Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Cell ; 167(2): 405-418.e13, 2016 Oct 06.
Article in English | MEDLINE | ID: mdl-27693350

ABSTRACT

The HVEM (TNFRSF14) receptor gene is among the most frequently mutated genes in germinal center lymphomas. We report that loss of HVEM leads to cell-autonomous activation of B cell proliferation and drives the development of GC lymphomas in vivo. HVEM-deficient lymphoma B cells also induce a tumor-supportive microenvironment marked by exacerbated lymphoid stroma activation and increased recruitment of T follicular helper (TFH) cells. These changes result from the disruption of inhibitory cell-cell interactions between the HVEM and BTLA (B and T lymphocyte attenuator) receptors. Accordingly, administration of the HVEM ectodomain protein (solHVEM(P37-V202)) binds BTLA and restores tumor suppression. To deliver solHVEM to lymphomas in vivo, we engineered CD19-targeted chimeric antigen receptor (CAR) T cells that produce solHVEM locally and continuously. These modified CAR-T cells show enhanced therapeutic activity against xenografted lymphomas. Hence, the HVEM-BTLA axis opposes lymphoma development, and our study illustrates the use of CAR-T cells as "micro-pharmacies" able to deliver an anti-cancer protein.


Subject(s)
Adoptive Transfer/methods , Lymphoma, Follicular/therapy , Receptors, Immunologic/metabolism , Receptors, Tumor Necrosis Factor, Member 14/genetics , T-Lymphocytes/immunology , Tumor Suppressor Proteins/genetics , Animals , Antigens, CD19/immunology , B-Lymphocytes/immunology , Cell Proliferation , Humans , Lymphocyte Activation , Lymphoma, Follicular/genetics , Mice , Neoplasms, Experimental/genetics , Neoplasms, Experimental/therapy , Protein Domains , Protein Engineering , Receptors, Tumor Necrosis Factor, Member 14/chemistry , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tumor Microenvironment , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism , Xenograft Model Antitumor Assays
2.
Nature ; 615(7954): 925-933, 2023 03.
Article in English | MEDLINE | ID: mdl-36922594

ABSTRACT

Whole-genome doubling (WGD) is a recurrent event in human cancers and it promotes chromosomal instability and acquisition of aneuploidies1-8. However, the three-dimensional organization of chromatin in WGD cells and its contribution to oncogenic phenotypes are currently unknown. Here we show that in p53-deficient cells, WGD induces loss of chromatin segregation (LCS). This event is characterized by reduced segregation between short and long chromosomes, A and B subcompartments and adjacent chromatin domains. LCS is driven by the downregulation of CTCF and H3K9me3 in cells that bypassed activation of the tetraploid checkpoint. Longitudinal analyses revealed that LCS primes genomic regions for subcompartment repositioning in WGD cells. This results in chromatin and epigenetic changes associated with oncogene activation in tumours ensuing from WGD cells. Notably, subcompartment repositioning events were largely independent of chromosomal alterations, which indicates that these were complementary mechanisms contributing to tumour development and progression. Overall, LCS initiates chromatin conformation changes that ultimately result in oncogenic epigenetic and transcriptional modifications, which suggests that chromatin evolution is a hallmark of WGD-driven cancer.


Subject(s)
Chromatin , Chromosome Aberrations , Chromosome Segregation , Chromosomes, Human , Genome, Human , Neoplasms , Humans , Chromatin/genetics , Chromatin/metabolism , Neoplasms/genetics , Chromosomes, Human/genetics , Genome, Human/genetics , Chromosome Segregation/genetics , Carcinogenesis/genetics , Epigenesis, Genetic , Disease Progression , Transcription, Genetic , Gene Expression Regulation, Neoplastic
3.
Nature ; 617(7959): 176-184, 2023 05.
Article in English | MEDLINE | ID: mdl-37100904

ABSTRACT

Physical interactions between proteins are essential for most biological processes governing life1. However, the molecular determinants of such interactions have been challenging to understand, even as genomic, proteomic and structural data increase. This knowledge gap has been a major obstacle for the comprehensive understanding of cellular protein-protein interaction networks and for the de novo design of protein binders that are crucial for synthetic biology and translational applications2-9. Here we use a geometric deep-learning framework operating on protein surfaces that generates fingerprints to describe geometric and chemical features that are critical to drive protein-protein interactions10. We hypothesized that these fingerprints capture the key aspects of molecular recognition that represent a new paradigm in the computational design of novel protein interactions. As a proof of principle, we computationally designed several de novo protein binders to engage four protein targets: SARS-CoV-2 spike, PD-1, PD-L1 and CTLA-4. Several designs were experimentally optimized, whereas others were generated purely in silico, reaching nanomolar affinity with structural and mutational characterization showing highly accurate predictions. Overall, our surface-centric approach captures the physical and chemical determinants of molecular recognition, enabling an approach for the de novo design of protein interactions and, more broadly, of artificial proteins with function.


Subject(s)
Computer Simulation , Deep Learning , Protein Binding , Proteins , Humans , Proteins/chemistry , Proteins/metabolism , Proteomics , Protein Interaction Maps , Binding Sites , Synthetic Biology
4.
Nat Chem Biol ; 2024 May 29.
Article in English | MEDLINE | ID: mdl-38811854

ABSTRACT

Cysteine cathepsins are a family of proteases that are relevant therapeutic targets for the treatment of different cancers and other diseases. However, no clinically approved drugs for these proteins exist, as their systemic inhibition can induce deleterious side effects. To address this problem, we developed a modular antibody-based platform for targeted drug delivery by conjugating non-natural peptide inhibitors (NNPIs) to antibodies. NNPIs were functionalized with reactive warheads for covalent inhibition, optimized with deep saturation mutagenesis and conjugated to antibodies to enable cell-type-specific delivery. Our antibody-peptide inhibitor conjugates specifically blocked the activity of cathepsins in different cancer cells, as well as osteoclasts, and showed therapeutic efficacy in vitro and in vivo. Overall, our approach allows for the rapid design of selective cathepsin inhibitors and can be generalized to inhibit a broad class of proteases in cancer and other diseases.

5.
Cell ; 147(3): 554-64, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-22036564

ABSTRACT

Insights into cancer genetics can lead to therapeutic opportunities. By cross-referencing chromosomal changes with an unbiased genetic screen we identify the ephrin receptor A7 (EPHA7) as a tumor suppressor in follicular lymphoma (FL). EPHA7 is a target of 6q deletions and inactivated in 72% of FLs. Knockdown of EPHA7 drives lymphoma development in a murine FL model. In analogy to its physiological function in brain development, a soluble splice variant of EPHA7 (EPHA7(TR)) interferes with another Eph-receptor and blocks oncogenic signals in lymphoma cells. Consistent with this drug-like activity, administration of the purified EPHA7(TR) protein produces antitumor effects against xenografted human lymphomas. Further, by fusing EPHA7(TR) to the anti-CD20 antibody (rituximab) we can directly target this tumor suppressor to lymphomas in vivo. Our study attests to the power of combining descriptive tumor genomics with functional screens and reveals EPHA7(TR) as tumor suppressor with immediate therapeutic potential.


Subject(s)
Genes, Tumor Suppressor , Lymphoma, Follicular/metabolism , Receptor, EphA7/metabolism , Animals , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Cell Line, Tumor , Chromosomes, Human, Pair 6 , Genomics , Humans , Lymphoma, Follicular/drug therapy , Lymphoma, Follicular/genetics , Male , Mice , Neoplasm Transplantation , RNA Interference , Rituximab , Transplantation, Heterologous
6.
Nat Rev Genet ; 24(7): 419, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36732639
7.
Blood ; 131(21): 2345-2356, 2018 05 24.
Article in English | MEDLINE | ID: mdl-29567799

ABSTRACT

In diffuse large B-cell lymphoma (DLBCL), activation of the B-cell receptor (BCR) promotes multiple oncogenic signals, which are essential for tumor proliferation. Inhibition of the Bruton's tyrosine kinase (BTK), a BCR downstream target, is therapeutically effective only in a subgroup of patients with DLBCL. Here, we used lymphoma cells isolated from patients with DLBCL to measure the effects of targeted therapies on BCR signaling and to anticipate response. In lymphomas resistant to BTK inhibition, we show that blocking BTK activity enhanced tumor dependencies from alternative oncogenic signals downstream of the BCR, converging on MYC upregulation. To completely ablate the activity of the BCR, we genetically and pharmacologically repressed the activity of the SRC kinases LYN, FYN, and BLK, which are responsible for the propagation of the BCR signal. Inhibition of these kinases strongly reduced tumor growth in xenografts and cell lines derived from patients with DLBCL independent of their molecular subtype, advancing the possibility to be relevant therapeutic targets in broad and diverse groups of DLBCL patients.


Subject(s)
Lymphoma, Non-Hodgkin/etiology , Lymphoma, Non-Hodgkin/metabolism , Protein Kinase Inhibitors/pharmacology , Receptors, Antigen, B-Cell/metabolism , Signal Transduction/drug effects , src-Family Kinases/antagonists & inhibitors , Adenine/analogs & derivatives , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Gene Expression , Genes, myc , Humans , Lymphoma, Non-Hodgkin/drug therapy , Lymphoma, Non-Hodgkin/pathology , Mice , Mice, Knockout , Piperidines , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Xenograft Model Antitumor Assays
8.
9.
Growth Factors ; 32(6): 214-22, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25494541

ABSTRACT

Recombinant antibody phage library technology provides multiple advantages, including that human antibodies can be generated against proteins that are highly conserved between species. We used this technology to isolate and characterize an anti-EphA2 single-chain antibody. We show that the antibody binds the antigen with 1:1 stoichiometry and has high specificity for EphA2. The crystal structure of the complex reveals that the antibody targets the same receptor surface cavity as the ephrin ligand. Specifically, a lengthy CDR-H3 loop protrudes deep into the ligand-binding cavity, with several hydrophobic residues at its tip forming an anchor-like structure buried within the hydrophobic Eph pocket, in a way similar to the ephrin receptor-binding loop in the Eph/ephrin structures. Consequently, the antibody blocks ephrin binding to EphA2. Furthermore, it induces apoptosis and reduces cell proliferation in lymphoma cells lines. Since Ephs are important mediators of tumorigenesis, such antibodies could have applications both in research and therapy.


Subject(s)
Receptor, EphA2/immunology , Single-Chain Antibodies/immunology , Amino Acid Sequence , Animals , Antibody Affinity , Binding Sites, Antibody , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , HEK293 Cells , Humans , Molecular Sequence Data , Single-Chain Antibodies/chemistry
10.
Curr Opin Hematol ; 20(4): 320-6, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23673338

ABSTRACT

PURPOSE OF REVIEW: To share the recent progress in research and new therapies against follicular lymphoma and highlight the exciting opportunities to improve the treatment of follicular lymphoma. RECENT FINDINGS: Follicular lymphoma has been somewhat neglected by the research community, but recent genomic studies have identified key genetic lesions in follicular lymphoma. In addition, a new murine model is available to explore the function of these lesions in the development, progression, and treatment of follicular lymphoma. Moreover, new small-molecule inhibitors are now available that target key pathways in follicular lymphoma including B-cell receptor signaling and histone modifiers. SUMMARY: Follicular lymphoma is a very common and still incurable form of lymphoma. However, recent genomic and in-vivo biological studies are beginning to unveil the molecular drivers of follicular lymphoma. This coincides with the development of effective small-molecule inhibitors against key targets. Together these developments suggest that we are at a long overdue watershed moment in the treatment of follicular lymphoma.


Subject(s)
Antineoplastic Agents/therapeutic use , Lymphoma, Follicular/drug therapy , Protein Kinase Inhibitors/therapeutic use , Animals , Disease Models, Animal , Humans , Mice , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors
11.
Cancer Discov ; 14(1): 36-48, 2024 01 12.
Article in English | MEDLINE | ID: mdl-38047596

ABSTRACT

Cancer cells adapt and survive through the acquisition and selection of molecular modifications. This process defines cancer evolution. Building on a theoretical framework based on heritable genetic changes has provided insights into the mechanisms supporting cancer evolution. However, cancer hallmarks also emerge via heritable nongenetic mechanisms, including epigenetic and chromatin topological changes, and interactions between tumor cells and the tumor microenvironment. Recent findings on tumor evolutionary mechanisms draw a multifaceted picture where heterogeneous forces interact and influence each other while shaping tumor progression. A comprehensive characterization of the cancer evolutionary toolkit is required to improve personalized medicine and biomarker discovery. SIGNIFICANCE: Tumor evolution is fueled by multiple enabling mechanisms. Importantly, genetic instability, epigenetic reprogramming, and interactions with the tumor microenvironment are neither alternative nor independent evolutionary mechanisms. As demonstrated by findings highlighted in this perspective, experimental and theoretical approaches must account for multiple evolutionary mechanisms and their interactions to ultimately understand, predict, and steer tumor evolution.


Subject(s)
Neoplasms , Humans , Neoplasms/genetics , Neoplasms/pathology , Epigenomics , Precision Medicine , Tumor Microenvironment/genetics
12.
Mol Cell Oncol ; 8(6): e969640, 2021.
Article in English | MEDLINE | ID: mdl-35419485

ABSTRACT

Follicular lymphoma (FL) is an incurable form of B-cell lymphoma. Genomic alterations that inactivate RB signaling are surprisingly common in indolent FL. We show that FLs that are positive for phosphorylated RB respond to dual CDK4/BCL2 inhibition. Our results imply that RB phosphorylation identifies patients likely to benefit from such dual intervention.

13.
Nat Commun ; 12(1): 2439, 2021 05 10.
Article in English | MEDLINE | ID: mdl-33972523

ABSTRACT

Chromatin compartmentalization reflects biological activity. However, inference of chromatin sub-compartments and compartment domains from chromosome conformation capture (Hi-C) experiments is limited by data resolution. As a result, these have been characterized only in a few cell types and systematic comparisons across multiple tissues and conditions are missing. Here, we present Calder, an algorithmic approach that enables the identification of multi-scale sub-compartments at variable data resolution. Calder allows to infer and compare chromatin sub-compartments and compartment domains in >100 cell lines. Our results reveal sub-compartments enriched for poised chromatin states and undergoing spatial repositioning during lineage differentiation and oncogenic transformation.

14.
Nat Genet ; 53(5): 650-662, 2021 05.
Article in English | MEDLINE | ID: mdl-33972799

ABSTRACT

In cancer cells, enhancer hijacking mediated by chromosomal alterations and/or increased deposition of acetylated histone H3 lysine 27 (H3K27ac) can support oncogene expression. However, how the chromatin conformation of enhancer-promoter interactions is affected by these events is unclear. In the present study, by comparing chromatin structure and H3K27ac levels in normal and lymphoma B cells, we show that enhancer-promoter-interacting regions assume different conformations according to the local abundance of H3K27ac. Genetic or pharmacological depletion of H3K27ac decreases the frequency and the spreading of these interactions, altering oncogene expression. Moreover, enhancer hijacking mediated by chromosomal translocations influences the epigenetic status of the regions flanking the breakpoint, prompting the formation of distinct intrachromosomal interactions in the two homologous chromosomes. These interactions are accompanied by allele-specific gene expression changes. Overall, our work indicates that H3K27ac dynamics modulates interaction frequency between regulatory regions and can lead to allele-specific chromatin configurations to sustain oncogene expression.


Subject(s)
Alleles , Chromatin/chemistry , Genetic Loci , Histones/metabolism , Nucleic Acid Conformation , Oncogenes , Acetylation , Base Pairing/genetics , Cell Line, Tumor , Enhancer Elements, Genetic , Epigenesis, Genetic , Gene Dosage , Humans , Lysine/metabolism , Promoter Regions, Genetic
15.
Cancer Discov ; 11(6): 1490-1507, 2021 06.
Article in English | MEDLINE | ID: mdl-33563664

ABSTRACT

Cancer evolution determines molecular and morphologic intratumor heterogeneity and challenges the design of effective treatments. In lung adenocarcinoma, disease progression and prognosis are associated with the appearance of morphologically diverse tumor regions, termed histologic patterns. However, the link between molecular and histologic features remains elusive. Here, we generated multiomics and spatially resolved molecular profiles of histologic patterns from primary lung adenocarcinoma, which we integrated with molecular data from >2,000 patients. The transition from indolent to aggressive patterns was not driven by genetic alterations but by epigenetic and transcriptional reprogramming reshaping cancer cell identity. A signature quantifying this transition was an independent predictor of patient prognosis in multiple human cohorts. Within individual tumors, highly multiplexed protein spatial profiling revealed coexistence of immune desert, inflamed, and excluded regions, which matched histologic pattern composition. Our results provide a detailed molecular map of lung adenocarcinoma intratumor spatial heterogeneity, tracing nongenetic routes of cancer evolution. SIGNIFICANCE: Lung adenocarcinomas are classified based on histologic pattern prevalence. However, individual tumors exhibit multiple patterns with unknown molecular features. We characterized nongenetic mechanisms underlying intratumor patterns and molecular markers predicting patient prognosis. Intratumor patterns determined diverse immune microenvironments, warranting their study in the context of current immunotherapies.This article is highlighted in the In This Issue feature, p. 1307.


Subject(s)
Adenocarcinoma of Lung/genetics , Lung Neoplasms/genetics , Disease Progression , Genetic Heterogeneity , Humans , Tumor Microenvironment
16.
Cancer Cell ; 37(5): 674-689.e12, 2020 05 11.
Article in English | MEDLINE | ID: mdl-32330455

ABSTRACT

Genomic alterations in cancer cells can influence the immune system to favor tumor growth. In non-Hodgkin lymphoma, physiological interactions between B cells and the germinal center microenvironment are coopted to sustain cancer cell proliferation. We found that follicular lymphoma patients harbor a recurrent hotspot mutation targeting tyrosine 132 (Y132D) in cathepsin S (CTSS) that enhances protein activity. CTSS regulates antigen processing and CD4+ and CD8+ T cell-mediated immune responses. Loss of CTSS activity reduces lymphoma growth by limiting communication with CD4+ T follicular helper cells while inducing antigen diversification and activation of CD8+ T cells. Overall, our results suggest that CTSS inhibition has non-redundant therapeutic potential to enhance anti-tumor immune responses in indolent and aggressive lymphomas.


Subject(s)
Antigen Presentation/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cathepsins/genetics , Lymphoma, Non-Hodgkin/immunology , Mutation , Tumor Microenvironment/immunology , Animals , Apoptosis , B-Lymphocytes/immunology , Cell Proliferation , Female , Germinal Center/immunology , Humans , Lymphocyte Activation/immunology , Lymphoma, Non-Hodgkin/genetics , Lymphoma, Non-Hodgkin/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , T-Lymphocytes, Helper-Inducer/immunology , Tumor Cells, Cultured
17.
Science ; 368(6492)2020 05 15.
Article in English | MEDLINE | ID: mdl-32409444

ABSTRACT

De novo protein design has been successful in expanding the natural protein repertoire. However, most de novo proteins lack biological function, presenting a major methodological challenge. In vaccinology, the induction of precise antibody responses remains a cornerstone for next-generation vaccines. Here, we present a protein design algorithm called TopoBuilder, with which we engineered epitope-focused immunogens displaying complex structural motifs. In both mice and nonhuman primates, cocktails of three de novo-designed immunogens induced robust neutralizing responses against the respiratory syncytial virus. Furthermore, the immunogens refocused preexisting antibody responses toward defined neutralization epitopes. Overall, our design approach opens the possibility of targeting specific epitopes for the development of vaccines and therapeutic antibodies and, more generally, will be applicable to the design of de novo proteins displaying complex functional motifs.


Subject(s)
Antibodies, Neutralizing/biosynthesis , Computational Biology/methods , Immunodominant Epitopes/chemistry , Protein Engineering/methods , Recombinant Fusion Proteins/chemistry , Respiratory Syncytial Virus Vaccines/chemistry , Respiratory Syncytial Virus, Human/immunology , Amino Acid Motifs , Humans , Immunodominant Epitopes/immunology , Protein Conformation , Recombinant Fusion Proteins/immunology , Respiratory Syncytial Virus Vaccines/immunology , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/immunology
18.
Nat Genet ; 51(3): 517-528, 2019 03.
Article in English | MEDLINE | ID: mdl-30692681

ABSTRACT

Chromatin is organized into topologically associating domains (TADs) enriched in distinct histone marks. In cancer, gain-of-function mutations in the gene encoding the enhancer of zeste homolog 2 protein (EZH2) lead to a genome-wide increase in histone-3 Lys27 trimethylation (H3K27me3) associated with transcriptional repression. However, the effects of these epigenetic changes on the structure and function of chromatin domains have not been explored. Here, we found a functional interplay between TADs and epigenetic and transcriptional changes mediated by mutated EZH2. Altered EZH2 (p.Tyr646* (EZH2Y646X)) led to silencing of entire domains, synergistically inactivating multiple tumor suppressors. Intra-TAD gene silencing was coupled with changes of interactions between gene promoter regions. Notably, gene expression and chromatin interactions were restored by pharmacological inhibition of EZH2Y646X. Our results indicate that EZH2Y646X alters the topology and function of chromatin domains to promote synergistic oncogenic programs.


Subject(s)
Chromatin/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Epigenesis, Genetic/genetics , Mutation/genetics , Transcription, Genetic/genetics , Animals , Cell Line, Tumor , DNA Methylation/genetics , Epigenomics/methods , Gene Expression Regulation, Neoplastic/genetics , Gene Silencing/physiology , Histones/genetics , Humans , Mice , Promoter Regions, Genetic/genetics
19.
Cancer Lett ; 263(1): 130-9, 2008 May 08.
Article in English | MEDLINE | ID: mdl-18282657

ABSTRACT

Ursolic acid (UA) is a pentacyclic triterpenoid compound that is widely distributed in the plant kingdom and has a broad range of biological effects. Here, we examined the effects of UA on the proliferation and differentiation of human tumor cell lines from melanoma (A375), glioblastoma (U87) and thyroid anaplastic carcinoma (ARO), and on the proliferation of a non-transformed human fibroblast cell line (WI-38). The results show that UA inhibits tumor cell proliferation in a dose- and time-dependent manner. Consistent with this finding, UA treatment promotes differentiation of all of the analyzed tumor cell lines. Interestingly, we found that UA inhibits the endogenous reverse transcriptase (RT) activity in tumor cells, which has recently been shown to be involved in the control of proliferation and differentiation of neoplastic cells. Considering these findings, we suggest that the observed anti-proliferative and differentiating effects of UA may be related to this target.


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , RNA-Directed DNA Polymerase/metabolism , Triterpenes/pharmacology , Cell Line, Tumor , Fluorescent Antibody Technique, Indirect , Humans , Reverse Transcriptase Polymerase Chain Reaction , Ursolic Acid
20.
Genome Biol ; 19(1): 217, 2018 12 10.
Article in English | MEDLINE | ID: mdl-30526631

ABSTRACT

BACKGROUND: Chromatin folding gives rise to structural elements among which are clusters of densely interacting DNA regions termed topologically associating domains (TADs). TADs have been characterized across multiple species, tissue types, and differentiation stages, sometimes in association with regulation of biological functions. The reliability and reproducibility of these findings are intrinsically related with the correct identification of these domains from high-throughput chromatin conformation capture (Hi-C) experiments. RESULTS: Here, we test and compare 22 computational methods to identify TADs across 20 different conditions. We find that TAD sizes and numbers vary significantly among callers and data resolutions, challenging the definition of an average TAD size, but strengthening the hypothesis that TADs are hierarchically organized domains, rather than disjoint structural elements. Performances of these methods differ based on data resolution and normalization strategy, but a core set of TAD callers consistently retrieve reproducible domains, even at low sequencing depths, that are enriched for TAD-associated biological features. CONCLUSIONS: This study provides a reference for the analysis of chromatin domains from Hi-C experiments and useful guidelines for choosing a suitable approach based on the experimental design, available data, and biological question of interest.


Subject(s)
Chromatin Assembly and Disassembly , Genomics/methods , Animals , Chromosomes , Humans , Mice
SELECTION OF CITATIONS
SEARCH DETAIL