Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.431
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 185(5): 777-793.e20, 2022 03 03.
Article in English | MEDLINE | ID: mdl-35196500

ABSTRACT

In development, lineage segregation is coordinated in time and space. An important example is the mammalian inner cell mass, in which the primitive endoderm (PrE, founder of the yolk sac) physically segregates from the epiblast (EPI, founder of the fetus). While the molecular requirements have been well studied, the physical mechanisms determining spatial segregation between EPI and PrE remain elusive. Here, we investigate the mechanical basis of EPI and PrE sorting. We find that rather than the differences in static cell surface mechanical parameters as in classical sorting models, it is the differences in surface fluctuations that robustly ensure physical lineage sorting. These differential surface fluctuations systematically correlate with differential cellular fluidity, which we propose together constitute a non-equilibrium sorting mechanism for EPI and PrE lineages. By combining experiments and modeling, we identify cell surface dynamics as a key factor orchestrating the correct spatial segregation of the founder embryonic lineages.


Subject(s)
Blastocyst , Embryo, Mammalian , Endoderm , Animals , Blastocyst/metabolism , Cell Differentiation/physiology , Cell Lineage/physiology , Cell Membrane/metabolism , Embryo, Mammalian/metabolism , Embryonic Development , Endoderm/metabolism , Mammals , Mice , Protein Transport
2.
Cell ; 182(6): 1384-1400, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32946781

ABSTRACT

Hematopoiesis has long served as a paradigm of stem cell biology and tissue homeostasis. In the past decade, the genomics revolution has ushered in powerful new methods for investigating the hematopoietic system that have provided transformative insights into its biology. As part of the advances in genomics, increasingly accurate deep sequencing and novel methods of cell tracking have revealed hematopoiesis to be more of a continuous and less of a discrete and punctuated process than originally envisioned. In part, this continuous nature of hematopoiesis is made possible by the emergent outcomes of vast, interconnected regulatory networks that influence cell fates and lineage commitment. It is also becoming clear how these mechanisms are modulated by genetic variation present throughout the population. This review describes how these recently uncovered complexities are reshaping our concept of tissue development and homeostasis while opening up a more comprehensive future understanding of hematopoiesis.


Subject(s)
Cell Lineage , Gene Expression Regulation, Developmental , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Animals , Cell Lineage/genetics , Cell Lineage/physiology , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Genomics , Homeostasis , Human Genetics , Humans , Transcription Factors/genetics , Transcription Factors/metabolism
3.
Cell ; 176(5): 1158-1173.e16, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30712869

ABSTRACT

Homeostatic regulation of the intestinal enteroendocrine lineage hierarchy is a poorly understood process. We resolved transcriptional changes during enteroendocrine differentiation in real time at single-cell level using a novel knockin allele of Neurog3, the master regulator gene briefly expressed at the onset of enteroendocrine specification. A bi-fluorescent reporter, Neurog3Chrono, measures time from the onset of enteroendocrine differentiation and enables precise positioning of single-cell transcriptomes along an absolute time axis. This approach yielded a definitive description of the enteroendocrine hierarchy and its sub-lineages, uncovered differential kinetics between sub-lineages, and revealed time-dependent hormonal plasticity in enterochromaffin and L cells. The time-resolved map of transcriptional changes predicted multiple novel molecular regulators. Nine of these were validated by conditional knockout in mice or CRISPR modification in intestinal organoids. Six novel candidate regulators (Sox4, Rfx6, Tox3, Myt1, Runx1t1, and Zcchc12) yielded specific enteroendocrine phenotypes. Our time-resolved single-cell transcriptional map presents a rich resource to unravel enteroendocrine differentiation.


Subject(s)
Cell Lineage/genetics , Enteroendocrine Cells/metabolism , Gene Expression Profiling/methods , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/genetics , Cell Lineage/physiology , Enteroendocrine Cells/physiology , Fluorescent Dyes , Homeodomain Proteins/genetics , Intestinal Mucosa/cytology , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Optical Imaging/methods , Organoids , Phenotype , Single-Cell Analysis/methods , Stem Cells , Transcription Factors/genetics , Transcriptome/genetics
4.
Cell ; 175(7): 1887-1901.e18, 2018 12 13.
Article in English | MEDLINE | ID: mdl-30550787

ABSTRACT

In early mammalian embryos, it remains unclear how the first cell fate bias is initially triggered and amplified toward cell fate segregation. Here, we report that a long noncoding RNA, LincGET, is transiently and asymmetrically expressed in the nucleus of two- to four-cell mouse embryos. Overexpression of LincGET in one of the two-cell blastomeres biases its progeny predominantly toward the inner cell mass (ICM) fate. Mechanistically, LincGET physically binds to CARM1 and promotes the nuclear localization of CARM1, which can further increase the level of H3 methylation at Arginine 26 (H3R26me), activate ICM-specific gene expression, upregulate transposons, and increase global chromatin accessibility. Simultaneous overexpression of LincGET and depletion of Carm1 no longer biased embryonic fate, indicating that the effect of LincGET in directing ICM lineage depends on CARM1. Thus, our data identify LincGET as one of the earliest known lineage regulators to bias cell fate in mammalian 2-cell embryos.


Subject(s)
Blastocyst/metabolism , Blastomeres/metabolism , Cell Lineage/physiology , Gene Expression Regulation, Developmental/physiology , RNA, Long Noncoding/biosynthesis , Animals , Blastocyst/cytology , Blastomeres/cytology , Female , Histones/metabolism , Methylation , Mice , Mice, Inbred ICR , Protein-Arginine N-Methyltransferases/biosynthesis , Protein-Arginine N-Methyltransferases/genetics , RNA, Long Noncoding/genetics
5.
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
6.
Immunity ; 54(7): 1433-1446.e5, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34062116

ABSTRACT

The extra-embryonic yolk sac contains the first definitive multipotent hematopoietic cells, denominated erythromyeloid progenitors. They originate in situ prior to the emergence of hematopoietic stem cells and give rise to erythroid, monocytes, granulocytes, mast cells and macrophages, the latter in a Myb transcription factor-independent manner. We uncovered here the heterogeneity of yolk sac erythromyeloid progenitors, at the single cell level, and discriminated multipotent from committed progenitors, prior to fetal liver colonization. We identified two temporally distinct megakaryocyte differentiation pathways. The first occurs in the yolk sac, bypasses intermediate bipotent megakaryocyte-erythroid progenitors and, similar to the differentiation of macrophages, is Myb-independent. By contrast, the second originates later, from Myb-dependent bipotent progenitors expressing Csf2rb and colonize the fetal liver, where they give rise to megakaryocytes and to large numbers of erythrocytes. Understanding megakaryocyte development is crucial as they play key functions during vascular development, in particular in separating blood and lymphatic networks.


Subject(s)
Cell Differentiation/physiology , Erythrocytes/cytology , Megakaryocytes/cytology , Myeloid Cells/cytology , Stem Cells/cytology , Yolk Sac/cytology , Animals , Cell Lineage/physiology , Cells, Cultured , Embryo, Mammalian/cytology , Female , Granulocytes/cytology , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Macrophages/cytology , Male , Mice , Mice, Inbred C57BL , Monocytes/cytology , Multipotent Stem Cells/cytology , Pregnancy
7.
Immunity ; 54(7): 1511-1526.e8, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34260887

ABSTRACT

Myeloid cells encounter stromal cells and their matrix determinants on a continual basis during their residence in any given organ. Here, we examined the impact of the collagen receptor LAIR1 on myeloid cell homeostasis and function. LAIR1 was highly expressed in the myeloid lineage and enriched in non-classical monocytes. Proteomic definition of the LAIR1 interactome identified stromal factor Colec12 as a high-affinity LAIR1 ligand. Proteomic profiling of LAIR1 signaling triggered by Collagen1 and Colec12 highlighted pathways associated with survival, proliferation, and differentiation. Lair1-/- mice had reduced frequencies of Ly6C- monocytes, which were associated with altered proliferation and apoptosis of non-classical monocytes from bone marrow and altered heterogeneity of interstitial macrophages in lung. Myeloid-specific LAIR1 deficiency promoted metastatic growth in a melanoma model and LAIR1 expression associated with improved clinical outcomes in human metastatic melanoma. Thus, monocytes and macrophages rely on LAIR1 sensing of stromal determinants for fitness and function, with relevance in homeostasis and disease.


Subject(s)
Homeostasis/physiology , Lung/metabolism , Macrophages, Alveolar/metabolism , Monocytes/metabolism , Receptors, Immunologic/metabolism , Animals , Apoptosis/physiology , Bone Marrow/metabolism , Bone Marrow/pathology , COS Cells , Cell Differentiation/physiology , Cell Line , Cell Line, Tumor , Cell Lineage/physiology , Cell Proliferation/physiology , Chlorocebus aethiops , Female , Humans , Lung/pathology , Macrophages, Alveolar/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/pathology , Myeloid Cells/metabolism , Myeloid Cells/pathology , Neoplasm Metastasis/pathology , Proteomics/methods , Signal Transduction/physiology
8.
Annu Rev Cell Dev Biol ; 31: 553-73, 2015.
Article in English | MEDLINE | ID: mdl-26359777

ABSTRACT

The respiratory endoderm develops from a small cluster of cells located on the ventral anterior foregut. This population of progenitors generates the myriad epithelial lineages required for proper lung function in adults through a complex and delicately balanced series of developmental events controlled by many critical signaling and transcription factor pathways. In the past decade, understanding of this process has grown enormously, helped in part by cell lineage fate analysis and deep sequencing of the transcriptomes of various progenitors and differentiated cell types. This review explores how these new techniques, coupled with more traditional approaches, have provided a detailed picture of development of the epithelial lineages in the lung and insight into how aberrant development can lead to lung disease.


Subject(s)
Endoderm/physiology , Gene Expression Regulation, Developmental/physiology , Lung/physiology , Morphogenesis/physiology , Animals , Cell Lineage/physiology , Humans , Organogenesis/physiology
9.
Mol Cell ; 81(12): 2583-2595.e6, 2021 06 17.
Article in English | MEDLINE | ID: mdl-33961797

ABSTRACT

53BP1 influences genome stability via two independent mechanisms: (1) regulating DNA double-strand break (DSB) repair and (2) enhancing p53 activity. We discovered a protein, Tudor-interacting repair regulator (TIRR), that associates with the 53BP1 Tudor domain and prevents its recruitment to DSBs. Here, we elucidate how TIRR affects 53BP1 function beyond its recruitment to DSBs and biochemically links the two distinct roles of 53BP1. Loss of TIRR causes an aberrant increase in the gene transactivation function of p53, affecting several p53-mediated cell-fate programs. TIRR inhibits the complex formation between the Tudor domain of 53BP1 and a dimethylated form of p53 (K382me2) that is poised for transcriptional activation of its target genes. TIRR mRNA expression levels negatively correlate with the expression of key p53 target genes in breast and prostate cancers. Further, TIRR loss is selectively not tolerated in p53-proficient tumors. Therefore, we establish that TIRR is an important inhibitor of the 53BP1-p53 complex.


Subject(s)
Cell Lineage/genetics , RNA-Binding Proteins/metabolism , Tumor Suppressor p53-Binding Protein 1/metabolism , Binding Sites , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Lineage/physiology , DNA/genetics , DNA Breaks, Double-Stranded , DNA Repair , Histones/metabolism , Humans , Protein Binding , RNA-Binding Proteins/physiology , Tudor Domain , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor p53-Binding Protein 1/physiology
10.
Annu Rev Neurosci ; 43: 163-186, 2020 07 08.
Article in English | MEDLINE | ID: mdl-32075518

ABSTRACT

Cells of the oligodendrocyte lineage express a wide range of Ca2+ channels and receptors that regulate oligodendrocyte progenitor cell (OPC) and oligodendrocyte formation and function. Here we define those key channels and receptors that regulate Ca2+ signaling and OPC development and myelination. We then discuss how the regulation of intracellular Ca2+ in turn affects OPC and oligodendrocyte biology in the healthy nervous system and under pathological conditions. Activation of Ca2+ channels and receptors in OPCs and oligodendrocytes by neurotransmitters converges on regulating intracellular Ca2+, making Ca2+ signaling a central candidate mediator of activity-driven myelination. Indeed, recent evidence indicates that localized changes in Ca2+ in oligodendrocytes can regulate the formation and remodeling of myelin sheaths and perhaps additional functions of oligodendrocytes and OPCs. Thus, decoding how OPCs and myelinating oligodendrocytes integrate and process Ca2+ signals will be important to fully understand central nervous system formation, health, and function.


Subject(s)
Calcium Signaling/physiology , Cell Lineage/physiology , Myelin Sheath/physiology , Neurogenesis/physiology , Oligodendroglia/physiology , Animals , Cell Differentiation/physiology , Humans , Oligodendroglia/cytology
11.
Development ; 151(3)2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38230563

ABSTRACT

An unanswered question in neurobiology is how are diverse neuron cell types generated from a small number of neural stem cells? In the Drosophila larval central brain, there are eight bilateral Type 2 neuroblast (T2NB) lineages that express a suite of early temporal factors followed by a different set of late temporal factors and generate the majority of the central complex (CX) neurons. The early-to-late switch is triggered by the orphan nuclear hormone receptor Seven-up (Svp), yet little is known about how this Svp-dependent switch is involved in specifying CX neuron identities. Here, we: (1) birth date the CX neurons P-EN and P-FN (early and late, respectively); (2) show that Svp is transiently expressed in all early T2NBs; and (3) show that loss of Svp expands the population of early born P-EN neurons at the expense of late born P-FN neurons. Furthermore, in the absence of Svp, T2NBs fail decommissioning and abnormally extend their lineage into week-old adults. We conclude that Svp is required to specify CX neuron identity, as well as to initiate T2NB decommissioning.


Subject(s)
Drosophila Proteins , Neural Stem Cells , Animals , Transcription Factors/metabolism , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Neural Stem Cells/metabolism , Neurons/metabolism , Drosophila/metabolism , Cell Lineage/physiology , Drosophila melanogaster/metabolism
12.
Mol Cell ; 69(2): 169-181, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29107536

ABSTRACT

The secretory capacity of a cell is constantly challenged by physiological demands and pathological perturbations. To adjust and match the protein-folding capacity of the endoplasmic reticulum (ER) to changing secretory needs, cells employ a dynamic intracellular signaling pathway known as the unfolded protein response (UPR). Homeostatic activation of the UPR enforces adaptive programs that modulate and augment key aspects of the entire secretory pathway, whereas maladaptive UPR outputs trigger apoptosis. Here, we discuss recent advances into how the UPR integrates information about the intensity and duration of ER stress stimuli in order to control cell fate. These findings are timely and significant because they inform an evolving mechanistic understanding of a wide variety of human diseases, including diabetes mellitus, neurodegeneration, and cancer, thus opening up the potential for new therapeutic modalities to treat these diverse diseases.


Subject(s)
Cell Lineage/physiology , Unfolded Protein Response/physiology , Activating Transcription Factor 6/metabolism , Animals , Apoptosis , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress , Endoribonucleases/metabolism , Homeostasis , Humans , Models, Biological , Protein Folding , Protein Serine-Threonine Kinases/metabolism , Secretory Pathway/physiology , Signal Transduction , eIF-2 Kinase/metabolism
13.
Development ; 149(20)2022 10 15.
Article in English | MEDLINE | ID: mdl-35993866

ABSTRACT

Embryogenesis is supported by dynamic loops of cellular interactions. Here, we create a partial mouse embryo model to elucidate the principles of epiblast (Epi) and extra-embryonic endoderm co-development (XEn). We trigger naive mouse embryonic stem cells to form a blastocyst-stage niche of Epi-like cells and XEn-like cells (3D, hydrogel free and serum free). Once established, these two lineages autonomously progress in minimal medium to form an inner pro-amniotic-like cavity surrounded by polarized Epi-like cells covered with visceral endoderm (VE)-like cells. The progression occurs through reciprocal inductions by which the Epi supports the primitive endoderm (PrE) to produce a basal lamina that subsequently regulates Epi polarization and/or cavitation, which, in return, channels the transcriptomic progression to VE. This VE then contributes to Epi bifurcation into anterior- and posterior-like states. Similarly, boosting the formation of PrE-like cells within blastoids supports developmental progression. We argue that self-organization can arise from lineage bifurcation followed by a pendulum of induction that propagates over time.


Subject(s)
Endoderm , Germ Layers , Animals , Blastocyst , Cell Differentiation , Cell Lineage/physiology , Embryo Implantation , Embryo, Mammalian , Mice
14.
Development ; 149(5)2022 03 01.
Article in English | MEDLINE | ID: mdl-35156684

ABSTRACT

Despite their medical and economic relevance, it remains largely unknown how suboptimal temperatures affect adult insect reproduction. Here, we report an in-depth analysis of how chronic adult exposure to suboptimal temperatures affects oogenesis using the model insect Drosophila melanogaster. In adult females maintained at 18°C (cold) or 29°C (warm), relative to females at the 25°C control temperature, egg production was reduced through distinct cellular mechanisms. Chronic 18°C exposure improved germline stem cell maintenance, survival of early germline cysts and oocyte quality, but reduced follicle growth with no obvious effect on vitellogenesis. By contrast, in females at 29°C, germline stem cell numbers and follicle growth were similar to those at 25°C, while early germline cyst death and degeneration of vitellogenic follicles were markedly increased and oocyte quality plummeted over time. Finally, we also show that these effects are largely independent of diet, male factors or canonical temperature sensors. These findings are relevant not only to cold-blooded organisms, which have limited thermoregulation, but also potentially to warm-blooded organisms, which are susceptible to hypothermia, heatstroke and fever.


Subject(s)
Cell Lineage/physiology , Drosophila melanogaster/physiology , Germ Cells/physiology , Oogenesis/physiology , Stem Cells/physiology , Animals , Cold Temperature , Female , Gene Expression Regulation, Developmental/physiology , Male , Oocytes/physiology , Ovarian Follicle/physiology , Ovary/physiology , Signal Transduction/physiology , Vitellogenesis/physiology
15.
Development ; 149(5)2022 03 01.
Article in English | MEDLINE | ID: mdl-35253855

ABSTRACT

During mammalian brain development, how different astrocytes are specified from progenitor cells is not well understood. In particular, whether astrocyte progenitor cells (APCs) start as a relatively homogenous population or whether there is early heterogeneity remains unclear. Here, we have dissected subpopulations of embryonic mouse forebrain progenitors using single-cell transcriptome analyses. Our sequencing data revealed two molecularly distinct APC subgroups at the start of gliogenesis from both dorsal and ventral forebrains. The two APC subgroups were marked, respectively, by specific expression of Sparc and Sparcl1, which are known to function in mature astrocytes with opposing activities for regulating synapse formation. Expression analyses showed that SPARC and SPARCL1 mark APC subgroups that display distinct temporal and spatial patterns, correlating with major waves of astrogliogenesis during development. Our results uncover an early molecular divergence of APCs in the mammalian brain and provide a useful transcriptome resource for the study of glial cell specification.


Subject(s)
Astrocytes/physiology , Mammals/physiology , Neurogenesis/physiology , Neuroglia/physiology , Stem Cells/physiology , Animals , Astrocytes/metabolism , Cell Differentiation/physiology , Cell Lineage/physiology , Cell Proliferation/physiology , Mammals/metabolism , Mice , Neural Stem Cells/metabolism , Neural Stem Cells/physiology , Neuroglia/metabolism , Osteonectin/metabolism , Prosencephalon/metabolism , Prosencephalon/physiology , Single-Cell Analysis/methods , Stem Cells/metabolism , Transcriptome/physiology
16.
Immunity ; 45(6): 1219-1231, 2016 12 20.
Article in English | MEDLINE | ID: mdl-27913094

ABSTRACT

Hematopoietic stem cells (HSCs) self-renew in bone marrow niches formed by mesenchymal progenitors and endothelial cells expressing the chemokine CXCL12, but whether a separate niche instructs multipotent progenitor (MPP) differentiation remains unclear. We show that MPPs resided in HSC niches, where they encountered lineage-instructive differentiation signals. Conditional deletion of the chemokine receptor CXCR4 in MPPs reduced differentiation into common lymphoid progenitors (CLPs), which decreased lymphopoiesis. CXCR4 was required for CLP positioning near Interleukin-7+ (IL-7) cells and for optimal IL-7 receptor signaling. IL-7+ cells expressed CXCL12 and the cytokine SCF, were mesenchymal progenitors capable of differentiation into osteoblasts and adipocytes, and comprised a minor subset of sinusoidal endothelial cells. Conditional Il7 deletion in mesenchymal progenitors reduced B-lineage committed CLPs, while conditional Cxcl12 or Scf deletion from IL-7+ cells reduced HSC and MPP numbers. Thus, HSC maintenance and multilineage differentiation are distinct cell lineage decisions that are both controlled by HSC niches.


Subject(s)
Cell Differentiation/physiology , Hematopoietic Stem Cells/cytology , Multipotent Stem Cells/cytology , Stem Cell Niche/physiology , Animals , Cell Lineage/physiology , Cell Separation , Chemokine CXCL2/metabolism , Flow Cytometry , Interleukin-7/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic
17.
Immunity ; 45(3): 597-609, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27590115

ABSTRACT

Hematopoietic stem cells (HSCs) sustain long-term reconstitution of hematopoiesis in transplantation recipients, yet their role in the endogenous steady-state hematopoiesis remains unclear. In particular, recent studies suggested that HSCs provide a relatively minor contribution to immune cell development in adults. We directed transgene expression in a fraction of HSCs that maintained reconstituting activity during serial transplantations. Inducible genetic labeling showed that transgene-expressing HSCs gave rise to other phenotypic HSCs, confirming their top position in the differentiation hierarchy. The labeled HSCs rapidly contributed to committed progenitors of all lineages and to mature myeloid cells and lymphocytes, but not to B-1a cells or tissue macrophages. Importantly, labeled HSCs gave rise to more than two-thirds of all myeloid cells and platelets in adult mice, and this contribution could be accelerated by an induced interferon response. Thus, classically defined HSCs maintain immune cell development in the steady state and during systemic cytokine responses.


Subject(s)
Cell Lineage/physiology , Hematopoiesis/physiology , Hematopoietic Stem Cells/physiology , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/physiology , Blood Platelets/metabolism , Blood Platelets/physiology , Cell Differentiation/physiology , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/metabolism , Interferons/metabolism , Macrophages/metabolism , Macrophages/physiology , Mice , Mice, Inbred C57BL , Myeloid Cells/metabolism , Myeloid Cells/physiology
18.
PLoS Comput Biol ; 20(4): e1012054, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38648250

ABSTRACT

Neural organoids model the development of the human brain and are an indispensable tool for studying neurodevelopment. Whole-organoid lineage tracing has revealed the number of progenies arising from each initial stem cell to be highly diverse, with lineage sizes ranging from one to more than 20,000 cells. This high variability exceeds what can be explained by existing stochastic models of corticogenesis and indicates the existence of an additional source of stochasticity. To explain this variability, we introduce the SAN model which distinguishes Symmetrically diving, Asymmetrically dividing, and Non-proliferating cells. In the SAN model, the additional source of stochasticity is the survival time of a lineage's pool of symmetrically dividing cells. These survival times result from neutral competition within the sub-population of all symmetrically dividing cells. We demonstrate that our model explains the experimentally observed variability of lineage sizes and derive the quantitative relationship between survival time and lineage size. We also show that our model implies the existence of a regulatory mechanism which keeps the size of the symmetrically dividing cell population constant. Our results provide quantitative insight into the clonal composition of neural organoids and how it arises. This is relevant for many applications of neural organoids, and similar processes may occur in other developing tissues both in vitro and in vivo.


Subject(s)
Organoids , Organoids/cytology , Humans , Cell Lineage/physiology , Computational Biology , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Stochastic Processes , Models, Biological , Neurons/physiology , Neurons/cytology , Brain/cytology , Brain/physiology , Cell Proliferation/physiology , Neurogenesis/physiology
19.
Nat Rev Mol Cell Biol ; 14(8): 489-502, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23860235

ABSTRACT

During embryonic and postnatal development, the different cells types that form adult tissues must be generated and specified in a precise temporal manner. During adult life, most tissues undergo constant renewal to maintain homeostasis. Lineage-tracing and genetic labelling technologies are beginning to shed light on the mechanisms and dynamics of stem and progenitor cell fate determination during development, tissue maintenance and repair, as well as their dysregulation in tumour formation. Statistical approaches, based on proliferation assays and clonal fate analyses, provide quantitative insights into cell kinetics and fate behaviour. These are powerful techniques to address new questions and paradigms in transgenic mouse models and other model systems.


Subject(s)
Cell Lineage/physiology , Cell Tracking/methods , Stem Cells/physiology , Adult , Animals , Humans , Kinetics , Mice , Mice, Transgenic , Models, Biological
20.
Nat Rev Mol Cell Biol ; 14(8): 467-73, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23839578

ABSTRACT

The field of stem cells and regenerative medicine offers considerable promise as a means of delivering new treatments for a wide range of diseases. In order to maximize the effectiveness of cell-based therapies - whether stimulating expansion of endogenous cells or transplanting cells into patients - it is essential to understand the environmental (niche) signals that regulate stem cell behaviour. One of those signals is from the extracellular matrix (ECM). New technologies have offered insights into how stem cells sense signals from the ECM and how they respond to these signals at the molecular level, which ultimately regulate their fate.


Subject(s)
Extracellular Matrix/physiology , Stem Cells/physiology , Animals , Cell Communication/genetics , Cell Communication/physiology , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Lineage/genetics , Cell Lineage/physiology , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Humans , Membrane Fluidity/genetics , Membrane Fluidity/physiology , Models, Biological , Stem Cell Niche/genetics , Stem Cell Niche/physiology , Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL