Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Proc Natl Acad Sci U S A ; 120(2): e2216216120, 2023 01 10.
Article in English | MEDLINE | ID: mdl-36595701

ABSTRACT

The rise of antibiotic-resistant bacterial infections poses a global threat. Antibiotic resistance development is generally studied in batch cultures which conceals the heterogeneity in cellular responses. Using single-cell imaging, we studied the growth response of Escherichia coli to sub-inhibitory and inhibitory concentrations of nine antibiotics. We found that the heterogeneity in growth increases more than what is expected from growth rate reduction for three out of the nine antibiotics tested. For two antibiotics (rifampicin and nitrofurantoin), we found that sub-populations were able to maintain growth at lethal antibiotic concentrations for up to 10 generations. This perseverance of growth increased the population size and led to an up to 40-fold increase in the frequency of antibiotic resistance mutations in gram-negative and gram-positive species. We conclude that antibiotic perseverance is a common phenomenon that has the potential to impact antibiotic resistance development across pathogenic bacteria.


Subject(s)
Anti-Bacterial Agents , Escherichia coli , Anti-Bacterial Agents/pharmacology , Microbial Sensitivity Tests , Rifampin/pharmacology , Mutation , Bacteria , Drug Resistance, Bacterial/genetics
2.
Nat Commun ; 13(1): 6215, 2022 10 20.
Article in English | MEDLINE | ID: mdl-36266330

ABSTRACT

Antimicrobial resistance is an increasing problem on a global scale. Rapid antibiotic susceptibility testing (AST) is urgently needed in the clinic to enable personalized prescriptions in high-resistance environments and to limit the use of broad-spectrum drugs. Current rapid phenotypic AST methods do not include species identification (ID), leaving time-consuming plating or culturing as the only available option when ID is needed to make the sensitivity call. Here we describe a method to perform phenotypic AST at the single-cell level in a microfluidic chip that allows subsequent genotyping by in situ FISH. By stratifying the phenotypic AST response on the species of individual cells, it is possible to determine the susceptibility profile for each species in a mixed sample in 2 h. In this proof-of-principle study, we demonstrate the operation with four antibiotics and mixed samples with combinations of seven species.


Subject(s)
Anti-Bacterial Agents , Microfluidics , Microbial Sensitivity Tests , Anti-Bacterial Agents/pharmacology , Microfluidics/methods
4.
Nature ; 597(7876): 426-429, 2021 09.
Article in English | MEDLINE | ID: mdl-34471288

ABSTRACT

Homologous recombination is essential for the accurate repair of double-stranded DNA breaks (DSBs)1. Initially, the RecBCD complex2 resects the ends of the DSB into 3' single-stranded DNA on which a RecA filament assembles3. Next, the filament locates the homologous repair template on the sister chromosome4. Here we directly visualize the repair of DSBs in single cells, using high-throughput microfluidics and fluorescence microscopy. We find that, in Escherichia coli, repair of DSBs between segregated sister loci is completed in 15 ± 5 min (mean ± s.d.) with minimal fitness loss. We further show that the search takes less than 9 ± 3 min (mean ± s.d) and is mediated by a thin, highly dynamic RecA filament that stretches throughout the cell. We propose that the architecture of the RecA filament effectively reduces search dimensionality. This model predicts a search time that is consistent with our measurement and is corroborated by the observation that the search time does not depend on the length of the cell or the amount of DNA. Given the abundance of RecA homologues5, we believe this model to be widely conserved across living organisms.


Subject(s)
DNA, Bacterial/metabolism , Escherichia coli/enzymology , Escherichia coli/genetics , Models, Biological , Rec A Recombinases/metabolism , Recombinational DNA Repair , Sequence Homology, Nucleic Acid , DNA Breaks, Double-Stranded , DNA, Single-Stranded/metabolism , Time Factors
5.
Commun Biol ; 4(1): 521, 2021 05 05.
Article in English | MEDLINE | ID: mdl-33953333

ABSTRACT

Many bacterial species and antibiotic classes exhibit heteroresistance, a phenomenon in which a susceptible bacterial isolate harbors a resistant subpopulation that can grow in the presence of an antibiotic and cause treatment failure. The resistant phenotype is often unstable and without antibiotic selection it reverts back to susceptibility. Here we studied the dynamics by which these resistant subpopulations are enriched in the presence of antibiotic and recede back to their baseline frequency in the absence of selection. An increasing understanding of this instability will allow more effective diagnostics and treatment of infections caused by heteroresistant bacteria. We show for clinical isolates of Escherichia coli and Salmonella enterica that different antibiotics at levels below the MIC of the susceptible main population can cause rapid enrichment of resistant subpopulations with increased copy number of genes that cause resistance. Modelling and growth rate measurements of bacteria with increased gene copy number in cultures and by microscopy of single-cells in a microfluidic chip show that the fitness cost of gene amplifications and their intrinsic instability drives their rapid loss in the absence of selection. Using a common antibiotic susceptibility test, we demonstrate that this test strongly underestimates the occurrence of heteroresistance in clinical isolates.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Drug Resistance, Multiple, Bacterial/genetics , Escherichia coli Infections/diagnosis , Escherichia coli/growth & development , Salmonella Infections/diagnosis , Salmonella enterica/growth & development , Escherichia coli/drug effects , Escherichia coli/isolation & purification , Escherichia coli Infections/genetics , Escherichia coli Infections/microbiology , Gene Expression Regulation, Bacterial , Humans , Microbial Sensitivity Tests , Salmonella Infections/genetics , Salmonella Infections/microbiology , Salmonella enterica/drug effects , Salmonella enterica/isolation & purification
6.
Proc Natl Acad Sci U S A ; 118(9)2021 03 02.
Article in English | MEDLINE | ID: mdl-33619089

ABSTRACT

The spread of antibiotic resistance is turning many of the currently used antibiotics less effective against common infections. To address this public health challenge, it is critical to enhance our understanding of the mechanisms of action of these compounds. Aminoglycoside drugs bind the bacterial ribosome, and decades of results from in vitro biochemical and structural approaches suggest that these drugs disrupt protein synthesis by inhibiting the ribosome's translocation on the messenger RNA, as well as by inducing miscoding errors. So far, however, we have sparse information about the dynamic effects of these compounds on protein synthesis inside the cell. In the present study, we measured the effect of the aminoglycosides apramycin, gentamicin, and paromomycin on ongoing protein synthesis directly in live Escherichia coli cells by tracking the binding of dye-labeled transfer RNAs to ribosomes. Our results suggest that the drugs slow down translation elongation two- to fourfold in general, and the number of elongation cycles per initiation event seems to decrease to the same extent. Hence, our results imply that none of the drugs used in this study cause severe inhibition of translocation.


Subject(s)
Aminoglycosides/pharmacology , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/pharmacology , Anti-Bacterial Agents/pharmacology , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli/metabolism , Microscopy, Fluorescence , Molecular Imaging/methods , RNA, Transfer/genetics , Ribosomes/metabolism , Single-Cell Analysis/methods
7.
EMBO Rep ; 22(2): e50218, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33369848

ABSTRACT

Cell signalling governs cellular behaviour and is therefore subject to tight spatiotemporal regulation. Signalling output is modulated by specialized cell membranes and vesicles which contain unique combinations of lipids and proteins. The phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2 ), an important component of the plasma membrane as well as other subcellular membranes, is involved in multiple processes, including signalling. However, which enzymes control the turnover of non-plasma membrane PI(4,5)P2 , and their impact on cell signalling and function at the organismal level are unknown. Here, we identify Paladin as a vascular PI(4,5)P2 phosphatase regulating VEGFR2 endosomal signalling and angiogenesis. Paladin is localized to endosomal and Golgi compartments and interacts with vascular endothelial growth factor receptor 2 (VEGFR2) in vitro and in vivo. Loss of Paladin results in increased internalization of VEGFR2, over-activation of extracellular regulated kinase 1/2, and hypersprouting of endothelial cells in the developing retina of mice. These findings suggest that inhibition of Paladin, or other endosomal PI(4,5)P2 phosphatases, could be exploited to modulate VEGFR2 signalling and angiogenesis, when direct and full inhibition of the receptor is undesirable.


Subject(s)
Neovascularization, Physiologic , Phosphoinositide Phosphatases , Phosphoprotein Phosphatases , Vascular Endothelial Growth Factor Receptor-2 , Animals , Endothelial Cells/metabolism , Mice , Phosphatidylinositol 4,5-Diphosphate , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
8.
Nat Methods ; 17(1): 86-92, 2020 01.
Article in English | MEDLINE | ID: mdl-31740817

ABSTRACT

Our ability to connect genotypic variation to biologically important phenotypes has been seriously limited by the gap between live-cell microscopy and library-scale genomic engineering. Here, we show how in situ genotyping of a library of strains after time-lapse imaging in a microfluidic device overcomes this problem. We determine how 235 different CRISPR interference knockdowns impact the coordination of the replication and division cycles of Escherichia coli by monitoring the location of replication forks throughout on average >500 cell cycles per knockdown. Subsequent in situ genotyping allows us to map each phenotype distribution to a specific genetic perturbation to determine which genes are important for cell cycle control. The single-cell time-resolved assay allows us to determine the distribution of single-cell growth rates, cell division sizes and replication initiation volumes. The technology presented in this study enables genome-scale screens of most live-cell microscopy assays.


Subject(s)
CRISPR-Cas Systems , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Image Processing, Computer-Assisted/methods , Metabolic Engineering/methods , Microfluidic Analytical Techniques/methods , Microscopy/methods , Cell Cycle , DNA Replication , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/genetics , Gene Expression Regulation, Bacterial , Gene Library , Genotype , Phenotype
9.
Genome Med ; 10(1): 2, 2018 01 04.
Article in English | MEDLINE | ID: mdl-29301589

ABSTRACT

BACKGROUND: The Ras pathway genes KRAS, BRAF, or ERBBs have somatic mutations in ~ 60% of human colorectal carcinomas. At present, it is unknown whether the remaining cases lack mutations activating the Ras pathway or whether they have acquired mutations in genes hitherto unknown to belong to the pathway. METHODS: To address the second possibility and extend the compendium of Ras pathway genes, we used genome-wide transposon mutagenesis of two human colorectal cancer cell systems deprived of their activating KRAS or BRAF allele to identify genes enabling growth in low glucose, a Ras pathway phenotype, when targeted. RESULTS: Of the 163 recurrently targeted genes in the two different genetic backgrounds, one-third were known cancer genes and one-fifth had links to the EGFR/Ras/MAPK pathway. When compared to cancer genome sequencing datasets, nine genes also mutated in human colorectal cancers were identified. Among these, stable knockdown of FOXO3, NCOA3, and TCF7L2 restored growth in low glucose but reduced MEK/MAPK phosphorylation, reduced anchorage-independent growth, and modulated expressions of GLUT1 and Ras pathway related proteins. Knockdown of NCOA3 and FOXO3 significantly decreased the sensitivity to cetuximab of KRAS mutant but not wild-type cells. CONCLUSIONS: This work establishes a proof-of-concept that human cell-based genome-wide forward genetic screens can assign genes to pathways with clinical importance in human colorectal cancer.


Subject(s)
Colorectal Neoplasms/genetics , Forkhead Box Protein O3/genetics , Genetic Testing , Genome, Human , Nuclear Receptor Coactivator 3/genetics , Signal Transduction/genetics , Transcription Factor 7-Like 2 Protein/genetics , ras Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cetuximab/pharmacology , Cetuximab/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , DNA Transposable Elements/genetics , Drug Resistance, Neoplasm/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Forkhead Box Protein O3/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Humans , Nuclear Receptor Coactivator 3/metabolism , Phenotype , Phosphorylation/drug effects , Proteomics , RNA, Small Interfering/metabolism , Transcription Factor 7-Like 2 Protein/metabolism
10.
Oncotarget ; 8(58): 98646-98659, 2017 Nov 17.
Article in English | MEDLINE | ID: mdl-29228717

ABSTRACT

The chromatin modifier PRDM2/RIZ1 is inactivated by mutation in several forms of cancer and is a putative tumor suppressor gene. Frameshift mutations in the C-terminal region of PRDM2, affecting (A)8 or (A)9 repeats within exon 8, are found in one third of colorectal cancers with microsatellite instability, but the contribution of these mutations to colorectal tumorigenesis is unknown. To model somatic mutations in microsatellite unstable tumors, we devised a general approach to perform genome editing while stabilizing the mutated nucleotide repeat. We then engineered isogenic cell systems where the PRDM2 c.4467delA mutation in human HCT116 colorectal cancer cells was corrected to wild-type by genome editing. Restored PRDM2 increased global histone 3 lysine 9 dimethylation and reduced migration, anchorage-independent growth and tumor growth in vivo. Gene set enrichment analysis revealed regulation of several hallmark cancer pathways, particularly of epithelial-to-mesenchymal transition (EMT), with VIM being the most significantly regulated gene. These observations provide direct evidence that PRDM2 c.4467delA is a driver mutation in colorectal cancer and confirms PRDM2 as a cancer gene, pointing to regulation of EMT as a central aspect of its tumor suppressive action.

11.
Mol Syst Biol ; 13(10): 947, 2017 10 17.
Article in English | MEDLINE | ID: mdl-29042431

ABSTRACT

In this work, we present a proof-of-principle experiment that extends advanced live cell microscopy to the scale of pool-generated strain libraries. We achieve this by identifying the genotypes for individual cells in situ after a detailed characterization of the phenotype. The principle is demonstrated by single-molecule fluorescence time-lapse imaging of Escherichia coli strains harboring barcoded plasmids that express a sgRNA which suppresses different genes in the E. coli genome through dCas9 interference. In general, the method solves the problem of characterizing complex dynamic phenotypes for diverse genetic libraries of cell strains. For example, it allows screens of how changes in regulatory or coding sequences impact the temporal expression, location, or function of a gene product, or how the altered expression of a set of genes impacts the intracellular dynamics of a labeled reporter.


Subject(s)
Escherichia coli/classification , Escherichia coli/genetics , Genotyping Techniques/methods , Escherichia coli Proteins/genetics , Gene Library , Genotype , Microfluidic Analytical Techniques , Phenotype , RNA, Guide, Kinetoplastida/genetics , Single Molecule Imaging/methods
12.
Clin Cancer Res ; 22(24): 6217-6227, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-26957556

ABSTRACT

PURPOSE: To identify resistance mechanisms for the chemotherapeutic drug fludarabine in chronic lymphocytic leukemia (CLL), as innate and acquired resistance to fludarabine-based chemotherapy represents a major challenge for long-term disease control. EXPERIMENTAL DESIGN: We used piggyBac transposon-mediated mutagenesis, combined with next-generation sequencing, to identify genes that confer resistance to fludarabine in a human CLL cell line. RESULTS: In total, this screen identified 782 genes with transposon integrations in fludarabine-resistant pools of cells. One of the identified genes is a known resistance mediator DCK (deoxycytidine kinase), which encodes an enzyme that is essential for the phosphorylation of the prodrug to the active metabolite. BMP2K, a gene not previously linked to CLL, was also identified as a modulator of response to fludarabine. In addition, 10 of 782 transposon-targeted genes had previously been implicated in treatment resistance based on somatic mutations seen in patients refractory to fludarabine-based therapy. Functional characterization of these genes supported a significant role for ARID5B and BRAF in fludarabine sensitivity. Finally, pathway analysis of transposon-targeted genes and RNA-seq profiling of fludarabine-resistant cells suggested deregulated MAPK signaling as involved in mediating drug resistance in CLL. CONCLUSIONS: To our knowledge, this is the first forward genetic screen for chemotherapy resistance in CLL. The screen pinpointed novel genes and pathways involved in fludarabine resistance along with previously known resistance mechanisms. Transposon screens can therefore aid interpretation of cancer genome sequencing data in the identification of genes modifying sensitivity to chemotherapy. Clin Cancer Res; 22(24); 6217-27. ©2016 AACR.


Subject(s)
DNA Transposable Elements/genetics , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Mutagenesis/genetics , Vidarabine/analogs & derivatives , Antineoplastic Agents/pharmacology , Humans , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Transcription Factors/metabolism , Tumor Cells, Cultured , Vidarabine/pharmacology
13.
J Exp Med ; 212(6): 833-43, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25987724

ABSTRACT

NF-κB is constitutively activated in chronic lymphocytic leukemia (CLL); however, the implicated molecular mechanisms remain largely unknown. Thus, we performed targeted deep sequencing of 18 core complex genes within the NF-κB pathway in a discovery and validation CLL cohort totaling 315 cases. The most frequently mutated gene was NFKBIE (21/315 cases; 7%), which encodes IκBε, a negative regulator of NF-κB in normal B cells. Strikingly, 13 of these cases carried an identical 4-bp frameshift deletion, resulting in a truncated protein. Screening of an additional 377 CLL cases revealed that NFKBIE aberrations predominated in poor-prognostic patients and were associated with inferior outcome. Minor subclones and/or clonal evolution were also observed, thus potentially linking this recurrent event to disease progression. Compared with wild-type patients, NFKBIE-deleted cases showed reduced IκBε protein levels and decreased p65 inhibition, along with increased phosphorylation and nuclear translocation of p65. Considering the central role of B cell receptor (BcR) signaling in CLL pathobiology, it is notable that IκBε loss was enriched in aggressive cases with distinctive stereotyped BcR, likely contributing to their poor prognosis, and leading to an altered response to BcR inhibitors. Because NFKBIE deletions were observed in several other B cell lymphomas, our findings suggest a novel common mechanism of NF-κB deregulation during lymphomagenesis.


Subject(s)
Gene Expression Regulation, Leukemic , I-kappa B Kinase/physiology , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , NF-kappa B/metabolism , Cell Nucleus/metabolism , Cell Survival , Chromosome Aberrations , Cohort Studies , Cytoplasm/metabolism , DNA Mutational Analysis , Frameshift Mutation , Gene Deletion , Gene Expression Profiling , Humans , I-kappa B Kinase/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell, Marginal Zone/metabolism , Lymphoma, Mantle-Cell/metabolism , Oligonucleotide Array Sequence Analysis , Receptors, Antigen, B-Cell/metabolism , Signal Transduction , Treatment Outcome
14.
Dev Cell ; 23(3): 587-99, 2012 Sep 11.
Article in English | MEDLINE | ID: mdl-22975327

ABSTRACT

Angiogenesis, the process by which new blood vessels arise from preexisting ones, is critical for embryonic development and is an integral part of many disease processes. Recent studies have provided detailed information on how angiogenic sprouts initiate, elongate, and branch, but less is known about how these processes cease. Here, we show that S1PR1, a receptor for the blood-borne bioactive lipid sphingosine-1-phosphate (S1P), is critical for inhibition of angiogenesis and acquisition of vascular stability. Loss of S1PR1 leads to increased endothelial cell sprouting and the formation of ectopic vessel branches. Conversely, S1PR1 signaling inhibits angiogenic sprouting and enhances cell-to-cell adhesion. This correlates with inhibition of vascular endothelial growth factor-A (VEGF-A)-induced signaling and stabilization of vascular endothelial (VE)-cadherin localization at endothelial junctions. Our data suggest that S1PR1 signaling acts as a vascular-intrinsic stabilization mechanism, protecting developing blood vessels against aberrant angiogenic responses.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Neovascularization, Physiologic , Receptors, Lysosphingolipid/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Cells, Cultured , Endothelial Cells/metabolism , Humans , Mice , Mice, Knockout , Mice, Transgenic , Receptors, Lysosphingolipid/deficiency , Sphingosine-1-Phosphate Receptors , Zebrafish
15.
Dev Dyn ; 241(4): 770-86, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22354871

ABSTRACT

BACKGROUND: Angiogenesis is implicated in many pathological conditions. The role of the proteins involved remains largely unknown, and few vascular-specific drug targets have been discovered. Previously, in a screen for angiogenesis regulators, we identified Paladin (mouse: X99384, human: KIAA1274), a protein containing predicted S/T/Y phosphatase domains. RESULTS: We present a mouse knockout allele for Paladin with a ß-galactosidase reporter, which in combination with Paladin antibodies demonstrate that Paladin is expressed in the vasculature. During mouse embryogenesis, Paladin is primarily expressed in capillary and venous endothelial cells. In adult mice Paladin is predominantly expressed in arterial pericytes and vascular smooth muscle cells. Paladin also displays vascular-restricted expression in human brain, astrocytomas, and glioblastomas. CONCLUSIONS: Paladin, a novel putative phosphatase, displays a dynamic expression pattern in the vasculature. During embryonic stages it is broadly expressed in endothelial cells, while in the adult it is selectively expressed in arterial smooth muscle cells.


Subject(s)
Blood Vessels/cytology , Blood Vessels/physiology , Endothelial Cells , Muscle, Smooth, Vascular , Phosphoprotein Phosphatases/physiology , Phosphoric Monoester Hydrolases/physiology , Animals , Blood Vessels/embryology , Cell Differentiation/physiology , Endothelial Cells/physiology , Humans , Mice , Muscle, Smooth, Vascular/embryology , Muscle, Smooth, Vascular/physiology , Neovascularization, Physiologic/physiology , Pericytes/cytology , Pericytes/physiology
16.
PLoS Genet ; 7(9): e1002285, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21949658

ABSTRACT

PMEL is an amyloidogenic protein that appears to be exclusively expressed in pigment cells and forms intralumenal fibrils within early stage melanosomes upon which eumelanins deposit in later stages. PMEL is well conserved among vertebrates, and allelic variants in several species are associated with reduced levels of eumelanin in epidermal tissues. However, in most of these cases it is not clear whether the allelic variants reflect gain-of-function or loss-of-function, and no complete PMEL loss-of-function has been reported in a mammal. Here, we have created a mouse line in which the Pmel gene has been inactivated (Pmel⁻/⁻). These mice are fully viable, fertile, and display no obvious developmental defects. Melanosomes within Pmel⁻/⁻ melanocytes are spherical in contrast to the oblong shape present in wild-type animals. This feature was documented in primary cultures of skin-derived melanocytes as well as in retinal pigment epithelium cells and in uveal melanocytes. Inactivation of Pmel has only a mild effect on the coat color phenotype in four different genetic backgrounds, with the clearest effect in mice also carrying the brown/Tyrp1 mutation. This phenotype, which is similar to that observed with the spontaneous silver mutation in mice, strongly suggests that other previously described alleles in vertebrates with more striking effects on pigmentation are dominant-negative mutations. Despite a mild effect on visible pigmentation, inactivation of Pmel led to a substantial reduction in eumelanin content in hair, which demonstrates that PMEL has a critical role for maintaining efficient epidermal pigmentation.


Subject(s)
Melanins/biosynthesis , Melanosomes/metabolism , Pigmentation/genetics , gp100 Melanoma Antigen/genetics , gp100 Melanoma Antigen/metabolism , Alleles , Animals , Cells, Cultured , Epidermal Cells , Epidermis/metabolism , Hair Color/genetics , HeLa Cells , Humans , Melanins/genetics , Melanosomes/ultrastructure , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Mutation , Oxidoreductases/metabolism , Phenotype , Skin/metabolism
17.
Mol Genet Genomics ; 280(3): 233-48, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18612650

ABSTRACT

We have screened a complete collection of yeast knockout mutants for sensitivity to monensin, an ionophore that interferes with intracellular transport. A total of 63 sensitive strains were found. Most of the strains were deleted for genes involved in post-Golgi traffic, with an emphasis on vacuolar biogenesis. A high correlation was thus seen with VPS and VAM genes, but there were also significant differences between the three sets of genes. A weaker correlation was seen with sensitivity to NaCl, in particular rate of growth effects. Interestingly, all 14 genes encoding subunits of the vacuolar H(+)-ATPase (V-ATPase) were absent in our screen, even though they appeared in the VPS or VAM screens. All monensin-sensitive mutants that could be tested interact synthetically with a deletion of the A subunit of the V-ATPase, Vma1. Synthetic lethality was limited to mutations affecting endocytosis or retrograde transport to Golgi. In addition, vma1 was epistatic over the monensin sensitivity of vacuolar transport mutants, but not endocytosis mutants. Deletions of the two isoforms of the V-ATPase a subunit, Vph1 and Stv1 had opposite effects on the monensin sensitivity of a ypt7 mutant. These findings are consistent with a model where monensin inhibits growth by interfering with the maintenance of an acidic pH in the late secretory pathway. The synthetic lethality of vma1 with mutations affecting retrograde transport to the Golgi further suggests that it is in the late Golgi that a low pH must be maintained.


Subject(s)
Genomics , Golgi Apparatus/metabolism , Monensin/pharmacology , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Vacuolar Proton-Translocating ATPases/metabolism , Vacuoles/enzymology , Biological Transport/drug effects , Cathepsin A/metabolism , Cytoskeleton/drug effects , Cytoskeleton/genetics , Endocytosis/drug effects , Endocytosis/genetics , Endosomes/drug effects , Endosomes/genetics , Epistasis, Genetic , Gene Expression Regulation, Fungal/drug effects , Genes, Fungal , Golgi Apparatus/genetics , Lipids/biosynthesis , Models, Biological , Mutation/genetics , Phosphatidylinositols/metabolism , Protein Processing, Post-Translational/drug effects , Saccharomyces cerevisiae/enzymology , Signal Transduction/drug effects , Signal Transduction/genetics , Sodium Chloride/pharmacology , Vacuolar Proton-Translocating ATPases/chemistry , Vacuolar Proton-Translocating ATPases/genetics , Vacuoles/drug effects , Vacuoles/genetics
18.
Growth Factors ; 24(3): 184-96, 2006 Sep.
Article in English | MEDLINE | ID: mdl-17079202

ABSTRACT

We have previously shown that platelet-derived growth factor AA (PDGF-AA) stimulates the expansion of neuronal progenitors from neural stem cells, but is unable to replace fibroblast-growth factor 2 (FGF-2) as a stem cell mitogen. In the present study, we compared gene expression in neural stem cells that were grown in the presence of FGF-2 and in cells cultured with PDGF-AA or in the absence of growth factor, which induces differentiation. The genetic program elicited by PDGF-AA (156 significantly regulated genes) was not unique, but an intermediate between the ones of FGF-2-cultured stem cells and differentiated cells. These observations are compatible with the hypothesis that PDGF-AA induces a partial differentiation of neural stem cells, which retain the ability to proliferate, rather than acting solely as an instructing agent for neuronal differentiation. Finally, the transcriptional signature of stem cells grown with FGF-2 included a large number of genes over-expressed in gliomas and a core set of conserved genes periodically expressed during the eukaryote cell cycle.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/metabolism , Fibroblast Growth Factor 2/physiology , Neocortex/embryology , Platelet-Derived Growth Factor/physiology , Animals , Cell Cycle/physiology , Cell Proliferation , Gene Expression Profiling , Neuroglia/cytology , Neuroglia/metabolism , Neurons/cytology , Neurons/metabolism , Rats , Rats, Sprague-Dawley
19.
Exp Cell Res ; 301(2): 201-10, 2004 Dec 10.
Article in English | MEDLINE | ID: mdl-15530856

ABSTRACT

Migration of neural cells to their final positions is crucial for the correct formation of the central nervous system. Several extrinsic factors are known to be involved in the regulation of neural migration. We asked if stem cell factor (SCF), well known as a chemoattractant and survival factor in the hematopoietic lineage, could elicit similar responses in neural stem cells. For that purpose, a microchemotaxis assay was used to study the effect of SCF on migration of neural stem cells from the embryonic rat cortex. Our results show that SCF-induced chemotaxis and that specific antibodies to SCF or tyrosine kinase inhibitors abolished the migratory response. The SCF-receptor, Kit, was expressed in neural stem cells and in their differentiated progeny. We also show that SCF is a survival factor, but not a mitogen or a differentiation factor for neural stem cells. These data suggest a role for SCF in cell migration and survival in the developing cortex.


Subject(s)
Cerebral Cortex/cytology , Chemotactic Factors/physiology , Stem Cell Factor/physiology , Stem Cells/cytology , Animals , Cell Differentiation , Cell Survival/drug effects , Cells, Cultured , Chemotactic Factors/pharmacology , Chemotaxis/drug effects , Embryo, Mammalian/cytology , Mice , Proto-Oncogene Proteins c-kit/biosynthesis , Rats , Rats, Sprague-Dawley , Stem Cell Factor/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...