Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Nature ; 614(7949): 742-751, 2023 02.
Article in English | MEDLINE | ID: mdl-36755098

ABSTRACT

Cell identity is governed by the complex regulation of gene expression, represented as gene-regulatory networks1. Here we use gene-regulatory networks inferred from single-cell multi-omics data to perform in silico transcription factor perturbations, simulating the consequent changes in cell identity using only unperturbed wild-type data. We apply this machine-learning-based approach, CellOracle, to well-established paradigms-mouse and human haematopoiesis, and zebrafish embryogenesis-and we correctly model reported changes in phenotype that occur as a result of transcription factor perturbation. Through systematic in silico transcription factor perturbation in the developing zebrafish, we simulate and experimentally validate a previously unreported phenotype that results from the loss of noto, an established notochord regulator. Furthermore, we identify an axial mesoderm regulator, lhx1a. Together, these results show that CellOracle can be used to analyse the regulation of cell identity by transcription factors, and can provide mechanistic insights into development and differentiation.


Subject(s)
Cell Differentiation , Computer Simulation , Gene Regulatory Networks , Transcription Factors , Animals , Humans , Mice , Cell Differentiation/genetics , Embryonic Development/genetics , Phenotype , Transcription Factors/metabolism , Zebrafish/embryology , Zebrafish/genetics , Mesoderm/enzymology , Mesoderm/metabolism , Hematopoiesis/genetics
2.
Nature ; 564(7735): 219-224, 2018 12.
Article in English | MEDLINE | ID: mdl-30518857

ABSTRACT

Direct lineage reprogramming involves the conversion of cellular identity. Single-cell technologies are useful for deconstructing the considerable heterogeneity that emerges during lineage conversion. However, lineage relationships are typically lost during cell processing, complicating trajectory reconstruction. Here we present 'CellTagging', a combinatorial cell-indexing methodology that enables parallel capture of clonal history and cell identity, in which sequential rounds of cell labelling enable the construction of multi-level lineage trees. CellTagging and longitudinal tracking of fibroblast to induced endoderm progenitor reprogramming reveals two distinct trajectories: one leading to successfully reprogrammed cells, and one leading to a 'dead-end' state, paths determined in the earliest stages of lineage conversion. We find that expression of a putative methyltransferase, Mettl7a1, is associated with the successful reprogramming trajectory; adding Mettl7a1 to the reprogramming cocktail increases the yield of induced endoderm progenitors. Together, these results demonstrate the utility of our lineage-tracing method for revealing the dynamics of direct reprogramming.


Subject(s)
Cell Lineage , Cell Tracking/methods , Cellular Reprogramming , Clone Cells/cytology , Single-Cell Analysis/methods , Animals , Cell Lineage/drug effects , Cell Separation , Cellular Reprogramming/drug effects , Clone Cells/drug effects , Endoderm/cytology , Endoderm/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , HEK293 Cells , Humans , Methyltransferases/metabolism , Mice , Stem Cells/cytology , Stem Cells/drug effects , Time Factors
3.
Int J Mol Sci ; 22(16)2021 Aug 09.
Article in English | MEDLINE | ID: mdl-34445257

ABSTRACT

The production of pancreatic ß cells is the most challenging step for curing diabetes using next-generation treatments. Adult pancreatic endocrine cells are thought to be maintained by the self-duplication of differentiated cells, and pancreatic endocrine neogenesis can only be observed when the tissue is severely damaged. Experimentally, this can be performed using a method named partial duct ligation (PDL). As the success rate of PDL surgery is low because of difficulties in identifying the pancreatic duct, we previously proposed a method for fluorescently labeling the duct in live animals. Using this method, we performed PDL on neurogenin3 (Ngn3)-GFP transgenic mice to determine the origin of endocrine precursor cells and evaluate their potential to differentiate into multiple cell types. Ngn3-activated cells, which were marked with GFP, appeared after PDL operation. Because some GFP-positive cells were aligned proximally to the duct, we hypothesized that Ngn3-positive cells arise from the pancreatic duct. Therefore, we next developed an in vitro pancreatic duct culture system using Ngn3-GFP mice and examined whether Ngn3-positive cells emerge from this duct. We observed GFP expressions in ductal organoid cultures. GFP expressions were correlated with Ngn3 expressions and endocrine cell lineage markers. Interestingly, tuft cell markers were also correlated with GFP expressions. Our results demonstrate that in adult mice, Ngn3-positive endocrine precursor cells arise from the pancreatic ducts both in vivo and in vitro experiments indicating that the pancreatic duct could be a potential donor for therapeutic use.


Subject(s)
Antigens, Differentiation/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Insulin-Secreting Cells/metabolism , Nerve Tissue Proteins/metabolism , Pancreatic Ducts/metabolism , Stem Cells/metabolism , Animals , Antigens, Differentiation/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Insulin-Secreting Cells/cytology , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Organoids/cytology , Organoids/metabolism , Pancreatic Ducts/cytology , Stem Cells/cytology
4.
J Biol Chem ; 293(17): 6214-6229, 2018 04 27.
Article in English | MEDLINE | ID: mdl-29523685

ABSTRACT

Under various conditions of liver injury, the intrahepatic biliary epithelium undergoes dynamic tissue expansion and remodeling, a process known as ductular reaction. Mouse models defective in inducing such a tissue-remodeling process are more susceptible to liver injury, suggesting a crucial role of this process in liver regeneration. However, the molecular mechanisms regulating the biliary epithelial cell (BEC) dynamics in the ductular reaction remain largely unclear. Here, we demonstrate that the transcription factor Krüppel-like factor 5 (Klf5) is highly enriched in mouse liver BECs and plays a key role in regulating the ductular reaction, specifically under cholestatic injury conditions. Although mice lacking Klf5 in the entire liver epithelium, including both hepatocytes and BECs (Klf5-LKO (liver epithelial-specific knockout) mice), did not exhibit any apparent phenotype in the hepatobiliary system under normal conditions, they exhibited significant defects in biliary epithelial tissue remodeling upon 3,5-diethoxycarbonyl-1,4-dihydrocollidine-induced cholangitis, concomitantly with exacerbated cholestasis and reduced survival rate. In contrast, mice lacking Klf5 solely in hepatocytes did not exhibit any such phenotypes, confirming Klf5's specific role in BECs. RNA-sequencing analyses of BECs isolated from the Klf5-LKO mouse livers revealed that the Klf5 deficiency primarily affected expression of cell cycle-related genes. Moreover, immunostaining analysis with the proliferation marker Ki67 disclosed that the Klf5-LKO mice had significantly reduced BEC proliferation levels upon injury. These results indicate that Klf5 plays a critical role in the ductular reaction and biliary epithelial tissue expansion and remodeling by inducing BEC proliferation and thereby contributing to liver regeneration.


Subject(s)
Bile Ducts, Intrahepatic/metabolism , Cholestasis/metabolism , Epithelial Cells/metabolism , Kruppel-Like Transcription Factors/biosynthesis , Liver Regeneration , Liver/metabolism , Animals , Bile Ducts, Intrahepatic/pathology , Cell Cycle/drug effects , Cholestasis/chemically induced , Cholestasis/genetics , Cholestasis/pathology , Epithelial Cells/pathology , Gene Expression Regulation/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Kruppel-Like Transcription Factors/genetics , Liver/injuries , Liver/pathology , Mice , Mice, Knockout , Pyridines/toxicity
5.
Hepatology ; 61(6): 2056-66, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25572923

ABSTRACT

UNLABELLED: Serving as the center for metabolism and detoxification, the liver is inherently susceptible to a wide variety of damage imposed by toxins or chemicals. Induction of cell populations with biliary epithelial phenotypes, which include progenitor-like cells and are referred to as liver progenitor cells, is often observed in histopathological examination of various liver diseases in both human patients and animal models and has been implicated in regeneration. However, the tissue dynamics underlying this phenomenon remains largely unclear. We have developed a simple imaging technique to reveal the global and fine-scale architecture of the biliary tract spreading in the mouse liver. Using this novel method, we show that the emergence and expansion of liver progenitor cells actually reflect structural transformation of the intrahepatic biliary tree in mouse liver injury models. The biliary branches expanded their area gradually and contiguously along with the course of chronic injury. Relevant regulatory signals known to be involved in liver progenitor cell regulation, including fibroblast growth factor 7 and tumor necrosis factor-like weak inducer of apoptosis, can modulate the dynamics of the biliary epithelium in different ways. Importantly, the structural transformations of the biliary tree were diverse and corresponded well with the parenchymal injury patterns. That is, when chronic hepatocyte damage was induced in the pericentral area, the biliary branches exhibited an extended structure from the periportal area with apparent tropism toward the distant injured area. CONCLUSION: The hepatobiliary system possesses a unique and unprecedented structural flexibility and can remodel dynamically and adaptively in response to various injury conditions; this type of tissue plasticity should constitute an essential component to maintain liver homeostasis.


Subject(s)
Bile Ducts, Intrahepatic/cytology , Bile Ducts, Intrahepatic/physiology , Stem Cells/physiology , Adaptation, Physiological , Animals , Homeostasis , Mice, Inbred C57BL
6.
Stem Cell Reports ; 18(1): 97-112, 2023 01 10.
Article in English | MEDLINE | ID: mdl-36584685

ABSTRACT

In direct lineage conversion, transcription factor (TF) overexpression reconfigures gene regulatory networks (GRNs) to reprogram cell identity. We previously developed CellOracle, a computational method to infer GRNs from single-cell transcriptome and epigenome data. Using inferred GRNs, CellOracle simulates gene expression changes in response to TF perturbation, enabling in silico interrogation of network reconfiguration. Here, we combine CellOracle analysis with lineage tracing of fibroblast to induced endoderm progenitor (iEP) conversion, a prototypical direct reprogramming paradigm. By linking early network state to reprogramming outcome, we reveal distinct network configurations underlying successful and failed fate conversion. Via in silico simulation of TF perturbation, we identify new factors to coax cells into successfully converting their identity, uncovering a central role for the AP-1 subunit Fos with the Hippo signaling effector, Yap1. Together, these results demonstrate the efficacy of CellOracle to infer and interpret cell-type-specific GRN configurations, providing new mechanistic insights into lineage reprogramming.


Subject(s)
Gene Expression Regulation , Gene Regulatory Networks , Transcription Factors/genetics , Transcriptome , Fibroblasts , Cellular Reprogramming/genetics
7.
Nat Biotechnol ; 2023 Sep 25.
Article in English | MEDLINE | ID: mdl-37749269

ABSTRACT

Complex gene regulatory mechanisms underlie differentiation and reprogramming. Contemporary single-cell lineage-tracing (scLT) methods use expressed, heritable DNA barcodes to combine cell lineage readout with single-cell transcriptomics. However, reliance on transcriptional profiling limits adaptation to other single-cell assays. With CellTag-multi, we present an approach that enables direct capture of heritable random barcodes expressed as polyadenylated transcripts, in both single-cell RNA sequencing and single-cell Assay for Transposase Accessible Chromatin using sequencing assays, allowing for independent clonal tracking of transcriptional and epigenomic cell states. We validate CellTag-multi to characterize progenitor cell lineage priming during mouse hematopoiesis. Additionally, in direct reprogramming of fibroblasts to endoderm progenitors, we identify core regulatory programs underlying on-target and off-target fates. Furthermore, we reveal the transcription factor Zfp281 as a regulator of reprogramming outcome, biasing cells toward an off-target mesenchymal fate. Our results establish CellTag-multi as a lineage-tracing method compatible with multiple single-cell modalities and demonstrate its utility in revealing fate-specifying gene regulatory changes across diverse paradigms of differentiation and reprogramming.

8.
Cancer Gene Ther ; 30(1): 38-50, 2023 01.
Article in English | MEDLINE | ID: mdl-35999358

ABSTRACT

The Philadelphia (Ph) chromosome was the first translocation identified in leukemia. It is supposed to be generated by aberrant ligation between two DNA double-strand breaks (DSBs) at the BCR gene located on chromosome 9q34 and the ABL1 gene located on chromosome 22q11. Thus, mimicking the initiation process of translocation, we induced CRISPR/Cas9-mediated DSBs simultaneously at the breakpoints of the BCR and ABL1 genes in a granulocyte-macrophage colony-stimulating factor (GM-CSF) dependent human leukemia cell line. After transfection of two single guide RNAs (sgRNAs) targeting intron 13 of the BCR gene and intron 1 of the ABL1 gene, a factor-independent subline was obtained. In the subline, p210 BCR::ABL1 and its reciprocal ABL1::BCR fusions were generated as a result of balanced translocation corresponding to the Ph chromosome. Another set of sgRNAs targeting intron 1 of the BCR gene and intron 1 of the ABL1 gene induced a factor-independent subline expressing p190 BCR::ABL1. Both p210 and p190 BCR::ABL1 induced factor-independent growth by constitutively activating intracellular signaling pathways for transcriptional regulation of cell cycle progression and cell survival that are usually regulated by GM-CSF. These observations suggested that simultaneous DSBs at the BCR and ABL1 gene breakpoints are initiation events for oncogenesis in Ph+ leukemia. (200/200 words).


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Philadelphia Chromosome , Humans , Fusion Proteins, bcr-abl/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , CRISPR-Cas Systems , Translocation, Genetic , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Carcinogenesis/genetics
9.
Nat Cardiovasc Res ; 2(4): 399-416, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37583573

ABSTRACT

Recovery of cardiac function is the holy grail of heart failure therapy yet is infrequently observed and remains poorly understood. In this study, we performed single-nucleus RNA sequencing from patients with heart failure who recovered left ventricular systolic function after left ventricular assist device implantation, patients who did not recover and non-diseased donors. We identified cell-specific transcriptional signatures of recovery, most prominently in macrophages and fibroblasts. Within these cell types, inflammatory signatures were negative predictors of recovery, and downregulation of RUNX1 was associated with recovery. In silico perturbation of RUNX1 in macrophages and fibroblasts recapitulated the transcriptional state of recovery. Cardiac recovery mediated by BET inhibition in mice led to decreased macrophage and fibroblast Runx1 expression and diminished chromatin accessibility within a Runx1 intronic peak and acquisition of human recovery signatures. These findings suggest that cardiac recovery is a unique biological state and identify RUNX1 as a possible therapeutic target to facilitate cardiac recovery.

10.
J Clin Invest ; 131(19)2021 10 01.
Article in English | MEDLINE | ID: mdl-34343135

ABSTRACT

Epithelial cells are charged with protection at barrier sites, but whether this normally beneficial response might sometimes become dysfunctional still needs definition. Here, we recognized a pattern of imbalance marked by basal epithelial cell growth and differentiation that replaced normal airspaces in a mouse model of progressive postviral lung disease due to the Sendai virus. Single-cell and lineage-tracing technologies identified a distinct subset of basal epithelial stem cells (basal ESCs) that extended into gas-exchange tissue to form long-term bronchiolar-alveolar remodeling regions. Moreover, this cell subset was selectively expanded by crossing a cell-growth and survival checkpoint linked to the nuclear-localized alarmin IL-33 that was independent of IL-33 receptor signaling and instead connected to autocrine chromatin accessibility. This mechanism creates an activated stem-progenitor cell lineage with potential for physiological or pathological function. Thus, conditional loss of Il33 gene function in basal epithelial cells disrupted the homeostasis of the epithelial barrier at skin and gut sites but also markedly attenuated postviral disease in the lung based on the downregulation of remodeling and inflammation. Thus, we define a basal ESC strategy to deploy innate immune machinery that appears to overshoot the primordial goal of self-defense. Our findings reveal new targets to stratify and correct chronic and often deadly postviral disease.


Subject(s)
Alarmins/physiology , Epithelial Cells/physiology , Interleukin-33/physiology , Lung Diseases/physiopathology , Respirovirus Infections/complications , Sendai virus , Stem Cells/physiology , Animals , Cell Differentiation , Interleukin-33/genetics , Mice , Single-Cell Analysis , Stem Cells/cytology
11.
Science ; 372(6542)2021 05 07.
Article in English | MEDLINE | ID: mdl-33958447

ABSTRACT

Deciphering how the human striatum develops is necessary for understanding the diseases that affect this region. To decode the transcriptional modules that regulate this structure during development, we compiled a catalog of 1116 long intergenic noncoding RNAs (lincRNAs) identified de novo and then profiled 96,789 single cells from the early human fetal striatum. We found that D1 and D2 medium spiny neurons (D1- and D2-MSNs) arise from a common progenitor and that lineage commitment is established during the postmitotic transition, across a pre-MSN phase that exhibits a continuous spectrum of fate determinants. We then uncovered cell type-specific gene regulatory networks that we validated through in silico perturbation. Finally, we identified human-specific lincRNAs that contribute to the phylogenetic divergence of this structure in humans. This work delineates the cellular hierarchies governing MSN lineage commitment.


Subject(s)
Atlases as Topic , Corpus Striatum/cytology , Corpus Striatum/embryology , Neurogenesis/genetics , RNA, Long Noncoding/genetics , Single-Cell Analysis , Transcription Factors/genetics , Fetus , GABAergic Neurons/metabolism , Humans , RNA-Seq , Transcription, Genetic
12.
Commun Biol ; 3(1): 289, 2020 06 05.
Article in English | MEDLINE | ID: mdl-32503996

ABSTRACT

Upon severe and/or chronic liver injury, ectopic emergence and expansion of atypical biliary epithelial-like cells in the liver parenchyma, known as the ductular reaction, is typically induced and implicated in organ regeneration. Although this phenomenon has long been postulated to represent activation of facultative liver stem/progenitor cells that give rise to new hepatocytes, recent lineage-tracing analyses have challenged this notion, thereby leaving the pro-regenerative role of the ductular reaction enigmatic. Here, we show that the expanded and remodelled intrahepatic biliary epithelia in the ductular reaction constituted functional and complementary bile-excreting conduit systems in injured parenchyma where hepatocyte bile canalicular networks were lost. The canalicular collapse was an incipient defect commonly associated with hepatocyte injury irrespective of cholestatic statuses, and could sufficiently provoke the ductular reaction when artificially induced. We propose a unifying model for the induction of the ductular reaction, where compensatory biliary epithelial tissue remodeling ensures bile-excreting network homeostasis.


Subject(s)
Bile Ducts, Intrahepatic/cytology , Chemical and Drug Induced Liver Injury/prevention & control , Disease Models, Animal , Epithelial Cells/cytology , Hepatocytes/cytology , Animals , Bile Ducts, Intrahepatic/physiology , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/pathology , Epithelial Cells/physiology , Female , Hepatocytes/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
13.
Nat Protoc ; 15(3): 750-772, 2020 03.
Article in English | MEDLINE | ID: mdl-32051617

ABSTRACT

Single-cell technologies are offering unparalleled insight into complex biology, revealing the behavior of rare cell populations that are masked in bulk population analyses. One current limitation of single-cell approaches is that lineage relationships are typically lost as a result of cell processing. We recently established a method, CellTagging, permitting the parallel capture of lineage information and cell identity via a combinatorial cell indexing approach. CellTagging integrates with high-throughput single-cell RNA sequencing, where sequential rounds of cell labeling enable the construction of multi-level lineage trees. Here, we provide a detailed protocol to (i) generate complex plasmid and lentivirus CellTag libraries for labeling of cells; (ii) sequentially CellTag cells over the course of a biological process; (iii) profile single-cell transcriptomes via high-throughput droplet-based platforms; and (iv) generate a CellTag expression matrix, followed by clone calling and lineage reconstruction. This lentiviral-labeling approach can be deployed in any organism or in vitro culture system that is amenable to viral transduction to simultaneously profile lineage and identity at single-cell resolution.


Subject(s)
Cell Lineage , Cell Tracking/methods , Fibroblasts/physiology , Animals , Cell Line , Escherichia coli , Gene Expression Regulation , Humans , Mice
14.
Methods Mol Biol ; 1905: 71-82, 2019.
Article in English | MEDLINE | ID: mdl-30536091

ABSTRACT

Recent technological advances have revealed the heterogeneity of cells and tissues. Existence of heterogeneity in hepatic progenitor cells is becoming apparent by various experimental approaches, and here we describe a series of techniques to investigate the proliferative heterogeneity of these cells. We have developed a new technique by combining genetic lineage tracking and three-dimensional imaging methods. The data obtained can be used in statistical analysis to quantitatively investigate the mechanisms underlying the heterogeneity of hepatic progenitor cells.


Subject(s)
Imaging, Three-Dimensional/methods , Liver/cytology , Stem Cells/cytology , Animals , Cell Differentiation , Cell Lineage , Cell Proliferation , Mice , Single-Cell Analysis
15.
Genome Biol ; 20(1): 90, 2019 05 09.
Article in English | MEDLINE | ID: mdl-31072405

ABSTRACT

High-throughput single-cell assays increasingly require special consideration in experimental design, sample multiplexing, batch effect removal, and data interpretation. Here, we describe a lentiviral barcode-based multiplexing approach, CellTag Indexing, which uses predefined genetic barcodes that are heritable, enabling cell populations to be tagged, pooled, and tracked over time in the same experimental replicate. We demonstrate the utility of CellTag Indexing by sequencing transcriptomes using a variety of cell types, including long-term tracking of cell engraftment and differentiation in vivo. Together, this presents CellTag Indexing as a broadly applicable genetic multiplexing tool that is complementary with existing single-cell technologies.


Subject(s)
Cell Tracking/methods , Genomics/methods , Single-Cell Analysis , Lentivirus , Transcriptome
16.
Cell Mol Gastroenterol Hepatol ; 8(3): 407-426, 2019.
Article in English | MEDLINE | ID: mdl-31195149

ABSTRACT

BACKGROUND & AIMS: The small intestine (SI) displays regionality in nutrient and immunological function. Following SI tissue loss (as occurs in short gut syndrome, or SGS), remaining SI must compensate, or "adapt"; the capacity of SI epithelium to reprogram its regional identity has not been described. Here, we apply single-cell resolution analyses to characterize molecular changes underpinning adaptation to SGS. METHODS: Single-cell RNA sequencing was performed on epithelial cells isolated from distal SI of mice following 50% proximal small bowel resection (SBR) vs sham surgery. Single-cell profiles were clustered based on transcriptional similarity, reconstructing differentiation events from intestinal stem cells (ISCs) through to mature enterocytes. An unsupervised computational approach to score cell identity was used to quantify changes in regional (proximal vs distal) SI identity, validated using immunofluorescence, immunohistochemistry, qPCR, western blotting, and RNA-FISH. RESULTS: Uniform Manifold Approximation and Projection-based clustering and visualization revealed differentiation trajectories from ISCs to mature enterocytes in sham and SBR. Cell identity scoring demonstrated segregation of enterocytes by regional SI identity: SBR enterocytes assumed more mature proximal identities. This was associated with significant upregulation of lipid metabolism and oxidative stress gene expression, which was validated via orthogonal analyses. Observed upstream transcriptional changes suggest retinoid metabolism and proximal transcription factor Creb3l3 drive proximalization of cell identity in response to SBR. CONCLUSIONS: Adaptation to proximal SBR involves regional reprogramming of ileal enterocytes toward a proximal identity. Interventions bolstering the endogenous reprogramming capacity of SI enterocytes-conceivably by engaging the retinoid metabolism pathway-merit further investigation, as they may increase enteral feeding tolerance, and obviate intestinal failure, in SGS.


Subject(s)
Gene Expression Profiling/methods , Gene Regulatory Networks , Intestine, Small/surgery , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Animals , Cellular Reprogramming , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Enterocytes/chemistry , Enterocytes/cytology , Intestine, Small/chemistry , Lipid Metabolism , Male , Mice , Oxidative Stress , RNA, Small Nuclear/pharmacology , Unsupervised Machine Learning , Up-Regulation
17.
Cell Syst ; 6(1): 5-7, 2018 01 24.
Article in English | MEDLINE | ID: mdl-29401450

ABSTRACT

Here, we outline p-Creode, a new algorithm to construct multi-branching cell lineage trajectories from single-cell data. Application of this platform to diverse sources of single-cell data demonstrates its robustness and scalability, while the discovery of a new origin for rare gut tuft cells showcases the utility of p-Creode.


Subject(s)
RNA , Single-Cell Analysis , Algorithms , Cell Lineage
18.
Tissue Eng Part C Methods ; 24(8): 480-485, 2018 08.
Article in English | MEDLINE | ID: mdl-29993334

ABSTRACT

Proper identification of pancreatic ducts is a major challenge for researchers performing partial duct ligation (PDL), because pancreatic ducts, which are covered with acinar cells, are translucent and thin. Although damage to pancreatic ducts may activate quiescent ductal stem cells, which may allow further investigation into ductal stem cells for therapeutic use, there is a lack of effective techniques to visualize pancreatic ducts. In this study, we report a new method for identifying pancreatic ducts. First, we aimed to visualize pancreatic ducts using black, waterproof fountain pen ink. We injected the ink into pancreatic ducts through the bile duct. The flow of ink was observed in pancreatic ducts, revealing their precise architecture. Next, to visualize pancreatic ducts in live animals, we injected fluorescein-labeled bile acid, cholyl-lysyl-fluorescein into the mouse tail vein. The fluorescent probe clearly marked not only the bile duct but also pancreatic ducts when observed with a fluorescent microscope. To confirm whether the pancreatic duct labeling was successful, we performed PDL on Neurogenin3 (Ngn3)-GFP transgenic mice. As a result, acinar tissue is lost. PDL tail pancreas becomes translucent almost completely devoid of acinar cells. Furthermore, strong activation of Ngn3 expression was observed in the ligated part of the adult mouse pancreas at 7 days after PDL.


Subject(s)
Pancreatic Ducts/physiology , Tissue Engineering/methods , Animals , Cholic Acids/metabolism , Fluoresceins/metabolism , Fluorescent Dyes/metabolism , Ligation , Mice, Inbred C57BL
19.
Elife ; 52016 07 19.
Article in English | MEDLINE | ID: mdl-27431614

ABSTRACT

Dynamic remodeling of the intrahepatic biliary epithelial tissue plays key roles in liver regeneration, yet the cellular basis for this process remains unclear. We took an unbiased approach based on in vivo clonal labeling and tracking of biliary epithelial cells in the three-dimensional landscape, in combination with mathematical simulation, to understand their mode of proliferation in a mouse liver injury model where the nascent biliary structure formed in a tissue-intrinsic manner. An apparent heterogeneity among biliary epithelial cells was observed: whereas most of the responders that entered the cell cycle upon injury exhibited a limited and tapering growth potential, a select population continued to proliferate, making a major contribution in sustaining the biliary expansion. Our study has highlighted a unique mode of epithelial tissue dynamics, which depends not on a hierarchical system driven by fixated stem cells, but rather, on a stochastically maintained progenitor population with persistent proliferative activity.


Subject(s)
Epithelial Cells/physiology , Epithelium/physiology , Liver/injuries , Liver/physiology , Regeneration , Animals , Disease Models, Animal , Mice , Models, Theoretical
SELECTION OF CITATIONS
SEARCH DETAIL