Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters











Publication year range
1.
Blood ; 137(16): 2139-2151, 2021 04 22.
Article in English | MEDLINE | ID: mdl-33667305

ABSTRACT

We studied a subset of hematopoietic stem cells (HSCs) that are defined by elevated expression of CD41 (CD41hi) and showed bias for differentiation toward megakaryocytes (Mks). Mouse models of myeloproliferative neoplasms (MPNs) expressing JAK2-V617F (VF) displayed increased frequencies and percentages of the CD41hi vs CD41lo HSCs compared with wild-type controls. An increase in CD41hi HSCs that correlated with JAK2-V617F mutant allele burden was also found in bone marrow from patients with MPN. CD41hi HSCs produced a higher number of Mk-colonies of HSCs in single-cell cultures in vitro, but showed reduced long-term reconstitution potential compared with CD41lo HSCs in competitive transplantations in vivo. RNA expression profiling showed an upregulated cell cycle, Myc, and oxidative phosphorylation gene signatures in CD41hi HSCs, whereas CD41lo HSCs showed higher gene expression of interferon and the JAK/STAT and TNFα/NFκB signaling pathways. Higher cell cycle activity and elevated levels of reactive oxygen species were confirmed in CD41hi HSCs by flow cytometry. Expression of Epcr, a marker for quiescent HSCs inversely correlated with expression of CD41 in mice, but did not show such reciprocal expression pattern in patients with MPN. Treatment with interferon-α further increased the frequency and percentage of CD41hi HSCs and reduced the number of JAK2-V617F+ HSCs in mice and patients with MPN. The shift toward the CD41hi subset of HSCs by interferon-α provides a possible mechanism of how interferon-α preferentially targets the JAK2 mutant clone.


Subject(s)
Interferon-alpha/therapeutic use , Janus Kinase 2/genetics , Megakaryocytes/metabolism , Myeloproliferative Disorders/genetics , Animals , Gene Knock-In Techniques , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Megakaryocytes/cytology , Mice , Mice, Transgenic , Myeloproliferative Disorders/drug therapy , Platelet Membrane Glycoprotein IIb/genetics , Point Mutation/drug effects
2.
Nature ; 573(7775): E5, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31515536

ABSTRACT

An Amendment to this paper has been published and can be accessed via a link at the top of the paper.

3.
Nature ; 573(7774): 426-429, 2019 09.
Article in English | MEDLINE | ID: mdl-31485073

ABSTRACT

Haematopoietic stem cells self-renew and differentiate into all blood lineages throughout life, and can repair damaged blood systems upon transplantation. Asymmetric cell division has previously been suspected to be a regulator of haematopoietic-stem-cell fate, but its existence has not directly been shown1. In asymmetric cell division, asymmetric fates of future daughter cells are prospectively determined by a mechanism that is linked to mitosis. This can be mediated by asymmetric inheritance of cell-extrinsic niche signals by, for example, orienting the divisional plane, or by the asymmetric inheritance of cell-intrinsic fate determinants. Observations of asymmetric inheritance or of asymmetric daughter-cell fates alone are not sufficient to demonstrate asymmetric cell division2. In both cases, sister-cell fates could be controlled by mechanisms that are independent of division. Here we demonstrate that the cellular degradative machinery-including lysosomes, autophagosomes, mitophagosomes and the protein NUMB-can be asymmetrically inherited into haematopoietic-stem-cell daughter cells. This asymmetric inheritance predicts the asymmetric future metabolic and translational activation and fates of haematopoietic-stem-cell daughter cells and their offspring. Therefore, our studies provide evidence for the existence of asymmetric cell division in haematopoietic stem cells.

4.
Bioinformatics ; 35(13): 2291-2299, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30452534

ABSTRACT

MOTIVATION: Recent imaging technologies allow for high-throughput tracking of cells as they migrate, divide, express fluorescent markers and change their morphology. The interpretation of these data requires unbiased, efficient statistical methods that model the dynamics of cell phenotypes. RESULTS: We introduce treeHFM, a probabilistic model which generalizes the theory of hidden Markov models to tree structured data. While accounting for the entire genealogy of a cell, treeHFM categorizes cells according to their primary phenotypic features. It models all relevant events in a cell's life, including cell division, and thereby enables the analysis of event order and cell fate heterogeneity. Simulations show higher accuracy in predicting correct state labels when modeling the more complex, tree-shaped dependency of samples over standard HMM modeling. Applying treeHFM to time lapse images of hematopoietic progenitor cell differentiation, we demonstrate that progenitor cells undergo a well-ordered sequence of differentiation events. AVAILABILITY AND IMPLEMENTATION: The treeHFM is implemented in C++. We provide wrapper functions for the programming languages R (CRAN package, https://CRAN.R-project.org/package=treeHFM) and Matlab (available at Mathworks Central, http://se.mathworks.com/matlabcentral/fileexchange/57575-treehfml). SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Time-Lapse Imaging , Cluster Analysis , Models, Statistical , Programming Languages , Software
5.
Nat Methods ; 14(4): 403-406, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28218899

ABSTRACT

Differentiation alters molecular properties of stem and progenitor cells, leading to changes in their shape and movement characteristics. We present a deep neural network that prospectively predicts lineage choice in differentiating primary hematopoietic progenitors using image patches from brightfield microscopy and cellular movement. Surprisingly, lineage choice can be detected up to three generations before conventional molecular markers are observable. Our approach allows identification of cells with differentially expressed lineage-specifying genes without molecular labeling.


Subject(s)
Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Image Processing, Computer-Assisted/methods , Neural Networks, Computer , Time-Lapse Imaging/methods , Animals , Area Under Curve , Biomarkers/metabolism , Cell Differentiation , Cell Lineage , Gene Knock-In Techniques , Machine Learning , Male , Mice, Mutant Strains , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
6.
Blood ; 129(12): 1691-1701, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28159742

ABSTRACT

Controlled regulation of lineage decisions is imperative for hematopoiesis. Yet, the molecular mechanisms underlying hematopoietic lineage choices are poorly defined. Colony-stimulating factor 1 (CSF-1), the cytokine acting as the principal regulator of monocyte/macrophage (M) development, has been shown to be able to instruct the lineage choice of uncommitted granulocyte M (GM) progenitors toward an M fate. However, the intracellular signaling pathways involved are unknown. CSF-1 activates a multitude of signaling pathways resulting in a pleiotropic cellular response. The precise role of individual pathways within this complex and redundant signaling network is dependent on cellular context, and is not well understood. Here, we address which CSF-1-activated pathways are involved in transmitting the lineage-instructive signal in primary bone marrow-derived GM progenitors. Although its loss is compensated for by alternative signaling activation mechanisms, Src family kinase (SFK) signaling is sufficient to transmit the CSF-1 lineage instructive signal. Moreover, c-Src activity is sufficient to drive M fate, even in nonmyeloid cells.


Subject(s)
Cell Lineage , Macrophage Colony-Stimulating Factor/physiology , Monocytes/cytology , Signal Transduction , src-Family Kinases/metabolism , Animals , Cells, Cultured , Granulocyte Precursor Cells/cytology , Hematopoiesis , Mice
7.
Sci Rep ; 6: 37462, 2016 11 28.
Article in English | MEDLINE | ID: mdl-27892478

ABSTRACT

Functionally distinct plasmacytoid and conventional dendritic cells (pDC and cDC) shape innate and adaptive immunity. They are derived from common dendritic cell progenitors (CDPs) in the murine bone marrow, which give rise to CD11c+ MHCII- precursors with early commitment to DC subpopulations. In this study, we dissect pDC development from CDP into an ordered sequence of differentiation events by monitoring the expression of CD11c, MHC class II, Siglec H and CCR9 in CDP cultures by continuous single cell imaging and tracking. Analysis of CDP genealogies revealed a stepwise differentiation of CDPs into pDCs in a part of the CDP colonies. This developmental pathway involved an early CD11c+ SiglecH- pre-DC stage and a Siglec H+ CCR9low precursor stage, which was followed rapidly by upregulation of CCR9 indicating final pDC differentiation. In the majority of the remaining CDP pedigrees however the Siglec H+ CCR9low precursor state was maintained for several generations. Thus, although a fraction of CDPs transits through precursor stages rapidly to give rise to a first wave of pDCs, the majority of CDP progeny differentiate more slowly and give rise to longer lived precursor cells which are poised to differentiate on demand.


Subject(s)
Bone Marrow Cells/cytology , Cell Lineage/immunology , Dendritic Cells/cytology , Single-Cell Analysis/methods , Stem Cells/cytology , Animals , Biomarkers/metabolism , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , CD11c Antigen/genetics , CD11c Antigen/immunology , Cell Differentiation , Dendritic Cells/immunology , Dendritic Cells/metabolism , Femur/cytology , Femur/immunology , Femur/metabolism , Flow Cytometry , Gene Expression , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Immunophenotyping , Mice , Mice, Inbred C57BL , Primary Cell Culture , Receptors, CCR/genetics , Receptors, CCR/immunology , Sialic Acid Binding Immunoglobulin-like Lectins/genetics , Sialic Acid Binding Immunoglobulin-like Lectins/immunology , Stem Cells/immunology , Stem Cells/metabolism , Tibia/cytology , Tibia/immunology , Tibia/metabolism
9.
Blood ; 128(9): 1181-92, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27365423

ABSTRACT

The maintenance of hematopoietic stem cells (HSCs) during ex vivo culture is an important prerequisite for their therapeutic manipulation. However, despite intense research, culture conditions for robust maintenance of HSCs are still missing. Cultured HSCs are quickly lost, preventing their improved analysis and manipulation. Identification of novel factors supporting HSC ex vivo maintenance is therefore necessary. Coculture with the AFT024 stroma cell line is capable of maintaining HSCs ex vivo long-term, but the responsible molecular players remain unknown. Here, we use continuous long-term single-cell observation to identify the HSC behavioral signature under supportive or nonsupportive stroma cocultures. We report early HSC survival as a major characteristic of HSC-maintaining conditions. Behavioral screening after manipulation of candidate molecules revealed that the extracellular matrix protein dermatopontin (Dpt) is involved in HSC maintenance. DPT knockdown in supportive stroma impaired HSC survival, whereas ectopic expression of the Dpt gene or protein in nonsupportive conditions restored HSC survival. Supplementing defined stroma- and serum-free culture conditions with recombinant DPT protein improved HSC clonogenicity. These findings illustrate a previously uncharacterized role of Dpt in maintaining HSCs ex vivo.


Subject(s)
Chondroitin Sulfate Proteoglycans/metabolism , Extracellular Matrix Proteins/metabolism , Hematopoietic Stem Cells/metabolism , Animals , Cell Culture Techniques , Cell Line , Cell Survival/drug effects , Cell Survival/genetics , Chondroitin Sulfate Proteoglycans/genetics , Chondroitin Sulfate Proteoglycans/pharmacology , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/pharmacology , Hematopoietic Stem Cells/cytology , Male , Mice , Mice, Transgenic , Stromal Cells/cytology , Stromal Cells/metabolism , Time Factors
10.
Nature ; 535(7611): 299-302, 2016 07 14.
Article in English | MEDLINE | ID: mdl-27411635

ABSTRACT

The mechanisms underlying haematopoietic lineage decisions remain disputed. Lineage-affiliated transcription factors with the capacity for lineage reprogramming, positive auto-regulation and mutual inhibition have been described as being expressed in uncommitted cell populations. This led to the assumption that lineage choice is cell-intrinsically initiated and determined by stochastic switches of randomly fluctuating cross-antagonistic transcription factors. However, this hypothesis was developed on the basis of RNA expression data from snapshot and/or population-averaged analyses. Alternative models of lineage choice therefore cannot be excluded. Here we use novel reporter mouse lines and live imaging for continuous single-cell long-term quantification of the transcription factors GATA1 and PU.1 (also known as SPI1). We analyse individual haematopoietic stem cells throughout differentiation into megakaryocytic-erythroid and granulocytic-monocytic lineages. The observed expression dynamics are incompatible with the assumption that stochastic switching between PU.1 and GATA1 precedes and initiates megakaryocytic-erythroid versus granulocytic-monocytic lineage decision-making. Rather, our findings suggest that these transcription factors are only executing and reinforcing lineage choice once made. These results challenge the current prevailing model of early myeloid lineage choice.


Subject(s)
Cell Differentiation , Cell Lineage , GATA1 Transcription Factor/metabolism , Myeloid Cells/cytology , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Animals , Erythrocytes/cytology , Feedback, Physiological , Female , Genes, Reporter , Granulocytes/cytology , Hematopoiesis , Hematopoietic Stem Cells/cytology , Male , Megakaryocytes/cytology , Mice , Models, Biological , Monocytes/cytology , Reproducibility of Results , Single-Cell Analysis , Stochastic Processes
11.
Nat Commun ; 7: 11903, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27301576

ABSTRACT

Stem cell identity depends on the integration of extrinsic and intrinsic signals, which directly influence the maintenance of their epigenetic state. Although Myc transcription factors play a major role in stem cell self-renewal and pluripotency, their integration with signalling pathways and epigenetic regulators remains poorly defined. We addressed this point by profiling the gene expression and epigenetic pattern in ESCs whose growth depends on conditional Myc activity. Here we show that Myc potentiates the Wnt/ß-catenin signalling pathway, which cooperates with the transcriptional regulatory network in sustaining ESC self-renewal. Myc activation results in the transcriptional repression of Wnt antagonists through the direct recruitment of PRC2 on these targets. The consequent potentiation of the autocrine Wnt/ß-catenin signalling induces the transcriptional activation of the endogenous Myc family members, which in turn activates a Myc-driven self-reinforcing circuit. Thus, our data unravel a Myc-dependent self-propagating epigenetic memory in the maintenance of ESC self-renewal capacity.


Subject(s)
Gene Regulatory Networks/genetics , Mouse Embryonic Stem Cells/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Animals , Cell Self Renewal/drug effects , Epigenesis, Genetic/drug effects , Feedback, Physiological/drug effects , Gene Regulatory Networks/drug effects , Leukemia Inhibitory Factor/pharmacology , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/drug effects , Polycomb-Group Proteins/metabolism , Transcription, Genetic/drug effects , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/genetics
12.
Nat Cell Biol ; 17(10): 1235-46, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26389663

ABSTRACT

Transcription factor (TF) networks are thought to regulate embryonic stem cell (ESC) pluripotency. However, TF expression dynamics and regulatory mechanisms are poorly understood. We use reporter mouse ESC lines allowing non-invasive quantification of Nanog or Oct4 protein levels and continuous long-term single-cell tracking and quantification over many generations to reveal diverse TF protein expression dynamics. For cells with low Nanog expression, we identified two distinct colony types: one re-expressed Nanog in a mosaic pattern, and the other did not re-express Nanog over many generations. Although both expressed pluripotency markers, they exhibited differences in their TF protein correlation networks and differentiation propensities. Sister cell analysis revealed that differences in Nanog levels are not necessarily accompanied by differences in the expression of other pluripotency factors. Thus, regulatory interactions of pluripotency TFs are less stringently implemented in individual self-renewing ESCs than assumed at present.


Subject(s)
Embryonic Stem Cells/metabolism , Gene Regulatory Networks , Pluripotent Stem Cells/metabolism , Transcription Factors/genetics , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Differentiation/genetics , Cell Tracking/methods , Cells, Cultured , Embryonic Stem Cells/cytology , Gene Expression , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Microscopy, Fluorescence , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/cytology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Single-Cell Analysis/methods , Time-Lapse Imaging/methods , Transcription Factors/metabolism , Transduction, Genetic , Red Fluorescent Protein
13.
Integr Biol (Camb) ; 7(2): 178-83, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25514157

ABSTRACT

The kinetics of stem and progenitor cell differentiation at the single-cell level provides essential clues to the complexity of the underlying decision-making circuits. In many hematopoietic progenitor cells, differentiation is accompanied by the expression of lineage-specific markers and by a transition from a non-adherent to an adherent state. Here, using the granulocyte-macrophage progenitor (GMP) as a model, we introduce a label-free approach that allows one to follow the course of this transition in hundreds of single cells in parallel. We trap single cells in patterned arrays of micro-wells and use phase-contrast time-lapse movies to distinguish non-adherent from adherent cells by an analysis of Brownian motion. This approach allowed us to observe the kinetics of induced differentiation of primary bone-marrow-derived GMPs into macrophages. The time lapse started 2 hours after addition of the cytokine M-CSF, and nearly 80% of the population had accomplished the transition within the first 20 h. The analysis of Brownian motion proved to be a sensitive and robust tool for monitoring the transition, and thus provides a high-throughput method for the study of cell differentiation at the single-cell level.


Subject(s)
Granulocyte-Macrophage Progenitor Cells/cytology , Lab-On-A-Chip Devices , Animals , Biophysical Phenomena , Cell Adhesion , Cell Differentiation , Cell Lineage , Equipment Design , Granulocyte-Macrophage Progenitor Cells/physiology , Mice , Microscopy, Phase-Contrast , Motion , Single-Cell Analysis/instrumentation , Time-Lapse Imaging/instrumentation
14.
Cell Stem Cell ; 15(5): 546-58, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25517464

ABSTRACT

Understanding the molecular control of cell fates is central to stem cell research. Such insight requires quantification of molecular and cellular behavior at the single-cell level. Recent advances now permit high-throughput molecular readouts from single cells as well as continuous, noninvasive observation of cell behavior over time. Here, we review current state-of-the-art approaches used to query stem cell fate at the single-cell level, including advances in lineage tracing, time-lapse imaging, and molecular profiling. We also offer our perspective on the advantages and drawbacks of available approaches, key technical limitations, considerations for data interpretation, and future innovation.


Subject(s)
Single-Cell Analysis/methods , Stem Cell Research , Animals , Cell Lineage , Genetic Techniques , Humans , Molecular Imaging , Stem Cell Transplantation
15.
Exp Cell Res ; 329(2): 207-13, 2014 Dec 10.
Article in English | MEDLINE | ID: mdl-25046868

ABSTRACT

Hematopoiesis is the cumulative consequence of finely tuned signaling pathways activated through extrinsic factors, such as local niche signals and systemic hematopoietic cytokines. Whether extrinsic factors actively instruct the lineage choice of hematopoietic stem and progenitor cells or are only selectively allowing survival and proliferation of already intrinsically lineage-committed cells has been debated over decades. Recent results demonstrated that cytokines can instruct lineage choice. However, the precise function of individual cytokine-triggered signaling molecules in inducing cellular events like proliferation, lineage choice, and differentiation remains largely elusive. Signal transduction pathways activated by different cytokine receptors are highly overlapping, but support the production of distinct hematopoietic lineages. Cellular context, signaling dynamics, and the crosstalk of different signaling pathways determine the cellular response of a given extrinsic signal. New tools to manipulate and continuously quantify signaling events at the single cell level are therefore required to thoroughly interrogate how dynamic signaling networks yield a specific cellular response.


Subject(s)
Cell Differentiation/drug effects , Cell Lineage/drug effects , Cytokines/pharmacology , Hematopoiesis/drug effects , Hematopoietic Stem Cells/drug effects , Signal Transduction/drug effects , Animals , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans
16.
Mol Biotechnol ; 56(8): 697-713, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24677035

ABSTRACT

Mammalian cells can be reprogrammed into induced pluripotent stem cells (iPSCs), a valuable tool for in vitro disease modeling and regenerative medicine. These applications demand for iPSCs devoid of reprogramming factor transgenes, but current procedures for the derivation of transgene-free iPSCs are inefficient and cumbersome. Here, we describe a new approach for the simple derivation of transgene-free iPSCs by the sequential use of two DNA recombinases, C31 Integrase and Cre, to control the genomic insertion and excision of a single, non-viral reprogramming vector. We show that such transgene-free iPSCs exhibit gene expression profiles and pluripotent developmental potential comparable to genuine, blastocyst-derived embryonic stem cells. As shown by a reporter iPSC line for the differentiation into midbrain dopaminergic neurons, the dual recombinase approach offers a simple and efficient way to derive transgene-free iPSCs for studying disease mechanisms and cell replacement therapies.


Subject(s)
Cellular Reprogramming , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Animals , Biotechnology , Cell Differentiation , Cells, Cultured , Cellular Reprogramming/genetics , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Female , Genes, Reporter , Genetic Vectors , Green Fluorescent Proteins/genetics , Induced Pluripotent Stem Cells/transplantation , Integrases/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Transcriptome
17.
J Clin Invest ; 123(1): 469-78, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23257354

ABSTRACT

The scaffold protein p62 (sequestosome 1; SQSTM1) is an emerging key molecular link among the metabolic, immune, and proliferative processes of the cell. Here, we report that adipocyte-specific, but not CNS-, liver-, muscle-, or myeloid-specific p62-deficient mice are obese and exhibit a decreased metabolic rate caused by impaired nonshivering thermogenesis. Our results show that p62 regulates energy metabolism via control of mitochondrial function in brown adipose tissue (BAT). Accordingly, adipocyte-specific p62 deficiency led to impaired mitochondrial function, causing BAT to become unresponsive to ß-adrenergic stimuli. Ablation of p62 leads to decreased activation of p38 targets, affecting signaling molecules that control mitochondrial function, such as ATF2, CREB, PGC1α, DIO2, NRF1, CYTC, COX2, ATP5ß, and UCP1. p62 ablation in HIB1B and BAT primary cells demonstrated that p62 controls thermogenesis in a cell-autonomous manner, independently of brown adipocyte development or differentiation. Together, our data identify p62 as a novel regulator of mitochondrial function and brown fat thermogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adipocytes, Brown/metabolism , Adipose Tissue, Brown/metabolism , Heat-Shock Proteins/metabolism , Mitochondria/metabolism , Thermogenesis/physiology , Adaptor Proteins, Signal Transducing/genetics , Adipocytes, Brown/cytology , Adipose Tissue, Brown/cytology , Animals , Cells, Cultured , Heat-Shock Proteins/genetics , MAP Kinase Signaling System/physiology , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Organ Specificity/genetics , Sequestosome-1 Protein , Transcription Factors/genetics , Transcription Factors/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
18.
Ann N Y Acad Sci ; 1266: 18-27, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22901252

ABSTRACT

Functional heterogeneity within stem and progenitor cells has been shown to influence cell fate decisions. Similarly, intracellular signaling activated by external stimuli is highly heterogeneous and its spatiotemporal activity is linked to future cell behavior. To quantify these heterogeneous states and link them to future cell fates, it is important to observe cell populations continuously with single cell resolution. Live cell imaging in combination with fluorescent biosensors for signaling activity serves as a powerful tool to study cellular and molecular heterogeneity and the long-term biological effects of signaling. Here, we describe these methodologies, their advantages over classical approaches, and we illustrate how they could be applied to improve our understanding of the importance of heterogeneous cellular and molecular responses to external signaling cues.


Subject(s)
Molecular Imaging/methods , Stem Cells/cytology , Stem Cells/metabolism , Animals , Biosensing Techniques , Cell Lineage , Cytokines/metabolism , Fluorescence Resonance Energy Transfer , Fluorescent Dyes , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Macrophage Colony-Stimulating Factor/metabolism , Mice , Models, Biological , Signal Transduction
19.
Pharmacogenet Genomics ; 19(10): 790-9, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19741568

ABSTRACT

OBJECTIVE: As a key player in modulating both human physiological and behavioural functions including anxiety, perception and in particular appetite, serotonin (5-hydroxytryptamine, 5-HT) is likely to be involved in the aetiology of eating disorders. Studies showing serotonin receptor type 3 (5-HT3) receptors to mediate food intake depression (anorexic response) have triggered our interest in investigating the putative role of variants in the 5-HT3 receptor genes, HTR3A and HTR3B, in the susceptibility to anorexia nervosa (AN) and bulimia nervosa (BN). METHODS: Two hundred and sixty-five patients with AN and 91 patients with BN as well as 191 healthy controls served as a pilot study group for mutational analysis by direct sequencing. Variants showing a significant association were subsequently genotyped in an independent Spanish cohort of 78 patients with AN and 119 patients with BN as well as 331 healthy controls for replication purposes. RESULTS: In the pilot study, we found the coding HTR3B variant, p.Y129S, (rs1176744, P = 0.004, odds ratio = 2.06) to be associated with the restrictive subtype of AN. The association was confirmed in the Spanish study group (P = 0.034, odds ratio = 2.26). CONCLUSION: Our study provides first evidence for an involvement of 5-HT3 variants in the aetiopathology of eating disorders in humans.


Subject(s)
Feeding and Eating Disorders/genetics , Genetic Variation , Receptors, Serotonin, 5-HT3/genetics , Adolescent , Female , Humans , Male , Models, Biological , Pilot Projects , Receptors, Serotonin/genetics , Receptors, Serotonin, 5-HT3/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL