Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
BMC Cancer ; 24(1): 369, 2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38519889

ABSTRACT

CD13 (APN) is an Alanyl-Aminopeptidase with diverse functions. The role of CD13 for gliomas is still unknown. In this study, data of glioma patients obtained by TCGA and CGGA databases were used to evaluate the survival rate and prognostic value of CD13 expression level. Protein expression of CD13 was confirmed by immunofluorescence staining of fresh patient tissues. Eight human glioblastoma cell lines were studied by RT-PCR, Western Blot, immunofluorescence staining and flow cytometry to define CD13 expression. Cell lines with different CD13 expression status were treated with a CD13 inhibitor, bestatin, and examined by MTT, scratch and colony formation assaysas well as by apoptosis assay and Western Blots. Bioinformatics analysis indicated that patients with high expression of CD13 had poor survival and prognosis. Additionally, CD13 protein expression was positively associated with clinical malignant characteristics. Investigated glioblastoma cell lines showed distinct expression levels and subcellular localization of CD13 with intracellular enrichment. Bestatin treatment reduced proliferation, migration and colony formation of glioma cells in a CD13-dependent manner while apoptosis was increased. In summary, CD13 has an impact on glioma patient survival and is important for the main function of specific glioma cells.


Subject(s)
Glioblastoma , Glioma , Humans , Apoptosis , CD13 Antigens/genetics , CD13 Antigens/metabolism , Cell Line, Tumor , Glioblastoma/genetics , Glioma/genetics
2.
Pain ; 164(8): 1718-1733, 2023 Aug 01.
Article in English | MEDLINE | ID: mdl-36727909

ABSTRACT

ABSTRACT: Induced pluripotent stem cells (iPSCs) have enabled the generation of various difficult-to-access cell types such as human nociceptors. A key challenge associated with human iPSC-derived nociceptors (hiPSCdNs) is their prolonged functional maturation. While numerous studies have addressed the expression of classic neuronal markers and ion channels in hiPSCdNs, the temporal development of key signaling cascades regulating nociceptor activity has remained largely unexplored. In this study, we used an immunocytochemical high-content imaging approach alongside electrophysiological staging to assess metabotropic and ionotropic signaling of large scale-generated hiPSCdNs across 70 days of in vitro differentiation. During this period, the resting membrane potential became more hyperpolarized, while rheobase, action potential peak amplitude, and membrane capacitance increased. After 70 days, hiPSCdNs exhibited robust physiological responses induced by GABA, pH shift, ATP, and capsaicin. Direct activation of protein kinase A type II (PKA-II) through adenylyl cyclase stimulation with forskolin resulted in PKA-II activation at all time points. Depolarization-induced activation of PKA-II emerged after 35 days of differentiation. However, effective inhibition of forskolin-induced PKA-II activation by opioid receptor agonists required 70 days of in vitro differentiation. Our results identify a pronounced time difference between early expression of functionally important ion channels and emergence of regulatory metabotropic sensitizing and desensitizing signaling only at advanced stages of in vitro cultivation, suggesting an independent regulation of ionotropic and metabotropic signaling. These data are relevant for devising future studies into the development and regulation of human nociceptor function and for defining time windows suitable for hiPSCdN-based drug discovery.


Subject(s)
Induced Pluripotent Stem Cells , Humans , Analgesics, Opioid , Colforsin/pharmacology , Nociception , Sensory Receptor Cells , Ion Channels
3.
Nat Neurosci ; 25(2): 168-179, 2022 02.
Article in English | MEDLINE | ID: mdl-34931070

ABSTRACT

Bacterial products can act on neurons to alter signaling and function. In the present study, we found that dorsal root ganglion (DRG) sensory neurons are enriched for ANTXR2, the high-affinity receptor for anthrax toxins. Anthrax toxins are composed of protective antigen (PA), which binds to ANTXR2, and the protein cargoes edema factor (EF) and lethal factor (LF). Intrathecal administration of edema toxin (ET (PA + EF)) targeted DRG neurons and induced analgesia in mice. ET inhibited mechanical and thermal sensation, and pain caused by formalin, carrageenan or nerve injury. Analgesia depended on ANTXR2 expressed by Nav1.8+ or Advillin+ neurons. ET modulated protein kinase A signaling in mouse sensory and human induced pluripotent stem cell-derived sensory neurons, and attenuated spinal cord neurotransmission. We further engineered anthrax toxins to introduce exogenous protein cargoes, including botulinum toxin, into DRG neurons to silence pain. Our study highlights interactions between a bacterial toxin and nociceptors, which may lead to the development of new pain therapeutics.


Subject(s)
Anthrax , Bacillus anthracis , Bacterial Toxins , Induced Pluripotent Stem Cells , Animals , Anthrax/microbiology , Anthrax/therapy , Bacillus anthracis/metabolism , Bacterial Toxins/metabolism , Ganglia, Spinal/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Nociceptors/metabolism , Pain , Receptors, Peptide/metabolism
5.
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
6.
Circ Res ; 128(3): 363-382, 2021 02 05.
Article in English | MEDLINE | ID: mdl-33301355

ABSTRACT

RATIONALE: Cerebrovascular function is critical for brain health, and endogenous vascular protective pathways may provide therapeutic targets for neurological disorders. S1P (Sphingosine 1-phosphate) signaling coordinates vascular functions in other organs, and S1P1 (S1P receptor-1) modulators including fingolimod show promise for the treatment of ischemic and hemorrhagic stroke. However, S1P1 also coordinates lymphocyte trafficking, and lymphocytes are currently viewed as the principal therapeutic target for S1P1 modulation in stroke. OBJECTIVE: To address roles and mechanisms of engagement of endothelial cell S1P1 in the naive and ischemic brain and its potential as a target for cerebrovascular therapy. METHODS AND RESULTS: Using spatial modulation of S1P provision and signaling, we demonstrate a critical vascular protective role for endothelial S1P1 in the mouse brain. With an S1P1 signaling reporter, we reveal that abluminal polarization shields S1P1 from circulating endogenous and synthetic ligands after maturation of the blood-neural barrier, restricting homeostatic signaling to a subset of arteriolar endothelial cells. S1P1 signaling sustains hallmark endothelial functions in the naive brain and expands during ischemia by engagement of cell-autonomous S1P provision. Disrupting this pathway by endothelial cell-selective deficiency in S1P production, export, or the S1P1 receptor substantially exacerbates brain injury in permanent and transient models of ischemic stroke. By contrast, profound lymphopenia induced by loss of lymphocyte S1P1 provides modest protection only in the context of reperfusion. In the ischemic brain, endothelial cell S1P1 supports blood-brain barrier function, microvascular patency, and the rerouting of blood to hypoperfused brain tissue through collateral anastomoses. Boosting these functions by supplemental pharmacological engagement of the endothelial receptor pool with a blood-brain barrier penetrating S1P1-selective agonist can further reduce cortical infarct expansion in a therapeutically relevant time frame and independent of reperfusion. CONCLUSIONS: This study provides genetic evidence to support a pivotal role for the endothelium in maintaining perfusion and microvascular patency in the ischemic penumbra that is coordinated by S1P signaling and can be harnessed for neuroprotection with blood-brain barrier-penetrating S1P1 agonists.


Subject(s)
Blood-Brain Barrier/metabolism , Cerebral Arteries/metabolism , Endothelial Cells/metabolism , Infarction, Middle Cerebral Artery/metabolism , Ischemic Attack, Transient/metabolism , Ischemic Stroke/metabolism , Lysophospholipids/metabolism , Sphingosine-1-Phosphate Receptors/metabolism , Sphingosine/analogs & derivatives , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/pathology , Blood-Brain Barrier/physiopathology , Cerebral Arteries/drug effects , Cerebral Arteries/pathology , Cerebral Arteries/physiopathology , Cerebrovascular Circulation , Disease Models, Animal , Endothelial Cells/pathology , Female , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Infarction, Middle Cerebral Artery/prevention & control , Ischemic Attack, Transient/pathology , Ischemic Attack, Transient/physiopathology , Ischemic Attack, Transient/prevention & control , Ischemic Stroke/pathology , Ischemic Stroke/physiopathology , Ischemic Stroke/prevention & control , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Microcirculation , Neuroprotective Agents/pharmacology , Signal Transduction , Sphingosine/metabolism , Sphingosine-1-Phosphate Receptors/agonists , Sphingosine-1-Phosphate Receptors/genetics , Vascular Patency
7.
Elife ; 92020 02 24.
Article in English | MEDLINE | ID: mdl-32091396

ABSTRACT

Despite the medical importance of G protein-coupled receptors (GPCRs), in vivo cellular heterogeneity of GPCR signaling and downstream transcriptional responses are not understood. We report the comprehensive characterization of transcriptomes (bulk and single-cell) and chromatin domains regulated by sphingosine 1-phosphate receptor-1 (S1PR1) in adult mouse aortic endothelial cells. First, S1PR1 regulates NFκB and nuclear glucocorticoid receptor pathways to suppress inflammation-related mRNAs. Second, S1PR1 signaling in the heterogenous endothelial cell (EC) subtypes occurs at spatially-distinct areas of the aorta. For example, a transcriptomically distinct arterial EC population at vascular branch points (aEC1) exhibits ligand-independent S1PR1/ß-arrestin coupling. In contrast, circulatory S1P-dependent S1PR1/ß-arrestin coupling was observed in non-branch point aEC2 cells that exhibit an inflammatory gene expression signature. Moreover, S1P/S1PR1 signaling regulates the expression of lymphangiogenic and inflammation-related transcripts in an adventitial lymphatic EC (LEC) population in a ligand-dependent manner. These insights add resolution to existing concepts of endothelial heterogeneity, GPCR signaling and S1P biology.


Subject(s)
Aorta/metabolism , Endothelium, Lymphatic/metabolism , Endothelium, Vascular/metabolism , Lysophospholipids/metabolism , Sphingosine/analogs & derivatives , Transcriptome , Animals , Green Fluorescent Proteins/metabolism , Mice , Mice, Transgenic , Sequence Analysis, RNA/methods , Signal Transduction , Single-Cell Analysis/methods , Sphingosine/metabolism , Sphingosine-1-Phosphate Receptors/genetics , Sphingosine-1-Phosphate Receptors/metabolism , beta-Arrestins/metabolism
8.
Blood Adv ; 3(11): 1702-1713, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31171507

ABSTRACT

The bioactive lipid mediator sphingosine 1-phosphate (S1P) was recently assigned critical roles in platelet biology: whereas S1P1 receptor-mediated S1P gradient sensing was reported to be essential for directing proplatelet extensions from megakaryocytes (MKs) toward bone marrow sinusoids, MK sphingosine kinase 2 (Sphk2)-derived S1P was reported to further promote platelet shedding through receptor-independent intracellular actions, and platelet aggregation through S1P1 Yet clinical use of S1P pathway modulators including fingolimod has not been associated with risk of bleeding or thrombosis. We therefore revisited the role of S1P in platelet biology in mice. Surprisingly, no reduction in platelet counts was observed when the vascular S1P gradient was ablated by impairing S1P provision to plasma or S1P degradation in interstitial fluids, nor when gradient sensing was impaired by S1pr1 deletion selectively in MKs. Moreover, S1P1 expression and signaling were both undetectable in mature MKs in situ, and MK S1pr1 deletion did not affect platelet aggregation or spreading. When S1pr1 deletion was induced in hematopoietic progenitor cells, platelet counts were instead significantly elevated. Isolated global Sphk2 deficiency was associated with thrombocytopenia, but this was not replicated by MK-restricted Sphk2 deletion and was reversed by compound deletion of either Sphk1 or S1pr2, suggesting that this phenotype arises from increased S1P export and S1P2 activation secondary to redistribution of sphingosine to Sphk1. Consistent with clinical observations, we thus observe no essential role for S1P1 in facilitating platelet production or activation. Instead, S1P restricts megakaryopoiesis through S1P1, and can further suppress thrombopoiesis through S1P2 when aberrantly secreted in the hematopoietic niche.


Subject(s)
Blood Platelets/metabolism , Lysophospholipids/metabolism , Megakaryocytes/metabolism , Signal Transduction , Sphingosine/analogs & derivatives , Stem Cell Niche , Thrombopoiesis , Animals , Blood Platelets/cytology , Lysophospholipids/genetics , Megakaryocytes/cytology , Mice , Mice, Knockout , Sphingosine/genetics , Sphingosine/metabolism , Sphingosine-1-Phosphate Receptors/genetics , Sphingosine-1-Phosphate Receptors/metabolism
9.
Sci Rep ; 7(1): 15453, 2017 11 13.
Article in English | MEDLINE | ID: mdl-29133847

ABSTRACT

Paladin (Pald1, mKIAA1274 or x99384) was identified in screens for vascular-specific genes and is a putative phosphatase. Paladin has also been proposed to be involved in various biological processes such as insulin signaling, innate immunity and neural crest migration. To determine the role of paladin we have now characterized the Pald1 knock-out mouse in a broad array of behavioral, physiological and biochemical tests. Here, we show that female, but not male, Pald1 heterozygous and homozygous knock-out mice display an emphysema-like histology with increased alveolar air spaces and impaired lung function with an obstructive phenotype. In contrast to many other tissues where Pald1 is restricted to the vascular compartment, Pald1 is expressed in both the epithelial and mesenchymal compartments of the postnatal lung. However, in Pald1 knock-out females, there is a specific increase in apoptosis and proliferation of endothelial cells, but not in non-endothelial cells. This results in a transient reduction of endothelial cells in the maturing lung. Our data suggests that Pald1 is required during lung vascular development and for normal function of the developing and adult lung in a sex-specific manner. To our knowledge, this is the first report of a sex-specific effect on endothelial cell apoptosis.


Subject(s)
Apoptosis/physiology , Emphysema/pathology , Endothelial Cells/pathology , Endothelium, Vascular/growth & development , Phosphoprotein Phosphatases/metabolism , Animals , Disease Models, Animal , Embryo, Mammalian , Emphysema/genetics , Female , Heterozygote , Humans , Lung/blood supply , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphoprotein Phosphatases/genetics , Sex Factors
10.
Sci Transl Med ; 8(335): 335ra56, 2016 04 20.
Article in English | MEDLINE | ID: mdl-27099175

ABSTRACT

In common with other chronic pain conditions, there is an unmet clinical need in the treatment of inherited erythromelalgia (IEM). TheSCN9Agene encoding the sodium channel Nav1.7 expressed in the peripheral nervous system plays a critical role in IEM. A gain-of-function mutation in this sodium channel leads to aberrant sensory neuronal activity and extreme pain, particularly in response to heat. Five patients with IEM were treated with a new potent and selective compound that blocked the Nav1.7 sodium channel resulting in a decrease in heat-induced pain in most of the patients. We derived induced pluripotent stem cell (iPSC) lines from four of five subjects and produced sensory neurons that emulated the clinical phenotype of hyperexcitability and aberrant responses to heat stimuli. When we compared the severity of the clinical phenotype with the hyperexcitability of the iPSC-derived sensory neurons, we saw a trend toward a correlation for individual mutations. The in vitro IEM phenotype was sensitive to Nav1.7 blockers, including the clinical test agent. Given the importance of peripherally expressed sodium channels in many pain conditions, our approach may have broader utility for a wide range of pain and sensory conditions.


Subject(s)
Erythromelalgia/drug therapy , Induced Pluripotent Stem Cells/cytology , Pain/drug therapy , Pain/metabolism , Phenyl Ethers/therapeutic use , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Sulfonamides/therapeutic use , Adult , Erythromelalgia/genetics , Female , Humans , Male , Mutation/genetics , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Sensory Receptor Cells/cytology
11.
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
12.
Nat Methods ; 9(4): 360-2, 2012 Feb 12.
Article in English | MEDLINE | ID: mdl-22327834

ABSTRACT

Whereas methods to comprehensively study cellular roles of protein-coding genes are available, techniques to systematically investigate long noncoding RNAs (lncRNAs), which have been implicated in diverse biological pathways, are limited. Here we report combined knockdown and localization analysis of noncoding RNAs (c-KLAN) that merges functional characterization and localization approaches to study lncRNAs. Using this technique we identified transcripts that regulate mouse embryonic stem cell identity.


Subject(s)
RNA Interference , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Animals , Embryonic Stem Cells/metabolism , Gene Knockdown Techniques , In Situ Hybridization, Fluorescence , Mice
13.
PLoS One ; 6(5): e19470, 2011 May 12.
Article in English | MEDLINE | ID: mdl-21589869

ABSTRACT

For self-renewal, embryonic stem cells (ESCs) require the expression of specific transcription factors accompanied by a particular chromosome organization to maintain a balance between pluripotency and the capacity for rapid differentiation. However, how transcriptional regulation is linked to chromosome organization in ESCs is not well understood. Here we show that the cohesin component RAD21 exhibits a functional role in maintaining ESC identity through association with the pluripotency transcriptional network. ChIP-seq analyses of RAD21 reveal an ESC specific cohesin binding pattern that is characterized by CTCF independent co-localization of cohesin with pluripotency related transcription factors Oct4, Nanog, Sox2, Esrrb and Klf4. Upon ESC differentiation, most of these binding sites disappear and instead new CTCF independent RAD21 binding sites emerge, which are enriched for binding sites of transcription factors implicated in early differentiation. Furthermore, knock-down of RAD21 causes expression changes that are similar to expression changes after Nanog depletion, demonstrating the functional relevance of the RAD21--pluripotency transcriptional network association. Finally, we show that Nanog physically interacts with the cohesin or cohesin interacting proteins STAG1 and WAPL further substantiating this association. Based on these findings we propose that a dynamic placement of cohesin by pluripotency transcription factors contributes to a chromosome organization supporting the ESC expression program.


Subject(s)
Embryonic Stem Cells/cytology , Nuclear Proteins/physiology , Phosphoproteins/physiology , Pluripotent Stem Cells/cytology , Transcription Factors/physiology , Animals , Binding Sites , Cell Cycle Proteins/physiology , Cells, Cultured , Chromatin Immunoprecipitation , Chromosomal Proteins, Non-Histone/physiology , DNA-Binding Proteins , Gene Expression Profiling , Homeodomain Proteins/physiology , Kruppel-Like Factor 4 , Mice , Nanog Homeobox Protein , Cohesins
14.
PLoS One ; 6(4): e18709, 2011 Apr 14.
Article in English | MEDLINE | ID: mdl-21533193

ABSTRACT

The Notch signaling pathway is essential for normal development due to its role in control of cell differentiation, proliferation and survival. It is also critically involved in tumorigenesis and cancer progression. A key enzyme in the activation of Notch signaling is the gamma-secretase protein complex and therefore, gamma-secretase inhibitors (GSIs)--originally developed for Alzheimer's disease--are now being evaluated in clinical trials for human malignancies. It is also clear that Notch plays an important role in angiogenesis driven by Vascular Endothelial Growth Factor A (VEGF-A)--a process instrumental for tumor growth and metastasis. The effect of GSIs on tumor vasculature has not been conclusively determined. Here we report that Compound X (CX), a GSI previously reported to potently inhibit Notch signaling in vitro and in vivo, promotes angiogenic sprouting in vitro and during developmental angiogenesis in mice. Furthermore, CX treatment suppresses tumor growth in a mouse model of renal carcinoma, leads to the formation of abnormal vessels and an increased tumor vascular density. Using a rabbit model of VEGF-A-driven angiogenesis in skeletal muscle, we demonstrate that CX treatment promotes abnormal blood vessel growth characterized by vessel occlusion, disrupted blood flow, and increased vascular leakage. Based on these findings, we propose a model for how GSIs and other Notch inhibitors disrupt tumor blood vessel perfusion, which might be useful for understanding this new class of anti-cancer agents.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Blood Vessels/growth & development , Enzyme Inhibitors/pharmacology , Neovascularization, Pathologic/prevention & control , Vascular Endothelial Growth Factor A/physiology , Animals , Humans , Mice , Rabbits
15.
Cell Stem Cell ; 4(5): 403-15, 2009 May 08.
Article in English | MEDLINE | ID: mdl-19345177

ABSTRACT

Pluripotent embryonic stem cells (ESCs) maintain self-renewal while ensuring a rapid response to differentiation cues. The identification of genes maintaining ESC identity is important to develop these cells for their potential therapeutic use. Here we report a genome-scale RNAi screen for a global survey of genes affecting ESC identity via alteration of Oct4 expression. Factors with the strongest effect on Oct4 expression included components of the Paf1 complex, a protein complex associated with RNA polymerase II. Using a combination of proteomics, expression profiling, and chromatin immunoprecipitation, we demonstrate that the Paf1C binds to promoters of key pluripotency genes, where it is required to maintain a transcriptionally active chromatin structure. The Paf1C is developmentally regulated and blocks ESC differentiation upon overexpression, and the knockdown in ESCs causes expression changes similar to Oct4 or Nanog depletions. We propose that the Paf1C plays an important role in maintaining ESC identity.


Subject(s)
Embryonic Stem Cells/cytology , Nuclear Proteins/metabolism , Octamer Transcription Factor-3/genetics , RNA Interference , Transcription Factors/metabolism , Animals , Cell Lineage , Cell Proliferation , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Embryonic Stem Cells/metabolism , Gene Regulatory Networks , Genome , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Nanog Homeobox Protein , Nuclear Proteins/genetics , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , RNA Polymerase II/genetics , RNA Polymerase II/metabolism
16.
Nat Methods ; 5(5): 409-15, 2008 May.
Article in English | MEDLINE | ID: mdl-18391959

ABSTRACT

The interpretation of genome sequences requires reliable and standardized methods to assess protein function at high throughput. Here we describe a fast and reliable pipeline to study protein function in mammalian cells based on protein tagging in bacterial artificial chromosomes (BACs). The large size of the BAC transgenes ensures the presence of most, if not all, regulatory elements and results in expression that closely matches that of the endogenous gene. We show that BAC transgenes can be rapidly and reliably generated using 96-well-format recombineering. After stable transfection of these transgenes into human tissue culture cells or mouse embryonic stem cells, the localization, protein-protein and/or protein-DNA interactions of the tagged protein are studied using generic, tag-based assays. The same high-throughput approach will be generally applicable to other model systems.


Subject(s)
Chromosomes, Artificial, Bacterial/genetics , Genomics/methods , Mammals/genetics , Mammals/metabolism , Proteins/metabolism , Transgenes/genetics , Animals , Anti-Bacterial Agents/pharmacology , Cell Line , Drug Resistance , Gene Expression Regulation , Gene Library , Genetic Engineering , Genome , Protein Array Analysis , Protein Binding , Protein Transport , Proteins/genetics
17.
Nat Biotechnol ; 25(6): 669-74, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17546028

ABSTRACT

The HIV-1 RNase H can be prematurely activated by oligodeoxynucleotides targeting the highly conserved polypurine tract required for second strand DNA synthesis. This inhibits retroviral replication in cell-free HIV particles and newly infected cells. Here we extend these studies to an in vivo model of retroviral replication. Mice that are chronically infected with the spleen focus-forming virus and treated with oligodeoxynucleotides that target the polypurine tract, exhibit either transient or long-term reductions in plasma virus titer, depending on the therapeutic regimen. Treatment prior to, during or shortly after infection can delay disease progression, increase survival rates and prevent viral infection. This strategy destroys viral RNA template in virus particles in serum as well as early retroviral replication intermediates in infected cells. As it targets events common to the replication cycle of all retroviruses, this approach may be broadly applicable to retroviruses of medical and agricultural importance.


Subject(s)
Gene Silencing , Gene Targeting/methods , HIV Infections/therapy , HIV Infections/virology , HIV-1/genetics , Retroviridae/genetics , Ribonuclease H/genetics , Animals , HIV Infections/genetics , Mice , NIH 3T3 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...