , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/metabolism
8.
Nat Methods ; 16(5): 397-400, 2019 05.
Article in English | MEDLINE | ID: mdl-30962623

ABSTRACT

We present cisTopic, a probabilistic framework used to simultaneously discover coaccessible enhancers and stable cell states from sparse single-cell epigenomics data ( http://github.com/aertslab/cistopic ). Using a compendium of single-cell ATAC-seq datasets from differentiating hematopoietic cells, brain and transcription factor perturbations, we demonstrate that topic modeling can be exploited for robust identification of cell types, enhancers and relevant transcription factors. cisTopic provides insight into the mechanisms underlying regulatory heterogeneity in cell populations.


Subject(s)
Epigenomics/methods , Gene Expression Profiling/methods , Models, Theoretical , Single-Cell Analysis/methods , Animals , Blood Cells/metabolism , Brain/metabolism , Cells, Cultured , Cluster Analysis , Gene Regulatory Networks/genetics , Humans , Mice , Regulatory Sequences, Nucleic Acid/genetics , Sequence Analysis, RNA , Workflow
9.
Nature ; 531(7595): 518-22, 2016 Mar 24.
Article in English | MEDLINE | ID: mdl-27008969

ABSTRACT

Focal amplifications of chromosome 3p13-3p14 occur in about 10% of melanomas and are associated with a poor prognosis. The melanoma-specific oncogene MITF resides at the epicentre of this amplicon. However, whether other loci present in this amplicon also contribute to melanomagenesis is unknown. Here we show that the recently annotated long non-coding RNA (lncRNA) gene SAMMSON is consistently co-gained with MITF. In addition, SAMMSON is a target of the lineage-specific transcription factor SOX10 and its expression is detectable in more than 90% of human melanomas. Whereas exogenous SAMMSON increases the clonogenic potential in trans, SAMMSON knockdown drastically decreases the viability of melanoma cells irrespective of their transcriptional cell state and BRAF, NRAS or TP53 mutational status. Moreover, SAMMSON targeting sensitizes melanoma to MAPK-targeting therapeutics both in vitro and in patient-derived xenograft models. Mechanistically, SAMMSON interacts with p32, a master regulator of mitochondrial homeostasis and metabolism, to increase its mitochondrial targeting and pro-oncogenic function. Our results indicate that silencing of the lineage addiction oncogene SAMMSON disrupts vital mitochondrial functions in a cancer-cell-specific manner; this silencing is therefore expected to deliver highly effective and tissue-restricted anti-melanoma therapeutic responses.


Subject(s)
Melanoma/genetics , Melanoma/pathology , Oncogenes/genetics , RNA, Long Noncoding/genetics , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Carrier Proteins , Cell Lineage , Cell Proliferation , Cell Survival , Chromosomes, Human, Pair 3/genetics , Clone Cells/metabolism , Clone Cells/pathology , Female , Gene Amplification/genetics , Gene Knockdown Techniques , Humans , Melanoma/therapy , Mice , Microphthalmia-Associated Transcription Factor/genetics , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Proteins/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Molecular Targeted Therapy , RNA, Long Noncoding/therapeutic use , SOXE Transcription Factors/metabolism , Xenograft Model Antitumor Assays
10.
Nat Methods ; 14(11): 1083-1086, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28991892

ABSTRACT

We present SCENIC, a computational method for simultaneous gene regulatory network reconstruction and cell-state identification from single-cell RNA-seq data (http://scenic.aertslab.org). On a compendium of single-cell data from tumors and brain, we demonstrate that cis-regulatory analysis can be exploited to guide the identification of transcription factors and cell states. SCENIC provides critical biological insights into the mechanisms driving cellular heterogeneity.


Subject(s)
Gene Regulatory Networks , Single-Cell Analysis , Algorithms , Animals , Brain/metabolism , Cluster Analysis , Gene Expression Profiling , Humans , Mice
11.
J Pathol ; 248(1): 66-76, 2019 05.
Article in English | MEDLINE | ID: mdl-30584802

ABSTRACT

Ductular reaction (DR) represents the activation of hepatic progenitor cells (HPCs) and has been associated with features of advanced chronic liver disease; yet it is not clear whether these cells contribute to disease progression and how the composition of their micro-environment differs depending on the aetiology. This study aimed to identify HPC-associated signalling pathways relevant in different chronic liver diseases using a high-throughput sequencing approach. DR/HPCs were isolated using laser microdissection from patient samples diagnosed with HCV or primary sclerosing cholangitis (PSC), as models for hepatocellular or biliary regeneration. Key signals were validated at the protein level for a cohort of 56 patients (20 early and 36 advanced stage). In total, 330 genes were significantly differentially expressed between the HPCs in HCV and PSC. Recruitment and homing of inflammatory cells were distinctly different depending on the aetiology. HPCs in PSC were characterised by a response to oxidative stress (e.g. JUN, VNN1) and neutrophil-attractant chemokines (CXCL5, CXCL6, IL-8), whereas HPCs in HCV were identified by T- and B-lymphocyte infiltration. Moreover, we found that communication between HPCs and macrophages was aetiology driven. In PSC, a high frequency of CCL28-positive macrophages was observed in the portal infiltrate, already in early disease in the absence of advanced fibrosis, while in HCV, HPCs showed a strong expression of the macrophage scavenger receptor MARCO. Interestingly, DR/HPCs in PSC showed more deposition of ECM (e.g. FN1, LAMC2, collagens) compared to HCV, where an increase of pro-invasive genes (e.g. PDGFRA, IGF2) was observed. Additionally, endothelial cells in the vicinity of DR/HPCs showed differential immunopositivity (e.g. IGF2 and INHBA expression). In conclusion, our data shine light on the role of DR/HPCs in immune signalling, fibrogenesis and angiogenesis in chronic liver disease. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Hepatocytes/pathology , Liver Diseases/pathology , Stem Cells/pathology , Cholangitis, Sclerosing/genetics , Cholangitis, Sclerosing/immunology , Cholangitis, Sclerosing/pathology , Chronic Disease , Extracellular Matrix/genetics , Extracellular Matrix/pathology , Gene Expression Regulation/physiology , Hepatitis C, Chronic/genetics , Hepatitis C, Chronic/immunology , Hepatitis C, Chronic/pathology , High-Throughput Nucleotide Sequencing/methods , Humans , Liver Diseases/genetics , Liver Diseases/immunology , Neovascularization, Pathologic/pathology , Regulatory Elements, Transcriptional/physiology , Signal Transduction , Stem Cell Niche
12.
Br J Cancer ; 116(1): 58-65, 2017 01 03.
Article in English | MEDLINE | ID: mdl-27884016

ABSTRACT

BACKGROUND: Tumour budding, described as the presence of single cells or small clusters of up to five tumour cells at the invasive margin, is established as a prognostic marker in colorectal carcinoma. In the present study, we aimed to investigate the molecular signature of tumour budding cells and the corresponding tumour bulk. METHODS: Tumour bulk and budding areas were microdissected and processed for RNA-sequencing. As little RNA was obtained from budding cells, a special low-input mRNA library preparation protocol was used. Gene expression profiles of budding as compared with tumour bulk were investigated for established EMT signatures, consensus molecular subtype (CMS), gene set enrichment and pathway analysis. RESULTS: A total of 296 genes were differentially expressed with an FDR <0.05 and a twofold change between tumour bulk and budding regions. Genes that were upregulated in the budding signature were mainly involved in cell migration and survival while downregulated genes were important for cell proliferation. Supervised clustering according to an established EMT gene signature categorised budding regions as EMT-positive, whereas tumour bulk was considered EMT-negative. Furthermore, a shift from CMS2 (epithelial) to CMS4 (mesenchymal) was observed as tumour cells transit from the tumour bulk to the budding regions. CONCLUSIONS: Tumour budding regions are characterised by a phenotype switch compared with the tumour bulk, involving the acquisition of migratory characteristics and a decrease in cell proliferation. In particular, most tumour budding signatures were EMT-positive and switched from an epithelial subtype (CMS2) in the tumour bulk to a mesenchymal subtype (CMS4) in budding cells.


Subject(s)
Cell Division/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition/genetics , Genes, Switch/genetics , Transcriptome , Adult , Aged , Aged, 80 and over , Cell Proliferation/genetics , Colorectal Neoplasms/metabolism , Female , Gene Expression Profiling , Humans , Male , Margins of Excision , Middle Aged , Neoplasm Invasiveness , Phenotype , Tissue Array Analysis
13.
BMC Med ; 15(1): 101, 2017 06 05.
Article in English | MEDLINE | ID: mdl-28578692

ABSTRACT

BACKGROUND: Cutaneous melanoma is the deadliest skin cancer, with an increasing incidence and mortality rate. Currently, staging of patients with primary melanoma is performed using histological biomarkers such as tumor thickness and ulceration. As disruption of the epigenomic landscape is recognized as a widespread feature inherent in tumor development and progression, we aimed to identify novel biomarkers providing additional clinical information over current factors using unbiased genome-wide DNA methylation analyses. METHODS: We performed a comprehensive DNA methylation analysis during all progression stages of melanoma using Infinium HumanMethylation450 BeadChips on a discovery cohort of benign nevi (n = 14) and malignant melanoma from both primary (n = 33) and metastatic (n = 28) sites, integrating the DNA methylome with gene expression data. We validated the discovered biomarkers in three independent validation cohorts by pyrosequencing and immunohistochemistry. RESULTS: We identified and validated biomarkers for, and pathways involved in, melanoma development (e.g., HOXA9 DNA methylation) and tumor progression (e.g., TBC1D16 DNA methylation). In addition, we determined a prognostic signature with potential clinical applicability and validated PON3 DNA methylation and OVOL1 protein expression as biomarkers with prognostic information independent of tumor thickness and ulceration. CONCLUSIONS: Our data underscores the importance of epigenomic regulation in triggering metastatic dissemination through the inactivation of central cancer-related pathways. Inactivation of cell-adhesion and differentiation unleashes dissemination, and subsequent activation of inflammatory and immune system programs impairs anti-tumoral defense pathways. Moreover, we identify several markers of tumor development and progression previously unrelated to melanoma, and determined a prognostic signature with potential clinical utility.


Subject(s)
DNA Methylation , DNA, Neoplasm/metabolism , Melanoma/genetics , Melanoma/physiopathology , Skin Neoplasms/genetics , Skin Neoplasms/physiopathology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Disease Progression , Female , Humans , Male , Middle Aged , Prognosis , Melanoma, Cutaneous Malignant
14.
J Hepatol ; 64(3): 609-17, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26592953

ABSTRACT

BACKGROUND & AIMS: Cancer stem cells (CSCs) are thought to be persistent in tumours due to their chemoresistance and to cause relapse and metastasis. Hepatic carcinomas displaying hepatic progenitor cell (HPC) features have been associated with a poor prognosis, though it remains unclear how CSCs relate to these different histological subtypes. METHODS: Candidate CSCs were isolated using the side population (SP) technique from primary tissue samples diagnosed as keratin(K)19-negative or -positive hepatocellular carcinoma (HCC) or as combined hepatocellular/cholangiocarcinoma and analysed for gene and protein expression. The effect of laminin-332 was analysed in vitro by using HCC cell lines and in vivo using a xenograft mouse model. RESULTS: The size of the SP correlated with the degree of HPC features found in human hepatic cancer, and also showed an elevated mRNA expression of biliary/HPC markers and the extracellular matrix marker LAMC2, the gene encoding the laminin γ2-chain. Immunopositivity for the γ2-chain of laminin-332 was seen in the extracellular matrix surrounding small HPC-like tumour cells with a low proliferation rate. In vitro, laminin-332 increased K19 expression, phosphorylated mTOR and decreased phospho-histone H3 expression, indicating reduced cell mitosis. The effect of laminin-332 was enhanced upon mTORC1 inhibition and diminished when inhibiting mTORC1+C2. Resistance to doxorubicin and sorafenib treatment, and the SP fraction increased in the coated condition. In vivo, laminin-332 reduced tumour growth and sustained K19 expression. CONCLUSIONS: In this study we identified a prominent role for laminin-332 as part of the specialised CSC niche in maintaining and supporting cell 'stemness', which leads to chemoresistance and quiescence.


Subject(s)
Cell Adhesion Molecules/pharmacology , Liver Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Stem Cell Niche/drug effects , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Keratin-19/analysis , Liver Neoplasms/chemistry , Liver Neoplasms/pathology , Mice , Neoplastic Stem Cells/chemistry , TOR Serine-Threonine Kinases/physiology , Kalinin
15.
Mod Pathol ; 29(4): 347-58, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26867783

ABSTRACT

Melanoma is not only one of the most immunogenic cancers but also one of the most effective cancers at subverting host immunity. The role of T lymphocytes in tumor immunity has been extensively studied in melanoma, whereas less is known about the importance of B lymphocytes. The effects of plasma cells (PCs), in particular, are still obscure. The aim of this study was to characterize pathological features and clinical outcome of primary cutaneous melanomas associated with PCs. Moreover, we investigated the origins of the melanoma-associated PCs. Finally, we studied the outcome of patients with primary melanomas with PCs. We reviewed 710 melanomas to correlate the presence of PCs with histological prognostic markers. Immunohistochemistry for CD138 and heavy and light chains was performed in primary melanomas (PM) and in loco-regional lymph nodes (LN), both metastatic and not metastatic. In three PM and nine LN with frozen material, VDJ-rearrangement was analyzed by Gene Scan Analysis. Survival analysis was performed on a group of 85 primary melanomas >2 mm in thickness. Forty-one cases (3.7%) showed clusters/sheets of PCs. PC-rich melanomas occurred at an older age and were thicker, more often ulcerated and more mitotically active (P<0.05). PCs were polyclonal and often expressed IgA in addition to IgG. In LN, clusters/sheets of IgA+ PCs were found both in the sinuses and subcapsular areas. Analysis of VDJ-rearrangements showed the IgA to be oligoclonal. Melanomas with clusters/sheets of PCs had a significantly worse survival compared with melanomas without PCs while, interestingly, melanomas with sparse PCs were associated with a better clinical outcome (P=0.002). In conclusion, melanomas with sheets/clusters of PCs are associated with worse prognosis. IgG and IgA are the isotypes predominantly produced by these PCs. IgA oligoclonality suggests an antigen-driven response that facilitates melanoma progression by a hitherto unknown mechanism.


Subject(s)
Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/immunology , Plasma Cells/immunology , Skin Neoplasms/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Child , Female , Humans , Immunoglobulin A , Immunohistochemistry , Kaplan-Meier Estimate , Male , Melanoma/mortality , Melanoma/pathology , Middle Aged , Prognosis , Reverse Transcriptase Polymerase Chain Reaction , Skin Neoplasms/mortality , Skin Neoplasms/pathology , Young Adult
16.
Oncologist ; 20(10): 1121-2, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26330458

ABSTRACT

BACKGROUND: This first-in-human proof-of-concept study aimed to check whether safety and preclinical results obtained by intratumoral administration of BQ788, an endothelin receptor B (EDNRB) antagonist, can be repeated in human melanoma patients. METHODS: Three patients received a single intralesional BQ788 application of 3 mg. After 3-7 days, the lesions were measured and removed for analysis. The administered dose was increased to a cumulative dosage of 8 mg in patient 4 (4 × 2.0 mg, days 0-3; lesion removed on day 4) and to 10 mg in patient 5 (3 × 3.3 mg, days 0, 3, and 10; lesion removed after 14 days). Control lesions were simultaneously treated with phosphate-buffered saline (PBS). All samples were processed and analyzed without knowledge of the clinical findings. RESULTS: No statistical evaluation was possible because of the number of patients (n = 5) and the variability in the mode of administration. No adverse events were observed, regardless of administered dose. All observations were in accordance with results obtained in preclinical studies. Accordingly, no difference in degree of tumor necrosis was detected between BQ788- and PBS-treated samples. In addition, both EDNRB and Ki67 showed decreased expression in patients 2 and 5 and, to a lesser extent, in patient 1. Similarly, decreased expression of EDNRB mRNA in patients 2 and 5 and of BCL2A1 and/or PARP3 in patients 2, 3, and 5 was found. Importantly, semiquantitatively scored immunohistochemistry for CD31 and CD3 revealed more blood vessels and lymphocytes, respectively, in BQ788-treated tumors of patients 2 and 4. Also, in all patients, we observed inverse correlation in expression levels between EDNRB and HIF1A. Finally, in patient 5 (the only patient treated for longer than 1 week), we observed inhibition in lesion growth, as shown by size measurement. CONCLUSION: The intralesional applications of BQ788 were well tolerated and showed signs of directly and indirectly reducing the viability of melanoma cells.


Subject(s)
Antineoplastic Agents/administration & dosage , Endothelin B Receptor Antagonists/administration & dosage , Melanoma/drug therapy , Oligopeptides/administration & dosage , Piperidines/administration & dosage , Skin Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Cell Cycle Proteins/genetics , Drug Administration Schedule , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Injections, Intralesional , Male , Melanoma/secondary , Middle Aged , Minor Histocompatibility Antigens , Poly(ADP-ribose) Polymerases/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Receptor, Endothelin B/genetics , Receptor, Endothelin B/metabolism , Skin Neoplasms/secondary , Treatment Outcome
17.
Gut ; 63(4): 674-85, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23958557

ABSTRACT

OBJECTIVE: Keratin (K)19, a biliary/hepatic progenitor cell (HPC) marker, is expressed in a subset of hepatocellular carcinomas (HCC) with poor prognosis. The underlying mechanisms driving this phenotype of K19-positive HCC remain elusive. DESIGN: Clinicopathological value of K19 was compared with EpCAM, and α-fetoprotein, in a Caucasian cohort of 242 consecutive patients (167 surgical specimens, 75 needle biopsies) with different underlying aetiologies. Using microarrays and microRNA profiling the molecular phenotype of K19-positive HCCs was identified. Clinical primary HCC samples were submitted to in vitro invasion assays and to side population analysis. HCC cell lines were transfected with synthetic siRNAs against KRT19 and submitted to invasion and cytotoxicity assays. RESULTS: In the cohort of surgical specimens, K19 expression showed the strongest correlation with increased tumour size (p<0.01), decreased tumour differentiation (p<0.001), metastasis (p<0.05) and microvascular invasion (p<0.001). The prognostic value of K19 was also confirmed in a set of 75 needle biopsies. Profiling showed that K19-positive HCCs highly express invasion-related/metastasis-related markers (eg, VASP, TACSTD2, LAMB1, LAMC2, PDGFRA), biliary/HPC markers (eg, CD133, GSTP1, NOTCH2, JAG1) and members of the miRNA family 200 (eg, miR-141, miR-200c). In vitro, primary human K19-positive tumour cells showed increased invasiveness, and reside in the chemoresistant side population. Functionally, K19/KRT19 knockdown results in reduced invasion, loss of invadopodia formation and decreased resistance to doxorubicin, 5-fluorouracil and sorafenib. CONCLUSIONS: Giving the distinct invasive properties, the different molecular profile and the poor prognostic outcome, K19-positive HCCs should be considered as a seperate entity of HCCs.


Subject(s)
Carcinoma, Hepatocellular/physiopathology , Keratin-19/physiology , Liver Neoplasms/physiopathology , Antigens, Neoplasm/physiology , Biomarkers/analysis , Carcinoma, Hepatocellular/chemistry , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/pathology , Cell Adhesion Molecules/physiology , Cell Line, Tumor , Epithelial Cell Adhesion Molecule , Gene Knockdown Techniques , Humans , Keratin-19/analysis , Liver Neoplasms/chemistry , Liver Neoplasms/diagnosis , Liver Neoplasms/pathology , Neoplasm Invasiveness/physiopathology , Oligonucleotide Array Sequence Analysis , Prognosis , Real-Time Polymerase Chain Reaction , alpha-Fetoproteins/physiology
18.
Mod Pathol ; 27(8): 1088-100, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24390218

ABSTRACT

In many human cancers, the epithelial-to-mesenchymal transition has an important role in the induction of cancer stem-like cells, and hence, in the causation of intratumoral heterogeneity. This process, also referred to as mesenchymal mimicry, is, however, only poorly understood in melanoma and histological correlation is still lacking. In an immunohistochemical analysis of a large prospective series of 220 primary and metastatic melanomas for the well-known epithelial-to-mesenchymal transition marker FN1, we observed melanoma cells with high FN1 expression in metastases with ischemic necrosis, but rarely or not at all in samples lacking evidence of hypoxia. In a blinded, retrospective series of 82 melanoma metastases with 10-year follow-up, the presence of clusters of these FN1(high) melanoma cells correlated significantly with shortened melanoma-specific survival, highlighting the prognostic value of their presence. We describe in detail the unique light- and electron-microscopic features of these FN1(high) melanoma cells, enabling their identification in routinely hematoxylin-and-eosin-stained sections. In addition, by laser microdissection and subsequent gene expression analysis and immunohistochemistry, we highlight their distinctive, molecular phenotype that includes expression of various markers of the epithelial-to-mesenchymal transition (eg, ZEB1) and of melanoma stem-like cells (eg, NGFR), and lack of immunoreactivity for the melanocytic marker MITF. This phenotype could be reproduced in vitro by culturing melanoma cells under hypoxic conditions. Functionally, the hypoxic microenvironment was shown to induce a more migratory and invasive cell type. In conclusion, we identified a novel clinically relevant FN1(high)MITF(low) cell type in melanoma associated with ischemic necrosis, and propose that these cells reside at the crossroad of the epithelial-to-mesenchymal transition and stem-like cell induction, plausibly triggered by the hypoxic environment.


Subject(s)
Biomarkers, Tumor/metabolism , Fibronectins/metabolism , Melanoma/metabolism , Microphthalmia-Associated Transcription Factor/metabolism , Neoplastic Stem Cells/metabolism , Skin Neoplasms/metabolism , Tumor Microenvironment , Biomarkers, Tumor/genetics , Cell Hypoxia , Cell Line, Tumor , Cell Movement , Epithelial-Mesenchymal Transition , Fibronectins/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Laser Capture Microdissection , Melanoma/genetics , Melanoma/mortality , Melanoma/secondary , Melanoma/ultrastructure , Microphthalmia-Associated Transcription Factor/genetics , Microscopy, Electron, Transmission , Necrosis , Neoplasm Invasiveness , Neoplastic Stem Cells/pathology , Phenotype , Prognosis , Proportional Hazards Models , Prospective Studies , Retrospective Studies , Skin Neoplasms/genetics , Skin Neoplasms/mortality , Skin Neoplasms/ultrastructure , Time Factors
19.
J Neural Eng ; 19(1)2022 02 09.
Article in English | MEDLINE | ID: mdl-35078163

ABSTRACT

Objective. We present a framework to objectively test and compare stimulation artefact removal techniques in the context of neural spike sorting.Approach. To this end, we used realistic hybrid ground-truth spiking data, with superimposed artefacts fromin vivorecordings. We used the framework to evaluate and compare several techniques: blanking, template subtraction by averaging, linear regression, and a multi-channel Wiener filter (MWF).Main results. Our study demonstrates that blanking and template subtraction result in a poorer spike sorting performance than linear regression and MWF, while the latter two perform similarly. Finally, to validate the conclusions found from the hybrid evaluation framework, we also performed a qualitative analysis onin vivorecordings without artificial manipulations.Significance. Our framework allows direct quantification of the impact of the residual artefact on the spike sorting accuracy, thereby allowing for a more objective and more relevant comparison compared to indirect signal quality metrics that are estimated from the signal statistics. Furthermore, the availability of a ground truth in the form of single-unit spiking activity also facilitates a better estimation of such signal quality metrics.


Subject(s)
Artifacts , Models, Neurological , Action Potentials/physiology , Algorithms , Neurons/physiology , Signal Processing, Computer-Assisted
20.
Elife ; 112022 02 23.
Article in English | MEDLINE | ID: mdl-35195064

ABSTRACT

Single-cell RNA-seq and single-cell assay for transposase-accessible chromatin (ATAC-seq) technologies are used extensively to create cell type atlases for a wide range of organisms, tissues, and disease processes. To increase the scale of these atlases, lower the cost and pave the way for more specialized multiome assays, custom droplet microfluidics may provide solutions complementary to commercial setups. We developed HyDrop, a flexible and open-source droplet microfluidic platform encompassing three protocols. The first protocol involves creating dissolvable hydrogel beads with custom oligos that can be released in the droplets. In the second protocol, we demonstrate the use of these beads for HyDrop-ATAC, a low-cost noncommercial scATAC-seq protocol in droplets. After validating HyDrop-ATAC, we applied it to flash-frozen mouse cortex and generated 7996 high-quality single-cell chromatin accessibility profiles in a single run. In the third protocol, we adapt both the reaction chemistry and the capture sequence of the barcoded hydrogel bead to capture mRNA, and demonstrate a significant improvement in throughput and sensitivity compared to previous open-source droplet-based scRNA-seq assays (Drop-seq and inDrop). Similarly, we applied HyDrop-RNA to flash-frozen mouse cortex and generated 9508 single-cell transcriptomes closely matching reference single-cell gene expression data. Finally, we leveraged HyDrop-RNA's high capture rate to analyze a small population of fluorescence-activated cell sorted neurons from the Drosophila brain, confirming the protocol's applicability to low input samples and small cells. HyDrop is currently capable of generating single-cell data in high throughput and at a reduced cost compared to commercial methods, and we envision that HyDrop can be further developed to be compatible with novel (multi) omics protocols.


Scientists are now able to determine the order of chemical blocks, or nucleic acids, that make up the genetic code. These sequencing tools can be used to identify which genes are active within a biological sample. They do this by extracting and analysing open chromatin (regions of DNA that are accessible to the cell's machinery), or sequences of RNA (the molecular templates cells use to translate genes into working proteins). Initially, most sequencing tools could only provide an 'averaged-out' profile of the genes activated in bulk pieces of tissue which contain multiple types of cell. However, advances in technology have led to new methods that can extract and analyse open chromatin or RNA from individual cells. First, the cells are separated, via a technique called microfluidics, into tiny droplets of water along with a single bead that carries a unique barcode. The cell is then broken apart inside the droplet and the barcode within the bead gets released and attaches itself to the genetic material extracted from the cell. All the genetic material inside the droplets is then pooled together and sequenced. Researchers then use the barcode tags to identify which bits of RNA or DNA belong to each cell. Single-cell sequencing has many advantages, including being able to pinpoint precise genetic differences between healthy and abnormal cells, and to create cell atlases of whole organisms, tissues and microbial communities. But existing methods for extracting chromatin are very expensive, and there were no openly available tools for processing thousands of cells at speed. Furthermore, while several single-cell RNA sequencing tools are already freely available, they are not very sensitive or practical to use. Here, De Rop et al. have developed a new open-source platform called HyDrop that overcomes these barriers. The method entails a new type of barcoded bead and optimised elements of existing microfluidics protocols using open-source reagents. These changes created a more user-friendly workflow and increased sensitivity of sequencing at no additional cost. De Rop et al. used their new platform to screen the RNA and open chromatin of thousands of individuals cells from the brains of mice and flies. HyDrop outperformed other open-source methods when working in RNA-sequencing mode. It also provides the first open-source tool for sequencing open chromatin in single cells. Further improvements are expected as researchers tweak the platform, which for now provides an affordable alternative to existing methods.


Subject(s)
Chromatin Immunoprecipitation Sequencing , High-Throughput Nucleotide Sequencing , Animals , Chromatin , High-Throughput Nucleotide Sequencing/methods , Hydrogels , Mice , RNA , RNA-Seq , Single-Cell Analysis
SELECTION OF CITATIONS
SEARCH DETAIL