Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 191
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 180(2): 233-247.e21, 2020 01 23.
Article in English | MEDLINE | ID: mdl-31978343

ABSTRACT

Wnt dependency and Lgr5 expression define multiple mammalian epithelial stem cell types. Under defined growth factor conditions, such adult stem cells (ASCs) grow as 3D organoids that recapitulate essential features of the pertinent epithelium. Here, we establish long-term expanding venom gland organoids from several snake species. The newly assembled transcriptome of the Cape coral snake reveals that organoids express high levels of toxin transcripts. Single-cell RNA sequencing of both organoids and primary tissue identifies distinct venom-expressing cell types as well as proliferative cells expressing homologs of known mammalian stem cell markers. A hard-wired regional heterogeneity in the expression of individual venom components is maintained in organoid cultures. Harvested venom peptides reflect crude venom composition and display biological activity. This study extends organoid technology to reptilian tissues and describes an experimentally tractable model system representing the snake venom gland.


Subject(s)
Cell Culture Techniques/methods , Organoids/growth & development , Snake Venoms/metabolism , Adult Stem Cells/metabolism , Animals , Coral Snakes/metabolism , Gene Expression Profiling/methods , Organoids/metabolism , Salivary Glands/metabolism , Snake Venoms/genetics , Snakes/genetics , Snakes/growth & development , Stem Cells/metabolism , Toxins, Biological/genetics , Transcriptome/genetics
2.
Cell ; 179(2): 527-542.e19, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31585086

ABSTRACT

Much of current molecular and cell biology research relies on the ability to purify cell types by fluorescence-activated cell sorting (FACS). FACS typically relies on the ability to label cell types of interest with antibodies or fluorescent transgenic constructs. However, antibody availability is often limited, and genetic manipulation is labor intensive or impossible in the case of primary human tissue. To date, no systematic method exists to enrich for cell types without a priori knowledge of cell-type markers. Here, we propose GateID, a computational method that combines single-cell transcriptomics with FACS index sorting to purify cell types of choice using only native cellular properties such as cell size, granularity, and mitochondrial content. We validate GateID by purifying various cell types from zebrafish kidney marrow and the human pancreas to high purity without resorting to specific antibodies or transgenes.


Subject(s)
Cell Separation/methods , Flow Cytometry/methods , Software , Transcriptome , Animals , Humans , Kidney/cytology , Pancreas/cytology , Single-Cell Analysis , Zebrafish/anatomy & histology
3.
Nat Rev Mol Cell Biol ; 20(12): 753-765, 2019 12.
Article in English | MEDLINE | ID: mdl-31690888

ABSTRACT

Tracking the progeny of single cells is necessary for building lineage trees that recapitulate processes such as embryonic development and stem cell differentiation. In classical lineage tracing experiments, cells are fluorescently labelled to allow identification by microscopy of a limited number of cell clones. To track a larger number of clones in complex tissues, fluorescent proteins are now replaced by heritable DNA barcodes that are read using next-generation sequencing. In prospective lineage tracing, unique DNA barcodes are introduced into single cells through genetic manipulation (using, for example, Cre-mediated recombination or CRISPR-Cas9-mediated editing) and tracked over time. Alternatively, in retrospective lineage tracing, naturally occurring somatic mutations can be used as endogenous DNA barcodes. Finally, single-cell mRNA-sequencing datasets that capture different cell states within a developmental or differentiation trajectory can be used to recapitulate lineages. In this Review, we discuss methods for prospective or retrospective lineage tracing and demonstrate how trajectory reconstruction algorithms can be applied to single-cell mRNA-sequencing datasets to infer developmental or differentiation tracks. We discuss how these approaches are used to understand cell fate during embryogenesis, cell differentiation and tissue regeneration.


Subject(s)
CRISPR-Cas Systems , Cell Differentiation/physiology , Cell Lineage/physiology , Embryonic Development/physiology , Gene Expression Regulation, Developmental/physiology , Regeneration/physiology , Animals , High-Throughput Nucleotide Sequencing , Humans
4.
Cell ; 167(7): 1883-1896.e15, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27984734

ABSTRACT

In multicellular organisms, dedicated regulatory circuits control cell type diversity and responses. The crosstalk and redundancies within these circuits and substantial cellular heterogeneity pose a major research challenge. Here, we present CRISP-seq, an integrated method for massively parallel single-cell RNA sequencing (RNA-seq) and clustered regularly interspaced short palindromic repeats (CRISPR)-pooled screens. We show that profiling the genomic perturbation and transcriptome in the same cell enables us to simultaneously elucidate the function of multiple factors and their interactions. We applied CRISP-seq to probe regulatory circuits of innate immunity. By sampling tens of thousands of perturbed cells in vitro and in mice, we identified interactions and redundancies between developmental and signaling-dependent factors. These include opposing effects of Cebpb and Irf8 in regulating the monocyte/macrophage versus dendritic cell lineages and differential functions for Rela and Stat1/2 in monocyte versus dendritic cell responses to pathogens. This study establishes CRISP-seq as a broadly applicable, comprehensive, and unbiased approach for elucidating mammalian regulatory circuits.


Subject(s)
Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Animals , CCAAT-Enhancer-Binding Protein-beta/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats , Dendritic Cells/metabolism , Inflammation/metabolism , Macrophages/metabolism , Mice, Inbred C57BL , Monocytes/metabolism
5.
Cell ; 163(4): 799-810, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26544934

ABSTRACT

Recent advances in single-cell sequencing hold great potential for exploring biological systems with unprecedented resolution. Sequencing the genome of individual cells can reveal somatic mutations and allows the investigation of clonal dynamics. Single-cell transcriptome sequencing can elucidate the cell type composition of a sample. However, single-cell sequencing comes with major technical challenges and yields complex data output. In this Primer, we provide an overview of available methods and discuss experimental design and single-cell data analysis. We hope that these guidelines will enable a growing number of researchers to leverage the power of single-cell sequencing.


Subject(s)
Cells/metabolism , Gene Expression Profiling , Single-Cell Analysis , Animals , Cells/classification , Guidelines as Topic , Humans , Sequence Analysis, DNA , Sequence Analysis, RNA
6.
Cell ; 163(1): 134-47, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26365489

ABSTRACT

Mammalian interphase chromosomes interact with the nuclear lamina (NL) through hundreds of large lamina-associated domains (LADs). We report a method to map NL contacts genome-wide in single human cells. Analysis of nearly 400 maps reveals a core architecture consisting of gene-poor LADs that contact the NL with high cell-to-cell consistency, interspersed by LADs with more variable NL interactions. The variable contacts tend to be cell-type specific and are more sensitive to changes in genome ploidy than the consistent contacts. Single-cell maps indicate that NL contacts involve multivalent interactions over hundreds of kilobases. Moreover, we observe extensive intra-chromosomal coordination of NL contacts, even over tens of megabases. Such coordinated loci exhibit preferential interactions as detected by Hi-C. Finally, the consistency of NL contacts is inversely linked to gene activity in single cells and correlates positively with the heterochromatic histone modification H3K9me3. These results highlight fundamental principles of single-cell chromatin organization. VIDEO ABSTRACT.


Subject(s)
Chromatin/metabolism , Nuclear Lamina/metabolism , Single-Cell Analysis/methods , Cell Line, Tumor , Chromatin/chemistry , Chromosomes/chemistry , Chromosomes/metabolism , Genome-Wide Association Study , Humans , In Situ Hybridization, Fluorescence , Interphase
7.
Cell ; 161(4): 933-45, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25957691

ABSTRACT

In Rspondin-based 3D cultures, Lgr5 stem cells from multiple organs form ever-expanding epithelial organoids that retain their tissue identity. We report the establishment of tumor organoid cultures from 20 consecutive colorectal carcinoma (CRC) patients. For most, organoids were also generated from adjacent normal tissue. Organoids closely recapitulate several properties of the original tumor. The spectrum of genetic changes within the "living biobank" agrees well with previous large-scale mutational analyses of CRC. Gene expression analysis indicates that the major CRC molecular subtypes are represented. Tumor organoids are amenable to high-throughput drug screens allowing detection of gene-drug associations. As an example, a single organoid culture was exquisitely sensitive to Wnt secretion (porcupine) inhibitors and carried a mutation in the negative Wnt feedback regulator RNF43, rather than in APC. Organoid technology may fill the gap between cancer genetics and patient trials, complement cell-line- and xenograft-based drug studies, and allow personalized therapy design. PAPERCLIP.


Subject(s)
Biological Specimen Banks , Colorectal Neoplasms/pathology , Drug Screening Assays, Antitumor/methods , Organoids , Colorectal Neoplasms/drug therapy , DNA-Binding Proteins/metabolism , Humans , Oncogene Proteins/metabolism , Organ Culture Techniques , Organoids/drug effects , Precision Medicine , Ubiquitin-Protein Ligases
8.
Cell ; 157(1): 8-11, 2014 Mar 27.
Article in English | MEDLINE | ID: mdl-24679522

ABSTRACT

Single-cell analyses have provided invaluable insights into studying heterogenity, signaling, and stochastic gene expression. Recent technological advances now open the door to genome-wide single-cell studies.


Subject(s)
Single-Cell Analysis , Animals , Cell Physiological Phenomena , Genome-Wide Association Study , Humans , Sequence Analysis, RNA
9.
Cell ; 159(3): 662-75, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25417113

ABSTRACT

Advancing our understanding of embryonic development is heavily dependent on identification of novel pathways or regulators. Although genome-wide techniques such as RNA sequencing are ideally suited for discovering novel candidate genes, they are unable to yield spatially resolved information in embryos or tissues. Microscopy-based approaches, using in situ hybridization, for example, can provide spatial information about gene expression, but are limited to analyzing one or a few genes at a time. Here, we present a method where we combine traditional histological techniques with low-input RNA sequencing and mathematical image reconstruction to generate a high-resolution genome-wide 3D atlas of gene expression in the zebrafish embryo at three developmental stages. Importantly, our technique enables searching for genes that are expressed in specific spatial patterns without manual image annotation. We envision broad applicability of RNA tomography as an accurate and sensitive approach for spatially resolved transcriptomics in whole embryos and dissected organs.


Subject(s)
Embryo, Nonmammalian/metabolism , Gene Expression Profiling , Sequence Analysis, RNA , Tomography/methods , Zebrafish/embryology , Animals , Imaging, Three-Dimensional
10.
Annu Rev Cell Dev Biol ; 31: 317-45, 2015.
Article in English | MEDLINE | ID: mdl-26566114

ABSTRACT

Correct and timely lineage decisions are critical for normal embryonic development and homeostasis of adult tissues. Therefore, the search for fundamental principles that underlie lineage decision-making lies at the heart of developmental biology. Here, we review attempts to understand lineage decision-making as the interplay of single-cell heterogeneity and gene regulation. Fluctuations at the single-cell level are an important driving force behind cell-state transitions and the creation of cell-type diversity. Gene regulatory networks amplify such fluctuations and define stable cell types. They also mediate the influence of signaling inputs on the lineage decision. In this review, we focus on insights gleaned from in vitro differentiation of embryonic stem cells. We discuss emerging concepts, with an emphasis on transcriptional regulation, dynamical aspects of differentiation, and functional single-cell heterogeneity. We also highlight some novel tools to study lineage decision-making in vitro.


Subject(s)
Cell Lineage/genetics , Gene Expression Regulation, Developmental/genetics , Gene Regulatory Networks/genetics , Animals , Cell Differentiation/genetics , Embryonic Development/genetics , Embryonic Stem Cells/physiology , Humans , Signal Transduction/genetics
11.
Cell ; 155(4): 869-80, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24209624

ABSTRACT

Variability in gene expression contributes to phenotypic heterogeneity even in isogenic populations. Here, we used the stereotyped, Wnt signaling-dependent development of the Caenorhabditis elegans Q neuroblast to probe endogenous mechanisms that control gene expression variability. We found that the key Hox gene that orients Q neuroblast migration exhibits increased gene expression variability in mutants in which Wnt pathway activity has been perturbed. Distinct features of the gene expression distributions prompted us on a systematic search for regulatory interactions, revealing a network of interlocked positive and negative feedback loops. Interestingly, positive feedback appeared to cooperate with negative feedback to reduce variability while keeping the Hox gene expression at elevated levels. A minimal model correctly predicts the increased gene expression variability across mutants. Our results highlight the influence of gene network architecture on expression variability and implicate feedback regulation as an effective mechanism to ensure developmental robustness.


Subject(s)
Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Genetic Variation , Wnt Signaling Pathway , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans Proteins/genetics , Cell Movement , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Feedback, Physiological , Frizzled Receptors/genetics , Frizzled Receptors/metabolism , Gene Regulatory Networks , Glycoproteins/genetics , Homeodomain Proteins/genetics , Transcription Factors/genetics , Wnt Proteins
12.
EMBO J ; 42(17): e113280, 2023 09 04.
Article in English | MEDLINE | ID: mdl-37522872

ABSTRACT

Embryo implantation into the uterus marks a key transition in mammalian development. In mice, implantation is mediated by the trophoblast and is accompanied by a morphological transition from the blastocyst to the egg cylinder. However, the roles of trophoblast-uterine interactions in embryo morphogenesis during implantation are poorly understood due to inaccessibility in utero and the remaining challenges to recapitulate it ex vivo from the blastocyst. Here, we engineer a uterus-like microenvironment to recapitulate peri-implantation development of the whole mouse embryo ex vivo and reveal essential roles of the physical embryo-uterine interaction. We demonstrate that adhesion between the trophoblast and the uterine matrix is required for in utero-like transition of the blastocyst to the egg cylinder. Modeling the implanting embryo as a wetting droplet links embryo shape dynamics to the underlying changes in trophoblast adhesion and suggests that the adhesion-mediated tension release facilitates egg cylinder formation. Light-sheet live imaging and the experimental control of the engineered uterine geometry and trophoblast velocity uncovers the coordination between trophoblast motility and embryo growth, where the trophoblast delineates space for embryo morphogenesis.


Subject(s)
Blastocyst , Embryo Implantation , Female , Mice , Animals , Trophoblasts , Uterus , Embryonic Development , Mammals
13.
Nat Methods ; 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38886577

ABSTRACT

In a human cell, thousands of replication forks simultaneously coordinate duplication of the entire genome. The rate at which this process occurs might depend on the epigenetic state of the genome and vary between, or even within, cell types. To accurately measure DNA replication speeds, we developed single-cell 5-ethynyl-2'-deoxyuridine sequencing to detect nascent replicated DNA. We observed that the DNA replication speed is not constant but increases during S phase of the cell cycle. Using genetic and pharmacological perturbations we were able to alter this acceleration of replication and conclude that DNA damage inflicted by the process of transcription limits the speed of replication during early S phase. In late S phase, during which less-transcribed regions replicate, replication accelerates and approaches its maximum speed.

14.
Immunity ; 49(4): 627-639.e6, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30314756

ABSTRACT

The non-hematopoietic cell fraction of the bone marrow (BM) is classically identified as CD45- Ter119- CD31- (herein referred to as triple-negative cells or TNCs). Although TNCs are believed to contain heterogeneous stromal cell populations, they remain poorly defined. Here we showed that the vast majority of TNCs (∼85%) have a hematopoietic rather than mesenchymal origin. Single cell RNA-sequencing revealed erythroid and lymphoid progenitor signatures among CD51- TNCs. Ly6D+ CD44+ CD51- TNCs phenotypically and functionally resembled CD45+ pro-B lymphoid cells, whereas Ly6D- CD44+ CD51- TNCs were enriched in previously unappreciated stromal-dependent erythroid progenitors hierarchically situated between preCFU-E and proerythroblasts. Upon adoptive transfer, CD44+ CD51- TNCs contributed to repopulate the B-lymphoid and erythroid compartments. CD44+ CD51- TNCs also expanded during phenylhydrazine-induced acute hemolysis or in a model of sickle cell anemia. These findings thus uncover physiologically relevant new classes of stromal-associated functional CD45- hematopoietic progenitors.


Subject(s)
Bone Marrow Cells/immunology , Erythroid Cells/immunology , Lymphoid Progenitor Cells/immunology , Stromal Cells/immunology , Adoptive Transfer/methods , Animals , Blood Group Antigens/immunology , Blood Group Antigens/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Differentiation/immunology , Cells, Cultured , Erythroid Cells/cytology , Erythroid Cells/metabolism , Leukocyte Common Antigens/immunology , Leukocyte Common Antigens/metabolism , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Platelet Endothelial Cell Adhesion Molecule-1/immunology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Stromal Cells/cytology , Stromal Cells/metabolism
15.
Cell ; 148(3): 608-19, 2012 Feb 03.
Article in English | MEDLINE | ID: mdl-22304925

ABSTRACT

Intestinal crypts in mammals are comprised of long-lived stem cells and shorter-lived progenies. These two populations are maintained in specific proportions during adult life. Here, we investigate the design principles governing the dynamics of these proportions during crypt morphogenesis. Using optimal control theory, we show that a proliferation strategy known as a "bang-bang" control minimizes the time to obtain a mature crypt. This strategy consists of a surge of symmetric stem cell divisions, establishing the entire stem cell pool first, followed by a sharp transition to strictly asymmetric stem cell divisions, producing nonstem cells with a delay. We validate these predictions using lineage tracing and single-molecule fluorescence in situ hybridization of intestinal crypts in infant mice, uncovering small crypts that are entirely composed of Lgr5-labeled stem cells, which become a minority as crypts continue to grow. Our approach can be used to uncover similar design principles in other developmental systems.


Subject(s)
Cell Lineage , Intestine, Small/growth & development , Morphogenesis , Animals , Cell Proliferation , In Situ Hybridization, Fluorescence/methods , Intestine, Small/cytology , Intestine, Small/embryology , Mice , Receptors, G-Protein-Coupled/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Time Factors
16.
Cell ; 150(6): 1170-81, 2012 Sep 14.
Article in English | MEDLINE | ID: mdl-22959267

ABSTRACT

The cell-fate decision leading to gametogenesis is essential for sexual reproduction. In S. cerevisiae, only diploid MATa/α but not haploid MATa or MATα cells undergo gametogenesis, known as sporulation. We find that transcription of two long noncoding RNAs (lncRNAs) mediates mating-type control of sporulation. In MATa or MATα haploids, expression of IME1, the central inducer of gametogenesis, is inhibited in cis by transcription of the lncRNA IRT1, located in the IME1 promoter. IRT1 transcription recruits the Set2 histone methyltransferase and the Set3 histone deacetylase complex to establish repressive chromatin at the IME1 promoter. Inhibiting expression of IRT1 and an antisense transcript that antagonizes the expression of the meiotic regulator IME4 allows cells expressing the haploid mating type to sporulate with kinetics that are indistinguishable from that of MATa/α diploids. Conversely, expression of the two lncRNAs abolishes sporulation in MATa/α diploids. Thus, transcription of two lncRNAs governs mating-type control of gametogenesis in yeast.


Subject(s)
Gene Expression Regulation, Fungal , Genes, Mating Type, Fungal , RNA, Fungal/genetics , RNA, Long Noncoding/genetics , Saccharomyces cerevisiae/genetics , Transcription, Genetic , Gametogenesis , Nuclear Proteins/genetics , Promoter Regions, Genetic , Repressor Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/physiology , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Spores, Fungal , Transcription Factors/genetics
17.
Cell ; 150(6): 1209-22, 2012 Sep 14.
Article in English | MEDLINE | ID: mdl-22980981

ABSTRACT

During cellular reprogramming, only a small fraction of cells become induced pluripotent stem cells (iPSCs). Previous analyses of gene expression during reprogramming were based on populations of cells, impeding single-cell level identification of reprogramming events. We utilized two gene expression technologies to profile 48 genes in single cells at various stages during the reprogramming process. Analysis of early stages revealed considerable variation in gene expression between cells in contrast to late stages. Expression of Esrrb, Utf1, Lin28, and Dppa2 is a better predictor for cells to progress into iPSCs than expression of the previously suggested reprogramming markers Fbxo15, Fgf4, and Oct4. Stochastic gene expression early in reprogramming is followed by a late hierarchical phase with Sox2 being the upstream factor in a gene expression hierarchy. Finally, downstream factors derived from the late phase, which do not include Oct4, Sox2, Klf4, c-Myc, and Nanog, can activate the pluripotency circuitry.


Subject(s)
Cellular Reprogramming , Induced Pluripotent Stem Cells/metabolism , Single-Cell Analysis , Transcriptome , Animals , Cell Line , Embryo, Mammalian/cytology , Embryonic Stem Cells , Fibroblasts/cytology , Fibroblasts/metabolism , Genetic Markers , Induced Pluripotent Stem Cells/cytology , Kruppel-Like Factor 4 , Mice , Microfluidic Analytical Techniques , SOXB1 Transcription Factors/metabolism , Transcription Factors/metabolism
18.
Cell ; 148(5): 1015-28, 2012 Mar 02.
Article in English | MEDLINE | ID: mdl-22385965

ABSTRACT

Regulatory networks orchestrated by key transcription factors (TFs) have been proposed to play a central role in the determination of stem cell states. However, the master transcriptional regulators of adult stem cells are poorly understood. We have identified two TFs, Slug and Sox9, that act cooperatively to determine the mammary stem cell (MaSC) state. Inhibition of either Slug or Sox9 blocks MaSC activity in primary mammary epithelial cells. Conversely, transient coexpression of exogenous Slug and Sox9 suffices to convert differentiated luminal cells into MaSCs with long-term mammary gland-reconstituting ability. Slug and Sox9 induce MaSCs by activating distinct autoregulatory gene expression programs. We also show that coexpression of Slug and Sox9 promotes the tumorigenic and metastasis-seeding abilities of human breast cancer cells and is associated with poor patient survival, providing direct evidence that human breast cancer stem cells are controlled by key regulators similar to those operating in normal murine MaSCs.


Subject(s)
Breast Neoplasms/metabolism , Mammary Glands, Human/cytology , SOX9 Transcription Factor/metabolism , Transcription Factors/metabolism , Animals , Cells, Cultured , Female , Gene Knockdown Techniques , Humans , Mammary Glands, Human/metabolism , Mice , SOX9 Transcription Factor/genetics , Snail Family Transcription Factors , Transcription Factors/genetics
19.
Nature ; 597(7877): 561-565, 2021 09.
Article in English | MEDLINE | ID: mdl-34497418

ABSTRACT

Single-cell sequencing methods have enabled in-depth analysis of the diversity of cell types and cell states in a wide range of organisms. These tools focus predominantly on sequencing the genomes1, epigenomes2 and transcriptomes3 of single cells. However, despite recent progress in detecting proteins by mass spectrometry with single-cell resolution4, it remains a major challenge to measure translation in individual cells. Here, building on existing protocols5-7, we have substantially increased the sensitivity of these assays to enable ribosome profiling in single cells. Integrated with a machine learning approach, this technology achieves single-codon resolution. We validate this method by demonstrating that limitation for a particular amino acid causes ribosome pausing at a subset of the codons encoding the amino acid. Of note, this pausing is only observed in a sub-population of cells correlating to its cell cycle state. We further expand on this phenomenon in non-limiting conditions and detect pronounced GAA pausing during mitosis. Finally, we demonstrate the applicability of this technique to rare primary enteroendocrine cells. This technology provides a first step towards determining the contribution of the translational process to the remarkable diversity between seemingly identical cells.


Subject(s)
Cell Cycle/genetics , Codon/genetics , Protein Biosynthesis , RNA-Seq/methods , Ribosomes/metabolism , Single-Cell Analysis , Amino Acids/deficiency , Amino Acids/pharmacology , Animals , Cell Cycle/drug effects , Cell Line , Female , Humans , Machine Learning , Male , Mice , Peptide Chain Elongation, Translational , Peptide Chain Initiation, Translational , Peptide Chain Termination, Translational , Protein Biosynthesis/drug effects , Reproducibility of Results , Ribosomes/drug effects
20.
Cell ; 144(6): 910-25, 2011 Mar 18.
Article in English | MEDLINE | ID: mdl-21414483

ABSTRACT

Cellular decision making is the process whereby cells assume different, functionally important and heritable fates without an associated genetic or environmental difference. Such stochastic cell fate decisions generate nongenetic cellular diversity, which may be critical for metazoan development as well as optimized microbial resource utilization and survival in a fluctuating, frequently stressful environment. Here, we review several examples of cellular decision making from viruses, bacteria, yeast, lower metazoans, and mammals, highlighting the role of regulatory network structure and molecular noise. We propose that cellular decision making is one of at least three key processes underlying development at various scales of biological organization.


Subject(s)
Cell Differentiation , Models, Biological , Animals , Humans
SELECTION OF CITATIONS
SEARCH DETAIL