Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
Curr Opin Cell Biol ; 87: 102323, 2024 04.
Article in English | MEDLINE | ID: mdl-38301378

ABSTRACT

The process of embryonic development involves remarkable cellular plasticity, which governs the coordination between cells necessary to build an organism. One role of this plasticity is to ensure that when aberrant cells are eliminated, growth adjustment occurs so that the size of the tissue is maintained. An important regulator of cellular plasticity that ensures cellular cooperation is a fitness-sensing mechanism termed cell competition. During cell competition, cells with defects that lower fitness but do not affect viability, such as those that cause impaired signal transduction, slower cellular growth, mitochondrial dysregulation or impaired protein homeostasis, are killed when surrounded by fitter cells. This is accompanied by the compensatory proliferation of the surviving cells. The underlying factors and mechanisms that demarcate certain cells as less fit than their neighbouring cells and losers of cell competition are still relatively unknown. Recent evidence has pointed to mitochondrial defects and proteotoxic stress as important hallmarks of these loser cells. Here, we review recent advances in this area, focussing on the role of mitochondrial activity and protein homeostasis as major mechanisms determining competitive cell fitness during development and the importance of cell proteostasis in determining cell fitness.


Subject(s)
Cell Competition , Proteostasis , Signal Transduction/physiology , Cell Proliferation , Mitochondria
2.
Development ; 151(2)2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38131530

ABSTRACT

During development, the rate of tissue growth is determined by the relative balance of cell division and cell death. Cell competition is a fitness quality-control mechanism that contributes to this balance by eliminating viable cells that are less fit than their neighbours. The mutations that confer cells with a competitive advantage and the dynamics of the interactions between winner and loser cells are not well understood. Here, we show that embryonic cells lacking the tumour suppressor p53 are 'super-competitors' that eliminate their wild-type neighbours through the direct induction of apoptosis. This elimination is context dependent, as it does not occur when cells are pluripotent and it is triggered by the onset of differentiation. Furthermore, by combining mathematical modelling and cell-based assays we show that the elimination of wild-type cells is not through competition for space or nutrients, but instead is mediated by short-range interactions that are dependent on the local cell neighbourhood. This highlights the importance of the local cell neighbourhood and the competitive interactions within this neighbourhood for the regulation of proliferation during early embryonic development.


Subject(s)
Cell Communication , Pluripotent Stem Cells , Cell Communication/physiology , Tumor Suppressor Protein p53/genetics , Mutation/genetics , Apoptosis/genetics
3.
Cells ; 12(15)2023 07 28.
Article in English | MEDLINE | ID: mdl-37566036

ABSTRACT

MicroRNAs (miRNAs) are important regulators of embryonic stem cell (ESC) biology, and their study has identified key regulatory mechanisms. To find novel pathways regulated by miRNAs in ESCs, we undertook a bioinformatics analysis of gene pathways differently expressed in the absence of miRNAs due to the deletion of Dicer, which encodes an RNase that is essential for the synthesis of miRNAs. One pathway that stood out was Ca2+ signaling. Interestingly, we found that Dicer-/- ESCs had no difference in basal cytoplasmic Ca2+ levels but were hyperresponsive when Ca2+ import into the endoplasmic reticulum (ER) was blocked by thapsigargin. Remarkably, the increased Ca2+ response to thapsigargin in ESCs resulted in almost no increase in apoptosis and no differences in stress response pathways, despite the importance of miRNAs in the stress response of other cell types. The increased Ca2+ response in Dicer-/- ESCs was also observed during purinergic receptor activation, demonstrating a physiological role for the miRNA regulation of Ca2+ signaling pathways. In examining the mechanism of increased Ca2+ responsiveness to thapsigargin, neither store-operated Ca2+ entry nor Ca2+ clearance mechanisms from the cytoplasm appeared to be involved. Rather, it appeared to involve an increase in the expression of one isoform of the IP3 receptors (Itpr2). miRNA regulation of Itpr2 expression primarily appeared to be indirect, with transcriptional regulation playing a major role. Therefore, the miRNA regulation of Itpr2 expression offers a unique mechanism to regulate Ca2+ signaling pathways in the physiology of pluripotent stem cells.


Subject(s)
MicroRNAs , Animals , Mice , MicroRNAs/metabolism , Thapsigargin/pharmacology , Cell Differentiation/genetics , Embryonic Stem Cells , Homeostasis
4.
Cells ; 12(9)2023 05 07.
Article in English | MEDLINE | ID: mdl-37174738

ABSTRACT

The induction of pluripotency by enforced expression of different sets of genes in somatic cells has been achieved with reprogramming technologies first described by Yamanaka's group. Methodologies for generating induced pluripotent stem cells are as varied as the combinations of genes used. It has previously been reported that the adenoviral E1a gene can induce the expression of two of the Yamanaka factors (c-Myc and Oct-4) and epigenetic changes. Here, we demonstrate that the E1a-12S over-expression is sufficient to induce pluripotent-like characteristics closely to epiblast stem cells in mouse embryonic fibroblasts through the activation of the pluripotency gene regulatory network. These findings provide not only empirical evidence that the expression of one single factor is sufficient for partial reprogramming but also a potential mechanistic explanation for how viral infection could lead to neoplasia if they are surrounded by the appropriate environment or the right medium, as happens with the tumorogenic niche.


Subject(s)
Cellular Reprogramming , Induced Pluripotent Stem Cells , Animals , Mice , Cellular Reprogramming/genetics , Cell Differentiation , Fibroblasts/metabolism , Kruppel-Like Factor 4 , Induced Pluripotent Stem Cells/metabolism
5.
Cell Stem Cell ; 29(7): 1018-1030, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35803224

ABSTRACT

The mammalian embryo exhibits a remarkable plasticity that allows it to correct for the presence of aberrant cells, adjust its growth so that its size is in accordance with its developmental stage, or integrate cells of another species to form fully functional organs. Here, we will discuss the contribution that cell competition, a quality control that eliminates viable cells that are less fit than their neighbors, makes to this plasticity. We will do this by reviewing the roles that cell competition plays in the early mammalian embryo and how they contribute to ensure normal development of the embryo.


Subject(s)
Cell Competition , Embryo, Mammalian , Animals , Embryonic Development , Mammals
6.
Dev Cell ; 57(11): 1316-1330.e7, 2022 06 06.
Article in English | MEDLINE | ID: mdl-35597240

ABSTRACT

The changes that drive differentiation facilitate the emergence of abnormal cells that need to be removed before they contribute to further development or the germline. Consequently, in mice in the lead-up to gastrulation, ∼35% of embryonic cells are eliminated. This elimination is caused by hypersensitivity to apoptosis, but how it is regulated is poorly understood. Here, we show that upon exit of naive pluripotency, mouse embryonic stem cells lower their mitochondrial apoptotic threshold, and this increases their sensitivity to cell death. We demonstrate that this enhanced apoptotic response is induced by a decrease in mitochondrial fission due to a reduction in the activity of dynamin-related protein 1 (DRP1). Furthermore, we show that in naive pluripotent cells, DRP1 prevents apoptosis by promoting mitophagy. In contrast, during differentiation, reduced mitophagy levels facilitate apoptosis. Together, these results indicate that during early mammalian development, DRP1 regulation of mitophagy determines the apoptotic response.


Subject(s)
Dynamins/metabolism , Mitophagy , Animals , Apoptosis/physiology , Mammals/metabolism , Mice , Mitochondria/metabolism , Mitochondrial Dynamics/physiology , Mitophagy/physiology
7.
Diabetes ; 71(7): 1472-1489, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35472764

ABSTRACT

Mitochondrial glucose metabolism is essential for stimulated insulin release from pancreatic ß-cells. Whether mitofusin gene expression, and hence, mitochondrial network integrity, is important for glucose or incretin signaling has not previously been explored. Here, we generated mice with ß-cell-selective, adult-restricted deletion knock-out (dKO) of the mitofusin genes Mfn1 and Mfn2 (ßMfn1/2 dKO). ßMfn1/2-dKO mice displayed elevated fed and fasted glycemia and a more than fivefold decrease in plasma insulin. Mitochondrial length, glucose-induced polarization, ATP synthesis, and cytosolic and mitochondrial Ca2+ increases were all reduced in dKO islets. In contrast, oral glucose tolerance was more modestly affected in ßMfn1/2-dKO mice, and glucagon-like peptide 1 or glucose-dependent insulinotropic peptide receptor agonists largely corrected defective glucose-stimulated insulin secretion through enhanced EPAC-dependent signaling. Correspondingly, cAMP increases in the cytosol, as measured with an Epac-camps-based sensor, were exaggerated in dKO mice. Mitochondrial fusion and fission cycles are thus essential in the ß-cell to maintain normal glucose, but not incretin, sensing. These findings broaden our understanding of the roles of mitofusins in ß-cells, the potential contributions of altered mitochondrial dynamics to diabetes development, and the impact of incretins on this process.


Subject(s)
GTP Phosphohydrolases , Glucose , Incretins , Insulin-Secreting Cells , Animals , GTP Phosphohydrolases/genetics , Glucose/metabolism , Glucose/pharmacology , Guanine Nucleotide Exchange Factors/metabolism , Incretins/metabolism , Incretins/pharmacology , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Mice , Mice, Knockout
9.
Dev Cell ; 56(17): 2455-2470.e10, 2021 09 13.
Article in English | MEDLINE | ID: mdl-34407428

ABSTRACT

The appearance of genetic changes in human pluripotent stem cells (hPSCs) presents a concern for their use in research and regenerative medicine. Variant hPSCs that harbor recurrent culture-acquired aneuploidies display growth advantages over wild-type diploid cells, but the mechanisms that yield a drift from predominantly wild-type to variant cell populations remain poorly understood. Here, we show that the dominance of variant clones in mosaic cultures is enhanced through competitive interactions that result in the elimination of wild-type cells. This elimination occurs through corralling and mechanical compression by faster-growing variants, causing a redistribution of F-actin and sequestration of yes-associated protein (YAP) in the cytoplasm that induces apoptosis in wild-type cells. YAP overexpression or promotion of YAP nuclear localization in wild-type cells alleviates their "loser" phenotype. Our results demonstrate that hPSC fate is coupled to mechanical cues imposed by neighboring cells and reveal that hijacking this mechanism allows variants to achieve clonal dominance in cultures.


Subject(s)
Cell Competition/genetics , Cell Differentiation/genetics , Cell Proliferation/genetics , Pluripotent Stem Cells/cytology , YAP-Signaling Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Cells, Cultured , Cytoplasm/metabolism , Humans , Transcription Factors/metabolism
11.
Nat Metab ; 3(8): 1091-1108, 2021 08.
Article in English | MEDLINE | ID: mdl-34253906

ABSTRACT

Cell competition is emerging as a quality-control mechanism that eliminates unfit cells in a wide range of settings from development to the adult. However, the nature of the cells normally eliminated by cell competition and what triggers their elimination remains poorly understood. In mice, 35% of epiblast cells are eliminated before gastrulation. Here we show that cells with mitochondrial defects are eliminated by cell competition during early mouse development. Using single-cell transcriptional profiling of eliminated mouse epiblast cells, we identify hallmarks of cell competition and mitochondrial defects. We demonstrate that mitochondrial defects are common to a range of different loser cell types and that manipulating mitochondrial function triggers cell competition. Moreover, we show that in the mouse embryo, cell competition eliminates cells with sequence changes in mt-Rnr1 and mt-Rnr2, and that even non-pathological changes in mitochondrial DNA sequences can induce cell competition. Our results suggest that cell competition is a purifying selection that optimizes mitochondrial performance before gastrulation.


Subject(s)
Cell Competition , Embryo, Mammalian , Embryonic Development , Mitochondria/genetics , Mitochondria/metabolism , Animals , Biomarkers , Embryonic Development/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental , Mice , Single-Cell Analysis/methods
12.
Genes Dev ; 35(7-8): 433-448, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33861719

ABSTRACT

p53 is an important tumor suppressor, and the complexities of p53 function in regulating cancer cell behaviour are well established. Many cancers lose or express mutant forms of p53, with evidence that the type of alteration affecting p53 may differentially impact cancer development and progression. It is also clear that in addition to cell-autonomous functions, p53 status also affects the way cancer cells interact with each other. In this review, we briefly examine the impact of different p53 mutations and focus on how heterogeneity of p53 status can affect relationships between cells within a tumor.


Subject(s)
Cell Communication/genetics , Mutation/genetics , Neoplasms/genetics , Neoplasms/physiopathology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Carcinogenesis/genetics , Cell Competition/genetics , Embryonic Development/genetics , Humans
13.
Curr Biol ; 31(5): R255-R257, 2021 03 08.
Article in English | MEDLINE | ID: mdl-33689726

ABSTRACT

During cell competition fitter cells eliminate the weaker ones. New work identifies FGF21 as a factor that is secreted by the prospective loser cells of this competition and that acts to attract the winners towards them.


Subject(s)
Cell Competition , Drosophila melanogaster , Animals , Apoptosis , Prospective Studies
14.
Genes Dev ; 34(23-24): 1599-1604, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33184225

ABSTRACT

There is increasing evidence demonstrating that adult neural stem cells (NSCs) are a cell of origin of glioblastoma. Here we analyzed the interaction between transformed and wild-type NSCs isolated from the adult mouse subventricular zone niche. We found that transformed NSCs are refractory to quiescence-inducing signals. Unexpectedly, we also demonstrated that these cells induce quiescence in surrounding wild-type NSCs in a cell-cell contact and Notch signaling-dependent manner. Our findings therefore suggest that oncogenic mutations are propagated in the stem cell niche not just through cell-intrinsic advantages, but also by outcompeting neighboring stem cells through repression of their proliferation.


Subject(s)
Glioblastoma/physiopathology , Neoplastic Stem Cells/physiology , Neural Stem Cells/cytology , Receptors, Notch/genetics , Signal Transduction/physiology , Animals , Cell Communication/physiology , Cell Proliferation/physiology , Cells, Cultured , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Lateral Ventricles/cytology , Mice , Neoplastic Stem Cells/cytology , Neural Stem Cells/physiology
15.
Semin Cancer Biol ; 63: 36-43, 2020 06.
Article in English | MEDLINE | ID: mdl-31102668

ABSTRACT

The maintenance of tissue homeostasis and health relies on the efficient removal of damaged or otherwise suboptimal cells. One way this is achieved is through cell competition, a fitness quality control mechanism that eliminates cells that are less fit than their neighbours. Through this process, cell competition has been shown to play diverse roles in development and in the adult, including in homeostasis and tumour suppression. However, over the last few years it has also become apparent that certain oncogenic mutations can provide cells with a competitive advantage that promotes their expansion via the elimination of surrounding wild-type cells. Thus, understanding how this process is initiated and regulated will provide important insights with relevance to a number of different research areas. A key question in cell competition is what determines the competitive fitness of a cell. Here, we will review what is known about this question by focussing on two non-mutually exclusive possibilities; first, that the activity of a subset of transcription factors determines competitive fitness, and second, that the outcome of cell competition is determined by the relative cellular metabolic status.


Subject(s)
Neoplasms/genetics , Neoplasms/pathology , Animals , Cell Communication/physiology , Genetic Fitness , Humans , Mutation , Neoplasms/metabolism , Oncogenes , Selection, Genetic , Transcription, Genetic
16.
Stem Cell Reports ; 13(6): 970-979, 2019 12 10.
Article in English | MEDLINE | ID: mdl-31761678

ABSTRACT

The role of the homeobox transcriptional repressor HESX1 in embryonic stem cells (ESCs) remains mostly unknown. Here, we show that Hesx1 is expressed in the preimplantation mouse embryo, where it is required during developmental diapause. Absence of Hesx1 leads to reduced expression of epiblast and primitive endoderm determinants and failure of diapaused embryos to resume embryonic development after implantation. Genetic deletion of Hesx1 impairs self-renewal and promotes differentiation toward epiblast by reducing the expression of pluripotency factors and decreasing the activity of LIF/STAT3 signaling. We reveal that Hesx1-deficient ESCs show elevated ERK pathway activation, resulting in accelerated differentiation toward primitive endoderm, which can be prevented by overexpression of Hesx1. Together, our data provide evidence for a novel role of Hesx1 in the control of self-renewal and maintenance of the undifferentiated state in ESCs and mouse embryos.


Subject(s)
Cell Differentiation/genetics , Cell Self Renewal/genetics , Diapause/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Deletion , Repressor Proteins/deficiency , Animals , Biomarkers , Embryonic Development , Fluorescent Antibody Technique , Gene Expression Regulation , Homeodomain Proteins , Leukemia Inhibitory Factor/metabolism , MAP Kinase Signaling System , Mice , Models, Biological , Phenotype , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Signal Transduction
17.
Development ; 146(13)2019 07 05.
Article in English | MEDLINE | ID: mdl-31278123

ABSTRACT

The process of cell competition results in the 'elimination of cells that are viable but less fit than surrounding cells'. Given the highly heterogeneous nature of our tissues, it seems increasingly likely that cells are engaged in a 'survival of the fittest' battle throughout life. The process has a myriad of positive roles in the organism: it selects against mutant cells in developing tissues, prevents the propagation of oncogenic cells and eliminates damaged cells during ageing. However, 'super-fit' cancer cells can exploit cell competition mechanisms to expand and spread. Here, we review the regulation, roles and risks of cell competition in organism development, ageing and disease.


Subject(s)
Cell Communication/physiology , Cell Physiological Phenomena , Competitive Behavior/physiology , Genetic Fitness/physiology , Selection, Genetic/physiology , Aging/physiology , Animals , Cell Physiological Phenomena/genetics , Cellular Microenvironment/physiology , Humans , Reproduction/physiology
18.
Nat Commun ; 9(1): 3123, 2018 08 02.
Article in English | MEDLINE | ID: mdl-30072790

ABSTRACT

The original version of this article contained an error in the spelling of Juan Pedro Martinez-Barbera, which was incorrectly given as Juan Pedro Martinez Barbera. This error has now been corrected in both the PDF and HTML versions of the Article.

19.
Nat Commun ; 9(1): 1763, 2018 05 02.
Article in English | MEDLINE | ID: mdl-29720666

ABSTRACT

Ensuring the fitness of the pluripotent cells that will contribute to future development is important both for the integrity of the germline and for proper embryogenesis. Consequently, it is becoming increasingly apparent that pluripotent cells can compare their fitness levels and signal the elimination of those cells that are less fit than their neighbours. In mammals the nature of the pathways that communicate fitness remain largely unknown. Here we identify that in the early mouse embryo and upon exit from naive pluripotency, the confrontation of cells with different fitness levels leads to an inhibition of mTOR signalling in the less fit cell type, causing its elimination. We show that during this process, p53 acts upstream of mTOR and is required to repress its activity. Finally, we demonstrate that during normal development around 35% of cells are eliminated by this pathway, highlighting the importance of this mechanism for embryonic development.


Subject(s)
Embryo, Mammalian/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cell Communication/genetics , Cell Differentiation/genetics , Cell Line , Cell Proliferation/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryonic Development/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , TOR Serine-Threonine Kinases/genetics , Tumor Suppressor Protein p53/genetics
20.
Curr Top Dev Biol ; 128: 339-363, 2018.
Article in English | MEDLINE | ID: mdl-29477168

ABSTRACT

From fertilization until the onset of gastrulation the early mammalian embryo undergoes a dramatic series of changes that converts a single fertilized cell into a remarkably complex organism. Much attention has been given to the molecular changes occurring during this process, but here we will review what is known about the changes affecting the mitochondria and how they impact on the energy metabolism and apoptotic response of the embryo. We will also focus on understanding what quality control mechanisms ensure optimal mitochondrial activity in the embryo, and in this way provide an overview of the importance of the mitochondria in determining cell fitness during early mammalian development.


Subject(s)
Embryonic Development , Mammals/embryology , Mitochondria/metabolism , Animals , Apoptosis , DNA, Mitochondrial/genetics , Humans , Organelle Biogenesis
SELECTION OF CITATIONS
SEARCH DETAIL
...