Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Natalia Naumova is active.

Publication


Featured researches published by Natalia Naumova.


Science | 2013

Organization of the Mitotic Chromosome

Natalia Naumova; Maxim Imakaev; Geoffrey Fudenberg; Ye Zhan; Bryan R. Lajoie; Leonid A. Mirny; Job Dekker

Chromosome Conundrum The three-dimensional organization of chromosomal DNA within the cell nucleus plays an important role in gene regulation. Naumova et al. (p. 948, published online 7 November; see the Perspective by Kleckner et al.) used chromosome conformation capture-based methods in human tissue culture cells to analyze the higher order folding of human chromosomes across the cell cycle. During interphase the chromosomes showed locus-specific compart-mentalization. In mitotic cells, on the other hand, the chromosome organization was more linear, consistent with arrays of consecutive chromatin loops. Chromosome conformation changes dramatically during the cell cycle and is unlikely to carry epigenetic information. [Also see Perspective by Kleckner et al.] Mitotic chromosomes are among the most recognizable structures in the cell, yet for over a century their internal organization remains largely unsolved. We applied chromosome conformation capture methods, 5C and Hi-C, across the cell cycle and revealed two distinct three-dimensional folding states of the human genome. We show that the highly compartmentalized and cell type–specific organization described previously for nonsynchronous cells is restricted to interphase. In metaphase, we identified a homogenous folding state that is locus-independent, common to all chromosomes, and consistent among cell types, suggesting a general principle of metaphase chromosome organization. Using polymer simulations, we found that metaphase Hi-C data are inconsistent with classic hierarchical models and are instead best described by a linearly organized longitudinally compressed array of consecutive chromatin loops.


Nature Methods | 2012

Iterative correction of Hi-C data reveals hallmarks of chromosome organization.

Maxim Imakaev; Geoffrey Fudenberg; Rachel Patton McCord; Natalia Naumova; Anton Goloborodko; Bryan R. Lajoie; Job Dekker; Leonid A. Mirny

Extracting biologically meaningful information from chromosomal interactions obtained with genome-wide chromosome conformation capture (3C) analyses requires the elimination of systematic biases. We present a computational pipeline that integrates a strategy to map sequencing reads with a data-driven method for iterative correction of biases, yielding genome-wide maps of relative contact probabilities. We validate this ICE (iterative correction and eigenvector decomposition) technique on published data obtained by the high-throughput 3C method Hi-C, and we demonstrate that eigenvector decomposition of the obtained maps provides insights into local chromatin states, global patterns of chromosomal interactions, and the conserved organization of human and mouse chromosomes.


Methods | 2012

Hi–C: A comprehensive technique to capture the conformation of genomes

Jon-Matthew Belton; Rachel Patton McCord; Johan H. Gibcus; Natalia Naumova; Ye Zhan; Job Dekker

We describe a method, Hi-C, to comprehensively detect chromatin interactions in the mammalian nucleus. This method is based on Chromosome Conformation Capture, in which chromatin is crosslinked with formaldehyde, then digested, and re-ligated in such a way that only DNA fragments that are covalently linked together form ligation products. The ligation products contain the information of not only where they originated from in the genomic sequence but also where they reside, physically, in the 3D organization of the genome. In Hi-C, a biotin-labeled nucleotide is incorporated at the ligation junction, enabling selective purification of chimeric DNA ligation junctions followed by deep sequencing. The compatibility of Hi-C with next generation sequencing platforms makes it possible to detect chromatin interactions on an unprecedented scale. This advance gives Hi-C the power to both explore the biophysical properties of chromatin as well as the implications of chromatin structure for the biological functions of the nucleus. A massively parallel survey of chromatin interaction provides the previously missing dimension of spatial context to other genomic studies. This spatial context will provide a new perspective to studies of chromatin and its role in genome regulation in normal conditions and in disease.


Methods | 2012

Analysis of long-range chromatin interactions using Chromosome Conformation Capture

Natalia Naumova; Emily Smith; Ye Zhan; Job Dekker

Chromosome Conformation Capture, or 3C, is a pioneering method for investigating the three-dimensional structure of chromatin. 3C is used to analyze long-range looping interactions between any pair of selected genomic loci. Most 3C studies focus on defined genomic regions of interest that can be up to several hundred Kb in size. The method has become widely adopted and has been modified to increase throughput to allow unbiased genome-wide analysis. These large-scale adaptations are presented in other articles in this issue of Methods. Here we describe the 3C procedure in detail, including the appropriate use of the technology, the experimental set-up, an optimized protocol and troubleshooting guide, and considerations for data analysis. The protocol described here contains previously unpublished improvements, which save time and reduce labor. We pay special attention to primer design, appropriate controls and data analysis. We include notes and discussion based on our extensive experience to help researchers understand the principles of 3C-based techniques and to avoid common pitfalls and mistakes. This paper represents a complete resource and detailed guide for anyone who desires to perform 3C.


Journal of Cell Science | 2010

Integrating one-dimensional and three-dimensional maps of genomes

Natalia Naumova; Job Dekker

Genomes exist in vivo as complex physical structures, and their functional output (i.e. the gene expression profile of a cell) is related to their spatial organization inside the nucleus as well as to local chromatin status. Chromatin modifications and chromosome conformation are distinct in different tissues and cell types, which corresponds closely with the diversity in gene-expression patterns found in different tissues of the body. The biological processes and mechanisms driving these general correlations are currently the topic of intense study. An emerging theme is that genome compartmentalization – both along the linear length of chromosomes, and in three dimensions by the spatial colocalization of chromatin domains and genomic loci from across the genome – is a crucial parameter in regulating genome expression. In this Commentary, we propose that a full understanding of genome regulation requires integrating three different types of data: first, one-dimensional data regarding the state of local chromatin – such as patterns of protein binding along chromosomes; second, three-dimensional data that describe the population-averaged folding of chromatin inside cells and; third, single-cell observations of three-dimensional spatial colocalization of genetic loci and trans factors that reveal information about their dynamics and frequency of colocalization.


Science | 2018

A pathway for mitotic chromosome formation

Johan H. Gibcus; Kumiko Samejima; Anton Goloborodko; Itaru Samejima; Natalia Naumova; Johannes Nuebler; Masato T. Kanemaki; Linfeng Xie; James R. Paulson; William C. Earnshaw; Leonid A. Mirny; Job Dekker

Tracking mitotic chromosome formation How cells pack DNA into fully compact, rod-shaped chromosomes during mitosis has fascinated cell biologists for more than a century. Gibcus et al. delineated the conformational transition trajectory from interphase chromatin to mitotic chromosomes minute by minute during the cell cycle. The mitotic chromosome is organized in a spiral staircase architecture in which chromatin loops emanate radially from a centrally located helical scaffold. The molecular machines condensin I and II play distinct roles in these processes: Condensin II is essential for helical winding, whereas condensin I modulates the organization within each helical turn. Science, this issue p. eaao6135 Mitotic chromosome folding involves formation of increasingly compacted helically arranged nested loop arrays. INTRODUCTION During mitosis, cells compact their chromosomes into dense rod-shaped structures to ensure their reliable transmission to daughter cells. Our work explores how cells achieve this compaction. We integrate genetic, genomic, and computational approaches to characterize the key steps in mitotic chromosome formation from the G2 nucleus to metaphase, and we identify roles of specific molecular machines, condensin I and II, in these major conformational transitions. RATIONALE We used chicken DT-40 cells expressing an analog-sensitive CDK1 to produce cell cultures that synchronously enter mitosis. We collected cells at key time points during mitotic entry; analyzed chromosome organization by microscopy, chromosome conformation capture, and polymer simulations; and delineated a pathway of mitotic chromosome formation. We used engineered cell lines to study the function of condensin complexes, which are critical for mitotic chromosome formation. We fused condensin I and II subunits to plant auxin-inducible degron domains, thus enabling their rapid depletion in late G2 just before mitotic entry. These cell lines allowed us to determine the roles of condensin I and II in specific steps of the mitotic chromosome morphogenesis pathway. RESULTS Our analysis of G2 chromosomes reveals hallmarks of interphase chromosomes, including topologically associating domains and compartments. Upon entry into prophase, this organization is lost within minutes, and by late prophase, chromosomes are folded as arrays of consecutive loops condensed around a central axis. These loops project with random but mutually correlated angles from the axis. During prometaphase, the loop array undergoes two major reorganizations. First, it acquires a helical arrangement of loops. Polymer simulations of Hi-C data show that the centrally located axis acquires a helical twist so that consecutive loops emanate as the steps of a spiral staircase. Second, the chromatin loops become nested with ~400-kb outer loops split up by ~80-kb inner loops. As prometaphase proceeds, chromosomes shorten through progressive helical winding, with the numbers of loops per turn increasing. As a result, the size of a helical turn grows from ~3 Mb (~40 loops) to ~12 Mb (~150 loops). Depletion of condensin I or II before mitotic entry revealed their differing roles in mitotic chromosome formation. Either condensin can mediate loop array formation. However, condensin II is required for the helical twisting of the scaffold from which loops emanate, whereas condensin I modulates the size and arrangement of nested inner loops. CONCLUSION We describe a pathway of mitotic chromosome folding that unifies many previous observations. In prophase, condensins mediate the loss of interphase organization and the formation of arrays of consecutive loops. In prometaphase, chromosomes adopt a spiral staircase–like structure with a helically arranged axial scaffold of condensin II at the bases of chromatin loops. The condensin II loops are further compacted by condensin I into clusters of smaller nested loops that are additionally collapsed by chromatin-to-chromatin attractions. The combination of nested loops distributed around a helically twisted axis plus dense chromatin packing achieves the 10,000-fold compaction of chromatin into linearly organized chromosomes that is required for accurate chromosome segregation when cells divide. A pathway for mitotic chromosome formation. In prophase, condensins mediate the loss of interphase chromosome conformation, and loop arrays are formed. In prometaphase, the combined action of condensin I (blue spheres in the bottom diagram) and II (red spheres) results in helically arranged nested loop arrays. Mitotic chromosomes fold as compact arrays of chromatin loops. To identify the pathway of mitotic chromosome formation, we combined imaging and Hi-C analysis of synchronous DT40 cell cultures with polymer simulations. Here we show that in prophase, the interphase organization is rapidly lost in a condensin-dependent manner, and arrays of consecutive 60-kilobase (kb) loops are formed. During prometaphase, ~80-kb inner loops are nested within ~400-kb outer loops. The loop array acquires a helical arrangement with consecutive loops emanating from a central “spiral staircase” condensin scaffold. The size of helical turns progressively increases to ~12 megabases during prometaphase. Acute depletion of condensin I or II shows that nested loops form by differential action of the two condensins, whereas condensin II is required for helical winding.


Methods | 2012

From cells to chromatin: Capturing snapshots of genome organization with 5C technology

Maria Ferraiuolo; Amartya Sanyal; Natalia Naumova; Job Dekker; Josée Dostie

In eukaryotes, genome organization can be observed on many levels and at different scales. This organization is important not only to reduce chromosome length but also for the proper execution of various biological processes. High-resolution mapping of spatial chromatin structure was made possible by the development of the chromosome conformation capture (3C) technique. 3C uses chemical cross-linking followed by proximity-based ligation of fragmented DNA to capture frequently interacting chromatin segments in cell populations. Several 3C-related methods capable of higher chromosome conformation mapping throughput were reported afterwards. These techniques include the 3C-carbon copy (5C) approach, which offers the advantage of being highly quantitative and reproducible. We provide here an updated reference protocol for the production of 5C libraries analyzed by next-generation sequencing or onto microarrays. A procedure used to verify that 3C library templates bear the high quality required to produce superior 5C libraries is also described. We believe that this detailed protocol will help guide researchers in probing spatial genome organization and its role in various biological processes.


bioRxiv | 2017

Mitotic chromosomes fold by condensin-dependent helical winding of chromatin loop arrays

Johan H. Gibcus; Kumiko Samejima; Anton Goloborodko; Itaru Samejima; Natalia Naumova; Masato T. Kanemaki; Linfeng Xie; James R. Paulson; William C. Earnshaw; Leonid A. Mirny; Job Dekker

During mitosis, chromosomes fold into compacted rod shaped structures. We combined imaging and Hi-C of synchronous DT40 cell cultures with polymer simulations to determine how interphase chromosomes are converted into compressed arrays of loops characteristic of mitotic chromosomes. We found that the interphase organization is disassembled within minutes of prophase entry and by late prophase chromosomes are already folded as arrays of consecutive loops. During prometaphase, this array reorganizes to form a helical arrangement of nested loops. Polymer simulations reveal that Hi-C data are inconsistent with solenoidal coiling of the entire chromatid, but instead suggest a centrally located helically twisted axis from which consecutive loops emanate as in a spiral staircase. Chromosomes subsequently shorten through progressive helical winding, with the numbers of loops per turn increasing so that the size of a helical turn grows from around 3 Mb (~40 loops) to ~12 Mb (~150 loops) in fully condensed metaphase chromosomes. Condensin is essential to disassemble the interphase chromatin conformation. Analysis of mutants revealed differing roles for condensin I and II during these processes. Either condensin can mediate formation of loop arrays. However, condensin II was required for helical winding during prometaphase, whereas condensin I modulated the size and arrangement of loops inside the helical turns. These observations identify a mitotic chromosome morphogenesis pathway in which folding of linear loop arrays produces long thin chromosomes during prophase that then shorten by progressive growth of loops and helical winding during prometaphase. One Sentence Summary Mitotic chromosome morphogenesis occurs through condensin-mediated disassembly of the interphase conformation and formation of extended prophase loop arrays that then shorten by loop growth and condensin-dependent helical winding.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Crystal structure of the DNA binding domain of the transcription factor T-bet suggests simultaneous recognition of distant genome sites

Ce Feng Liu; Gabriel S. Brandt; Quyen Q. Hoang; Natalia Naumova; Vanja Lazarevic; Eun Sook Hwang; Job Dekker; Laurie H. Glimcher; Dagmar Ringe; Gregory A. Petsko

Significance The transcription factor T-bet (Tbox protein expressed in T cells), a master regulator of T-cell lineage commitment, is a member of the Tbox family but coordinately regulates many more genes than other Tbox proteins. How T-bet simultaneously recognizes distant elements that may be thousands of base pairs apart is unknown. We have determined the crystal structure of the Tbox DNA binding domain of T-bet complexed with a 24-bp palindromic DNA. The structure shows a dimer where each monomer binds simultaneously to two independent DNA molecules. Fluorescence-based assays show T-bet can synapse two DNA molecules in solution. Chromosome conformation capture assays confirm that T-bet can directly mediate the formation of chromatin loops at the IFN-γ gene locus in the absence of other transcription-related proteins. The transcription factor T-bet (Tbox protein expressed in T cells) is one of the master regulators of both the innate and adaptive immune responses. It plays a central role in T-cell lineage commitment, where it controls the TH1 response, and in gene regulation in plasma B-cells and dendritic cells. T-bet is a member of the Tbox family of transcription factors; however, T-bet coordinately regulates the expression of many more genes than other Tbox proteins. A central unresolved question is how T-bet is able to simultaneously recognize distant Tbox binding sites, which may be located thousands of base pairs away. We have determined the crystal structure of the Tbox DNA binding domain (DBD) of T-bet in complex with a palindromic DNA. The structure shows a quaternary structure in which the T-bet dimer has its DNA binding regions splayed far apart, making it impossible for a single dimer to bind both sites of the DNA palindrome. In contrast to most other Tbox proteins, a single T-bet DBD dimer binds simultaneously to identical half-sites on two independent DNA. A fluorescence-based assay confirms that T-bet dimers are able to bring two independent DNA molecules into close juxtaposition. Furthermore, chromosome conformation capture assays confirm that T-bet functions in the direct formation of chromatin loops in vitro and in vivo. The data are consistent with a looping/synapsing model for transcriptional regulation by T-bet in which a single dimer of the transcription factor can recognize and coalesce distinct genetic elements, either a promoter plus a distant regulatory element, or promoters on two different genes.


Biophysical Journal | 2017

Polymer Models Integrate Inverted Nuclear Geometry with Conventional HI-C Compartmentalization

Martin Falk; Natalia Naumova; Geoffrey Fudenberg; Yana Feodorova; Job Dekker; Leonid A. Mirny; Irina Solovei

Collaboration


Dive into the Natalia Naumova's collaboration.

Top Co-Authors

Avatar

Job Dekker

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Leonid A. Mirny

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Anton Goloborodko

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Bryan R. Lajoie

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Geoffrey Fudenberg

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Rachel Patton McCord

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Ye Zhan

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Johan H. Gibcus

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Amartya Sanyal

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

James R. Paulson

University of Wisconsin–Oshkosh

View shared research outputs
Researchain Logo
Decentralizing Knowledge