Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
J Theor Biol ; 582: 111743, 2024 04 07.
Article in English | MEDLINE | ID: mdl-38307450

ABSTRACT

OBJECTIVE: Owing to the heterogeneity in the evolution of cancer, distinguishing between diverse growth patterns and predicting long-term outcomes based on short-term measurements poses a great challenge. METHODS: A novel multiscale framework is proposed to unravel the connections between the population dynamics of cancer growth (i.e., aggressive, bounded, and indolent) and the cellular-subclonal dynamics of cancer evolution. This framework employs the non-negative lasso (NN-LASSO) algorithm to forge a link between an ordinary differential equation (ODE)-based population model and a cellular evolution model. RESULTS: The findings of our current work not only affirm the impact of subclonal composition on growth dynamics but also identify two significant subclones within heterogeneous growth patterns. Moreover, the subclonal compositions at the initial time are able to accurately discriminate diverse growth patterns through a machine learning algorithm. CONCLUSION: The proposed multiscale framework successfully delineates the intricate landscape of cancer evolution, bridging the gap between long-term growth dynamics and short-term measurements, both in simulated and real-world data. This methodology provides a novel avenue for thorough exploration into the realm of cancer evolution.


Subject(s)
Neoplasms , Humans , Neoplasms/genetics , Algorithms , Polymorphism, Single Nucleotide , High-Throughput Nucleotide Sequencing/methods
2.
Genome Biol ; 24(1): 166, 2023 Jul 13.
Article in English | MEDLINE | ID: mdl-37443062

ABSTRACT

BACKGROUND: The oocyte-to-embryo transition (OET) converts terminally differentiated gametes into a totipotent embryo and is critically controlled by maternal mRNAs and proteins, while the genome is silent until zygotic genome activation. How the transcriptome, translatome, and proteome are coordinated during this critical developmental window remains poorly understood. RESULTS: Utilizing a highly sensitive and quantitative mass spectrometry approach, we obtain high-quality proteome data spanning seven mouse stages, from full-grown oocyte (FGO) to blastocyst, using 100 oocytes/embryos at each stage. Integrative analyses reveal distinct proteome reprogramming compared to that of the transcriptome or translatome. FGO to 8-cell proteomes are dominated by FGO-stockpiled proteins, while the transcriptome and translatome are more dynamic. FGO-originated proteins frequently persist to blastocyst while corresponding transcripts are already downregulated or decayed. Improved concordance between protein and translation or transcription is observed for genes starting translation upon meiotic resumption, as well as those transcribed and translated only in embryos. Concordance between protein and transcription/translation is also observed for proteins with short half-lives. We built a kinetic model that predicts protein dynamics by incorporating both initial protein abundance in FGOs and translation kinetics across developmental stages. CONCLUSIONS: Through integrative analyses of datasets generated by ultrasensitive methods, our study reveals that the proteome shows distinct dynamics compared to the translatome and transcriptome during mouse OET. We propose that the remarkably stable oocyte-originated proteome may help save resources to accommodate the demanding needs of growing embryos. This study will advance our understanding of mammalian OET and the fundamental principles governing gene expression.


Subject(s)
Proteome , Transcriptome , Animals , Mice , Proteome/metabolism , Embryo, Mammalian/metabolism , Blastocyst/metabolism , Oocytes/metabolism , Gene Expression Regulation, Developmental , Mammals/metabolism
3.
BMC Cancer ; 23(1): 712, 2023 Jul 31.
Article in English | MEDLINE | ID: mdl-37525139

ABSTRACT

BACKGROUND: Endometrial Cancer (EC) is one of the most prevalent malignancies that affect the female population globally. In the context of immunotherapy, Tumor Mutation Burden (TMB) in the DNA polymerase epsilon (POLE) subtype of this cancer holds promise as a viable therapeutic target. METHODS: We devised a method known as NEM-TIE to forecast the TMB status of patients with endometrial cancer. This approach utilized a combination of the Network Evolution Model, Transfer Information Entropy, Clique Percolation (CP) methodology, and Support Vector Machine (SVM) classification. To construct the Network Evolution Model, we employed an adjacency matrix that utilized transfer information entropy to assess the information gain between nodes of radiomic-clinical features. Subsequently, using the CP algorithm, we unearthed potentially pivotal modules in the Network Evolution Model. Finally, the SVM classifier extracted essential features from the module set. RESULTS: Upon analyzing the importance of modules, we discovered that the dependence count energy in tumor volumes-of-interest holds immense significance in distinguishing TMB statuses among patients with endometrial cancer. Using the 13 radiomic-clinical features extracted via NEM-TIE, we demonstrated that the area under the receiver operating characteristic curve (AUROC) in the test set is 0.98 (95% confidence interval: 0.95-1.00), surpassing the performance of existing techniques such as the mRMR and Laplacian methods. CONCLUSIONS: Our study proposed the NEM-TIE method as a means to identify the TMB status of patients with endometrial cancer. The integration of radiomic-clinical data utilizing the NEM-TIE method may offer a novel technology for supplementary diagnosis.


Subject(s)
Brain Neoplasms , Endometrial Neoplasms , Humans , Female , Magnetic Resonance Imaging/methods , Brain Neoplasms/genetics , ROC Curve , Endometrial Neoplasms/diagnostic imaging , Endometrial Neoplasms/genetics , Mutation , Retrospective Studies
4.
Nature ; 618(7966): 808-817, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37344645

ABSTRACT

Niche signals maintain stem cells in a prolonged quiescence or transiently activate them for proper regeneration1. Altering balanced niche signalling can lead to regenerative disorders. Melanocytic skin nevi in human often display excessive hair growth, suggesting hair stem cell hyperactivity. Here, using genetic mouse models of nevi2,3, we show that dermal clusters of senescent melanocytes drive epithelial hair stem cells to exit quiescence and change their transcriptome and composition, potently enhancing hair renewal. Nevus melanocytes activate a distinct secretome, enriched for signalling factors. Osteopontin, the leading nevus signalling factor, is both necessary and sufficient to induce hair growth. Injection of osteopontin or its genetic overexpression is sufficient to induce robust hair growth in mice, whereas germline and conditional deletions of either osteopontin or CD44, its cognate receptor on epithelial hair cells, rescue enhanced hair growth induced by dermal nevus melanocytes. Osteopontin is overexpressed in human hairy nevi, and it stimulates new growth of human hair follicles. Although broad accumulation of senescent cells, such as upon ageing or genotoxic stress, is detrimental for the regenerative capacity of tissue4, we show that signalling by senescent cell clusters can potently enhance the activity of adjacent intact stem cells and stimulate tissue renewal. This finding identifies senescent cells and their secretome as an attractive therapeutic target in regenerative disorders.


Subject(s)
Hair , Melanocytes , Signal Transduction , Animals , Mice , Hair/cytology , Hair/growth & development , Hair Follicle/cytology , Hair Follicle/physiology , Hyaluronan Receptors/metabolism , Melanocytes/cytology , Melanocytes/metabolism , Nevus/metabolism , Nevus/pathology , Osteopontin/metabolism , Stem Cells/cytology
5.
Math Biosci Eng ; 20(2): 2890-2907, 2023 01.
Article in English | MEDLINE | ID: mdl-36899563

ABSTRACT

Radiomics, providing quantitative data extracted from medical images, has emerged as a critical role in diagnosis and classification of diseases such as glioma. One main challenge is how to uncover key disease-relevant features from the large amount of extracted quantitative features. Many existing methods suffer from low accuracy or overfitting. We propose a new method, Multiple-Filter and Multi-Objective-based method (MFMO), to identify predictive and robust biomarkers for disease diagnosis and classification. This method combines a multi-filter feature extraction with a multi-objective optimization-based feature selection model, which identifies a small set of predictive radiomic biomarkers with less redundancy. Taking magnetic resonance imaging (MRI) images-based glioma grading as a case study, we identify 10 key radiomic biomarkers that can accurately distinguish low-grade glioma (LGG) from high-grade glioma (HGG) on both training and test datasets. Using these 10 signature features, the classification model reaches training Area Under the receiving operating characteristic Curve (AUC) of 0.96 and test AUC of 0.95, which shows superior performance over existing methods and previously identified biomarkers.


Subject(s)
Brain Neoplasms , Glioma , Humans , Brain Neoplasms/pathology , Retrospective Studies , Glioma/pathology , Magnetic Resonance Imaging/methods
6.
Nat Commun ; 14(1): 46, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36596814

ABSTRACT

Spinal motor neurons (MNs) integrate sensory stimuli and brain commands to generate movements. In vertebrates, the molecular identities of the cardinal MN types such as those innervating limb versus trunk muscles are well elucidated. Yet the identities of finer subtypes within these cell populations that innervate individual muscle groups remain enigmatic. Here we investigate heterogeneity in mouse MNs using single-cell transcriptomics. Among limb-innervating MNs, we reveal a diverse neuropeptide code for delineating putative motor pool identities. Additionally, we uncover that axial MNs are subdivided into three molecularly distinct subtypes, defined by mediolaterally-biased Satb2, Nr2f2 or Bcl11b expression patterns with different axon guidance signatures. These three subtypes are present in chicken and human embryos, suggesting a conserved axial MN expression pattern across higher vertebrates. Overall, our study provides a molecular resource of spinal MN types and paves the way towards deciphering how neuronal subtypes evolved to accommodate vertebrate motor behaviors.


Subject(s)
Motor Neurons , Transcriptome , Animals , Mice , Humans , Transcriptome/genetics , Motor Neurons/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Muscle, Skeletal/metabolism , Embryo, Mammalian/metabolism , Spinal Cord/metabolism , Mammals/metabolism , Repressor Proteins/metabolism , Tumor Suppressor Proteins/metabolism
7.
Nucleic Acids Res ; 50(21): 12112-12130, 2022 11 28.
Article in English | MEDLINE | ID: mdl-36440766

ABSTRACT

Although single-cell sequencing has provided a powerful tool to deconvolute cellular heterogeneity of diseases like cancer, extrapolating clinical significance or identifying clinically-relevant cells remains challenging. Here, we propose a novel computational method scAB, which integrates single-cell genomics data with clinically annotated bulk sequencing data via a knowledge- and graph-guided matrix factorization model. Once combined, scAB provides a coarse- and fine-grain multiresolution perspective of phenotype-associated cell states and prognostic signatures previously not visible by single-cell genomics. We use scAB to enhance live cancer single-cell RNA-seq data, identifying clinically-relevant previously unrecognized cancer and stromal cell subsets whose signatures show a stronger poor-survival association. The identified fine-grain cell subsets are associated with distinct cancer hallmarks and prognosis power. Furthermore, scAB demonstrates its utility as a biomarker identification tool, with the ability to predict immunotherapy, drug responses and survival when applied to melanoma single-cell RNA-seq datasets and glioma single-cell ATAC-seq datasets. Across multiple single-cell and bulk datasets from different cancer types, we also demonstrate the superior performance of scAB in generating prognosis signatures and survival predictions over existing models. Overall, scAB provides an efficient tool for prioritizing clinically-relevant cell subsets and predictive signatures, utilizing large publicly available databases to improve prognosis and treatments.


Subject(s)
Gene Expression Profiling , Melanoma , Humans , Gene Expression Profiling/methods , Clinical Relevance , Genomics , Prognosis , Melanoma/genetics , Single-Cell Analysis/methods
8.
Cell Rep ; 40(5): 111155, 2022 08 02.
Article in English | MEDLINE | ID: mdl-35926463

ABSTRACT

Delayed and often impaired wound healing in the elderly presents major medical and socioeconomic challenges. A comprehensive understanding of the cellular/molecular changes that shape complex cell-cell communications in aged skin wounds is lacking. Here, we use single-cell RNA sequencing to define the epithelial, fibroblast, immune cell types, and encompassing heterogeneities in young and aged skin during homeostasis and identify major changes in cell compositions, kinetics, and molecular profiles during wound healing. Our comparative study uncovers a more pronounced inflammatory phenotype in aged skin wounds, featuring neutrophil persistence and higher abundance of an inflammatory/glycolytic Arg1Hi macrophage subset that is more likely to signal to fibroblasts via interleukin (IL)-1 than in young counterparts. We predict systems-level differences in the number, strength, route, and signaling mediators of putative cell-cell communications in young and aged skin wounds. Our study exposes numerous cellular/molecular targets for functional interrogation and provides a hypothesis-generating resource for future wound healing studies.


Subject(s)
Fibroblasts , Wound Healing , Cell Communication , Fibroblasts/metabolism , Macrophages/metabolism , Signal Transduction , Skin
9.
JCI Insight ; 7(13)2022 07 08.
Article in English | MEDLINE | ID: mdl-35653192

ABSTRACT

Vitiligo is an autoimmune skin disease characterized by the destruction of melanocytes by autoreactive CD8+ T cells. Melanocyte destruction in active vitiligo is mediated by CD8+ T cells, but the persistence of white patches in stable disease is poorly understood. The interaction between immune cells, melanocytes, and keratinocytes in situ in human skin has been difficult to study due to the lack of proper tools. We combine noninvasive multiphoton microscopy (MPM) imaging and single-cell RNA-Seq (scRNA-Seq) to identify subpopulations of keratinocytes in stable vitiligo patients. We show that, compared with nonlesional skin, some keratinocyte subpopulations are enriched in lesional vitiligo skin and shift their energy utilization toward oxidative phosphorylation. Systematic investigation of cell-to-cell communication networks show that this small population of keratinocyte secrete CXCL9 and CXCL10 to potentially drive vitiligo persistence. Pseudotemporal dynamics analyses predict an alternative differentiation trajectory that generates this new population of keratinocytes in vitiligo skin. Further MPM imaging of patients undergoing punch grafting treatment showed that keratinocytes favoring oxidative phosphorylation persist in nonresponders but normalize in responders. In summary, we couple advanced imaging with transcriptomics and bioinformatics to discover cell-to-cell communication networks and keratinocyte cell states that can perpetuate inflammation and prevent repigmentation.


Subject(s)
Vitiligo , CD8-Positive T-Lymphocytes , Humans , Keratinocytes , Melanocytes , Skin
10.
Cell Rep Methods ; 2(5): 100207, 2022 05 23.
Article in English | MEDLINE | ID: mdl-35637911

ABSTRACT

In vivo calcium imaging enables simultaneous recording of large neuronal ensembles engaged in complex operations. Many experiments require monitoring and identification of cell populations across multiple sessions. Population cell tracking across multiple sessions is complicated by non-rigid transformations induced by cell movement and imaging field shifts. We introduce SCOUT (Single-Cell spatiOtemporal longitUdinal Tracking), a fast, robust cell-tracking method utilizing multiple cell-cell similarity metrics, probabilistic inference, and an adaptive clustering methodology, to perform cell identification across multiple sessions. By comparing SCOUT with earlier cell-tracking algorithms on simulated, 1-photon, and 2-photon recordings, we show that our approach significantly improves cell-tracking quality, particularly when recordings exhibit spatial footprint movement between sessions or sub-optimal neural extraction quality.


Subject(s)
Calcium, Dietary , Neurons , Neurons/physiology , Algorithms , Movement
11.
Biochem Soc Trans ; 50(1): 297-308, 2022 02 28.
Article in English | MEDLINE | ID: mdl-35191953

ABSTRACT

Tissue development and homeostasis require coordinated cell-cell communication. Recent advances in single-cell sequencing technologies have emerged as a revolutionary method to reveal cellular heterogeneity with unprecedented resolution. This offers a great opportunity to explore cell-cell communication in tissues systematically and comprehensively, and to further identify signaling mechanisms driving cell fate decisions and shaping tissue phenotypes. Using gene expression information from single-cell transcriptomics, several computational tools have been developed for inferring cell-cell communication, greatly facilitating analysis and interpretation. However, in single-cell transcriptomics, spatial information of cells is inherently lost. Given that most cell signaling events occur within a limited distance in tissues, incorporating spatial information into cell-cell communication analysis is critical for understanding tissue organization and function. Spatial transcriptomics provides spatial location of cell subsets along with their gene expression, leading to new directions for leveraging spatial information to develop computational approaches for cell-cell communication inference and analysis. These computational approaches have been successfully applied to uncover previously unrecognized mechanisms of intercellular communication within various contexts and across organ systems, including the skin, a formidable model to study mechanisms of cell-cell communication due to the complex interactions between the different cell populations that comprise it. Here, we review emergent cell-cell communication inference tools using single-cell transcriptomics and spatial transcriptomics, and highlight the biological insights gained by applying these computational tools to exploring cellular communication in skin development, homeostasis, disease and aging, as well as discuss future potential research avenues.


Subject(s)
Cell Communication , Transcriptome , Cell Differentiation , Communication , Computational Biology/methods , Signal Transduction , Single-Cell Analysis/methods
12.
Front Genet ; 12: 751158, 2021.
Article in English | MEDLINE | ID: mdl-34858473

ABSTRACT

Identification of intercellular signaling changes across multiple single-cell RNA-sequencing (scRNA-seq) datasets as well as how intercellular communications affect intracellular transcription factors (TFs) to regulate target genes is crucial in understanding how distinct cell states respond to evolution, perturbations, and diseases. Here, we first generalized our previously developed tool CellChat, enabling flexible comparison analysis of cell-cell communication networks across any number of scRNA-seq datasets from interrelated biological conditions. This greatly facilitates the ready detection of signaling changes of cell-cell communication in response to any biological perturbations. We then investigated how intercellular communications affect intracellular signaling response by inferring a multiscale signaling network which bridges the intercellular communications at the population level and the cell state-specific intracellular signaling network at the molecular level. The latter is constructed by integrating receptor-TF interactions collected from public databases and TF-target gene regulations inferred from a network-regularized regression model. By applying our approaches to three scRNA-seq datasets from skin development, spinal cord injury, and COVID-19, we demonstrated the capability of our approaches in identifying the predominant signaling changes across conditions and the critical signaling mechanisms regulating target gene expression. Together, our work will facilitate the identification of both intercellular and intracellular dysregulated signaling mechanisms responsible for biological perturbations in diverse tissues.

13.
Curr Opin Syst Biol ; 26: 12-23, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33969247

ABSTRACT

Cell-cell communication is a fundamental process that shapes biological tissue. Historically, studies of cell-cell communication have been feasible for one or two cell types and a few genes. With the emergence of single-cell transcriptomics, we are now able to examine the genetic profiles of individual cells at unprecedented scale and depth. The availability of such data presents an exciting opportunity to construct a more comprehensive description of cell-cell communication. This review discusses the recent explosion of methods that have been developed to infer cell-cell communication from non-spatial and spatial single-cell transcriptomics, two promising technologies which have complementary strengths and limitations. We propose several avenues to propel this rapidly expanding field forward in meaningful ways.

14.
Nat Commun ; 12(1): 1088, 2021 02 17.
Article in English | MEDLINE | ID: mdl-33597522

ABSTRACT

Understanding global communications among cells requires accurate representation of cell-cell signaling links and effective systems-level analyses of those links. We construct a database of interactions among ligands, receptors and their cofactors that accurately represent known heteromeric molecular complexes. We then develop CellChat, a tool that is able to quantitatively infer and analyze intercellular communication networks from single-cell RNA-sequencing (scRNA-seq) data. CellChat predicts major signaling inputs and outputs for cells and how those cells and signals coordinate for functions using network analysis and pattern recognition approaches. Through manifold learning and quantitative contrasts, CellChat classifies signaling pathways and delineates conserved and context-specific pathways across different datasets. Applying CellChat to mouse and human skin datasets shows its ability to extract complex signaling patterns. Our versatile and easy-to-use toolkit CellChat and a web-based Explorer ( http://www.cellchat.org/ ) will help discover novel intercellular communications and build cell-cell communication atlases in diverse tissues.


Subject(s)
Cell Communication/genetics , Computational Biology/methods , Sequence Analysis, RNA/methods , Signal Transduction/genetics , Single-Cell Analysis/methods , Algorithms , Animals , Gene Expression Profiling/methods , Humans , Internet , Mice , Models, Theoretical , Skin/cytology , Skin/embryology , Skin/metabolism , Software
15.
Nat Commun ; 11(1): 5434, 2020 10 28.
Article in English | MEDLINE | ID: mdl-33116143

ABSTRACT

The interfollicular epidermis (IFE) forms a water-tight barrier that is often disrupted in inflammatory skin diseases. During homeostasis, the IFE is replenished by stem cells in the basal layer that differentiate as they migrate toward the skin surface. Conventionally, IFE differentiation is thought to be stepwise as reflected in sharp boundaries between its basal, spinous, granular and cornified layers. The transcription factor GRHL3 regulates IFE differentiation by transcriptionally activating terminal differentiation genes. Here we use single cell RNA-seq to show that murine IFE differentiation is best described as a single step gradualistic process with a large number of transition cells between the basal and spinous layer. RNA-velocity analysis identifies a commitment point that separates the plastic basal and transition cell state from unidirectionally differentiating cells. We also show that in addition to promoting IFE terminal differentiation, GRHL3 is essential for suppressing epidermal stem cell expansion and the emergence of an abnormal stem cell state by suppressing Wnt signaling in stem cells.


Subject(s)
DNA-Binding Proteins/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Epidermal Cells/cytology , Epidermal Cells/metabolism , Transcription Factors/metabolism , Animals , Animals, Newborn , Cell Differentiation , Cell Lineage , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Epidermis/embryology , Epidermis/metabolism , Female , Gene Expression Profiling , Gestational Age , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Single-Cell Analysis , Transcription Factors/deficiency , Transcription Factors/genetics
16.
Proc Natl Acad Sci U S A ; 117(11): 5761-5771, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32132203

ABSTRACT

The circadian clock coordinates a variety of immune responses with signals from the external environment to promote survival. We investigated the potential reciprocal relationship between the circadian clock and skin inflammation. We treated mice topically with the Toll-like receptor 7 (TLR7) agonist imiquimod (IMQ) to activate IFN-sensitive gene (ISG) pathways and induce psoriasiform inflammation. IMQ transiently altered core clock gene expression, an effect mirrored in human patient psoriatic lesions. In mouse skin 1 d after IMQ treatment, ISGs, including the key ISG transcription factor IFN regulatory factor 7 (Irf7), were more highly induced after treatment during the day than the night. Nuclear localization of phosphorylated-IRF7 was most prominently time-of-day dependent in epidermal leukocytes, suggesting that these cell types play an important role in the diurnal ISG response to IMQ. Mice lacking Bmal1 systemically had exacerbated and arrhythmic ISG/Irf7 expression after IMQ. Furthermore, daytime-restricted feeding, which affects the phase of the skin circadian clock, reverses the diurnal rhythm of IMQ-induced ISG expression in the skin. These results suggest a role for the circadian clock, driven by BMAL1, as a negative regulator of the ISG response, and highlight the finding that feeding time can modulate the skin immune response. Since the IFN response is essential for the antiviral and antitumor effects of TLR activation, these findings are consistent with the time-of-day-dependent variability in the ability to fight microbial pathogens and tumor initiation and offer support for the use of chronotherapy for their treatment.


Subject(s)
Circadian Rhythm , Immunity, Innate/genetics , Interferons/genetics , Membrane Glycoproteins/genetics , Skin/metabolism , Toll-Like Receptor 7/genetics , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Animals , CLOCK Proteins/genetics , CLOCK Proteins/metabolism , Imiquimod/pharmacology , Interferon Inducers/pharmacology , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/metabolism , Interferons/metabolism , Male , Membrane Glycoproteins/agonists , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Skin/drug effects , Toll-Like Receptor 7/agonists , Toll-Like Receptor 7/metabolism
17.
Cell Rep ; 30(11): 3932-3947.e6, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32187560

ABSTRACT

Our knowledge of transcriptional heterogeneities in epithelial stem and progenitor cell compartments is limited. Epidermal basal cells sustain cutaneous tissue maintenance and drive wound healing. Previous studies have probed basal cell heterogeneity in stem and progenitor potential, but a comprehensive dissection of basal cell dynamics during differentiation is lacking. Using single-cell RNA sequencing coupled with RNAScope and fluorescence lifetime imaging, we identify three non-proliferative and one proliferative basal cell state in homeostatic skin that differ in metabolic preference and become spatially partitioned during wound re-epithelialization. Pseudotemporal trajectory and RNA velocity analyses predict a quasi-linear differentiation hierarchy where basal cells progress from Col17a1Hi/Trp63Hi state to early-response state, proliferate at the juncture of these two states, or become growth arrested before differentiating into spinous cells. Wound healing induces plasticity manifested by dynamic basal-spinous interconversions at multiple basal transcriptional states. Our study provides a systematic view of epidermal cellular dynamics, supporting a revised "hierarchical-lineage" model of homeostasis.


Subject(s)
Epidermis/metabolism , Epidermis/pathology , Gene Expression Profiling , Homeostasis/genetics , Single-Cell Analysis , Wound Healing/genetics , Animals , Cell Movement/genetics , Female , Inflammation/genetics , Inflammation/pathology , Mice, Inbred C57BL , Mice, Transgenic , Up-Regulation/genetics
18.
Genome Biol ; 21(1): 25, 2020 02 03.
Article in English | MEDLINE | ID: mdl-32014031

ABSTRACT

Simultaneous measurements of transcriptomic and epigenomic profiles in the same individual cells provide an unprecedented opportunity to understand cell fates. However, effective approaches for the integrative analysis of such data are lacking. Here, we present a single-cell aggregation and integration (scAI) method to deconvolute cellular heterogeneity from parallel transcriptomic and epigenomic profiles. Through iterative learning, scAI aggregates sparse epigenomic signals in similar cells learned in an unsupervised manner, allowing coherent fusion with transcriptomic measurements. Simulation studies and applications to three real datasets demonstrate its capability of dissecting cellular heterogeneity within both transcriptomic and epigenomic layers and understanding transcriptional regulatory mechanisms.


Subject(s)
Epigenome , Gene Expression Profiling/methods , Genomics/methods , Single-Cell Analysis/methods , Transcriptome , Unsupervised Machine Learning , A549 Cells , Genetic Heterogeneity , Humans
19.
IEEE Trans Biomed Eng ; 67(5): 1418-1428, 2020 05.
Article in English | MEDLINE | ID: mdl-31449003

ABSTRACT

Single cell technologies provide an unprecedented opportunity to explore the heterogeneity in a biological process at the level of single cells. One major challenge in analyzing single cell data is to identify cell subpopulations, stable cell states, and cells in transition between states. To elucidate the transition mechanisms in cell fate dynamics, it is highly desirable to quantitatively characterize cellular states and intermediate states. Here, we present scRCMF, an unsupervised method that identifies stable cell states and transition cells by adopting a nonlinear optimization model that infers the latent substructures from a gene-cell matrix. We incorporate a random coefficient matrix-based regularization into the standard nonnegative matrix decomposition model to improve the reliability and stability of estimating latent substructures. To quantify the transition capability of each cell, we propose two new measures: single-cell transition entropy (scEntropy) and transition probability (scTP). When applied to two simulated and three published scRNA-seq datasets, scRCMF not only successfully captures multiple subpopulations and transition processes in large-scale data, but also identifies transition states and some known marker genes associated with cell state transitions and subpopulations. Furthermore, the quantity scEntropy is found to be significantly higher for transition cells than other cellular states during the global differentiation, and the scTP predicts the "fate decisions" of transition cells within the transition. The present study provides new insights into transition events during differentiation and development.


Subject(s)
Single-Cell Analysis , Transcriptome , Cell Differentiation , Nonlinear Dynamics , Reproducibility of Results
20.
Nat Neurosci ; 22(11): 1857-1870, 2019 11.
Article in English | MEDLINE | ID: mdl-31548723

ABSTRACT

Recent anatomical evidence suggests a functionally significant back-projection pathway from the subiculum to the CA1. Here we show that the afferent circuitry of CA1-projecting subicular neurons is biased by inputs from CA1 inhibitory neurons and the visual cortex, but lacks input from the entorhinal cortex. Efferents of the CA1-projecting subiculum neurons also target the perirhinal cortex, an area strongly implicated in object-place learning. We identify a critical role for CA1-projecting subicular neurons in object-location learning and memory, and show that this projection modulates place-specific activity of CA1 neurons and their responses to displaced objects. Together, these experiments reveal a novel pathway by which cortical inputs, particularly those from the visual cortex, reach the hippocampal output region CA1. Our findings also implicate this circuitry in the formation of complex spatial representations and learning of object-place associations.


Subject(s)
CA1 Region, Hippocampal/physiology , Entorhinal Cortex/physiology , Hippocampus/physiology , Learning/physiology , Perirhinal Cortex/physiology , Visual Cortex/physiology , Animals , Female , Male , Mice , Mice, Transgenic , Neural Pathways/physiology , Neurons/physiology , Space Perception/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...