Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 185(17): 3169-3185.e20, 2022 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-35908548

RESUMO

Mice deficient for all ten-eleven translocation (TET) genes exhibit early gastrulation lethality. However, separating cause and effect in such embryonic failure is challenging. To isolate cell-autonomous effects of TET loss, we used temporal single-cell atlases from embryos with partial or complete mutant contributions. Strikingly, when developing within a wild-type embryo, Tet-mutant cells retain near-complete differentiation potential, whereas embryos solely comprising mutant cells are defective in epiblast to ectoderm transition with degenerated mesoderm potential. We map de-repressions of early epiblast factors (e.g., Dppa4 and Gdf3) and failure to activate multiple signaling from nascent mesoderm (Lefty, FGF, and Notch) as likely cell-intrinsic drivers of TET loss phenotypes. We further suggest loss of enhancer demethylation as the underlying mechanism. Collectively, our work demonstrates an unbiased approach for defining intrinsic and extrinsic embryonic gene function based on temporal differentiation atlases and disentangles the intracellular effects of the demethylation machinery from its broader tissue-level ramifications.


Assuntos
Gastrulação , Mesoderma , Animais , Diferenciação Celular/genética , Embrião de Mamíferos/metabolismo , Gastrulação/genética , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Proteínas Nucleares/metabolismo , Transdução de Sinais
2.
Cell ; 184(11): 2825-2842.e22, 2021 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-33932341

RESUMO

Mouse embryonic development is a canonical model system for studying mammalian cell fate acquisition. Recently, single-cell atlases comprehensively charted embryonic transcriptional landscapes, yet inference of the coordinated dynamics of cells over such atlases remains challenging. Here, we introduce a temporal model for mouse gastrulation, consisting of data from 153 individually sampled embryos spanning 36 h of molecular diversification. Using algorithms and precise timing, we infer differentiation flows and lineage specification dynamics over the embryonic transcriptional manifold. Rapid transcriptional bifurcations characterize the commitment of early specialized node and blood cells. However, for most lineages, we observe combinatorial multi-furcation dynamics rather than hierarchical transcriptional transitions. In the mesoderm, dozens of transcription factors combinatorially regulate multifurcations, as we exemplify using time-matched chimeric embryos of Foxc1/Foxc2 mutants. Our study rejects the notion of differentiation being governed by a series of binary choices, providing an alternative quantitative model for cell fate acquisition.


Assuntos
Desenvolvimento Embrionário/fisiologia , Gastrulação/fisiologia , Animais , Diferenciação Celular , Linhagem da Célula , Embrião de Mamíferos/citologia , Desenvolvimento Embrionário/genética , Feminino , Expressão Gênica , Camundongos/embriologia , Camundongos Endogâmicos C57BL , Células-Tronco Embrionárias Murinas , Gravidez , Análise de Sequência de RNA/métodos , Análise de Célula Única/métodos
3.
Immunity ; 57(2): 271-286.e13, 2024 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-38301652

RESUMO

The immune system encodes information about the severity of a pathogenic threat in the quantity and type of memory cells it forms. This encoding emerges from lymphocyte decisions to maintain or lose self-renewal and memory potential during a challenge. By tracking CD8+ T cells at the single-cell and clonal lineage level using time-resolved transcriptomics, quantitative live imaging, and an acute infection model, we find that T cells will maintain or lose memory potential early after antigen recognition. However, following pathogen clearance, T cells may regain memory potential if initially lost. Mechanistically, this flexibility is implemented by a stochastic cis-epigenetic switch that tunably and reversibly silences the memory regulator, TCF1, in response to stimulation. Mathematical modeling shows how this flexibility allows memory T cell numbers to scale robustly with pathogen virulence and immune response magnitudes. We propose that flexibility and stochasticity in cellular decisions ensure optimal immune responses against diverse threats.


Assuntos
Linfócitos T CD8-Positivos , Células T de Memória , Epigênese Genética , Células Clonais , Memória Imunológica , Diferenciação Celular
4.
Immunity ; 54(9): 2005-2023.e10, 2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34525339

RESUMO

Cell fate decisions during early B cell activation determine the outcome of responses to pathogens and vaccines. We examined the early B cell response to T-dependent antigen in mice by single-cell RNA sequencing. Early after immunization, a homogeneous population of activated precursors (APs) gave rise to a transient wave of plasmablasts (PBs), followed a day later by the emergence of germinal center B cells (GCBCs). Most APs rapidly exited the cell cycle, giving rise to non-GC-derived early memory B cells (eMBCs) that retained an AP-like transcriptional profile. Rapid decline of antigen availability controlled these events; provision of excess antigen precluded cell cycle exit and induced a new wave of PBs. Fate mapping revealed a prominent contribution of eMBCs to the MBC pool. Quiescent cells with an MBC phenotype dominated the early response to immunization in primates. A reservoir of APs/eMBCs may enable rapid readjustment of the immune response when failure to contain a threat is manifested by increased antigen availability.


Assuntos
Linfócitos B/imunologia , Centro Germinativo/imunologia , Imunidade Humoral/imunologia , Memória Imunológica/imunologia , Ativação Linfocitária/imunologia , Animais , Apresentação de Antígeno/imunologia , Diferenciação Celular/imunologia , Camundongos , Plasmócitos/imunologia , Células Precursoras de Linfócitos B/imunologia
5.
Proc Natl Acad Sci U S A ; 121(2): e2304470121, 2024 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-38175868

RESUMO

Repeating patterns of synovial joints are a highly conserved feature of articulated digits, with variations in joint number and location resulting in diverse digit morphologies and limb functions across the tetrapod clade. During the development of the amniote limb, joints form iteratively within the growing digit ray, as a population of distal progenitors alternately specifies joint and phalanx cell fates to segment the digit into distinct elements. While numerous molecular pathways have been implicated in this fate choice, it remains unclear how they give rise to a repeating pattern. Here, using single-cell RNA sequencing and spatial gene expression profiling, we investigate the transcriptional dynamics of interphalangeal joint specification in vivo. Combined with mathematical modeling, we predict that interactions within the BMP signaling pathway-between the ligand GDF5, the inhibitor NOGGIN, and the intracellular effector pSMAD-result in a self-organizing Turing system that forms periodic joint patterns. Our model is able to recapitulate the spatiotemporal gene expression dynamics observed in vivo, as well as phenocopy digit malformations caused by BMP pathway perturbations. By contrasting in silico simulations with in vivo morphometrics of two morphologically distinct digits, we show how changes in signaling parameters and growth dynamics can result in variations in the size and number of phalanges. Together, our results reveal a self-organizing mechanism that underpins amniote digit segmentation and its evolvability and, more broadly, illustrate how Turing systems based on a single molecular pathway may generate complex repetitive patterns in a wide variety of organisms.


Assuntos
Padronização Corporal , Articulações , Animais , Padronização Corporal/genética , Extremidades , Transdução de Sinais , Aves , Mamíferos/genética
6.
Semin Cell Dev Biol ; 127: 46-58, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-34865988

RESUMO

The question of how the heart develops, and the genetic networks governing this process have become intense areas of research over the past several decades. This research is propelled by classical developmental studies and potential clinical applications to understand and treat congenital conditions in which cardiac development is disrupted. Discovery of the tinman gene in Drosophila, and examination of its vertebrate homolog Nkx2.5, along with other core cardiac transcription factors has revealed how cardiac progenitor differentiation and maturation drives heart development. Careful observation of cardiac morphogenesis along with lineage tracing approaches indicated that cardiac progenitors can be divided into two broad classes of cells, namely the first and second heart fields, that contribute to the heart in two distinct waves of differentiation. Ample evidence suggests that the fate of individual cardiac progenitors is restricted to distinct cardiac structures quite early in development, well before the expression of canonical cardiac progenitor markers like Nkx2.5. Here we review the initial specification of cardiac progenitors, discuss evidence for the early patterning of cardiac progenitors during gastrulation, and consider how early gene expression programs and epigenetic patterns can direct their development. A complete understanding of when and how the developmental potential of cardiac progenitors is determined, and their potential plasticity, is of great interest developmentally and also has important implications for both the study of congenital heart disease and therapeutic approaches based on cardiac stem cell programming.


Assuntos
Gastrulação , Mesoderma , Animais , Drosophila/genética , Regulação da Expressão Gênica no Desenvolvimento , Coração
7.
Stem Cells ; 41(5): 520-539, 2023 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-36945732

RESUMO

Epigenetic mechanisms regulate the multilineage differentiation capacity of hematopoietic stem cells (HSCs) into a variety of blood and immune cells. Mapping the chromatin dynamics of functionally defined cell populations will shed mechanistic insight into 2 major, unanswered questions in stem cell biology: how does epigenetic identity contribute to a cell type's lineage potential, and how do cascades of chromatin remodeling dictate ensuing fate decisions? Our recent work revealed evidence of multilineage gene priming in HSCs, where open cis-regulatory elements (CREs) exclusively shared between HSCs and unipotent lineage cells were enriched for DNA binding motifs of known lineage-specific transcription factors. Oligopotent progenitor populations operating between the HSCs and unipotent cells play essential roles in effecting hematopoietic homeostasis. To test the hypothesis that selective HSC-primed lineage-specific CREs remain accessible throughout differentiation, we used ATAC-seq to map the temporal dynamics of chromatin remodeling during progenitor differentiation. We observed epigenetic-driven clustering of oligopotent and unipotent progenitors into distinct erythromyeloid and lymphoid branches, with multipotent HSCs and MPPs associating with the erythromyeloid lineage. We mapped the dynamics of lineage-primed CREs throughout hematopoiesis and identified both unique and shared CREs as potential lineage reinforcement mechanisms at fate branch points. Additionally, quantification of genome-wide peak count and size revealed overall greater chromatin accessibility in HSCs, allowing us to identify HSC-unique peaks as putative regulators of self-renewal and multilineage potential. Finally, CRISPRi-mediated targeting of ATACseq-identified putative CREs in HSCs allowed us to demonstrate the functional role of selective CREs in lineage-specific gene expression. These findings provide insight into the regulation of stem cell multipotency and lineage commitment throughout hematopoiesis and serve as a resource to test functional drivers of hematopoietic lineage fate.


Assuntos
Cromatina , Hematopoese , Cromatina/genética , Cromatina/metabolismo , Hematopoese/genética , Células-Tronco Hematopoéticas/metabolismo , Diferenciação Celular/genética , Linhagem da Célula/genética
8.
Development ; 147(13)2020 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-32601056

RESUMO

Asymmetric cell division (ACD) is an evolutionarily conserved mechanism used by prokaryotes and eukaryotes alike to control cell fate and generate cell diversity. A detailed mechanistic understanding of ACD is therefore necessary to understand cell fate decisions in health and disease. ACD can be manifested in the biased segregation of macromolecules, the differential partitioning of cell organelles, or differences in sibling cell size or shape. These events are usually preceded by and influenced by symmetry breaking events and cell polarization. In this Review, we focus predominantly on cell intrinsic mechanisms and their contribution to cell polarization, ACD and binary cell fate decisions. We discuss examples of polarized systems and detail how polarization is established and, whenever possible, how it contributes to ACD. Established and emerging model organisms will be considered alike, illuminating both well-documented and underexplored forms of polarization and ACD.


Assuntos
Divisão Celular Assimétrica/fisiologia , Polaridade Celular/fisiologia , Animais , Divisão Celular Assimétrica/genética , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Membrana Celular/metabolismo , Polaridade Celular/genética , Humanos
9.
Dev Growth Differ ; 65(5): 245-254, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37190845

RESUMO

Cell fate decisions emerge as a consequence of a complex set of gene regulatory networks. Models of these networks are known to have more parameters than data can determine. Recent work, inspired by Waddington's metaphor of a landscape, has instead tried to understand the geometry of gene regulatory networks. Here, we describe recent results on the appropriate mathematical framework for constructing these landscapes. This allows the construction of minimally parameterized models consistent with cell behavior. We review existing examples where geometrical models have been used to fit experimental data on cell fate and describe how spatial interactions between cells can be understood geometrically.


Assuntos
Epigênese Genética , Redes Reguladoras de Genes , Diferenciação Celular/genética , Modelos Genéticos
10.
J Biol Phys ; 49(1): 1-27, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36580168

RESUMO

Cell fate decision processes are regulated by networks which contain different molecules and interactions. Different network topologies may exhibit synergistic or antagonistic effects on cellular functions. Here, we analyze six most common small networks with regulatory logic AND or OR, trying to clarify the relationship between network topologies and synergism (or antagonism) related to cell fate decisions. We systematically examine the contribution of both network topologies and regulatory logic to the cell fate synergism by bifurcation and combinatorial perturbation analysis. Initially, under a single set of parameters, the synergism of three types of networks with AND and OR logic is compared. Furthermore, to consider whether these results depend on the choices of parameter values, statistics on the synergism of five hundred parameter sets is performed. It is shown that the results are not sensitive to parameter variations, indicating that the synergy or antagonism mainly depends on the network topologies rather than the choices of parameter values. The results indicate that the topology with "Dual Inhibition" shows good synergism, while the topology with "Dual Promotion" or "Hybrid" shows antagonism. The results presented here may help us to design synergistic networks based on network structure and regulation combinations, which has promising implications for cell fate decisions and drug combinations.


Assuntos
Redes Reguladoras de Genes , Lógica , Interações Medicamentosas
11.
J Transl Med ; 20(1): 568, 2022 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-36474294

RESUMO

BACKGROUND: Mounting evidence has revealed the dynamic variations in the cellular status and phenotype of the smooth muscle cell (SMC) are vital for shaping the atherosclerotic plaque microenvironment and ultimately mapping onto heterogeneous clinical outcomes in coronary artery disease. Currently, the underlying clinical significance of SMC evolutions remains unexplored in atherosclerosis. METHODS: The dissociated cells from diseased segments within the right coronary artery of four cardiac transplant recipients and 1070 bulk samples with atherosclerosis from six bulk cohorts were retrieved. Following the SMC fate trajectory reconstruction, the MOVICS algorithm integrating the nearest template prediction was used to develop a stable and robust molecular classification. Subsequently, multi-dimensional potential biological implications, molecular features, and cell landscape heterogeneity among distinct clusters were decoded. RESULTS: We proposed an SMC cell fate decision signature (SCFDS)-based atherosclerosis stratification system and identified three SCFDS subtypes (C1-C3) with distinguishing features: (i) C1 (DNA-damage repair type), elevated base excision repair (BER), DNA replication, as well as oxidative phosphorylation status. (ii) C2 (immune-activated type), stronger immune activation, hyper-inflammatory state, the complex as well as varied lesion microenvironment, advanced stage, the most severe degree of coronary stenosis severity. (iii) C3 (stromal-rich type), abundant fibrous content, stronger ECM metabolism, immune-suppressed microenvironment. CONCLUSIONS: This study uncovered atherosclerosis complex cellular heterogeneity and a differentiated hierarchy of cell populations underlying SMC. The novel high-resolution stratification system could improve clinical outcomes and facilitate individualized management.


Assuntos
Miócitos de Músculo Liso
12.
Adv Exp Med Biol ; 1218: 1-7, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32060868

RESUMO

The attention of science first turned to the gene that later earned the name Notch over a century ago, when the American scientist John S. Dexter discovered in his laboratory at Olivet College the characteristic notched-wing phenotype (a nick or notch in the wingtip) in mutant fruit flies Drosophila melanogaster. At present, it is generally accepted that the Notch pathway governs tissue patterning and many key cell fate decisions and other core processes during embryonic development and in adult tissues. Not surprisingly, a broad variety of independent inherited diseases (including CADASIL, Alagille, Adams-Oliver, and Hajdu-Cheney syndromes) have now convincingly been linked to defective Notch signaling. In the second edition of the book entitled Notch Signaling in Embryology and Cancer, leading researchers provide a comprehensive, highly readable overview on molecular mechanisms of Notch signaling (Volume I), and notch's roles in embryology (Vol. II) and cancer (Vol. III). In these introductory pages of Vol. II, we give a short overview on its individual chapters, which are intended to provide both basic scientists and clinicians who seek today's clearest understanding of the broad role of Notch signaling in embryology with an authoritative day-to-day source.


Assuntos
Padronização Corporal , Receptores Notch/metabolismo , Transdução de Sinais , Animais , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriologia , Drosophila melanogaster/metabolismo , Humanos
13.
Adv Exp Med Biol ; 1218: 129-157, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32060875

RESUMO

Notch signalling is a major regulator of cell fate decisions and tissue patterning in metazoans. It is best known for its role in lateral inhibition, whereby Notch mediates competitive interactions between cells to limit adoption of a given developmental fate. However, it can also function by lateral induction, a cooperative mode of action that was originally described during the patterning of the Drosophila wing disc and creates boundaries or domains of cells of the same character. In this chapter, we introduce these two signalling modes and explain how they contribute to distinct aspects of the development and regeneration of the vertebrate inner ear, the organ responsible for the perception of sound and head movements. We discuss some of the factors that could influence the context-specific outcomes of Notch signalling in the inner ear and the ongoing efforts to target this pathway for the treatment of hearing loss and vestibular dysfunction.


Assuntos
Diferenciação Celular , Orelha Interna/embriologia , Orelha Interna/fisiologia , Receptores Notch/metabolismo , Regeneração , Transdução de Sinais , Animais , Orelha Interna/citologia , Orelha Interna/metabolismo , Perda Auditiva/metabolismo , Perda Auditiva/fisiopatologia , Humanos
14.
BMC Genomics ; 20(Suppl 2): 224, 2019 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-30967106

RESUMO

BACKGROUND: Time series single-cell RNA sequencing (scRNA-seq) data are emerging. However, the analysis of time series scRNA-seq data could be compromised by 1) distortion created by assorted sources of data collection and generation across time samples and 2) inheritance of cell-to-cell variations by stochastic dynamic patterns of gene expression. This calls for the development of an algorithm able to visualize time series scRNA-seq data in order to reveal latent structures and uncover dynamic transition processes. RESULTS: In this study, we propose an algorithm, termed time series elastic embedding (TSEE), by incorporating experimental temporal information into the elastic embedding (EE) method, in order to visualize time series scRNA-seq data. TSEE extends the EE algorithm by penalizing the proximal placement of latent points that correspond to data points otherwise separated by experimental time intervals. TSEE is herein used to visualize time series scRNA-seq datasets of embryonic developmental processed in human and zebrafish. We demonstrate that TSEE outperforms existing methods (e.g. PCA, tSNE and EE) in preserving local and global structures as well as enhancing the temporal resolution of samples. Meanwhile, TSEE reveals the dynamic oscillation patterns of gene expression waves during zebrafish embryogenesis. CONCLUSIONS: TSEE can efficiently visualize time series scRNA-seq data by diluting the distortions of assorted sources of data variation across time stages and achieve the temporal resolution enhancement by preserving temporal order and structure. TSEE uncovers the subtle dynamic structures of gene expression patterns, facilitating further downstream dynamic modeling and analysis of gene expression processes. The computational framework of TSEE is generalizable by allowing the incorporation of other sources of information.


Assuntos
Algoritmos , Biologia Computacional/métodos , Desenvolvimento Embrionário , Regulação da Expressão Gênica no Desenvolvimento , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Análise de Sequência de RNA/métodos , Análise de Célula Única/métodos , Animais , Perfilação da Expressão Gênica , Humanos , Fatores de Tempo , Peixe-Zebra/genética
15.
Dev Dyn ; 247(1): 47-53, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28643345

RESUMO

Mitochondria are classically known to be the cellular energy producers, but a renewed appreciation for these organelles has developed with the accumulating discoveries of additional functions. The importance of mitochondria within the brain has been long known, particularly given the high-energy demanding nature of neurons. The energy demands imposed by neurons require the well-orchestrated morphological adaptation and distribution of mitochondria. Recent studies now reveal the importance of mitochondrial dynamics not only in mature neurons but also during neural development, particularly during the process of neurogenesis and neural stem cell fate decisions. In this review, we will highlight the recent findings that illustrate the importance of mitochondrial dynamics in neurodevelopment and neural stem cell function. Developmental Dynamics 247:47-53, 2018. © 2017 Wiley Periodicals, Inc.


Assuntos
Encéfalo/metabolismo , Mitocôndrias/metabolismo , Dinâmica Mitocondrial/fisiologia , Neurogênese/fisiologia , Animais , Encéfalo/crescimento & desenvolvimento , Metabolismo Energético/fisiologia , Células-Tronco Neurais/metabolismo
16.
Development ; 142(13): 2250-60, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-26130756

RESUMO

The maintenance of pluripotency in embryonic stem cells (ESCs), its loss during lineage specification or its re-induction to generate induced pluripotent stem cells are central topics in stem cell biology. To uncover the molecular basis and the design principles of pluripotency control, a multitude of experimental, but also an increasing number of computational, studies have been published. Here, we consider recent reports that apply computational or mathematical modelling approaches to describe the regulatory processes that underlie cell fate decisions in mouse ESCs. We summarise the principles, the strengths and potentials but also the limitations of different computational strategies.


Assuntos
Linhagem da Célula , Simulação por Computador , Células-Tronco Embrionárias/citologia , Modelos Biológicos , Animais , Diferenciação Celular , Células-Tronco Embrionárias/metabolismo , Humanos , Células-Tronco Pluripotentes
17.
Stem Cells ; 35(2): 277-283, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27758015

RESUMO

Human embryonic stem cells (hESCs) present a fascinating and powerful system for generating specialized cell types of the human body. Culture and directed differentiation of these cells however requires an understanding of the pluripotent ground state and of how cell lineage decisions in this system are made. In this review, we highlight both these aspects in light of recent findings and technical progress. Hence, advances in culturing the human preimplantation embryo beyond the implantation barrier and in analyzing it at the single-cell level shed new light on the hESC tissue of origin. We argue that these findings have important implications for our view of hESC identity and we critically discuss recent efforts in converting these cells to a more primitive state. With an emphasis on the roles played by major signaling pathways, we furthermore attempt to infer key principles underlying cell fate control in hESCs from recently published work. This integrated model combines defined signaling pathway manipulation with the regulation of core hESC genes, to aid in controlling cell lineage allocation in a rational manner. Stem Cells 2017;35:277-283.


Assuntos
Linhagem da Célula , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Transdução de Sinais , Embrião de Mamíferos/citologia , Humanos , Modelos Biológicos
18.
Stem Cells ; 34(3): 699-710, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26731607

RESUMO

Hematopoietic stem cells (HSCs) maintain blood cell production life-long by their unique abilities of self-renewal and differentiation into all blood cell lineages. Growth arrest and DNA-damage-inducible 45 alpha (GADD45A) is induced by genotoxic stress in HSCs. GADD45A has been implicated in cell cycle control, cell death and senescence, as well as in DNA-damage repair. In general, GADD45A provides cellular stability by either arresting the cell cycle progression until DNA damage is repaired or, in cases of fatal damage, by inducing apoptosis. However, the function of GADD45A in hematopoiesis remains controversial. We revealed the changes in murine HSC fate control orchestrated by the expression of GADD45A at single cell resolution. In contrast to other cellular systems, GADD45A expression did not cause a cell cycle arrest or an alteration in the decision between cell survival and apoptosis in HSCs. Strikingly, GADD45A strongly induced and accelerated the differentiation program in HSCs. Continuous tracking of individual HSCs and their progeny via time-lapse microscopy elucidated that once GADD45A was expressed, HSCs differentiate into committed progenitors within 29 hours. GADD45A-expressing HSCs failed to long-term reconstitute the blood of recipients by inducing multilineage differentiation in vivo. Importantly, γ-irradiation of HSCs induced their differentiation by upregulating endogenous GADD45A. The differentiation induction by GADD45A was transmitted by activating p38 Mitogen-activated protein kinase (MAPK) signaling and allowed the generation of megakaryocytic-erythroid, myeloid, and lymphoid lineages. These data indicate that genotoxic stress-induced GADD45A expression in HSCs prevents their fatal transformation by directing them into differentiation and thereby clearing them from the system.


Assuntos
Pontos de Checagem do Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Diferenciação Celular/genética , Células-Tronco Hematopoéticas , Proteínas Nucleares/genética , Animais , Apoptose/genética , Proteínas de Ciclo Celular/biossíntese , Proliferação de Células/genética , Sobrevivência Celular/genética , Dano ao DNA/genética , Regulação da Expressão Gênica no Desenvolvimento , Hematopoese/genética , Humanos , Camundongos , Proteínas Nucleares/biossíntese , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/genética
19.
J Theor Biol ; 417: 84-99, 2017 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-28048969

RESUMO

Changes in gene expression are thought to regulate the cell differentiation process intrinsically through complex epigenetic mechanisms. In fundamental terms, however, this assumed regulation refers only to the intricate propagation of changes in gene expression or else leads to non-explanatory regresses. The developmental self-regulatory dynamics and evolution of individuated multicellular organisms also lack a unified and falsifiable description. To fill this gap, I computationally analyzed publicly available high-throughput data of histone H3 post-translational modifications and mRNA abundance for different Homo sapiens, Mus musculus, and Drosophila melanogaster cell-type/developmental-period samples. My analysis of genomic regions adjacent to transcription start sites generated a profile from pairwise partial correlations between histone modifications controlling for the respective mRNA levels for each cell-type/developmental-period dataset. I found that these profiles, while explicitly uncorrelated with the respective transcriptional "identities" by construction, associate strongly with cell differentiation states. This association is not expected if cell differentiation is, in effect, regulated by epigenetic mechanisms. Based on these results, I propose a general, falsifiable theory of individuated multicellularity, which relies on the synergistic coupling across the extracellular space of two explicitly uncorrelated "self-organizing" systems constraining histone modification states at the same sites. This theory describes how the simplest multicellular individual-understood as an intrinsic, higher-order constraint-emerges from proliferating undifferentiated cells, and could explain the intrinsic regulation of gene transcriptional changes for cell differentiation and the evolution of individuated multicellular organisms.


Assuntos
Evolução Biológica , Diferenciação Celular/genética , Epigênese Genética , Regulação da Expressão Gênica , Animais , Linhagem Celular , Biologia Computacional , Drosophila melanogaster , Histonas/metabolismo , Humanos , Camundongos , Processamento de Proteína Pós-Traducional , RNA Mensageiro/análise , Sítio de Iniciação de Transcrição
20.
EMBO Rep ; 16(3): 370-8, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25600117

RESUMO

Reprogramming to pluripotency is a low-efficiency process at the population level. Despite notable advances to molecularly characterize key steps, several fundamental aspects remain poorly understood, including when the potential to reprogram is first established. Here, we apply live-cell imaging combined with a novel statistical approach to infer when somatic cells become fated to generate downstream pluripotent progeny. By tracing cell lineages from several divisions before factor induction through to pluripotent colony formation, we find that pre-induction sister cells acquire similar outcomes. Namely, if one daughter cell contributes to a lineage that generates induced pluripotent stem cells (iPSCs), its paired sibling will as well. This result suggests that the potential to reprogram is predetermined within a select subpopulation of cells and heritable, at least over the short term. We also find that expanding cells over several divisions prior to factor induction does not increase the per-lineage likelihood of successful reprogramming, nor is reprogramming fate correlated to neighboring cell identity or cell-specific reprogramming factor levels. By perturbing the epigenetic state of somatic populations with Ezh2 inhibitors prior to factor induction, we successfully modulate the fraction of iPSC-forming lineages. Our results therefore suggest that reprogramming potential may in part reflect preexisting epigenetic heterogeneity that can be tuned to alter the cellular response to factor induction.


Assuntos
Linhagem da Célula/fisiologia , Reprogramação Celular/fisiologia , Epigênese Genética/fisiologia , Fibroblastos/citologia , Animais , Linhagem da Célula/efeitos dos fármacos , Reprogramação Celular/efeitos dos fármacos , Doxiciclina , Proteína Potenciadora do Homólogo 2 de Zeste , Proteínas de Fluorescência Verde/metabolismo , Processamento de Imagem Assistida por Computador , Camundongos , Análise em Microsséries , Células-Tronco Pluripotentes/citologia , Complexo Repressor Polycomb 2/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA