Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Proc Natl Acad Sci U S A ; 121(25): e2312499121, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38857395

ABSTRACT

Ex vivo expansion of human CD34+ hematopoietic stem and progenitor cells remains a challenge due to rapid differentiation after detachment from the bone marrow niche. In this study, we assessed the capacity of an inducible fusion protein to enable sustained ex vivo proliferation of hematopoietic precursors and their capacity to differentiate into functional phagocytes. We fused the coding sequences of an FK506-Binding Protein 12 (FKBP12)-derived destabilization domain (DD) to the myeloid/lymphoid lineage leukemia/eleven nineteen leukemia (MLL-ENL) fusion gene to generate the fusion protein DD-MLL-ENL and retrovirally expressed the protein switch in human CD34+ progenitors. Using Shield1, a chemical inhibitor of DD fusion protein degradation, we established large-scale and long-term expansion of late monocytic precursors. Upon Shield1 removal, the cells lost self-renewal capacity and spontaneously differentiated, even after 2.5 y of continuous ex vivo expansion. In the absence of Shield1, stimulation with IFN-γ, LPS, and GM-CSF triggered terminal differentiation. Gene expression analysis of the obtained phagocytes revealed marked similarity with naïve monocytes. In functional assays, the novel phagocytes migrated toward CCL2, attached to VCAM-1 under shear stress, produced reactive oxygen species, and engulfed bacterial particles, cellular particles, and apoptotic cells. Finally, we demonstrated Fcγ receptor recognition and phagocytosis of opsonized lymphoma cells in an antibody-dependent manner. Overall, we have established an engineered protein that, as a single factor, is useful for large-scale ex vivo production of human phagocytes. Such adjustable proteins have the potential to be applied as molecular tools to produce functional immune cells for experimental cell-based approaches.


Subject(s)
Cell Differentiation , Phagocytes , Humans , Phagocytes/metabolism , Hematopoietic Stem Cells/metabolism , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Myeloid-Lymphoid Leukemia Protein/genetics , Leukemia/genetics , Leukemia/pathology , Leukemia/metabolism , Protein Engineering/methods , Phagocytosis
3.
Angew Chem Int Ed Engl ; 63(21): e202401004, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38497898

ABSTRACT

The CRISPR/Cas9 system has emerged as a promising platform for gene editing; however, the lack of an efficient and safe delivery system to introduce it into cells continues to hinder clinical translation. Here, we report a rationally designed gene-editing nanoparticle (NP) formulation for brain applications: an sgRNA:Cas9 ribonucleoprotein complex is immobilized on the NP surface by oligonucleotides that are complementary to the sgRNA. Irradiation of the formulation with a near-infrared (NIR) laser generates heat in the NP, leading to the release of the ribonucleoprotein complex. The gene-editing potential of the formulation was demonstrated in vitro at the single-cell level. The safety and gene editing of the formulation were also demonstrated in the brains of reporter mice, specifically in the subventricular zone after intracerebral administration and in the olfactory bulb after intranasal administration. The formulation presented here offers a new strategy for the spatially controlled delivery of the CRISPR system to the brain.


Subject(s)
Brain , CRISPR-Cas Systems , Gene Editing , Infrared Rays , Gene Editing/methods , CRISPR-Cas Systems/genetics , Animals , Brain/metabolism , Mice , Ribonucleoproteins/metabolism , Ribonucleoproteins/chemistry , Ribonucleoproteins/genetics , Nanoparticles/chemistry , Humans
4.
Genes Dev ; 37(17-18): 779-780, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37821108

ABSTRACT

Imprinted gene clusters are confined genomic regions containing genes with parent-of-origin-dependent transcriptional activity. In this issue of Genes & Development, Loftus and colleagues (pp. 829-843) made use of an insightful combination of descriptive approaches, genetic manipulations, and epigenome-editing approaches to show that differences in nuclear topology precede the onset of imprinted expression at the Peg13-Kcnk9 locus. Furthermore, the investigators provide data in line with a model suggesting that parent-of-origin-specific topological differences could be responsible for parent-of-origin-specific enhancer activity and thus imprinted expression.


Subject(s)
DNA Methylation , Genomic Imprinting
5.
Mol Syst Biol ; 18(9): e11129, 2022 09.
Article in English | MEDLINE | ID: mdl-36106915

ABSTRACT

Despite the therapeutic promise of direct reprogramming, basic principles concerning fate erasure and the mechanisms to resolve cell identity conflicts remain unclear. To tackle these fundamental questions, we established a single-cell protocol for the simultaneous analysis of multiple cell fate conversion events based on combinatorial and traceable reprogramming factor expression: Collide-seq. Collide-seq revealed the lack of a common mechanism through which fibroblast-specific gene expression loss is initiated. Moreover, we found that the transcriptome of converting cells abruptly changes when a critical level of each reprogramming factor is attained, with higher or lower levels not contributing to major changes. By simultaneously inducing multiple competing reprogramming factors, we also found a deterministic system, in which titration of fates against each other yields dominant or colliding fates. By investigating one collision in detail, we show that reprogramming factors can disturb cell identity programs independent of their ability to bind their target genes. Taken together, Collide-seq has shed light on several fundamental principles of fate conversion that may aid in improving current reprogramming paradigms.


Subject(s)
Cellular Reprogramming , Fibroblasts , Cell Differentiation/genetics , Cellular Reprogramming/genetics , Fibroblasts/metabolism , Transcriptome/genetics
6.
Cells ; 11(3)2022 02 02.
Article in English | MEDLINE | ID: mdl-35159329

ABSTRACT

The oligodendrocyte progenitors (OPCs) are at the front of the glial reaction to the traumatic brain injury. However, regulatory pathways steering the OPC reaction as well as the role of reactive OPCs remain largely unknown. Here, we compared a long-lasting, exacerbated reaction of OPCs to the adult zebrafish brain injury with a timely restricted OPC activation to identify the specific molecular mechanisms regulating OPC reactivity and their contribution to regeneration. We demonstrated that the influx of the cerebrospinal fluid into the brain parenchyma after injury simultaneously activates the toll-like receptor 2 (Tlr2) and the chemokine receptor 3 (Cxcr3) innate immunity pathways, leading to increased OPC proliferation and thereby exacerbated glial reactivity. These pathways were critical for long-lasting OPC accumulation even after the ablation of microglia and infiltrating monocytes. Importantly, interference with the Tlr1/2 and Cxcr3 pathways after injury alleviated reactive gliosis, increased new neuron recruitment, and improved tissue restoration.


Subject(s)
Oligodendrocyte Precursor Cells , Animals , Brain , Gliosis/metabolism , Immunity, Innate , Oligodendrocyte Precursor Cells/metabolism , Zebrafish
10.
Physiol Rev ; 101(1): 177-211, 2021 01 01.
Article in English | MEDLINE | ID: mdl-32525760

ABSTRACT

Given the large amount of genome-wide data that have been collected during the last decades, a good understanding of how and why cells change during development, homeostasis, and disease might be expected. Unfortunately, the opposite is true; triggers that cause cellular state changes remain elusive, and the underlying molecular mechanisms are poorly understood. Although genes with the potential to influence cell states are known, the historic dependency on methods that manipulate gene expression outside the endogenous chromatin context has prevented us from understanding how cells organize, interpret, and protect cellular programs. Fortunately, recent methodological innovations are now providing options to answer these outstanding questions, by allowing to target and manipulate individual genomic and epigenomic loci. In particular, three experimental approaches are now feasible due to DNA targeting tools, namely, activation and/or repression of master transcription factors in their endogenous chromatin context; targeting transcription factors to endogenous, alternative, or inaccessible sites; and finally, functional manipulation of the chromatin context. In this article, we discuss the molecular basis of DNA targeting tools and review the potential of these new technologies before we summarize how these have already been used for the manipulation of cellular states and hypothesize about future applications.


Subject(s)
CRISPR-Cas Systems , Cell Physiological Phenomena/physiology , Epigenesis, Genetic , Gene Editing , Genetic Engineering/methods , Physiology/methods , Animals , Epigenomics , Humans , Transcription, Genetic
11.
Z Gerontol Geriatr ; 54(1): 28-36, 2021 Feb.
Article in German | MEDLINE | ID: mdl-33231762

ABSTRACT

The rehabilitation of people after suffering a stroke is a long-term process, in which patients, their families and friends are reliant on social support and assistance. Therefore, the individual promotion of available resources and autonomy of those affected represents an important task. The College for Health in Bochum and the German Stroke Help Foundation have developed the volunteer-supported model "Trained stroke helpers-a partnership-based model for local care (GeSa)" that educates stroke helpers as experts for individual local support of stroke patients and their relatives. The stroke helpers offer a patient-oriented care and individual assistance in the daily routine and therefore make a contribution to improvement of the health-related quality of life of those affected and their relatives. A training curriculum was developed and successfully tested and evaluated in a course with 21 stroke helpers. Up to summer 2020 a total of 480 stroke helpers have been trained at 16 locations in 37 courses. The experiences with the project have so far been consistently positive, which was confirmed by two scientific studies. The inclusion of cooperation partners for the coordination of the volunteers, which was already implemented in the model project, has been shown to be the most important success factor for the sustainability of the project. This article presents the results of the pilot study and the standardized process on implementation of a regional stroke helper project and describes the experiences with the previous course of the project.


Subject(s)
Aftercare , Stroke , Humans , Outpatients , Pilot Projects , Quality of Life , Stroke/diagnosis , Stroke/therapy
12.
Neurobiol Dis ; 148: 105174, 2021 01.
Article in English | MEDLINE | ID: mdl-33171228

ABSTRACT

The vulnerability of the mammalian brain is mainly due to its limited ability to generate new neurons once fully matured. Direct conversion of non-neuronal cells to neurons opens up a new avenue for therapeutic intervention and has made great strides also for in vivo applications in the injured brain. These great achievements raise the issue of adequate identity and chromatin hallmarks of the induced neurons. This may be particularly important, as aberrant epigenetic settings may reveal their adverse effects only in certain brain activity states. Therefore, we review here the knowledge about epigenetic memory and partially resetting of chromatin hallmarks from other reprogramming fields, before moving to the knowledge in direct neuronal reprogramming, which is still limited. Most importantly, novel tools are available now to manipulate specific epigenetic marks at specific sites of the genome. Applying these will eventually allow erasing aberrant epigenetic memory and paving the way towards new therapeutic approaches for brain repair.


Subject(s)
Cell Differentiation/genetics , Cellular Reprogramming Techniques , Cellular Reprogramming , Epigenesis, Genetic/genetics , Gene Expression Regulation , Neurons/metabolism , Animals , Cell Lineage , Chromatin , Humans , Neurogenesis/genetics
13.
Cell Stem Cell ; 28(3): 524-534.e7, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33202244

ABSTRACT

Astrocyte-to-neuron conversion is a promising avenue for neuronal replacement therapy. Neurons are particularly dependent on mitochondrial function, but how well mitochondria adapt to the new fate is unknown. Here, we determined the comprehensive mitochondrial proteome of cortical astrocytes and neurons, identifying about 150 significantly enriched mitochondrial proteins for each cell type, including transporters, metabolic enzymes, and cell-type-specific antioxidants. Monitoring their transition during reprogramming revealed late and only partial adaptation to the neuronal identity. Early dCas9-mediated activation of genes encoding mitochondrial proteins significantly improved conversion efficiency, particularly for neuron-enriched but not astrocyte-enriched antioxidant proteins. For example, Sod1 not only improves the survival of the converted neurons but also elicits a faster conversion pace, indicating that mitochondrial proteins act as enablers and drivers in this process. Transcriptional engineering of mitochondrial proteins with other functions improved reprogramming as well, demonstrating a broader role of mitochondrial proteins during fate conversion.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Mitochondrial Proteins , Astrocytes , Cells, Cultured , Mitochondrial Proteins/genetics , Neuroglia , Neurons
15.
EMBO J ; 38(17): e100481, 2019 09 02.
Article in English | MEDLINE | ID: mdl-31304985

ABSTRACT

Regulation of adult neural stem cell (NSC) number is critical for lifelong neurogenesis. Here, we identified a post-transcriptional control mechanism, centered around the microRNA 204 (miR-204), to control the maintenance of quiescent (q)NSCs. miR-204 regulates a spectrum of transcripts involved in cell cycle regulation, neuronal migration, and differentiation in qNSCs. Importantly, inhibition of miR-204 function reduced the number of qNSCs in the subependymal zone (SEZ) by inducing pre-mature activation and differentiation of NSCs without changing their neurogenic potential. Strikingly, we identified the choroid plexus of the mouse lateral ventricle as the major source of miR-204 that is released into the cerebrospinal fluid to control number of NSCs within the SEZ. Taken together, our results describe a novel mechanism to maintain adult somatic stem cells by a niche-specific miRNA repressing activation and differentiation of stem cells.


Subject(s)
Choroid Plexus/chemistry , MicroRNAs/genetics , Neural Stem Cells/cytology , Adult , Animals , Cell Cycle , Cell Differentiation , Cell Movement , Female , Gene Expression Regulation , Humans , Male , Mice , MicroRNAs/cerebrospinal fluid , Middle Aged , Neural Stem Cells/chemistry , Stem Cell Niche
16.
Nat Commun ; 10(1): 2119, 2019 05 09.
Article in English | MEDLINE | ID: mdl-31073172

ABSTRACT

Master transcription factors have the ability to direct and reverse cellular identities, and consequently their genes must be subject to particular transcriptional control. However, it is unclear which molecular processes are responsible for impeding their activation and safeguarding cellular identities. Here we show that the targeting of dCas9-VP64 to the promoter of the master transcription factor Sox1 results in strong transcript and protein up-regulation in neural progenitor cells (NPCs). This gene activation restores lost neuronal differentiation potential, which substantiates the role of Sox1 as a master transcription factor. However, despite efficient transactivator binding, major proportions of progenitor cells are unresponsive to the transactivating stimulus. By combining the transactivation domain with epigenome editing we find that among a series of euchromatic processes, the removal of DNA methylation (by dCas9-Tet1) has the highest potential to increase the proportion of cells activating foreign master transcription factors and thus breaking down cell identity barriers.


Subject(s)
Cell Differentiation/genetics , Cellular Reprogramming/genetics , Epigenesis, Genetic , Neural Stem Cells/physiology , SOXB1 Transcription Factors/metabolism , Animals , CRISPR-Cas Systems/genetics , Cell Line , DNA Methylation/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Editing/methods , Gene Expression Regulation , Mice , Neuroglia/cytology , Neuroglia/physiology , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism , SOXB1 Transcription Factors/genetics , Transcription, Genetic/genetics
17.
Trends Cancer ; 5(1): 5-7, 2019 01.
Article in English | MEDLINE | ID: mdl-30616756

ABSTRACT

Diabetes has long been associated with an increased risk of cancer. While many molecular connections likely exist between the diseases, a recent publication discovered a clear molecular link, demonstrating that a glucose-dependent destabilisation of the DNA demethylase TET2 can promote malignant transformation via an AMPK-dependent phosphoswitch.


Subject(s)
DNA-Binding Proteins/metabolism , Diabetes Mellitus/etiology , Diabetes Mellitus/metabolism , Disease Susceptibility , Glucose/metabolism , Neoplasms/etiology , Neoplasms/metabolism , Proto-Oncogene Proteins/metabolism , Diabetes Mellitus/therapy , Dioxygenases , Energy Metabolism , Humans , Neoplasms/therapy
18.
Cell Rep ; 25(12): 3241-3251.e5, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30566853

ABSTRACT

Zebrafish have a high capacity to replace lost neurons after brain injury. New neurons involved in repair are generated by a specific set of glial cells, known as ependymoglial cells. We analyze changes in the transcriptome of ependymoglial cells and their progeny after injury to infer the molecular pathways governing restorative neurogenesis. We identify the aryl hydrocarbon receptor (AhR) as a regulator of ependymoglia differentiation toward post-mitotic neurons. In vivo imaging shows that high AhR signaling promotes the direct conversion of a specific subset of ependymoglia into post-mitotic neurons, while low AhR signaling promotes ependymoglial proliferation. Interestingly, we observe the inactivation of AhR signaling shortly after injury followed by a return to the basal levels 7 days post injury. Interference with timely AhR regulation after injury leads to aberrant restorative neurogenesis. Taken together, we identify AhR signaling as a crucial regulator of restorative neurogenesis timing in the zebrafish brain.


Subject(s)
Neurogenesis , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction , Animals , Cell Differentiation , Cell Proliferation , Cell Survival , Ependymoglial Cells/cytology , Ependymoglial Cells/metabolism , Mitosis , Neurons/cytology , Time Factors , Zebrafish
19.
Genome Res ; 28(12): 1779-1790, 2018 12.
Article in English | MEDLINE | ID: mdl-30355600

ABSTRACT

Mosaic mutations present in the germline have important implications for reproductive risk and disease transmission. We previously demonstrated a phenomenon occurring in the male germline, whereby specific mutations arising spontaneously in stem cells (spermatogonia) lead to clonal expansion, resulting in elevated mutation levels in sperm over time. This process, termed "selfish spermatogonial selection," explains the high spontaneous birth prevalence and strong paternal age-effect of disorders such as achondroplasia and Apert, Noonan and Costello syndromes, with direct experimental evidence currently available for specific positions of six genes (FGFR2, FGFR3, RET, PTPN11, HRAS, and KRAS). We present a discovery screen to identify novel mutations and genes showing evidence of positive selection in the male germline, by performing massively parallel simplex PCR using RainDance technology to interrogate mutational hotspots in 67 genes (51.5 kb in total) in 276 biopsies of testes from five men (median age, 83 yr). Following ultradeep sequencing (about 16,000×), development of a low-frequency variant prioritization strategy, and targeted validation, we identified 61 distinct variants present at frequencies as low as 0.06%, including 54 variants not previously directly associated with selfish selection. The majority (80%) of variants identified have previously been implicated in developmental disorders and/or oncogenesis and include mutations in six newly associated genes (BRAF, CBL, MAP2K1, MAP2K2, RAF1, and SOS1), all of which encode components of the RAS-MAPK pathway and activate signaling. Our findings extend the link between mutations dysregulating the RAS-MAPK pathway and selfish selection, and show that the aging male germline is a repository for such deleterious mutations.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Mutation , Signal Transduction , Testis/metabolism , ras Proteins/metabolism , Aged , Aged, 80 and over , Genetic Variation , Humans , Male , Middle Aged
20.
PLoS One ; 13(4): e0196015, 2018.
Article in English | MEDLINE | ID: mdl-29702666

ABSTRACT

Novel applications based on the bacterial CRISPR system make genetic, genomic, transcriptional and epigenomic engineering widely accessible for the first time. A significant advantage of CRISPR over previous methods is its tremendous adaptability due to its bipartite nature. Cas9 or its engineered variants define the molecular effect, while short gRNAs determine the targeting sites. A majority of CRISPR approaches depend on the simultaneous delivery of multiple gRNAs into single cells, either as an essential precondition, to increase responsive cell populations or to enhance phenotypic outcomes. Despite these requirements, methods allowing the efficient generation and delivery of multiple gRNA expression units into single cells are still sparse. Here we present STAgR (String assembly gRNA cloning), a single step gRNA multiplexing system, that obtains its advantages by employing the N20 targeting sequences as necessary homologies for Gibson assembly. We show that STAgR allows reliable and cost-effective generation of vectors with high numbers of gRNAs enabling multiplexed CRISPR approaches. Moreover, STAgR is easily customizable, as vector backbones as well as gRNA structures, numbers and promoters can be freely chosen and combined. Finally, we demonstrate STAgR's widespread functionality, its efficiency in multi-targeting approaches, using it for both, genome and transcriptome editing, as well as applying it in vitro and in vivo.


Subject(s)
Genetic Engineering/methods , RNA, Guide, Kinetoplastida/genetics , CRISPR-Cas Systems , Gene Editing , HeLa Cells , Humans , Promoter Regions, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...