Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Mol Cancer Res ; 18(10): 1522-1533, 2020 10.
Article in English | MEDLINE | ID: mdl-32665429

ABSTRACT

Children suffering from neurologic cancers undergoing chemotherapy and radiotherapy are at high risk of reduced neurocognitive abilities likely via damage to proliferating neural stem cells (NSC). Therefore, strategies to protect NSCs are needed. We argue that induced cell-cycle arrest/quiescence in NSCs during cancer treatment can represent such a strategy. Here, we show that hyperpolarization-activated cyclic nucleotide-gated (HCN) ion channels are dynamically expressed over the cell cycle in NSCs, depolarize the membrane potential, underlie spontaneous calcium oscillations and are required to maintain NSCs in the actively proliferating pool. Hyperpolarizing pharmacologic inhibition of HCN channels during exposure to ionizing radiation protects NSCs cells in neurogenic brain regions of young mice. In contrast, brain tumor-initiating cells, which also express HCN channels, remain proliferative during HCN inhibition. IMPLICATIONS: Our finding that NSCs can be selectively rescued while cancer cells remain sensitive to the treatment, provide a foundation for reduction of cognitive impairment in children with neurologic cancers.


Subject(s)
Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Neoplasms/drug therapy , Neural Stem Cells/metabolism , Animals , Cell Proliferation , Humans , Mice
2.
Nat Commun ; 10(1): 2110, 2019 05 08.
Article in English | MEDLINE | ID: mdl-31068593

ABSTRACT

Ribosome biogenesis is a canonical hallmark of cell growth and proliferation. Here we show that execution of Epithelial-to-Mesenchymal Transition (EMT), a migratory cellular program associated with development and tumor metastasis, is fueled by upregulation of ribosome biogenesis during G1/S arrest. This unexpected EMT feature is independent of species and initiating signal, and is accompanied by release of the repressive nucleolar chromatin remodeling complex (NoRC) from rDNA, together with recruitment of the EMT-driving transcription factor Snai1 (Snail1), RNA Polymerase I (Pol I) and the Upstream Binding Factor (UBF). EMT-associated ribosome biogenesis is also coincident with increased nucleolar recruitment of Rictor, an essential component of the EMT-promoting mammalian target of rapamycin complex 2 (mTORC2). Inhibition of rRNA synthesis in vivo differentiates primary tumors to a benign, Estrogen Receptor-alpha (ERα) positive, Rictor-negative phenotype and reduces metastasis. These findings implicate the EMT-associated ribosome biogenesis program with cellular plasticity, de-differentiation, cancer progression and metastatic disease.


Subject(s)
Epithelial-Mesenchymal Transition/physiology , G1 Phase Cell Cycle Checkpoints/physiology , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Ribosomes/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Differentiation/physiology , Cell Line, Tumor/transplantation , Cell Movement/physiology , Cell Nucleolus/metabolism , Chick Embryo , Chromosomal Proteins, Non-Histone/metabolism , DNA, Ribosomal/metabolism , Disease Models, Animal , Female , Gene Expression Profiling , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Ribosomal/metabolism , Ribosomes/genetics
3.
PLoS One ; 12(12): e0188772, 2017.
Article in English | MEDLINE | ID: mdl-29228002

ABSTRACT

The cell cycle coordinates core functions such as replication and cell division. However, cell-cycle-regulated transcription in the control of non-core functions, such as cell identity maintenance through specific transcription factors (TFs) and signalling pathways remains unclear. Here, we provide a resource consisting of mapped transcriptomes in unsynchronized HeLa and U2OS cancer cells sorted for cell cycle phase by Fucci reporter expression. We developed a novel algorithm for data analysis that enables efficient visualization and data comparisons and identified cell cycle synchronization of Notch signalling and TFs associated with development. Furthermore, the cell cycle synchronizes with the circadian clock, providing a possible link between developmental transcriptional networks and the cell cycle. In conclusion we find that cell cycle synchronized transcriptional patterns are temporally compartmentalized and more complex than previously anticipated, involving genes, which control cell identity and development.


Subject(s)
Cell Cycle/genetics , Neoplasms/metabolism , Transcription Factors/metabolism , Transcriptome , Algorithms , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Humans , Neoplasms/genetics , Neoplasms/pathology
4.
Front Pharmacol ; 8: 726, 2017.
Article in English | MEDLINE | ID: mdl-29114221

ABSTRACT

Stem cells display a fundamentally different mechanism of proliferation control when compared to somatic cells. Uncovering these mechanisms would maximize the impact in drug discovery with a higher translational applicability. The unbiased approach used in phenotype-based drug discovery (PDD) programs can offer a unique opportunity to identify such novel biological phenomenon. Here, we describe an integrated phenotypic screening approach, employing a combination of in vitro and in vivo PDD models to identify a small molecule increasing stem cell proliferation. We demonstrate that a combination of both in vitro and in vivo screening models improves hit identification and reproducibility of effects across various PDD models. Using cell viability and colony size phenotype measurement we characterize the structure activity relationship of the lead molecule, and identify that the small molecule inhibits phosphorylation of ERK2 and promotes stem cell proliferation. This study demonstrates a PDD approach that employs combinatorial models to identify compounds promoting stem cell proliferation.

6.
Cancer Res ; 77(7): 1741-1752, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28087597

ABSTRACT

Glioma-initiating cells (GIC) are considered the underlying cause of recurrences of aggressive glioblastomas, replenishing the tumor population and undermining the efficacy of conventional chemotherapy. Here we report the discovery that inhibiting T-type voltage-gated Ca2+ and KCa channels can effectively induce selective cell death of GIC and increase host survival in an orthotopic mouse model of human glioma. At present, the precise cellular pathways affected by the drugs affecting these channels are unknown. However, using cell-based assays and integrated proteomics, phosphoproteomics, and transcriptomics analyses, we identified the downstream signaling events these drugs affect. Changes in plasma membrane depolarization and elevated intracellular Na+, which compromised Na+-dependent nutrient transport, were documented. Deficits in nutrient deficit acted in turn to trigger the unfolded protein response and the amino acid response, leading ultimately to nutrient starvation and GIC cell death. Our results suggest new therapeutic targets to attack aggressive gliomas. Cancer Res; 77(7); 1741-52. ©2017 AACR.


Subject(s)
Amino Acids/metabolism , Brain Neoplasms/drug therapy , Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/physiology , Glioma/drug therapy , Potassium Channels, Calcium-Activated/antagonists & inhibitors , Unfolded Protein Response/drug effects , Animals , Biological Transport , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Death , Cell Line, Tumor , Dihydropyridines/pharmacology , Glioma/metabolism , Glioma/pathology , Humans , Mice , Mycotoxins/pharmacology , Neoplastic Stem Cells/pathology , Proteomics , Sodium/metabolism
7.
Biochim Biophys Acta Gene Regul Mech ; 1860(2): 175-183, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27863263

ABSTRACT

The Oct4 gene codes for a transcription factor that plays a critical role in the maintenance of pluripotency in embryonic and cancer stem cells. Its expression thus has to be tightly regulated. We performed biophysical characterization of the promoter region using a combination of UV absorption, CD, and NMR spectroscopies, native PAGE and chemical probing, which was followed by functional studies involving luciferase reporter assays performed in osteosarcoma and human embryonic stem cell lines. We have shown that the evolutionarily conserved G-rich region close to the Oct4 transcription start site in the non-template strand forms a parallel G-quadruplex structure. We characterized its structure and stability upon point mutations in its primary structure. Functional studies then revealed that whereas the wild type quadruplex sequence ensures high reporter gene expression, the expression of mutated variants is significantly decreased proportionally to the destabilizing effect of the mutations on the quadruplex. A ligand, N-methyl mesoporphyrin IX that increases the stability of formed quadruplex rescued the reporter expression of single-mutated variants to the level of wild-type, but it has no effect on a mutated variant that cannot form quadruplex. These data indicate that the quadruplex acts as a strong, positive regulator of Oct4 expression and as such it might serve as a potential target for therapeutic intervention.


Subject(s)
Octamer Transcription Factor-3/genetics , Promoter Regions, Genetic/genetics , Cell Line, Tumor , Circular Dichroism/methods , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , G-Quadruplexes/drug effects , Genes, Reporter/genetics , Humans , Magnetic Resonance Imaging/methods , Mesoporphyrins/pharmacology , Mutation/genetics , Osteosarcoma/genetics , Promoter Regions, Genetic/drug effects , Transcription Initiation Site/drug effects , Transcription Initiation Site/physiology
8.
Oncotarget ; 7(45): 73200-73215, 2016 11 08.
Article in English | MEDLINE | ID: mdl-27689322

ABSTRACT

Glioblastoma multiforme (GBM, astrocytoma grade IV) is the most common malignant primary brain tumor in adults. Addressing the shortage of effective treatment options for this cancer, we explored repurposing of existing drugs into combinations with potent activity against GBM cells. We report that the phytoalexin pterostilbene is a potentiator of two drugs with previously reported anti-GBM activity, the EGFR inhibitor gefitinib and the antidepressant sertraline. Combinations of either of these two compounds with pterostilbene suppress cell growth, viability, sphere formation and inhibit migration in tumor GBM cell (GC) cultures. The potentiating effect of pterostilbene was observed to a varying degree across a panel of 41 patient-derived GCs, and correlated in a case specific manner with the presence of missense mutation of EGFR and PIK3CA and a focal deletion of the chromosomal region 1p32. We identify pterostilbene-induced cell cycle arrest, synergistic inhibition of MAPK activity and induction of Thioredoxin interacting protein (TXNIP) as possible mechanisms behind pterostilbene's effect. Our results highlight a nontoxic stilbenoid compound as a modulator of anticancer drug response, and indicate that pterostilbene might be used to modulate two anticancer compounds in well-defined sets of GBM patients.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Stilbenes/pharmacology , Aged , Aged, 80 and over , Antineoplastic Agents, Phytogenic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Copy Number Variations , Drug Synergism , Female , Gefitinib , Gene Expression Profiling , Gene Knockdown Techniques , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , MAP Kinase Signaling System/drug effects , Male , Middle Aged , Mutation , Phenotype , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Stilbenes/therapeutic use , Transcriptome
9.
Springerplus ; 5: 41, 2016.
Article in English | MEDLINE | ID: mdl-26835223

ABSTRACT

Pluripotent stem cells are the starting cell type of choice for the development of many cell-based regenerative therapies due to their rapid and unlimited proliferation and broad differentiation potential. The unique pluripotent cell cycle underlies both these properties. Hyperpolarization-activated cyclic nucleotide-gated cation (HCN) family channels have previously been reported to modulate mouse embryonic stem cell (ESC) proliferation and here we characterize the effects of HCN inhibitor ZD7288 on ESC proliferation and stem cell identity. The doubling time of cells treated with the HCN blocker increased by ~30 % due to longer G1 and S phases, resulting in a nearly twofold reduction in ESC numbers after 4 day serum-free culture. Slower progression through S phase was not accompanied by H2AX phosphorylation or cell stalling at transition points, although EdU incorporation in treated cells was reduced. Despite the drastic cell cycle perturbations, the pluripotent status of the cells was not compromised by treatment. Cultures treated with the HCN blocker in maintenance conditions maintained pluripotency marker expression on both RNA and protein level, although we observed a reversible effect on morphology and colony formation frequency. Addition of ZD7288 in differentiating media improved FBS-driven differentiation, but not directed differentiation to neuroectoderm, further indicating that altered cell cycle structure does not necessarily compromise pluripotency and drive ESCs to differentiation. The categorically different outcomes of ZD7288 use during differentiation indicate that cell culture context can be determinative for effects of ion-modulatory molecules and underscores the need for exploring their action in serum-free conditions demanded by potential clinical use.

10.
Oncotarget ; 6(35): 37083-97, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26397227

ABSTRACT

Radiotherapy in children causes debilitating cognitive decline, partly linked to impaired neurogenesis. Irradiation targets primarily cancer cells but also endogenous neural stem/progenitor cells (NSPCs) leading to cell death or cell cycle arrest. Here we evaluated the effects of lithium on proliferation, cell cycle and DNA damage after irradiation of young NSPCs in vitro.NSPCs were treated with 1 or 3 mM LiCl and we investigated proliferation capacity (neurosphere volume and bromodeoxyuridine (BrdU) incorporation). Using flow cytometry, we analysed apoptosis (annexin V), cell cycle (propidium iodide) and DNA damage (γH2AX) after irradiation (3.5 Gy) of lithium-treated NSPCs.Lithium increased BrdU incorporation and, dose-dependently, the number of cells in replicative phase as well as neurosphere growth. Irradiation induced cell cycle arrest in G1 and G2/M phases. Treatment with 3 mM LiCl was sufficient to increase NSPCs in S phase, boost neurosphere growth and reduce DNA damage. Lithium did not affect the levels of apoptosis, suggesting that it does not rescue NSPCs committed to apoptosis due to accumulated DNA damage.Lithium is a very promising candidate for protection of the juvenile brain from radiotherapy and for its potential to thereby improve the quality of life for those children who survive their cancer.


Subject(s)
Cell Cycle Checkpoints/drug effects , Hippocampus/cytology , Lithium Chloride/pharmacology , Neural Stem Cells/cytology , Neurogenesis/physiology , Animals , Animals, Newborn , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Cycle Checkpoints/radiation effects , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cells, Cultured , Cobalt Radioisotopes , DNA Damage/drug effects , DNA Damage/radiation effects , Female , Flow Cytometry , Gamma Rays , Hippocampus/drug effects , Hippocampus/radiation effects , In Vitro Techniques , Lithium Chloride/administration & dosage , Mice , Mice, Inbred C57BL , Neural Stem Cells/drug effects , Neural Stem Cells/radiation effects , Neurogenesis/drug effects , Neurogenesis/radiation effects
12.
PLoS One ; 9(12): e115698, 2014.
Article in English | MEDLINE | ID: mdl-25531110

ABSTRACT

Tumor-initiating cells are a subpopulation in aggressive cancers that exhibit traits shared with stem cells, including the ability to self-renew and differentiate, commonly referred to as stemness. In addition, such cells are resistant to chemo- and radiation therapy posing a therapeutic challenge. To uncover stemness-associated functions in glioma-initiating cells (GICs), transcriptome profiles were compared to neural stem cells (NSCs) and gene ontology analysis identified an enrichment of Ca2+ signaling genes in NSCs and the more stem-like (NSC-proximal) GICs. Functional analysis in a set of different GIC lines regarding sensitivity to disturbed homeostasis using A23187 and Thapsigargin, revealed that NSC-proximal GICs were more sensitive, corroborating the transcriptome data. Furthermore, Ca2+ drug sensitivity was reduced in GICs after differentiation, with most potent effect in the NSC-proximal GIC, supporting a stemness-associated Ca2+ sensitivity. NSCs and the NSC-proximal GIC line expressed a larger number of ion channels permeable to potassium, sodium and Ca2+. Conversely, a higher number of and higher expression levels of Ca2+ binding genes that may buffer Ca2+, were expressed in NSC-distal GICs. In particular, expression of the AMPA glutamate receptor subunit GRIA1, was found to associate with Ca2+ sensitive NSC-proximal GICs, and decreased as GICs differentiated along with reduced Ca2+ drug sensitivity. The correlation between high expression of Ca2+ channels (such as GRIA1) and sensitivity to Ca2+ drugs was confirmed in an additional nine novel GIC lines. Calcium drug sensitivity also correlated with expression of the NSC markers nestin (NES) and FABP7 (BLBP, brain lipid-binding protein) in this extended analysis. In summary, NSC-associated NES+/FABP7+/GRIA1+ GICs were selectively sensitive to disturbances in Ca2+ homeostasis, providing a potential target mechanism for eradication of an immature population of malignant cells.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/drug therapy , Calcium/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Glioma/drug therapy , Neoplastic Stem Cells/drug effects , Neural Stem Cells/drug effects , Blotting, Western , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Fluorescent Antibody Technique , Gene Expression Profiling , Glioma/genetics , Glioma/pathology , Humans , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Tumor Cells, Cultured
13.
Cell ; 157(2): 313-328, 2014 04 10.
Article in English | MEDLINE | ID: mdl-24656405

ABSTRACT

Glioblastoma multiforme (GBM) is the most aggressive form of brain cancer with marginal life expectancy. Based on the assumption that GBM cells gain functions not necessarily involved in the cancerous process, patient-derived glioblastoma cells (GCs) were screened to identify cellular processes amenable for development of targeted treatments. The quinine-derivative NSC13316 reliably and selectively compromised viability. Synthetic chemical expansion reveals delicate structure-activity relationship and analogs with increased potency, termed Vacquinols. Vacquinols stimulate death by membrane ruffling, cell rounding, massive macropinocytic vacuole accumulation, ATP depletion, and cytoplasmic membrane rupture of GCs. The MAP kinase MKK4, identified by a shRNA screen, represents a critical signaling node. Vacquinol-1 displays excellent in vivo pharmacokinetics and brain exposure, attenuates disease progression, and prolongs survival in a GBM animal model. These results identify a vulnerability to massive vacuolization that can be targeted by small molecules and point to the possible exploitation of this process in the design of anticancer therapies.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Glioblastoma/drug therapy , Glioblastoma/pathology , Piperidines/pharmacology , Quinolines/pharmacology , Small Molecule Libraries/pharmacology , Animals , Cell Death/drug effects , Heterografts , Humans , Hydroxyquinolines/pharmacology , MAP Kinase Kinase 4/metabolism , Mice , Neoplasm Transplantation , Pinocytosis/drug effects , Vacuoles/metabolism , Zebrafish
14.
PLoS One ; 8(8): e72409, 2013.
Article in English | MEDLINE | ID: mdl-23936540

ABSTRACT

The cell cycle progression in mouse embryonic stem cells (mESCs) is controlled by ion fluxes that alter cell volume [1]. This suggests that ion fluxes might control dynamic changes in morphology over the cell cycle, such as rounding up of the cell at mitosis. However, specific channels regulating such dynamic changes and the possible interactions with actomyosin complex have not been clearly identified. Following RNAseq transcriptome analysis of cell cycle sorted mESCs, we found that expression of the K(+) ion channel Erg1 peaked in G1 cell cycle phase, which was confirmed by immunostaining. Inhibition of Erg channel activity caused loss of G1 phase cells via non-apoptotic cell death. Cells first lost the ability of membrane blebbing, a typical feature of cultured embryonic stem cells. Continued Erg inhibition further increased cell volume and the cell eventually ruptured. In addition, atomic force measurements on live cells revealed a decreased cortical stiffness after treatment, suggesting alterations in actomyosin organization. When the intracellular osmotic pressure was experimentally decreased by hypertonic solution or block of K(+) ion import via the Na, K-ATPase, cell viability was restored and cells acquired normal volume and blebbing activity. Our results suggest that Erg channels have a critical function in K(+) ion homeostasis of mESCs over the cell cycle, and that cell death following Erg inhibition is a consequence of the inability to regulate cell volume.


Subject(s)
Cell Cycle/physiology , Cell Size , Embryonic Stem Cells/physiology , Ether-A-Go-Go Potassium Channels/metabolism , Animals , Apoptosis , Blotting, Western , Embryonic Stem Cells/cytology , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ether-A-Go-Go Potassium Channels/genetics , Flow Cytometry , Image Processing, Computer-Assisted , Mice , Microscopy, Atomic Force , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Time-Lapse Imaging
15.
Proc Natl Acad Sci U S A ; 110(16): E1524-32, 2013 Apr 16.
Article in English | MEDLINE | ID: mdl-23576737

ABSTRACT

Coherent network activity among assemblies of interconnected cells is essential for diverse functions in the adult brain. However, cellular networks before formations of chemical synapses are poorly understood. Here, embryonic stem cell-derived neural progenitors were found to form networks exhibiting synchronous calcium ion (Ca(2+)) activity that stimulated cell proliferation. Immature neural cells established circuits that propagated electrical signals between neighboring cells, thereby activating voltage-gated Ca(2+) channels that triggered Ca(2+) oscillations. These network circuits were dependent on gap junctions, because blocking prevented electrotonic transmission both in vitro and in vivo. Inhibiting connexin 43 gap junctions abolished network activity, suppressed proliferation, and affected embryonic cortical layer formation. Cross-correlation analysis revealed highly correlated Ca(2+) activities in small-world networks that followed a scale-free topology. Graph theory predicts that such network designs are effective for biological systems. Taken together, these results demonstrate that immature cells in the developing brain organize in small-world networks that critically regulate neural progenitor proliferation.


Subject(s)
Brain/embryology , Cell Proliferation , Nerve Net , Neural Stem Cells/physiology , Animals , Calcium/metabolism , Calcium Channels, L-Type/metabolism , Connexin 43/metabolism , Electrical Synapses/physiology , Mice , Mice, Inbred C57BL , Microscopy, Interference , Models, Neurological , Neural Stem Cells/cytology , Plasmids/genetics , RNA, Small Interfering/genetics
16.
Semin Cancer Biol ; 22(5-6): 369-73, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22659342

ABSTRACT

Epithelial to mesenchymal transition (EMT) during metastasis is initially a two-step process beginning with delamination of cells from the solid tumor followed by acquisition of a migratory phenotype. Several reports indicate that plasma membrane blebbing, associated with cell division, coincides with cell delamination during developmental EMT. This raises a speculative question if blebbing drives EMT in cancer cells in a similar way. Here, we review available data on factors and processes that may support such a connection.


Subject(s)
Epithelial-Mesenchymal Transition , Mitosis , Neoplasms/genetics , Neoplasms/pathology , Cell Movement , Humans , Ions/metabolism , Neoplasm Metastasis , Neoplasms/metabolism
17.
BMC Physiol ; 12: 3, 2012 Mar 26.
Article in English | MEDLINE | ID: mdl-22449203

ABSTRACT

BACKGROUND: Cardiovascular toxicity is a major limiting factor in drug development and requires multiple cost-effective models to perform toxicological evaluation. Zebrafish is an excellent model for many developmental, toxicological and regenerative studies. Using approaches like morpholino knockdown and electrocardiogram, researchers have demonstrated physiological and functional similarities between zebrafish heart and human heart. The close resemblance of the genetic cascade governing heart development in zebrafish to that of humans has propelled the zebrafish system as a cost-effective model to conduct various genetic and pharmacological screens on developing embryos and larvae. The current report describes a methodology for rapid isolation of adult zebrafish heart, maintenance ex vivo, and a setup to perform quick small molecule throughput screening, including an in-house implemented analysis script. RESULTS: Adult zebrafish were anesthetized and after rapid decapitation the hearts were isolated. The short time required for isolation of hearts allows dissection of multiple fishes, thereby obtaining a large sample size. The simple protocol for ex vivo culture allowed maintaining the beating heart for several days. The in-house developed script and spectral analyses allowed the readouts to be presented either in time domain or in frequency domain. Taken together, the current report offers an efficient platform for performing cardiac drug testing and pharmacological screens. CONCLUSION: The new methodology presents a fast, cost-effective, sensitive and reliable method for performing small molecule screening. The variety of readouts that can be obtained along with the in-house developed analyses script offers a powerful setup for performing cardiac toxicity evaluation by researchers from both academics and industry.


Subject(s)
Biological Assay/instrumentation , Drug Evaluation, Preclinical/instrumentation , Flow Cytometry/instrumentation , Heart/drug effects , Heart/physiology , Organ Culture Techniques/instrumentation , Zebrafish/physiology , Animals , Equipment Design , Equipment Failure Analysis
19.
Proc Natl Acad Sci U S A ; 108(14): 5837-42, 2011 Apr 05.
Article in English | MEDLINE | ID: mdl-21436033

ABSTRACT

Adult neural stem cell proliferation is dynamic and has the potential for massive self-renewal yet undergoes limited cell division in vivo. Here, we report an epigenetic mechanism regulating proliferation and self-renewal. The recruitment of the PI3K-related kinase signaling pathway and histone H2AX phosphorylation following GABA(A) receptor activation limits subventricular zone proliferation. As a result, NSC self-renewal and niche size is dynamic and can be directly modulated in both directions pharmacologically or by genetically targeting H2AX activation. Surprisingly, changes in proliferation have long-lasting consequences on stem cell numbers, niche size, and neuronal output. These results establish a mechanism that continuously limits proliferation and demonstrates its impact on adult neurogenesis. Such homeostatic suppression of NSC proliferation may contribute to the limited self-repair capacity of the damaged brain.


Subject(s)
Adult Stem Cells/physiology , Cell Cycle/physiology , Cell Proliferation , DNA Repair/physiology , Epigenesis, Genetic/physiology , Histones/metabolism , Neural Stem Cells/physiology , Signal Transduction/physiology , Animals , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Statistics, Nonparametric
20.
Curr Opin Neurobiol ; 18(3): 232-6, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18638551

ABSTRACT

The brain develops and functions in a complex ionic milieu, which is a prerequisite for neurotransmitter function and neuronal signaling. Neurotransmitters and ion fluxes are, however, important not only in neuronal signaling, but also in the control of neural differentiation, and in this review, we highlight the recent advances in our understanding of how the gamma-amino butyric acid (GABA) neurotransmitter and ion fluxes are relevant for cell cycle control and neural differentiation. Conversely, proteins previously associated with ion transport across membranes have been endowed with novel ion-independent functions, and we discuss this in the context of gap junctions in cell adhesion and of the neuron-specific K(+)-Cl(-) cotransporter KCC2 in dendritic spine development. Collectively, these findings provide a richer and more complex picture of when ion fluxes are needed in neural development and when they are not.


Subject(s)
Ion Transport/physiology , Neurons/physiology , Neurotransmitter Agents/metabolism , Signal Transduction/physiology , Animals , Cell Cycle/physiology , Cell Differentiation/physiology , gamma-Aminobutyric Acid/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...