Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
EMBO J ; 41(1): e110330, 2022 01 04.
Article in English | MEDLINE | ID: mdl-34981519

ABSTRACT

Looking back at the journal's first issue in January 1982 provides an opportunity to reflect on its historical development and to introduce upcoming initiatives.

2.
EMBO J ; 40(17): e109115, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34287984

ABSTRACT

The EMBO Journal highlights the multifaceted aspects of tumour biology in a series of complementary review articles published over the course of 2021.


Subject(s)
Neoplasms/metabolism , Animals , Humans , Neoplasms/genetics , Neoplasms/pathology , Tumor Microenvironment
3.
Blood ; 137(23): 3218-3224, 2021 06 10.
Article in English | MEDLINE | ID: mdl-33754628

ABSTRACT

Hematopoietic stem cells (HSCs) and distinct multipotent progenitor (MPP) populations (MPP1-4) contained within the Lin-Sca-1+c-Kit+ (LSK) compartment have previously been identified using diverse surface-marker panels. Here, we phenotypically define and functionally characterize MPP5 (LSK CD34+CD135-CD48-CD150-). Upon transplantation, MPP5 supports initial emergency myelopoiesis followed by stable contribution to the lymphoid lineage. MPP5, capable of generating MPP1-4 but not HSCs, represents a dynamic and versatile component of the MPP network. To characterize all hematopoietic stem and progenitor cells, we performed RNA-sequencing (RNA-seq) analysis to identify specific transcriptomic landscapes of HSCs and MPP1-5. This was complemented by single-cell RNA-seq analysis of LSK cells to establish the differentiation trajectories from HSCs to MPP1-5. In agreement with functional reconstitution activity, MPP5 is located immediately downstream of HSCs but upstream of the more committed MPP2-4. This study provides a comprehensive analysis of the LSK compartment, focusing on the functional and molecular characteristics of the newly defined MPP5 subset.


Subject(s)
Antigens, CD/metabolism , Hematopoietic Stem Cells/metabolism , Multipotent Stem Cells/metabolism , Animals , Mice
4.
Nat Commun ; 12(1): 608, 2021 01 27.
Article in English | MEDLINE | ID: mdl-33504783

ABSTRACT

Haematopoietic stem cells (HSCs) are characterized by their self-renewal potential associated to dormancy. Here we identify the cell surface receptor neogenin-1 as specifically expressed in dormant HSCs. Loss of neogenin-1 initially leads to increased HSC expansion but subsequently to loss of self-renewal and premature exhaustion in vivo. Its ligand netrin-1 induces Egr1 expression and maintains quiescence and function of cultured HSCs in a Neo1 dependent manner. Produced by arteriolar endothelial and periarteriolar stromal cells, conditional netrin-1 deletion in the bone marrow niche reduces HSC numbers, quiescence and self-renewal, while overexpression increases quiescence in vivo. Ageing associated bone marrow remodelling leads to the decline of netrin-1 expression in niches and a compensatory but reversible upregulation of neogenin-1 on HSCs. Our study suggests that niche produced netrin-1 preserves HSC quiescence and self-renewal via neogenin-1 function. Decline of netrin-1 production during ageing leads to the gradual decrease of Neo1 mediated HSC self-renewal.


Subject(s)
Hematopoietic Stem Cells/metabolism , Membrane Proteins/metabolism , Netrin-1/metabolism , Stem Cell Niche , Animals , Arterioles/metabolism , Cell Differentiation , Cell Proliferation , Cellular Senescence , Gene Deletion , Hematopoietic Stem Cell Transplantation , Mice, Mutant Strains , Mice, Transgenic , Signal Transduction
5.
Blood ; 136(13): 1507-1519, 2020 09 24.
Article in English | MEDLINE | ID: mdl-32556243

ABSTRACT

Acute myeloid leukemia is characterized by the accumulation of clonal myeloid blast cells unable to differentiate into mature leukocytes. Chemotherapy induces remission in the majority of patients, but relapse rates are high and lead to poor clinical outcomes. Because this is primarily caused by chemotherapy-resistant leukemic stem cells (LSCs), it is essential to eradicate LSCs to improve patient survival. LSCs have predominantly been studied at the transcript level, thus information about posttranscriptionally regulated genes and associated networks is lacking. Here, we extend our previous report on LSC proteomes to healthy age-matched hematopoietic stem and progenitor cells (HSPCs) and correlate the proteomes to the corresponding transcriptomes. By comparing LSCs to leukemic blasts and healthy HSPCs, we validate candidate LSC markers and highlight novel and potentially targetable proteins that are absent or only lowly expressed in HSPCs. In addition, our data provide strong evidence that LSCs harbor a characteristic energy metabolism, adhesion molecule composition, as well as RNA-processing properties. Furthermore, correlating proteome and transcript data of the same individual samples highlights the strength of proteome analyses, which are particularly potent in detecting alterations in metabolic pathways. In summary, our study provides a comprehensive proteomic and transcriptomic characterization of functionally validated LSCs, blasts, and healthy HSPCs, representing a valuable resource helping to design LSC-directed therapies.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , Neoplastic Stem Cells/metabolism , Animals , Energy Metabolism , Gene Expression Regulation, Leukemic , Humans , Leukemia, Myeloid, Acute/genetics , Mice , Proteome/genetics , Proteome/metabolism , Proteomics , Transcriptome
6.
Cell ; 169(5): 807-823.e19, 2017 May 18.
Article in English | MEDLINE | ID: mdl-28479188

ABSTRACT

Dormant hematopoietic stem cells (dHSCs) are atop the hematopoietic hierarchy. The molecular identity of dHSCs and the mechanisms regulating their maintenance or exit from dormancy remain uncertain. Here, we use single-cell RNA sequencing (RNA-seq) analysis to show that the transition from dormancy toward cell-cycle entry is a continuous developmental path associated with upregulation of biosynthetic processes rather than a stepwise progression. In addition, low Myc levels and high expression of a retinoic acid program are characteristic for dHSCs. To follow the behavior of dHSCs in situ, a Gprc5c-controlled reporter mouse was established. Treatment with all-trans retinoic acid antagonizes stress-induced activation of dHSCs by restricting protein translation and levels of reactive oxygen species (ROS) and Myc. Mice maintained on a vitamin A-free diet lose HSCs and show a disrupted re-entry into dormancy after exposure to inflammatory stress stimuli. Our results highlight the impact of dietary vitamin A on the regulation of cell-cycle-mediated stem cell plasticity. VIDEO ABSTRACT.


Subject(s)
Hematopoietic Stem Cells/cytology , Signal Transduction , Tretinoin/pharmacology , Vitamin A/administration & dosage , Animals , Biosynthetic Pathways , Cell Culture Techniques , Cell Cycle/drug effects , Cell Survival , Diet , Gene Expression Profiling , Hematopoietic Stem Cells/drug effects , Mice , Poly I-C/pharmacology , Reactive Oxygen Species/metabolism , Receptors, G-Protein-Coupled/metabolism , Single-Cell Analysis , Stress, Physiological , Vitamin A/pharmacology , Vitamins/administration & dosage , Vitamins/pharmacology
7.
Cell Stem Cell ; 17(4): 422-34, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26299573

ABSTRACT

Infections are associated with extensive platelet consumption, representing a high risk for health. However, the mechanism coordinating the rapid regeneration of the platelet pool during such stress conditions remains unclear. Here, we report that the phenotypic hematopoietic stem cell (HSC) compartment contains stem-like megakaryocyte-committed progenitors (SL-MkPs), a cell population that shares many features with multipotent HSCs and serves as a lineage-restricted emergency pool for inflammatory insults. During homeostasis, SL-MkPs are maintained in a primed but quiescent state, thus contributing little to steady-state megakaryopoiesis. Even though lineage-specific megakaryocyte transcripts are expressed, protein synthesis is suppressed. In response to acute inflammation, SL-MkPs become activated, resulting in megakaryocyte protein production from pre-existing transcripts and a maturation of SL-MkPs and other megakaryocyte progenitors. This results in an efficient replenishment of platelets that are lost during inflammatory insult. Thus, our study reveals an emergency machinery that counteracts life-threatening platelet depletions during acute inflammation.


Subject(s)
Blood Platelets/pathology , Inflammation/pathology , Megakaryocyte Progenitor Cells/pathology , Thrombopoiesis , Animals , Blood Platelets/physiology , Cell Lineage , Cell Proliferation , Hematopoietic Stem Cells/pathology , Hematopoietic Stem Cells/physiology , Megakaryocyte Progenitor Cells/physiology , Mice
8.
Cell Cycle ; 13(22): 3476-87, 2014.
Article in English | MEDLINE | ID: mdl-25483069

ABSTRACT

Epigenetic alterations during cellular differentiation are a key molecular mechanism which both instructs and reinforces the process of lineage commitment. Within the haematopoietic system, progressive changes in the DNA methylome of haematopoietic stem cells (HSCs) are essential for the effective production of mature blood cells. Inhibition or loss of function of the cellular DNA methylation machinery has been shown to lead to a severe perturbation in blood production and is also an important driver of malignant transformation. HSCs constitute a very rare cell population in the bone marrow, capable of life-long self-renewal and multi-lineage differentiation. The low abundance of HSCs has been a major technological barrier to the global analysis of the CpG methylation status within both HSCs and their immediate progeny, the multipotent progenitors (MPPs). Within this Extra View article, we review the current understanding of how the DNA methylome regulates normal and malignant hematopoiesis. We also discuss the current methodologies that are available for interrogating the DNA methylation status of HSCs and MPPs and describe a new data set that was generated using tagmentation-based whole genome bisulfite sequencing (TWGBS) in order to comprehensively map methylated cytosines using the limited amount of genomic DNA that can be harvested from rare cell populations. Extended analysis of this data set clearly demonstrates the added value of genome-wide sequencing of methylated cytosines and identifies novel important cis-acting regulatory regions that are dynamically remodeled during the first steps of haematopoietic differentiation.


Subject(s)
Cell Differentiation/genetics , DNA Methylation/genetics , Hematopoietic Stem Cells , Regulatory Sequences, Nucleic Acid/genetics , Epigenesis, Genetic , Genome , High-Throughput Nucleotide Sequencing , Humans
9.
Stem Cell Reports ; 3(5): 858-75, 2014 Nov 11.
Article in English | MEDLINE | ID: mdl-25418729

ABSTRACT

Hematopoietic stem cells possess lifelong self-renewal activity and generate multipotent progenitors that differentiate into lineage-committed and subsequently mature cells. We present a comparative transcriptome analysis of ex vivo isolated mouse multipotent hematopoietic stem/progenitor cells (Lin(neg)SCA-1(+)c-KIT(+)) and myeloid committed precursors (Lin(neg)SCA-1(neg)c-KIT(+)). Our data display dynamic transcriptional networks and identify a stem/progenitor gene expression pattern that is characterized by cell adhesion and immune response components including kallikrein-related proteases. We identify 498 expressed lncRNAs, which are potential regulators of multipotency or lineage commitment. By integrating these transcriptome with our recently reported proteome data, we found evidence for posttranscriptional regulation of processes including metabolism and response to oxidative stress. Finally, our study identifies a high number of genes with transcript isoform regulation upon lineage commitment. This in-depth molecular analysis outlines the enormous complexity of expressed coding and noncoding RNAs and posttranscriptional regulation during the early differentiation steps of hematopoietic stem cells toward the myeloid lineage.


Subject(s)
Cell Differentiation/genetics , Gene Expression Profiling , Hematopoietic Stem Cells/metabolism , Multipotent Stem Cells/metabolism , Myeloid Cells/metabolism , Animals , Cell Adhesion/genetics , Cell Cycle/genetics , Cell Lineage/genetics , Energy Metabolism/genetics , Female , Flow Cytometry , Gene Ontology , Gene Regulatory Networks , Hematopoietic Stem Cells/cytology , Immunity/genetics , Mice, Inbred C57BL , Multipotent Stem Cells/cytology , Myeloid Cells/cytology , Proteome/genetics , Proteome/metabolism , RNA, Long Noncoding/genetics , Reverse Transcriptase Polymerase Chain Reaction
10.
Cell Stem Cell ; 15(4): 507-522, 2014 Oct 02.
Article in English | MEDLINE | ID: mdl-25158935

ABSTRACT

In this study, we present integrated quantitative proteome, transcriptome, and methylome analyses of hematopoietic stem cells (HSCs) and four multipotent progenitor (MPP) populations. From the characterization of more than 6,000 proteins, 27,000 transcripts, and 15,000 differentially methylated regions (DMRs), we identified coordinated changes associated with early differentiation steps. DMRs show continuous gain or loss of methylation during differentiation, and the overall change in DNA methylation correlates inversely with gene expression at key loci. Our data reveal the differential expression landscape of 493 transcription factors and 682 lncRNAs and highlight specific expression clusters operating in HSCs. We also found an unexpectedly dynamic pattern of transcript isoform regulation, suggesting a critical regulatory role during HSC differentiation, and a cell cycle/DNA repair signature associated with multipotency in MPP2 cells. This study provides a comprehensive genome-wide resource for the functional exploration of molecular, cellular, and epigenetic regulation at the top of the hematopoietic hierarchy.


Subject(s)
DNA Methylation/genetics , Gene Regulatory Networks , Hematopoietic Stem Cells/metabolism , Proteome/metabolism , Transcriptome/genetics , Adult , Cell Differentiation/genetics , Cell Lineage/genetics , Cluster Analysis , Epigenesis, Genetic , Gene Expression Profiling , Genome, Human/genetics , Genomic Imprinting , Hematopoietic Stem Cells/cytology , Humans , Molecular Sequence Data , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Long Noncoding/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
11.
Mol Cell Proteomics ; 11(8): 286-302, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22454540

ABSTRACT

Regenerative tissues such as the skin epidermis, the intestinal mucosa or the hematopoietic system are organized in a hierarchical manner with stem cells building the top of this hierarchy. Somatic stem cells harbor the highest self-renewal activity and generate a series of multipotent progenitors which differentiate into lineage committed progenitors and subsequently mature cells. In this report, we applied an in-depth quantitative proteomic approach to analyze and compare the full proteomes of ex vivo isolated and FACS-sorted populations highly enriched for either multipotent hematopoietic stem/progenitor cells (HSPCs, Lin(neg)Sca-1(+)c-Kit(+)) or myeloid committed precursors (Lin(neg)Sca-1(-)c-Kit(+)). By employing stable isotope dimethyl labeling and high-resolution mass spectrometry, more than 5000 proteins were quantified. From biological triplicate experiments subjected to rigorous statistical evaluation, 893 proteins were found differentially expressed between multipotent and myeloid committed cells. The differential protein content in these cell populations points to a distinct structural organization of the cytoskeleton including remodeling activity. In addition, we found a marked difference in the expression of metabolic enzymes, including a clear shift of specific protein isoforms of the glycolytic pathway. Proteins involved in translation showed a collective higher expression in myeloid progenitors, indicating an increased translational activity. Strikingly, the data uncover a unique signature related to immune defense mechanisms, centering on the RIG-I and type-1 interferon response systems, which are installed in multipotent progenitors but not evident in myeloid committed cells. This suggests that specific, and so far unrecognized, mechanisms protect these immature cells before they mature. In conclusion, this study indicates that the transition of hematopoietic stem/progenitors toward myeloid commitment is accompanied by a profound change in processing of cellular resources, adding novel insights into the molecular mechanisms at the interface between multipotency and lineage commitment.


Subject(s)
Cell Differentiation , Hematopoietic Stem Cells/metabolism , Multipotent Stem Cells/metabolism , Myeloid Cells/metabolism , Proteomics/methods , Animals , Female , Flow Cytometry , Glycolysis , Hematopoiesis , Immunohistochemistry , Mice , Mice, Inbred C57BL , Proteome/analysis , Proteome/classification , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
12.
J Comp Neurol ; 515(5): 585-99, 2009 Aug 10.
Article in English | MEDLINE | ID: mdl-19480000

ABSTRACT

The chemosensory neuroepithelia of the vertebrate olfactory system share a life-long ability to regenerate. Novel neurons proliferate from basal stem cells that continuously replace old or damaged sensory neurons. The sensory neurons of the mouse and rat olfactory system specifically express bestrophin 2, a member of the bestrophin family of calcium-activated chloride channels. This channel was recently proposed to operate as a transduction channel in olfactory sensory cilia. We raised a polyclonal antibody against bestrophin 2 and characterized the expression pattern of this protein in the mouse main olfactory epithelium, septal organ of Masera, and vomeronasal organ. Comparison with the maturation markers growth-associated protein 43 and olfactory marker protein revealed that bestrophin 2 was expressed in developing sensory neurons of all chemosensory neuroepithelia, but was restricted to proximal cilia in mature sensory neurons. Our results suggest that bestrophin 2 plays a critical role during differentiation and growth of axons and cilia. In mature olfactory receptor neurons, it appears to support growth and function of sensory cilia.


Subject(s)
Chloride Channels/metabolism , Eye Proteins/metabolism , Neurogenesis/physiology , Olfactory Mucosa/physiology , Vomeronasal Organ/physiology , Animals , Axons/physiology , Bestrophins , Chloride Channels/genetics , Cilia/physiology , Eye Proteins/genetics , Humans , Mice , Olfactory Bulb/cytology , Olfactory Bulb/metabolism , Olfactory Marker Protein/genetics , Olfactory Marker Protein/metabolism , Olfactory Mucosa/cytology , Olfactory Perception/physiology , Olfactory Receptor Neurons/cytology , Olfactory Receptor Neurons/metabolism , Rats , Rats, Wistar , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Vomeronasal Organ/cytology
13.
Chem Senses ; 33(2): 145-62, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18032372

ABSTRACT

The cilia of mammalian olfactory receptor neurons (ORNs) represent the sensory interface that is exposed to the air within the nasal cavity. The cilia are the site where odorants bind to specific receptors and initiate olfactory transduction that leads to excitation of the neuron. This process involves a multitude of ciliary proteins that mediate chemoelectrical transduction, amplification, and adaptation of the primary sensory signal. Many of these proteins were initially identified by their enzymatic activities using a membrane protein preparation from olfactory cilia. This so-called "calcium-shock" preparation is a versatile tool for the exploration of protein expression, enzyme kinetics, regulatory mechanisms, and ciliary development. To support such studies, we present a first proteomic analysis of this membrane preparation. We subjected the cilia preparation to liquid chromatography-electrospray ionisation (LC-ESI-MS/MS) tandem mass spectrometry and identified 268 proteins, of which 49% are membrane proteins. A detailed analysis of their cellular and subcellular localization showed that the cilia preparation obtained by calcium shock not only is highly enriched in ORN proteins but also contains a significant amount of nonciliary material. Although our proteomic study does not identify the entire set of ciliary and nonciliary proteins, it provides the first estimate of the purity of the calcium-shock preparation and provides valuable biochemical information for further research.


Subject(s)
Cilia/chemistry , Membrane Proteins/analysis , Olfactory Mucosa/chemistry , Olfactory Receptor Neurons/chemistry , Proteins/analysis , Proteomics , Animals , Chromatography, Liquid , Computational Biology , Membrane Proteins/isolation & purification , Olfactory Mucosa/cytology , Olfactory Receptor Neurons/ultrastructure , Rats , Rats, Wistar , Smell , Spectrometry, Mass, Electrospray Ionization
SELECTION OF CITATIONS
SEARCH DETAIL
...