Network


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

Hotspot


Dive into the research topics where Ido Amit is active.

Publication


Featured researches published by Ido Amit.


Nature Biotechnology | 2011

Full-length transcriptome assembly from RNA-Seq data without a reference genome

Manfred Grabherr; Brian J. Haas; Moran Yassour; Joshua Z. Levin; Dawn Anne Thompson; Ido Amit; Xian Adiconis; Lin Fan; Raktima Raychowdhury; Qiandong Zeng; Zehua Chen; Evan Mauceli; Nir Hacohen; Andreas Gnirke; Nicholas Rhind; Federica Di Palma; Bruce Birren; Chad Nusbaum; Kerstin Lindblad-Toh; Nir Friedman; Aviv Regev

Massively parallel sequencing of cDNA has enabled deep and efficient probing of transcriptomes. Current approaches for transcript reconstruction from such data often rely on aligning reads to a reference genome, and are thus unsuitable for samples with a partial or missing reference genome. Here we present the Trinity method for de novo assembly of full-length transcripts and evaluate it on samples from fission yeast, mouse and whitefly, whose reference genome is not yet available. By efficiently constructing and analyzing sets of de Bruijn graphs, Trinity fully reconstructs a large fraction of transcripts, including alternatively spliced isoforms and transcripts from recently duplicated genes. Compared with other de novo transcriptome assemblers, Trinity recovers more full-length transcripts across a broad range of expression levels, with a sensitivity similar to methods that rely on genome alignments. Our approach provides a unified solution for transcriptome reconstruction in any sample, especially in the absence of a reference genome.Massively parallel sequencing of cDNA has enabled deep and efficient probing of transcriptomes. Current approaches for transcript reconstruction from such data often rely on aligning reads to a reference genome, and are thus unsuitable for samples with a partial or missing reference genome. Here we present the Trinity method for de novo assembly of full-length transcripts and evaluate it on samples from fission yeast, mouse and whitefly, whose reference genome is not yet available. By efficiently constructing and analyzing sets of de Bruijn graphs, Trinity fully reconstructs a large fraction of transcripts, including alternatively spliced isoforms and transcripts from recently duplicated genes. Compared with other de novo transcriptome assemblers, Trinity recovers more full-length transcripts across a broad range of expression levels, with a sensitivity similar to methods that rely on genome alignments. Our approach provides a unified solution for transcriptome reconstruction in any sample, especially in the absence of a reference genome.


Science | 2009

Comprehensive mapping of long range interactions reveals folding principles of the human genome

Erez Lieberman-Aiden; Nynke L. van Berkum; Louise Williams; Maxim Imakaev; Tobias Ragoczy; Agnes Telling; Ido Amit; Bryan R. Lajoie; Peter J. Sabo; Michael O. Dorschner; Richard Sandstrom; Bradley E. Bernstein; Michael Bender; Mark Groudine; Andreas Gnirke; John A. Stamatoyannopoulos; Leonid A. Mirny; Eric S. Lander

Chromosomal Mapping The conformation of the genome in the nucleus and contacts between both proximal and distal loci influence gene expression. In order to map genomic contacts, Lieberman-Aiden et al. (p. 289, see the cover) developed a technique to allow the detection of all interactions between genomic loci in the eukaryotic nucleus followed by deep sequencing. This technology was used to map the organization of the human genome and to examine the spatial proximity of chromosomal loci at one megabase resolution. The map suggests that the genome is partitioned into two spatial compartments that are related to local chromatin state and whose remodeling correlates with changes in the chromatin state. Chromosomes are organized in a fractal knot-free conformation that is densely packed while easily folded and unfolded. We describe Hi-C, a method that probes the three-dimensional architecture of whole genomes by coupling proximity-based ligation with massively parallel sequencing. We constructed spatial proximity maps of the human genome with Hi-C at a resolution of 1 megabase. These maps confirm the presence of chromosome territories and the spatial proximity of small, gene-rich chromosomes. We identified an additional level of genome organization that is characterized by the spatial segregation of open and closed chromatin to form two genome-wide compartments. At the megabase scale, the chromatin conformation is consistent with a fractal globule, a knot-free, polymer conformation that enables maximally dense packing while preserving the ability to easily fold and unfold any genomic locus. The fractal globule is distinct from the more commonly used globular equilibrium model. Our results demonstrate the power of Hi-C to map the dynamic conformations of whole genomes.


Nature | 2009

Chromatin signature reveals over a thousand highly conserved large non-coding RNAs in mammals

Mitchell Guttman; Ido Amit; Manuel Garber; Courtney French; Michael F. Lin; David M. Feldser; Maite Huarte; Or Zuk; Bryce W. Carey; John P. Cassady; Moran N. Cabili; Rudolf Jaenisch; Tarjei S. Mikkelsen; Tyler Jacks; Nir Hacohen; Bradley E. Bernstein; Manolis Kellis; Aviv Regev; John L. Rinn; Eric S. Lander

There is growing recognition that mammalian cells produce many thousands of large intergenic transcripts. However, the functional significance of these transcripts has been particularly controversial. Although there are some well-characterized examples, most (>95%) show little evidence of evolutionary conservation and have been suggested to represent transcriptional noise. Here we report a new approach to identifying large non-coding RNAs using chromatin-state maps to discover discrete transcriptional units intervening known protein-coding loci. Our approach identified ∼1,600 large multi-exonic RNAs across four mouse cell types. In sharp contrast to previous collections, these large intervening non-coding RNAs (lincRNAs) show strong purifying selection in their genomic loci, exonic sequences and promoter regions, with greater than 95% showing clear evolutionary conservation. We also developed a functional genomics approach that assigns putative functions to each lincRNA, demonstrating a diverse range of roles for lincRNAs in processes from embryonic stem cell pluripotency to cell proliferation. We obtained independent functional validation for the predictions for over 100 lincRNAs, using cell-based assays. In particular, we demonstrate that specific lincRNAs are transcriptionally regulated by key transcription factors in these processes such as p53, NFκB, Sox2, Oct4 (also known as Pou5f1) and Nanog. Together, these results define a unique collection of functional lincRNAs that are highly conserved and implicated in diverse biological processes.


Cell | 2014

Tissue-Resident Macrophage Enhancer Landscapes Are Shaped by the Local Microenvironment

Yonit Lavin; Deborah R. Winter; Ronnie Blecher-Gonen; Eyal David; Hadas Keren-Shaul; Miriam Merad; Steffen Jung; Ido Amit

Macrophages are critical for innate immune defense and also control organ homeostasis in a tissue-specific manner. They provide a fitting model to study the impact of ontogeny and microenvironment on chromatin state and whether chromatin modifications contribute to macrophage identity. Here, we profile the dynamics of four histone modifications across seven tissue-resident macrophage populations. We identify 12,743 macrophage-specific enhancers and establish that tissue-resident macrophages have distinct enhancer landscapes beyond what can be explained by developmental origin. Combining our enhancer catalog with gene expression profiles and open chromatin regions, we show that a combination of tissue- and lineage-specific transcription factors form the regulatory networks controlling chromatin specification in tissue-resident macrophages. The environment is capable of shaping the chromatin landscape of transplanted bone marrow precursors, and even differentiated macrophages can be reprogrammed when transferred into a new microenvironment. These results provide a comprehensive view of macrophage regulatory landscape and highlight the importance of the microenvironment, along with pioneer factors in orchestrating identity and plasticity.


Science | 2014

Massively parallel single cell RNA-Seq for marker-free decomposition of tissues into cell types

Diego Jaitin; Ephraim Kenigsberg; Hadas Keren-Shaul; Naama Elefant; Franziska Paul; Irina Zaretsky; Alexander Mildner; Nadav Cohen; Steffen Jung; Amos Tanay; Ido Amit

Sequencing of RNA from thousands of individual immune cells allows unbiased identification of cellular subtypes. In multicellular organisms, biological function emerges when heterogeneous cell types form complex organs. Nevertheless, dissection of tissues into mixtures of cellular subpopulations is currently challenging. We introduce an automated massively parallel single-cell RNA sequencing (RNA-seq) approach for analyzing in vivo transcriptional states in thousands of single cells. Combined with unsupervised classification algorithms, this facilitates ab initio cell-type characterization of splenic tissues. Modeling single-cell transcriptional states in dendritic cells and additional hematopoietic cell types uncovers rich cell-type heterogeneity and gene-modules activity in steady state and after pathogen activation. Cellular diversity is thereby approached through inference of variable and dynamic pathway activity rather than a fixed preprogrammed cell-type hierarchy. These data demonstrate single-cell RNA-seq as an effective tool for comprehensive cellular decomposition of complex tissues. Introducing MARS-Seq Immune cells are typically differentiated by surface markers; however, this designation is somewhat crude and does not allow for fine distinctions that might be characterized by their RNA transcripts. Jaitin et al. (p. 776) used massively parallel single-cell RNA-sequencing (MARS-Seq) analysis to explore cellular heterogeneity within the immune system by assembling an automated experimental platform that enables RNA profiling of cells sorted from tissues using flow cytometry. More than 1000 cells could be sequenced, and unsupervised clustering analysis of the RNA profiles revealed distinct cellular groupings that corresponded to B cells, macrophages, and dendritic cells. This approach provides the ability to perform a bottom-up characterization of in vivo cell-type landscapes independent of cell markers or prior knowledge.


Nature | 2013

Derivation of novel human ground state naive pluripotent stem cells

Ohad Gafni; Leehee Weinberger; Abed AlFatah Mansour; Yair S. Manor; Elad Chomsky; Dalit Ben-Yosef; Yael Kalma; Sergey Viukov; Itay Maza; Asaf Zviran; Yoach Rais; Zohar Shipony; Zohar Mukamel; Vladislav Krupalnik; Mirie Zerbib; Shay Geula; Inbal Caspi; Dan Schneir; Tamar Shwartz; Shlomit Gilad; Daniela Amann-Zalcenstein; Sima Benjamin; Ido Amit; Amos Tanay; Rada Massarwa; Noa Novershtern; Jacob Hanna

Mouse embryonic stem (ES) cells are isolated from the inner cell mass of blastocysts, and can be preserved in vitro in a naive inner-cell-mass-like configuration by providing exogenous stimulation with leukaemia inhibitory factor (LIF) and small molecule inhibition of ERK1/ERK2 and GSK3β signalling (termed 2i/LIF conditions). Hallmarks of naive pluripotency include driving Oct4 (also known as Pou5f1) transcription by its distal enhancer, retaining a pre-inactivation X chromosome state, and global reduction in DNA methylation and in H3K27me3 repressive chromatin mark deposition on developmental regulatory gene promoters. Upon withdrawal of 2i/LIF, naive mouse ES cells can drift towards a primed pluripotent state resembling that of the post-implantation epiblast. Although human ES cells share several molecular features with naive mouse ES cells, they also share a variety of epigenetic properties with primed murine epiblast stem cells (EpiSCs). These include predominant use of the proximal enhancer element to maintain OCT4 expression, pronounced tendency for X chromosome inactivation in most female human ES cells, increase in DNA methylation and prominent deposition of H3K27me3 and bivalent domain acquisition on lineage regulatory genes. The feasibility of establishing human ground state naive pluripotency in vitro with equivalent molecular and functional features to those characterized in mouse ES cells remains to be defined. Here we establish defined conditions that facilitate the derivation of genetically unmodified human naive pluripotent stem cells from already established primed human ES cells, from somatic cells through induced pluripotent stem (iPS) cell reprogramming or directly from blastocysts. The novel naive pluripotent cells validated herein retain molecular characteristics and functional properties that are highly similar to mouse naive ES cells, and distinct from conventional primed human pluripotent cells. This includes competence in the generation of cross-species chimaeric mouse embryos that underwent organogenesis following microinjection of human naive iPS cells into mouse morulas. Collectively, our findings establish new avenues for regenerative medicine, patient-specific iPS cell disease modelling and the study of early human development in vitro and in vivo.


Nature Neuroscience | 2015

Host microbiota constantly control maturation and function of microglia in the CNS

Daniel Erny; Anna Lena Hrabě de Angelis; Diego Jaitin; Peter Wieghofer; Ori Staszewski; Eyal David; Hadas Keren-Shaul; Tanel Mahlakõiv; Kristin Jakobshagen; Thorsten Buch; Vera Schwierzeck; Olaf Utermöhlen; Eunyoung Chun; Wendy S. Garrett; Kathy D. McCoy; Andreas Diefenbach; Peter Staeheli; Bärbel Stecher; Ido Amit; Marco Prinz

As the tissue macrophages of the CNS, microglia are critically involved in diseases of the CNS. However, it remains unknown what controls their maturation and activation under homeostatic conditions. We observed substantial contributions of the host microbiota to microglia homeostasis, as germ-free (GF) mice displayed global defects in microglia with altered cell proportions and an immature phenotype, leading to impaired innate immune responses. Temporal eradication of host microbiota severely changed microglia properties. Limited microbiota complexity also resulted in defective microglia. In contrast, recolonization with a complex microbiota partially restored microglia features. We determined that short-chain fatty acids (SCFA), microbiota-derived bacterial fermentation products, regulated microglia homeostasis. Accordingly, mice deficient for the SCFA receptor FFAR2 mirrored microglia defects found under GF conditions. These findings suggest that host bacteria vitally regulate microglia maturation and function, whereas microglia impairment can be rectified to some extent by complex microbiota.


Science | 2009

Unbiased Reconstruction of a Mammalian Transcriptional Network Mediating Pathogen Responses

Ido Amit; Manuel Garber; Nicolas Chevrier; Ana Paula Leite; Yoni Donner; Thomas Eisenhaure; Mitchell Guttman; Jennifer K. Grenier; Weibo Li; Or Zuk; Lisa A. Schubert; Brian Birditt; Tal Shay; Alon Goren; Xiaolan Zhang; Zachary D. Smith; Raquel P. Deering; Rebecca C. McDonald; Moran N. Cabili; Bradley E. Bernstein; John L. Rinn; Alexander Meissner; David E. Root; Nir Hacohen; Aviv Regev

Peeking at Pathogen Response Networks Networks controlling gene expression serve as key decision-making circuits in cells, but the regulatory networks that control dynamic and specific gene expression responses to stimuli are often not well understood. This is particularly true for immune dendritic cells (DCs), which respond to pathogens by mounting elaborate transcriptional responses, and are centrally involved in infectious diseases, autoimmunity, and vaccines. Amit et al. (p. 257, published online 3 September) explored the transcriptional response of dendritic cells to specific classes of pathogens. The transcriptional subnetworks responsible for mammalian dendritic cell responses to different pathogens were identified, and the function of 100 regulators clarified. Inflammatory and antiviral programs in dendritic cells are controlled and tuned by a network of regulators. Models of mammalian regulatory networks controlling gene expression have been inferred from genomic data but have largely not been validated. We present an unbiased strategy to systematically perturb candidate regulators and monitor cellular transcriptional responses. We applied this approach to derive regulatory networks that control the transcriptional response of mouse primary dendritic cells to pathogens. Our approach revealed the regulatory functions of 125 transcription factors, chromatin modifiers, and RNA binding proteins, which enabled the construction of a network model consisting of 24 core regulators and 76 fine-tuners that help to explain how pathogen-sensing pathways achieve specificity. This study establishes a broadly applicable, comprehensive, and unbiased approach to reveal the wiring and functions of a regulatory network controlling a major transcriptional response in primary mammalian cells.


Nature | 2013

Deterministic direct reprogramming of somatic cells to pluripotency

Yoach Rais; Asaf Zviran; Shay Geula; Ohad Gafni; Elad Chomsky; Sergey Viukov; Abed AlFatah Mansour; Inbal Caspi; Vladislav Krupalnik; Mirie Zerbib; Itay Maza; Nofar Mor; Dror Baran; Leehee Weinberger; Diego Jaitin; David Lara-Astiaso; Ronnie Blecher-Gonen; Zohar Shipony; Zohar Mukamel; Tzachi Hagai; Shlomit Gilad; Daniela Amann-Zalcenstein; Amos Tanay; Ido Amit; Noa Novershtern; Jacob Hanna

Somatic cells can be inefficiently and stochastically reprogrammed into induced pluripotent stem (iPS) cells by exogenous expression of Oct4 (also called Pou5f1), Sox2, Klf4 and Myc (hereafter referred to as OSKM). The nature of the predominant rate-limiting barrier(s) preventing the majority of cells to successfully and synchronously reprogram remains to be defined. Here we show that depleting Mbd3, a core member of the Mbd3/NuRD (nucleosome remodelling and deacetylation) repressor complex, together with OSKM transduction and reprogramming in naive pluripotency promoting conditions, result in deterministic and synchronized iPS cell reprogramming (near 100% efficiency within seven days from mouse and human cells). Our findings uncover a dichotomous molecular function for the reprogramming factors, serving to reactivate endogenous pluripotency networks while simultaneously directly recruiting the Mbd3/NuRD repressor complex that potently restrains the reactivation of OSKM downstream target genes. Subsequently, the latter interactions, which are largely depleted during early pre-implantation development in vivo, lead to a stochastic and protracted reprogramming trajectory towards pluripotency in vitro. The deterministic reprogramming approach devised here offers a novel platform for the dissection of molecular dynamics leading to establishing pluripotency at unprecedented flexibility and resolution.


Nature Genetics | 2007

A module of negative feedback regulators defines growth factor signaling.

Ido Amit; Tal Shay; Yiling Lu; Menachem Katz; Fan Zhang; Gabi Tarcic; Doris R. Siwak; John P. Lahad; Jasmine Jacob-Hirsch; Ninette Amariglio; Nora Vaisman; Eran Segal; Gideon Rechavi; Uri Alon; Gordon B. Mills; Eytan Domany; Yosef Yarden

Signaling pathways invoke interplays between forward signaling and feedback to drive robust cellular response. In this study, we address the dynamics of growth factor signaling through profiling of protein phosphorylation and gene expression, demonstrating the presence of a kinetically defined cluster of delayed early genes that function to attenuate the early events of growth factor signaling. Using epidermal growth factor receptor signaling as the major model system and concentrating on regulation of transcription and mRNA stability, we demonstrate that a number of genes within the delayed early gene cluster function as feedback regulators of immediate early genes. Consistent with their role in negative regulation of cell signaling, genes within this cluster are downregulated in diverse tumor types, in correlation with clinical outcome. More generally, our study proposes a mechanistic description of the cellular response to growth factors by defining architectural motifs that underlie the function of signaling networks.

Collaboration


Dive into the Ido Amit's collaboration.

Top Co-Authors

Avatar

Eyal David

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Diego Jaitin

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Hadas Keren-Shaul

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Aviv Regev

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Deborah R. Winter

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Amir Giladi

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Ronnie Blecher-Gonen

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Assaf Weiner

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Michal Schwartz

Weizmann Institute of Science

View shared research outputs
Researchain Logo
Decentralizing Knowledge