Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 968, 2024 Feb 06.
Article in English | MEDLINE | ID: mdl-38320988

ABSTRACT

Tumor microtubes (TMs) connect glioma cells to a network with considerable relevance for tumor progression and therapy resistance. However, the determination of TM-interconnectivity in individual tumors is challenging and the impact on patient survival unresolved. Here, we establish a connectivity signature from single-cell RNA-sequenced (scRNA-Seq) xenografted primary glioblastoma (GB) cells using a dye uptake methodology, and validate it with recording of cellular calcium epochs and clinical correlations. Astrocyte-like and mesenchymal-like GB cells have the highest connectivity signature scores in scRNA-sequenced patient-derived xenografts and patient samples. In large GB cohorts, TM-network connectivity correlates with the mesenchymal subtype and dismal patient survival. CHI3L1 gene expression serves as a robust molecular marker of connectivity and functionally influences TM networks. The connectivity signature allows insights into brain tumor biology, provides a proof-of-principle that tumor cell TM-connectivity is relevant for patients' prognosis, and serves as a robust prognostic biomarker.


Subject(s)
Brain Neoplasms , Glioblastoma , Glioma , Humans , Glioblastoma/genetics , Glioma/genetics , Brain Neoplasms/genetics , Chitinase-3-Like Protein 1
2.
Nature ; 613(7942): 179-186, 2023 01.
Article in English | MEDLINE | ID: mdl-36517594

ABSTRACT

Diffuse gliomas, particularly glioblastomas, are incurable brain tumours1. They are characterized by networks of interconnected brain tumour cells that communicate via Ca2+ transients2-6. However, the networks' architecture and communication strategy and how these influence tumour biology remain unknown. Here we describe how glioblastoma cell networks include a small, plastic population of highly active glioblastoma cells that display rhythmic Ca2+ oscillations and are particularly connected to others. Their autonomous periodic Ca2+ transients preceded Ca2+ transients of other network-connected cells, activating the frequency-dependent MAPK and NF-κB pathways. Mathematical network analysis revealed that glioblastoma network topology follows scale-free and small-world properties, with periodic tumour cells frequently located in network hubs. This network design enabled resistance against random damage but was vulnerable to losing its key hubs. Targeting of autonomous rhythmic activity by selective physical ablation of periodic tumour cells or by genetic or pharmacological interference with the potassium channel KCa3.1 (also known as IK1, SK4 or KCNN4) strongly compromised global network communication. This led to a marked reduction of tumour cell viability within the entire network, reduced tumour growth in mice and extended animal survival. The dependency of glioblastoma networks on periodic Ca2+ activity generates a vulnerability7 that can be exploited for the development of novel therapies, such as with KCa3.1-inhibiting drugs.


Subject(s)
Brain Neoplasms , Glioblastoma , Animals , Mice , Brain/metabolism , Brain/pathology , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , NF-kappa B/metabolism , MAP Kinase Signaling System , Calcium Signaling , Cell Death , Survival Analysis , Calcium/metabolism
3.
Stem Cell Res ; 66: 102989, 2023 02.
Article in English | MEDLINE | ID: mdl-36473250

ABSTRACT

Brain-derived neurotrophic factor (BDNF) has been implicated in a multitude of neurodevelopmental processes including neuronal differentiation, axonal outgrowth, synaptic plasticity, or survival. One human-specific single nucleotide polymorphism (rs6265) in the BDNF gene causes a substitution of valine (Val) to methionine (Met) at codon 66 in the pro domain of the protein (Val66Met). This substitution is associated to reduced hippocampal volumes, poor performance on hippocampal-dependent memory tasks, and some mental disorders such as schizophrenia, depression or Alzheimer's disease. Here we generated three iPSC lines from healthy donors, either homozygous (Val/Val and Met/Met) or heterozygous (Val/Met) for the polymorphism.


Subject(s)
Induced Pluripotent Stem Cells , Schizophrenia , Humans , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Induced Pluripotent Stem Cells/metabolism , Polymorphism, Single Nucleotide , Schizophrenia/genetics , Hippocampus/metabolism , Methionine/genetics , Racemethionine
4.
Cell Death Dis ; 13(10): 887, 2022 10 21.
Article in English | MEDLINE | ID: mdl-36270985

ABSTRACT

In humans, most neurons are born during embryonic development and have to persist throughout the entire lifespan of an individual. Thus, human neurons have to develop elaborate survival strategies to protect against accidental cell death. We set out to decipher the developmental adaptations resulting in neuronal resilience. We demonstrate that, during the time course of maturation, human neurons install a complex and complementary anti-apoptotic signaling network. This includes i.) a downregulation of central proteins of the intrinsic apoptosis pathway including several caspases, ii.) a shift in the ratio of pro- and anti-apoptotic BCL-2 family proteins, and iii.) an elaborate regulatory network resulting in upregulation of the inhibitor of apoptosis protein (IAP) XIAP. Together, these adaptations strongly increase the threshold for apoptosis initiation when confronted with a wide range of cellular stressors. Our results highlight how human neurons are endowed with complex and redundant preemptive strategies to protect against stress and cell death.


Subject(s)
Induced Pluripotent Stem Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Caspases/metabolism , Apoptosis/physiology , Cell Death , Inhibitor of Apoptosis Proteins/metabolism , Neurons/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , X-Linked Inhibitor of Apoptosis Protein/metabolism
5.
Anal Chem ; 94(25): 8847-8856, 2022 06 28.
Article in English | MEDLINE | ID: mdl-35713335

ABSTRACT

Depression is quickly becoming one of the world's most pressing public health crises, and there is an urgent need for better diagnostics and therapeutics. Behavioral models in animals and humans have not adequately addressed the diagnosis and treatment of depression, and biomarkers of mental illnesses remain ill-defined. It has been very difficult to identify biomarkers of depression because of in vivo measurement challenges. While our group has made important strides in developing in vivo tools to measure such biomarkers (e.g., serotonin) in mice using voltammetry, these tools cannot be easily applied for depression diagnosis and drug screening in humans due to the inaccessibility of the human brain. In this work, we take a chemical approach, ex vivo, to introduce a human-derived system to investigate brain serotonin. We utilize human induced pluripotent stem cells differentiated into serotonin neurons and establish a new ex vivo model of real-time serotonin neurotransmission measurements. We show that evoked serotonin release responds to stimulation intensity and tryptophan preloading, and that serotonin release and reuptake kinetics resemble those found in vivo in rodents. Finally, after selective serotonin reuptake inhibitor (SSRI) exposure, we find dose-dependent internalization of the serotonin reuptake transporters (a signature of the in vivo response to SSRI). Our new human-derived chemical model has great potential to provide an ex vivo chemical platform as a translational tool for in vivo neuropsychopharmacology.


Subject(s)
Induced Pluripotent Stem Cells , Serotonin , Animals , Biomarkers , Humans , Mice , Neurons , Serotonin/pharmacology , Serotonin Plasma Membrane Transport Proteins , Selective Serotonin Reuptake Inhibitors/pharmacology
6.
EMBO Rep ; 23(5): e54027, 2022 05 04.
Article in English | MEDLINE | ID: mdl-35289477

ABSTRACT

Malformations of human cortical development (MCD) can cause severe disabilities. The lack of human-specific models hampers our understanding of the molecular underpinnings of the intricate processes leading to MCD. Here, we use cerebral organoids derived from patients and genome edited-induced pluripotent stem cells to address pathophysiological changes associated with a complex MCD caused by mutations in the echinoderm microtubule-associated protein-like 1 (EML1) gene. EML1-deficient organoids display ectopic neural rosettes at the basal side of the ventricular zone areas and clusters of heterotopic neurons. Single-cell RNA sequencing shows an upregulation of basal radial glial (RG) markers and human-specific extracellular matrix components in the ectopic cell population. Gene ontology and molecular analyses suggest that ectopic progenitor cells originate from perturbed apical RG cell behavior and yes-associated protein 1 (YAP1)-triggered expansion. Our data highlight a progenitor origin of EML1 mutation-induced MCD and provide new mechanistic insight into the human disease pathology.


Subject(s)
Induced Pluripotent Stem Cells , Organoids , Cerebral Cortex/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mutation , Neurons/metabolism , Organoids/metabolism
7.
J Steroid Biochem Mol Biol ; 182: 119-126, 2018 09.
Article in English | MEDLINE | ID: mdl-29751108

ABSTRACT

Glucocorticoids (GC) have first been shown to originate from the adrenal glands where synthesis and release is controlled by the hypothalamic-pituitary-adrenal (HPA) axis. Recently, it was shown that GC and other steroid hormones are also synthesized in the central nervous system, so-called neurosteroids. GC bind to specific GC receptors (GR) which function as ligand-activated transcription factors. GR are expressed in nearly all cell types in the brain, and therefore GC have a strong impact on neuronal development. Most knowledge of the influence of GC on neurodevelopment has been obtained from animal research. Recent advances in stem cell technology made it possible to generate neuronal precursor cells (NPCs) and neurons from human induced pluripotent stem cells (hiPSCs). To explore the cellular mechanism of GC affecting human neuronal development, we quantified the proliferation and differentiation of hiPSCs-derived NPCs in the absence and presence of the selective high-affinity GR agonist dexamethasone and the selective GR antagonist mifepristone, respectively. Our results show that inhibition of GR significantly reduced proliferation of NPCs and promoted differentiation whereas GR activation suppressed neuronal differentiation. This implies that neuronal GC must be present in NPCs for proliferation. Consequently we identified the presence of 11-ß-hydroxylase CYP11B1, which hydroxylates the respective steroid precursors to bioactive GC, in NPCs. We propose that hiPSC technology offers an ideal system to get more insight into the synthesising and regulatory pathways in steroidogenesis in human neurons and to differentiate between the mechanism by which adrenal GC and neuronal GC impact on neurodevelopment.


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation/drug effects , Induced Pluripotent Stem Cells/cytology , Neurogenesis , Neurons/cytology , Receptors, Glucocorticoid/metabolism , Anti-Inflammatory Agents/pharmacology , Cells, Cultured , Dexamethasone/pharmacology , Hormone Antagonists/pharmacology , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Mifepristone/pharmacology , Neurons/drug effects , Neurons/metabolism , Receptors, Glucocorticoid/genetics , Signal Transduction
8.
Dis Model Mech ; 10(4): 451-461, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28167616

ABSTRACT

The research domain criteria (RDoC) matrix has been developed to reorient psychiatric research towards measurable behavioral dimensions and underlying mechanisms. Here, we used a new genetic rat model with a loss-of-function point mutation in the dopamine transporter (DAT) gene (Slc6a3_N157K) to systematically study the RDoC matrix. First, we examined the impact of the Slc6a3_N157K mutation on monoaminergic signaling. We then performed behavioral tests representing each of the five RDoC domains: negative and positive valence systems, cognitive, social and arousal/regulatory systems. The use of RDoC may be particularly helpful for drug development. We studied the effects of a novel pharmacological approach metabotropic glutamate receptor mGluR2/3 antagonism, in DAT mutants in a comparative way with standard medications. Loss of DAT functionality in mutant rats not only elevated subcortical extracellular dopamine concentration but also altered the balance of monoaminergic transmission. DAT mutant rats showed deficits in all five RDoC domains. Thus, mutant rats failed to show conditioned fear responses, were anhedonic, were unable to learn stimulus-reward associations, showed impaired cognition and social behavior, and were hyperactive. Hyperactivity in mutant rats was reduced by amphetamine and atomoxetine, which are well-established medications to reduce hyperactivity in humans. The mGluR2/3 antagonist LY341495 also normalized hyperactivity in DAT mutant rats without affecting extracellular dopamine levels. We systematically characterized an altered dopamine system within the context of the RDoC matrix and studied mGluR2/3 antagonism as a new pharmacological strategy to treat mental disorders with underlying subcortical dopaminergic hyperactivity.


Subject(s)
Dopamine Plasma Membrane Transport Proteins/genetics , Mental Disorders/diagnosis , Mental Disorders/genetics , Animals , Behavior, Animal , Disease Models, Animal , Dopamine , HEK293 Cells , Humans , Loss of Function Mutation , Male , Mutant Proteins/metabolism , Phenotype , Point Mutation , Psychiatry , Rats, Inbred F344
9.
Stem Cell Res ; 15(1): 30-41, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26001168

ABSTRACT

Human induced pluripotent stem cells (hiPSCs) are a suitable tool to study basic molecular and cellular mechanisms of neurodevelopment. The directed differentiation of hiPSCs via the generation of a self-renewable neuronal precursor cell line allows the standardization of defined differentiation protocols. Here, we have investigated whether preconditioning with retinoic acid during early neural induction impacts on morphological and functional characteristics of the neuronal culture after terminal differentiation. For this purpose we have analyzed neuronal and glial cell markers, neuronal outgrowth, soma size, depolarization-induced distal shifts of the axon initial segment as well as glutamate-evoked calcium influx. Retinoic acid preconditioning led to a higher yield of neurons vs. glia cells and longer axons than unconditioned controls. In contrast, glutamatergic activation and depolarization induced structural plasticity were unchanged. Our results show that the treatment of neuroectodermal cells with retinoic acid during early development, i.e. during the neurulation phase, increases the yield of neuronal phenotypes, but does not impact on the functionality of terminally differentiated neuronal cells.


Subject(s)
Cell Shape/drug effects , Induced Pluripotent Stem Cells/cytology , Neurons/cytology , Tretinoin/pharmacology , Axons/drug effects , Axons/metabolism , Calcium/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Gene Expression Profiling , Glutamic Acid/pharmacology , Humans , Imaging, Three-Dimensional , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neurons/drug effects , Neurons/metabolism , Phenotype , Up-Regulation/drug effects
10.
Neuroreport ; 21(3): 185-90, 2010 Feb 17.
Article in English | MEDLINE | ID: mdl-20087235

ABSTRACT

We have examined the potency of two methods for the neuronal differentiation of embryonic stem cells on the generation of neuronal cells from human blood cells. A mixture of mononuclear cells from peripheral blood cells expressing monocytic, hematopoietic, and mesenchymal cell surface markers were exposed to all-trans retinoic acid, epidermal growth factor, and basic fibroblast growth factor (method A), or epidermal growth factor, fibroblast growth factor 8b, sonic hedgehog and ascorbic acid (method B). Both methods led to the generation of neuronal cells as judged by changes in morphology and the expression of the neuronal markers microtubule-associated protein type 2, tau, and beta-tubulin III. Differentiation according to method B favoured the development of neurons also expressing the dopamine transporter.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation/physiology , Leukocytes, Mononuclear/cytology , Neurons/cytology , Ascorbic Acid/pharmacology , Cell Differentiation/drug effects , Cell Separation , ErbB Receptors/metabolism , Fibroblast Growth Factor 2/pharmacology , Flow Cytometry , Fluorescent Antibody Technique , Hedgehog Proteins/pharmacology , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Leukocytes, Mononuclear/drug effects , Neurons/drug effects , Tretinoin/pharmacology
11.
Neurochem Int ; 54(3-4): 271-6, 2009.
Article in English | MEDLINE | ID: mdl-19121357

ABSTRACT

In the central nervous system serotonergic neurotransmission is terminated by the rapid removal of serotonin (5-hydroxytryptamine, 5HT) out of the extra-cellular space back into the presynaptic neuron. This task is fulfilled by a specific serotonin transporter (SERT) protein which controls the concentration of extra-cellular 5HT. Consequently, one mechanism to regulate the efficacy of serotonergic neurotransmission is via modulation of the density of SERT molecules on the cell membrane. In this regard it has been shown, that chronic activation of the p38 mitogen-activated protein kinase (p38 MAPK) leads to enhanced SERT surface expression whereas activation of protein kinase C (PKC) reduces SERT surface expression. In addition, it has been reported that exposure to selective serotonin re-uptake inhibitors (SSRIs) leads to a down-regulation of SERT expression in vivo and in vitro in different cellular systems. Here, we have studied interactions between kinase- and SSRI-induced SERT internalization in mouse stem cell-derived serotonergic neurons expressing the native SERT allele in its natural surroundings. Therefore we established a method to quantify the amount of cell surface-expressed SERT molecules on individual cells by antibody detection combined with confocal laser scanning microscopy. Using this methodology we could show that activation of PKC, inhibition of the p38 MAPK as well as exposure to the SSRI citalopram each induced a significant reduction of cell surface-expressed SERT over time. Combinations of PKC activation, p38 MAPK inhibition and SSRI exposure led to a more pronounced down-regulation of SERT surface expression depending on the time of drug exposure.


Subject(s)
Endocytosis/physiology , Neurons/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Serotonin/metabolism , Stem Cells/metabolism , Animals , Cell Differentiation/physiology , Cell Line , Cell Lineage/physiology , Citalopram/pharmacology , Down-Regulation/drug effects , Down-Regulation/physiology , Enzyme Inhibitors/pharmacology , Extracellular Space/metabolism , Mice , Microscopy, Confocal , Neurogenesis/physiology , Neurons/cytology , Phosphorylation/drug effects , Protein Kinase C/drug effects , Protein Kinase C/metabolism , Protein Transport/physiology , Selective Serotonin Reuptake Inhibitors/pharmacology , Stem Cells/cytology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
12.
FASEB J ; 22(6): 1702-14, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18216289

ABSTRACT

A deficiency of serotonergic signaling is thought to be involved in the etiology of depression. Thus, drugs blocking the reuptake of serotonin back into the neurons are widely used in treatment of this disease; however, their delayed effect in remission of patients suggests that the clinical response does not rely on simple serotonin uptake inhibition but may include further regulatory mechanisms. We have analyzed cellular serotonin transporter (SERT) expression on exposure to the selective serotonin reuptake inhibitor citalopram in serotonergic neurons expressing the native SERT allele in its natural surroundings. Biotinylation of SERT-expressing HEK293 cells, as well as confocal microscopy analysis in these cells and in serotonergic neurons, revealed that exposure to citalopram time dependently reduces the amount of cell surface-expressed SERT. Furthermore, in serotonergic neurons, longer exposure to citalopram not only caused an internalization of SERT proteins from the cell surface but also induced a redistribution of SERT from neurite extensions into the soma. This process was reversible on drug removal. Microarray analysis performed on citalopram-treated serotonergic neurons revealed that antidepressant treatment does not alter SERT mRNA expression, suggesting that SERT trafficking from and to the cell membrane is regulated on the posttranscriptional level.


Subject(s)
Endocytosis/drug effects , Neurons/chemistry , Selective Serotonin Reuptake Inhibitors/pharmacology , Serotonin Plasma Membrane Transport Proteins/analysis , Antidepressive Agents , Cell Line , Citalopram/pharmacology , Humans , Kinetics , Neurites/chemistry , Protein Transport/drug effects , RNA, Messenger/analysis , Serotonin Plasma Membrane Transport Proteins/metabolism , Tissue Distribution
13.
Neurochem Int ; 52(4-5): 770-5, 2008.
Article in English | MEDLINE | ID: mdl-17949855

ABSTRACT

Compelling evidence has been provided that Na(+) and Cl(-)-dependent neurotransmitter transporter proteins form oligomeric complexes. Specific helix-helix interactions in lipid bilayers are thought to promote the assembly of integral membrane proteins to oligomeric structures. These interactions are determined by selective transmembrane helix packing motifs one of which is the Glycophorin A motif (GxxxG). This motif is present in the sixth transmembrane domain of most transporter proteins. In order to investigate, whether this motif is important for proper expression and function of the serotonin transporter (SERT), we have analysed the effect of mutating the respective glycine residues Gly338 and Gly342 to valine upon transient expression of the respective cDNAs in HEK293 cells. As revealed by western blotting, wildtype SERT is found in monomeric and dimeric forms while both mutants are expressed as monomers solely. Confocal microscopy revealed that the wildtype SERT is expressed at the cell surface, whereas both mutant proteins are localised in intracellular compartments. Failure of integration into the cell membrane is responsible for a total loss of [(3)H]5HT uptake capability by the mutants. These findings show that in the SERT protein the integrity of the GxxxG motif is essential for dimerisation and proper targeting of the transporter complex to the cell surface.


Subject(s)
Glycine/metabolism , Membrane Glycoproteins/biosynthesis , Serotonin Plasma Membrane Transport Proteins/biosynthesis , Amino Acid Substitution/genetics , Amino Acid Substitution/physiology , Blotting, Western , Cell Line, Tumor , Cell Membrane/metabolism , Citalopram/metabolism , Data Interpretation, Statistical , Dimerization , Electrophoresis, Polyacrylamide Gel , Fluorescent Antibody Technique , Glycophorins/metabolism , Green Fluorescent Proteins/metabolism , Humans , Microscopy, Confocal , Mutation , Radioligand Assay , Serotonin Plasma Membrane Transport Proteins/genetics , Selective Serotonin Reuptake Inhibitors/metabolism , Valine/metabolism
14.
J Neurochem ; 86(4): 958-65, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12887693

ABSTRACT

Re-uptake of the neurotransmitters serotonin and noradrenaline out of the synaptic cleft is mediated by selective transporter proteins, the serotonin transporter and the noradrenaline transporter respectively. Both are integral membrane proteins that are have a high degree of homology and represent members of a larger neurotransmitter transporter superfamily. Several studies have indicated that the serotonin transporter has an an oligomeric structure. To determine whether monoamine transporters can also function in oligomeric structures in situ, we constructed a concatenate consisting of one molecule of serotonin transporter covalently linked to one molecule of noradrenaline transporter. Heterologous expression of this hybrid construct allowed us to analyse the function, i.e. transport activity, and the structure, i.e. the molecular weight of the total construct and of its single components, at the same time. We showed that serotonin-noradrenaline transporter fusion proteins are fully active and exhibit the pharmacological profile of both their individual components. These findings support the hypothesis that monoamine transporters are expressed and may function as oligomeric proteins composed of non-interacting monomers.


Subject(s)
Carrier Proteins/metabolism , Fluoxetine/analogs & derivatives , Membrane Glycoproteins/metabolism , Membrane Transport Proteins , Nerve Tissue Proteins , Symporters/metabolism , Amino Acid Sequence , Animals , Antidepressive Agents/pharmacokinetics , Binding, Competitive/drug effects , Carrier Proteins/genetics , Cell Line , Cell Membrane/metabolism , Citalopram/pharmacokinetics , Fluoxetine/pharmacokinetics , Humans , Imipramine/pharmacokinetics , Immunoblotting , Kidney/cytology , Kidney/metabolism , Membrane Glycoproteins/genetics , Molecular Sequence Data , Norepinephrine/metabolism , Norepinephrine/pharmacokinetics , Norepinephrine Plasma Membrane Transport Proteins , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Serotonin/metabolism , Serotonin/pharmacokinetics , Serotonin Plasma Membrane Transport Proteins , Symporters/genetics , Transfection , Tritium
SELECTION OF CITATIONS
SEARCH DETAIL
...