Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Dev Cell ; 58(18): 1801-1818.e15, 2023 09 25.
Article in English | MEDLINE | ID: mdl-37751684

ABSTRACT

Approaches to study human pharyngeal foregut endoderm-a developmental intermediate that is linked to various human syndromes involving pharynx development and organogenesis of tissues such as thymus, parathyroid, and thyroid-have been hampered by scarcity of tissue access and cellular models. We present an efficient stepwise differentiation method to generate human pharyngeal foregut endoderm from pluripotent stem cells. We determine dose and temporal requirements of signaling pathway engagement for optimized differentiation and characterize the differentiation products on cellular and integrated molecular level. We present a computational classification tool, "CellMatch," and transcriptomic classification of differentiation products on an integrated mouse scRNA-seq developmental roadmap confirms cellular maturation. Integrated transcriptomic and chromatin analyses infer differentiation stage-specific gene regulatory networks. Our work provides the method and integrated multiomic resource for the investigation of disease-relevant loci and gene regulatory networks and their role in developmental defects affecting the pharyngeal endoderm and its derivatives.


Subject(s)
Pharynx , Pluripotent Stem Cells , Humans , Animals , Mice , Endoderm/metabolism , Digestive System , Cell Differentiation/genetics , Gene Expression Regulation, Developmental
2.
Nat Methods ; 18(9): 1046-1055, 2021 09.
Article in English | MEDLINE | ID: mdl-34480151

ABSTRACT

Chromosome conformation capture (3C) assays are used to map chromatin interactions genome-wide. Chromatin interaction maps provide insights into the spatial organization of chromosomes and the mechanisms by which they fold. Hi-C and Micro-C are widely used 3C protocols that differ in key experimental parameters including cross-linking chemistry and chromatin fragmentation strategy. To understand how the choice of experimental protocol determines the ability to detect and quantify aspects of chromosome folding we have performed a systematic evaluation of 3C experimental parameters. We identified optimal protocol variants for either loop or compartment detection, optimizing fragment size and cross-linking chemistry. We used this knowledge to develop a greatly improved Hi-C protocol (Hi-C 3.0) that can detect both loops and compartments relatively effectively. In addition to providing benchmarked protocols, this work produced ultra-deep chromatin interaction maps using Micro-C, conventional Hi-C and Hi-C 3.0 for key cell lines used by the 4D Nucleome project.


Subject(s)
Chromatin/chemistry , Chromosomes, Human/chemistry , Cross-Linking Reagents/chemistry , Genetic Techniques , Cell Line , Chromatin/metabolism , Databases, Factual , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/physiology , Humans
3.
Proc Natl Acad Sci U S A ; 116(36): 17970-17979, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31420514

ABSTRACT

Single-cell sequencing technologies have revealed an unexpectedly broad repertoire of cells required to mediate complex functions in multicellular organisms. Despite the multiple roles of adipose tissue in maintaining systemic metabolic homeostasis, adipocytes are thought to be largely homogenous with only 2 major subtypes recognized in humans so far. Here we report the existence and characteristics of 4 distinct human adipocyte subtypes, and of their respective mesenchymal progenitors. The phenotypes of these distinct adipocyte subtypes are differentially associated with key adipose tissue functions, including thermogenesis, lipid storage, and adipokine secretion. The transcriptomic signature of "brite/beige" thermogenic adipocytes reveals mechanisms for iron accumulation and protection from oxidative stress, necessary for mitochondrial biogenesis and respiration upon activation. Importantly, this signature is enriched in human supraclavicular adipose tissue, confirming that these cells comprise thermogenic depots in vivo, and explain previous findings of a rate-limiting role of iron in adipose tissue browning. The mesenchymal progenitors that give rise to beige/brite adipocytes express a unique set of cytokines and transcriptional regulators involved in immune cell modulation of adipose tissue browning. Unexpectedly, we also find adipocyte subtypes specialized for high-level expression of the adipokines adiponectin or leptin, associated with distinct transcription factors previously implicated in adipocyte differentiation. The finding of a broad adipocyte repertoire derived from a distinct set of mesenchymal progenitors, and of the transcriptional regulators that can control their development, provides a framework for understanding human adipose tissue function and role in metabolic disease.


Subject(s)
Adipocytes, Beige/metabolism , Adiponectin/biosynthesis , Leptin/blood , Mesenchymal Stem Cells/metabolism , Thermogenesis , Transcriptome , Adipocytes, Beige/cytology , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/metabolism , Female , Gene Expression Profiling , Humans , Male , Mesenchymal Stem Cells/cytology
4.
Cell Rep ; 27(3): 708-718.e10, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30995470

ABSTRACT

Studies in vertebrates have outlined conserved molecular control of definitive endoderm (END) development. However, recent work also shows that key molecular aspects of human END regulation differ even from rodents. Differentiation of human embryonic stem cells (ESCs) to END offers a tractable system to study the molecular basis of normal and defective human-specific END development. Here, we interrogated dynamics in chromatin accessibility during differentiation of ESCs to END, predicting DNA-binding proteins that may drive this cell fate transition. We then combined single-cell RNA-seq with parallel CRISPR perturbations to comprehensively define the loss-of-function phenotype of those factors in END development. Following a few candidates, we revealed distinct impairments in the differentiation trajectories for mediators of TGFß signaling and expose a role for the FOXA2 transcription factor in priming human END competence for human foregut and hepatic END specification. Together, this single-cell functional genomics study provides high-resolution insight on human END development.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats/genetics , RNA, Guide, Kinetoplastida/metabolism , Transcription Factors/metabolism , Cell Differentiation , Chromatin/metabolism , Endoderm/cytology , Endoderm/metabolism , Hepatocyte Nuclear Factor 3-beta/antagonists & inhibitors , Hepatocyte Nuclear Factor 3-beta/genetics , Hepatocyte Nuclear Factor 3-beta/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Humans , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , RNA Interference , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism , Signal Transduction , Single-Cell Analysis , Smad4 Protein/genetics , Smad4 Protein/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transforming Growth Factor beta/metabolism
5.
Nat Biotechnol ; 37(4): 461-468, 2019 04.
Article in English | MEDLINE | ID: mdl-30936567

ABSTRACT

Recent single-cell RNA-sequencing studies have suggested that cells follow continuous transcriptomic trajectories in an asynchronous fashion during development. However, observations of cell flux along trajectories are confounded with population size effects in snapshot experiments and are therefore hard to interpret. In particular, changes in proliferation and death rates can be mistaken for cell flux. Here we present pseudodynamics, a mathematical framework that reconciles population dynamics with the concepts underlying developmental trajectories inferred from time-series single-cell data. Pseudodynamics models population distribution shifts across trajectories to quantify selection pressure, population expansion, and developmental potentials. Applying this model to time-resolved single-cell RNA-sequencing of T-cell and pancreatic beta cell maturation, we characterize proliferation and apoptosis rates and identify key developmental checkpoints, data inaccessible to existing approaches.


Subject(s)
Cell Differentiation/genetics , Sequence Analysis, RNA/statistics & numerical data , Single-Cell Analysis/statistics & numerical data , Animals , Apoptosis/genetics , Biotechnology , Cell Proliferation/genetics , Female , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Likelihood Functions , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Time Factors
6.
Immunity ; 48(6): 1258-1270.e6, 2018 06 19.
Article in English | MEDLINE | ID: mdl-29884461

ABSTRACT

Thymus development is critical to the adaptive immune system, yet a comprehensive transcriptional framework capturing thymus organogenesis at single-cell resolution is still needed. We applied single-cell RNA sequencing (RNA-seq) to capture 8 days of thymus development, perturbations of T cell receptor rearrangement, and in vitro organ cultures, producing profiles of 24,279 cells. We resolved transcriptional heterogeneity of developing lymphocytes, and genetic perturbation confirmed T cell identity of conventional and non-conventional lymphocytes. We characterized maturation dynamics of thymic epithelial cells in vivo, classified cell maturation state in a thymic organ culture, and revealed the intrinsic capacity of thymic epithelium to preserve transcriptional regularity despite exposure to exogenous retinoic acid. Finally, by integrating the cell atlas with human genome-wide association study (GWAS) data and autoimmune-disease-related genes, we implicated embryonic thymus-resident cells as possible participants in autoimmune disease etiologies. This resource provides a single-cell transcriptional framework for biological discovery and molecular analysis of thymus organogenesis.


Subject(s)
Cell Differentiation/immunology , Sequence Analysis, RNA/methods , T-Lymphocytes/immunology , Thymus Gland/embryology , Animals , Autoimmune Diseases/immunology , Embryo, Mammalian , Gene Expression Profiling/methods , Genome-Wide Association Study , Humans , Mice , Organogenesis/immunology , T-Lymphocytes/cytology , Thymus Gland/cytology
7.
Methods ; 101: 36-42, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26525193

ABSTRACT

The ability to manipulate transcription in human pluripotent stem cells (hPSCs) is fundamental for the discovery of key genes and mechanisms governing cellular state and differentiation. Recently developed CRISPR-effector systems provide a systematic approach to rapidly test gene function in mammalian cells, including hPSCs. In this review, we discuss recent advances in CRISPR-effector technologies that have been employed to control transcription through gene activation, gene repression, and epigenome engineering. We describe an application of CRISPR-effector mediated transcriptional regulation in hPSCs by targeting a synthetic promoter driving a GFP transgene, demonstrating the ease and effectiveness of CRISPR-effector mediated transcriptional regulation in hPSCs.


Subject(s)
CRISPR-Cas Systems , Pluripotent Stem Cells/physiology , Transcription, Genetic , Animals , Base Sequence , Cell Culture Techniques , HEK293 Cells , Humans , RNA, Guide, Kinetoplastida/genetics
8.
Nat Methods ; 12(5): 401-403, 2015 May.
Article in English | MEDLINE | ID: mdl-25775043

ABSTRACT

Understanding of mammalian enhancers is limited by the lack of a technology to rapidly and thoroughly test the cell type-specific function. Here, we use a nuclease-deficient Cas9 (dCas9)-histone demethylase fusion to functionally characterize previously described and new enhancer elements for their roles in the embryonic stem cell state. Further, we distinguish the mechanism of action of dCas9-LSD1 at enhancers from previous dCas9-effectors.


Subject(s)
Caspase 9/metabolism , Enhancer Elements, Genetic/physiology , Histone Demethylases/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Caspase 9/drug effects , Cells, Cultured , Embryonic Stem Cells/metabolism , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Enzymologic , Genes, Reporter , Histone Demethylases/drug effects , Histones/metabolism , Mice , Neisseria meningitidis/enzymology , Recombinant Proteins/metabolism
9.
Elife ; 32014 Jun 16.
Article in English | MEDLINE | ID: mdl-24935936

ABSTRACT

Gene expression and metabolism are coupled at numerous levels. Cells must sense and respond to nutrients in their environment, and specialized cells must synthesize metabolic products required for their function. Pluripotent stem cells have the ability to differentiate into a wide variety of specialized cells. How metabolic state contributes to stem cell differentiation is not understood. In this study, we show that RNA-binding by the stem cell translation regulator Musashi-1 (MSI1) is allosterically inhibited by 18-22 carbon ω-9 monounsaturated fatty acids. The fatty acid binds to the N-terminal RNA Recognition Motif (RRM) and induces a conformational change that prevents RNA association. Musashi proteins are critical for development of the brain, blood, and epithelium. We identify stearoyl-CoA desaturase-1 as a MSI1 target, revealing a feedback loop between ω-9 fatty acid biosynthesis and MSI1 activity. We propose that other RRM proteins could act as metabolite sensors to couple gene expression changes to physiological state.


Subject(s)
Nerve Tissue Proteins/metabolism , Oleic Acid/chemistry , RNA-Binding Proteins/metabolism , Stem Cells/cytology , Allosteric Site , Amino Acid Motifs , Animals , Cell Differentiation , Cell Line, Tumor , Gene Expression Profiling , Gene Expression Regulation , Mice , Molecular Dynamics Simulation , Pluripotent Stem Cells/cytology , Protein Structure, Tertiary , Recombinant Proteins/metabolism , Stearoyl-CoA Desaturase/metabolism , Structure-Activity Relationship
10.
Development ; 141(1): 219-23, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24346702

ABSTRACT

The identification of the trans-acting factors and cis-regulatory modules that are involved in human pluripotent stem cell (hPSC) maintenance and differentiation is necessary to dissect the operating regulatory networks in these processes and thereby identify nodes where signal input will direct desired cell fate decisions in vitro or in vivo. To deconvolute these networks, we established a method to influence the differentiation state of hPSCs with a CRISPR-associated catalytically inactive dCas9 fused to an effector domain. In human embryonic stem cells, we find that the dCas9 effectors can exert positive or negative regulation on the expression of developmentally relevant genes, which can influence cell differentiation status when impinging on a key node in the regulatory network that governs the cell state. This system provides a platform for the interrogation of the underlying regulators governing specific differentiation decisions, which can then be employed to direct cellular differentiation down desired pathways.


Subject(s)
Bacterial Proteins/genetics , CRISPR-Associated Proteins/genetics , CRISPR-Cas Systems , Cell Differentiation/genetics , Endonucleases/genetics , Gene Regulatory Networks/genetics , Pluripotent Stem Cells/metabolism , Transcriptional Activation/genetics , Amino Acid Sequence , CRISPR-Associated Protein 9 , Cell Lineage/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , HEK293 Cells , Humans , Molecular Sequence Data , Octamer Transcription Factor-3/genetics , Pluripotent Stem Cells/cytology , SOXF Transcription Factors/biosynthesis , Transcription, Genetic
11.
Stem Cell Res ; 11(3): 1003-12, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23917481

ABSTRACT

Anterior foregut endoderm (AFE) gives rise to therapeutically relevant cell types in tissues such as the esophagus, salivary glands, lung, thymus, parathyroid and thyroid. Despite its importance, reports describing the generation of AFE from pluripotent stem cells (PSCs) by directed differentiation have mainly focused on the Nkx2.1(+) lung and thyroid lineages. Here, we describe a novel protocol to derive a subdomain of AFE, identified by expression of Pax9, from PSCs using small molecules and defined media conditions. We generated a reporter PSC line for isolation and characterization of Pax9(+) AFE cells, which when transplanted in vivo, can form several distinct complex AFE-derived epithelia, including mucosal glands and stratified squamous epithelium. Finally, we show that the directed differentiation protocol can be used to generate AFE from human PSCs. Thus, this work both broadens the range of PSC-derived AFE tissues and creates a platform enabling the study of AFE disorders.


Subject(s)
Epithelial Cells/cytology , Epithelial Cells/drug effects , Pluripotent Stem Cells/drug effects , Small Molecule Libraries/pharmacology , Animals , Cell Differentiation/drug effects , Cell Line , Cell Lineage/drug effects , Culture Media/pharmacology , Embryonic Stem Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/transplantation , Hepatocyte Nuclear Factor 3-beta/metabolism , Humans , Mice , Nuclear Proteins/metabolism , PAX9 Transcription Factor/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , SOXB1 Transcription Factors/metabolism , Thyroid Nuclear Factor 1 , Transcription Factors/metabolism , Transcriptome
12.
Blood ; 117(20): 5514-22, 2011 May 19.
Article in English | MEDLINE | ID: mdl-21389323

ABSTRACT

Hemophilia A and B are caused by deficiencies in coagulation factor VIII (FVIII) and factor IX, respectively, resulting in deficient blood coagulation via the intrinsic pathway. The extrinsic coagulation pathway, mediated by factor VIIa and tissue factor (TF), remains intact but is negatively regulated by tissue factor pathway inhibitor (TFPI), which inhibits both factor VIIa and its product, factor Xa. This inhibition limits clot initiation via the extrinsic pathway, whereas factor deficiency in hemophilia limits clot propagation via the intrinsic pathway. ARC19499 is an aptamer that inhibits TFPI, thereby enabling clot initiation and propagation via the extrinsic pathway. The core aptamer binds tightly and specifically to TFPI. ARC19499 blocks TFPI inhibition of both factor Xa and the TF/factor VIIa complex. ARC19499 corrects thrombin generation in hemophilia A and B plasma and restores clotting in FVIII-neutralized whole blood. In the present study, using a monkey model of hemophilia, FVIII neutralization resulted in prolonged clotting times as measured by thromboelastography and prolonged saphenous-vein bleeding times, which are consistent with FVIII deficiency. ARC19499 restored thromboelastography clotting times to baseline levels and corrected bleeding times. These results demonstrate that ARC19499 inhibition of TFPI may be an effective alternative to current treatments of bleeding associated with hemophilia.


Subject(s)
Aptamers, Nucleotide/pharmacology , Blood Coagulation/drug effects , Hemostasis/drug effects , Lipoproteins/antagonists & inhibitors , Animals , Aptamers, Nucleotide/chemistry , Bleeding Time , Disease Models, Animal , Factor VIII/metabolism , Factor VIIa/metabolism , Factor Xa/metabolism , Hemophilia A/blood , Hemophilia A/drug therapy , Hemophilia B/blood , Hemophilia B/drug therapy , Humans , In Vitro Techniques , Macaca fascicularis , Recombinant Proteins/antagonists & inhibitors , Thrombin/biosynthesis , Thromboplastin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...