Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Front Neurosci ; 15: 621076, 2021.
Article in English | MEDLINE | ID: mdl-33841077

ABSTRACT

The ventricular-subventricular zone (V-SVZ) is the principal neurogenic niche in the adult mammalian forebrain. Neural stem/progenitor cell (NSPC) activity within the V-SVZ is controlled by numerous of extrinsic factors, whose downstream effects on NSPC proliferation, survival and differentiation are transduced via a limited number of intracellular signaling pathways. Here, we investigated the relationship between age-related changes in NSPC output and activity of signaling pathways downstream of the epidermal growth factor receptor (EGFR), a major regulator of NSPC activity. Biochemical experiments indicated that age-related decline of NSPC activity in vivo is accompanied by selective deficits amongst various EGFR-induced signal pathways within the V-SVZ niche. Pharmacological loss-of-function signaling experiments with cultured NSPCs revealed both overlap and selectivity in the biological functions modulated by the EGFR-induced PI3K/AKT, MEK/ERK and mTOR signaling modules. Specifically, while all three modules promoted EGFR-mediated NSPC proliferation, only mTOR contributed to NSPC survival and only MEK/ERK repressed NSPC differentiation. Using a gain-of-function in vivo genetic approach, we electroporated a constitutively active EGFR construct into a subpopulation of quiescent, EGFR-negative neural stem cells (qNSCs); this ectopic activation of EGFR signaling enabled qNSCs to divide in 3-month-old early adult mice, but not in mice at middle-age or carrying familial Alzheimer disease mutations. Thus, (i) individual EGFR-induced signaling pathways have dissociable effects on NSPC proliferation, survival, and differentiation, (ii) activation of EGFR signaling is sufficient to stimulate qNSC cell cycle entry during early adulthood, and (iii) the proliferative effects of EGFR-induced signaling are dominantly overridden by anti-proliferative signals associated with aging and Alzheimer's disease.

2.
Life Sci Alliance ; 3(7)2020 07.
Article in English | MEDLINE | ID: mdl-32482782

ABSTRACT

The ventricular epithelium of the adult forebrain is a heterogeneous cell population that is a source of both quiescent and activated neural stem cells (qNSCs and aNSCs, respectively). We genetically targeted a subset of ventricle-contacting, glial fibrillary acidic protein (GFAP)-expressing cells, to study their involvement in qNSC/aNSC-mediated adult neurogenesis. Ventricle-contacting GFAP+ cells were lineage-traced beginning in early adulthood using adult brain electroporation and produced small numbers of olfactory bulb neuroblasts until at least 21 mo of age. Notably, electroporated GFAP+ neurogenic precursors were distinct from both qNSCs and aNSCs: they did not give rise to neurosphere-forming aNSCs in vivo or after extended passaging in vitro and they were not recruited during niche regeneration. GFAP+ cells with these properties included a FoxJ1+GFAP+ subset, as they were also present in an inducible FoxJ1 transgenic lineage-tracing model. Transiently overexpressing Mash1 increased the neurogenic output of electroporated GFAP+ cells in vivo, identifying them as a potentially recruitable population. We propose that the qNSC/aNSC lineage of the adult forebrain coexists with a distinct, minimally expanding subset of GFAP+ neurogenic precursors.


Subject(s)
Cerebral Ventricles/metabolism , Epithelium/metabolism , Gene Targeting , Nerve Growth Factors/genetics , Neural Stem Cells/metabolism , Prosencephalon/metabolism , Adult , Adult Stem Cells/metabolism , Animals , Biomarkers , Cell Differentiation/genetics , Fluorescent Antibody Technique , Gene Expression , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Humans , Mice , Mice, Transgenic , Nerve Growth Factors/metabolism , Neural Stem Cells/cytology , Neurogenesis/genetics , Neurons/cytology , Neurons/metabolism , Olfactory Bulb/cytology , Olfactory Bulb/metabolism , Stem Cell Niche/genetics
3.
Front Neurosci ; 9: 407, 2015.
Article in English | MEDLINE | ID: mdl-26576147

ABSTRACT

A single asymmetric division by an adult neural stem cell (NSC) ultimately generates dozens of differentiated progeny, a feat made possible by the proliferative expansion of transit-amplifying progenitor cells (TAPs). Although NSC activation and TAP expansion is determined by pro- and anti-proliferative signals found within the niche, remarkably little is known about how these cells integrate simultaneous conflicting signals. We investigated this question focusing on the subventricular zone (SVZ) niche of the adult murine forebrain. Using primary cultures of SVZ cells, we demonstrate that Epidermal Growth Factor (EGF) and Bone Morphogenetic Protein (BMP)-2 are particularly powerful pro- and anti-proliferative factors for SVZ-derived neural precursors. Dose-response experiments showed that when simultaneously exposed to both signals, BMP dominantly suppressed EGF-induced proliferation; moreover, this dominance extended to all parameters of neural precursor behavior tested, including inhibition of proliferation, modulation of cell cycle, promotion of differentiation, and increase of cell death. BMP's anti-proliferative effect did not involve inhibition of mTORC1 or ERK signaling, key mediators of EGF-induced proliferation, and had distinct stage-specific consequences, promoting TAP differentiation but NSC quiescence. In line with these in vitro data, in vivo experiments showed that exogenous BMP limits EGF-induced proliferation of TAPs while inhibition of BMP-SMAD signaling promotes activation of quiescent NSCs. These findings clarify the stage-specific effects of BMPs on SVZ neural precursors, and support a hierarchical model in which the anti-proliferative effects of BMP dominate over EGF proliferation signaling to constitutively drive TAP differentiation and NSC quiescence.

4.
Protein Sci ; 24(7): 1114-28, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25970557

ABSTRACT

Biotechnological applications of enzymes can involve the use of these molecules under nonphysiological conditions. Thus, it is of interest to understand how environmental variables affect protein structure and dynamics and how this ultimately modulates enzyme function. NADH oxidase (NOX) from Thermus thermophilus exemplifies how enzyme activity can be tuned by reaction conditions, such as temperature, cofactor substitution, and the addition of cosolutes. This enzyme catalyzes the oxidation of reduced NAD(P)H to NAD(P)(+) with the concurrent reduction of O2 to H2O2, with relevance to biosensing applications. It is thermophilic, with an optimum temperature of approximately 65°C and sevenfold lower activity at 25°C. Moderate concentrations (≈1M) of urea and other chaotropes increase NOX activity by up to a factor of 2.5 at room temperature. Furthermore, it is a flavoprotein that accepts either FMN or the much larger FAD as cofactor. We have used nuclear magnetic resonance (NMR) titration and (15)N spin relaxation experiments together with isothermal titration calorimetry to study how NOX structure and dynamics are affected by changes in temperature, the addition of urea and the substitution of the FMN cofactor with FAD. The majority of signals from NOX are quite insensitive to changes in temperature, cosolute addition, and cofactor substitution. However, a small cluster of residues surrounding the active site shows significant changes. These residues are implicated in coupling changes in the solution conditions of the enzyme to changes in catalytic activity.


Subject(s)
Flavin Mononucleotide/metabolism , Flavin-Adenine Dinucleotide/metabolism , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , NADH, NADPH Oxidoreductases/chemistry , NADH, NADPH Oxidoreductases/metabolism , Thermus thermophilus/enzymology , Urea/metabolism , Binding Sites , Catalytic Domain , Flavin Mononucleotide/chemistry , Flavin-Adenine Dinucleotide/chemistry , Models, Molecular , NAD/metabolism , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Conformation , Temperature , Thermus thermophilus/chemistry , Thermus thermophilus/metabolism
5.
Mol Cancer ; 12: 84, 2013 Jul 31.
Article in English | MEDLINE | ID: mdl-23902727

ABSTRACT

BACKGROUND: The Graffi murine retrovirus is a powerful tool to find leukemia associated oncogenes. Using DNA microarrays, we recently identified several genes specifically deregulated in T- and B-leukemias induced by this virus. RESULTS: In the present study, probsets associated with T-CD8+ leukemias were analyzed and we validated the expression profile of the Parm-1 gene. PARM-1 is a member of the mucin family. We showed that human PARM-1 is an intact secreted protein accumulating predominantly, such as murine PARM-1, at the Golgi and in the early and late endosomes. PARM-1 colocalization with α-tubulin suggests that its trafficking within the cell involves the microtubule cytoskeleton. Also, the protein co-localizes with caveolin-1 which probably mediates its internalization. Transient transfection of both mouse and human Parm-1 cDNAs conferred anchorage- and serum-independent growth and enhanced cell proliferation. Moreover, deletion mutants of human PARM-1 without either extracellular or cytoplasmic portions seem to retain the ability to induce anchorage-independent growth of NIH/3T3 cells. In addition, PARM-1 increases ERK1/2, but more importantly AKT and STAT3 phosphorylation. CONCLUSIONS: Our results strongly suggest the oncogenic potential of PARM-1.


Subject(s)
Androgen-Binding Protein/genetics , Oncogenes , Androgen-Binding Protein/metabolism , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Caveolin 1 , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Endosomes/metabolism , Gene Expression , Gene Expression Profiling , Golgi Apparatus/metabolism , Humans , Leukemia, T-Cell/genetics , Leukemia, T-Cell/metabolism , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mutation , NIH 3T3 Cells , Protein Binding , Protein Transport , Proto-Oncogene Proteins c-akt/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Tubulin/metabolism
6.
Cytokine ; 63(2): 187-93, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23688618

ABSTRACT

Human bone marrow-derived mesenchymal stromal cells (MSCs) express Toll-like receptors (TLRs) and produce cytokines and chemokines, all of which contribute to these cells' immunomodulatory and proangiogenic properties. Among the secreted cytokines, colony-stimulating factors (CSFs) regulate angiogenesis through activation of endothelial cell proliferation and migration. Since MSC are recruited within hypoxic tumors where they signal paracrine-regulated angiogenesis, the aim of this study was to evaluate which CSF members are expressed and are inducible in activated MSC. Furthermore, we investigated the JAK/STAT signal transducing pathway that may impact on CSF transcription. MSC were activated with Concanavalin-A (ConA), a TLR-2/6 agonist as well as a membrane type-1 matrix metalloproteinase (MT1-MMP) inducer, and we found increased transcription of granulocyte macrophage-CSF (GM-CSF, CSF-2), granulocyte CSF (G-CSF, CSF-3), and MT1-MMP. Gene silencing of either STAT3 or MT1-MMP prevented ConA-induced phosphorylation of STAT3, and reversed ConA effects on CSF-2 and CSF-3. Treatment with the Janus Kinase (JAK)2 inhibitor AG490 antagonized the ConA induction of MT1-MMP and CSF-2, while the pan-JAK inhibitor Tofacitinib reversed ConA-induced CSF-2 and -3 gene expression. Silencing of JAK2 prevented the ConA-mediated increase of CSF-2, while silencing of JAK1, JAK3 and TYK2 prevented the increase in CSF-3. Given that combined TLR-activation and locally-produced CSF-2 and CSF-3 could regulate immunomodulation and neovascularization, pharmacological targeting of TLR-2/6-induced MT1-MMP/JAK/STAT3 signalling pathway may prevent MSC contribution to tumor development.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Janus Kinases/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Mesenchymal Stem Cells/metabolism , STAT3 Transcription Factor/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Concanavalin A , Enzyme Inhibitors/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Janus Kinases/antagonists & inhibitors , Janus Kinases/genetics , Macrophage Colony-Stimulating Factor/genetics , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , Neovascularization, Physiologic , Phosphorylation , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , RNA Interference , RNA, Small Interfering , STAT3 Transcription Factor/genetics , Signal Transduction , TYK2 Kinase/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 6/metabolism , Transcription, Genetic , Tyrphostins/pharmacology
7.
Mol Neurobiol ; 48(3): 669-80, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23715769

ABSTRACT

Apolipoprotein D (ApoD) gene expression is increased in several neurological disorders such as Alzheimer's disease (AD) and multiple sclerosis. We previously showed that transgenic mice that overexpress human ApoD show a better resistance against paraquat or OC43 coronavirus-induced neurodegeneration. Here, we identified several nuclear factors from the cortex of control and OC43-infected mice which bind a fragment of the proximal ApoD promoter in vitro. Of interest, we detected apolipoprotein E (ApoE). Human ApoE consists of three isoforms (E2, E3, and E4) with the E4 and E2 alleles representing a greater and a lower risk for developping AD, respectively. Our results show that ApoE is located in the nucleus and on the ApoD promoter in human hepatic and glioblastoma cells lines. Furthermore, overexpression of ApoE3 and ApoE4 isoforms but not ApoE2 significantly inhibited the ApoD promoter activity in U87 cells (E3/E3 genotype) cultured under normal or different stress conditions while ApoE knock-down by siRNA had a converse effect. Consistent with these results, we also demonstrated by ChIP assay that E3 and E4 isoforms, but not E2, bind the ApoD promoter. Moreover, using the Allen Brain Atlas in situ hybridization database, we observed an inverse correlation between ApoD and ApoE mRNA expression during development and in several regions of the mouse brain, notably in the cortex, hippocampus, plexus choroid, and cerebellum. This negative correlation was also observed for cortex layers IV-VI based on a new Transcriptomic Atlas of the Mouse Neocortical Layers. These findings reveal a new function for ApoE by regulating ApoD gene expression.


Subject(s)
Apolipoprotein E3/metabolism , Apolipoprotein E4/metabolism , Apolipoproteins D/genetics , Promoter Regions, Genetic , Animals , Base Sequence , Cell Line, Tumor , Cell Nucleus/metabolism , Cerebral Cortex/metabolism , Gene Expression Regulation , Humans , Mass Spectrometry , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Nuclear Proteins/isolation & purification , Nuclear Proteins/metabolism , Protein Binding , Protein Isoforms/metabolism , Protein Transport
8.
Blood ; 117(6): 1899-910, 2011 Feb 10.
Article in English | MEDLINE | ID: mdl-21135260

ABSTRACT

The Graffi murine leukemia virus induces a large spectrum of leukemias in mice and thus provides a good model to compare the transcriptome of all types of leukemias. We analyzed the gene expression profiles of both T and B leukemias induced by the virus with DNA microarrays. Given that we considered that a 4-fold change in expression level was significant, 388 probe sets were associated to B, to T, or common to both leukemias. Several of them were not yet associated with lymphoid leukemia. We confirmed specific deregulation of Fmn2, Arntl2, Bfsp2, Gfra2, Gpm6a, and Gpm6b in B leukemia, of Nln, Fbln1, and Bmp7 in T leukemias, and of Etv5 in both leukemias. More importantly, we show that the mouse Fmn2 induced an anchorage-independent growth, a drastic modification in cell shape with a concomitant disruption of the actin cytoskeleton. Interestingly, we found that human FMN2 is overexpressed in approximately 95% of pre-B acute lymphoblastic leukemia with the highest expression levels in patients with a TEL/AML1 rearrangement. These results, surely related to the role of FMN2 in meiotic spindle maintenance, suggest its important role in leukemogenesis. Finally, we propose a new panel of genes potentially involved in T and/or B leukemias.


Subject(s)
Leukemia Virus, Murine/pathogenicity , Leukemia, Experimental/genetics , Microfilament Proteins/genetics , Nuclear Proteins/genetics , Oncogenes , Retroviridae Infections/genetics , Tumor Virus Infections/genetics , Adult , Animals , Biomarkers, Tumor/genetics , Child , Core Binding Factor Alpha 2 Subunit/genetics , Formins , Gene Expression Profiling , Genetic Markers , Humans , Leukemia, Experimental/metabolism , Leukemia, T-Cell/genetics , Mice , Microfilament Proteins/metabolism , Nerve Tissue Proteins/genetics , Nuclear Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Oncogene Proteins, Fusion/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Retroviridae Infections/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Virus Infections/metabolism
9.
Biochim Biophys Acta ; 1803(9): 1062-71, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20493910

ABSTRACT

Human Apolipoprotein D (apoD) is upregulated under several stress conditions and pathological situations such as neurodegenerative diseases and cancers. We previously showed that apoD mRNA expression is induced in growth-arrested cells and demonstrated the specific binding of nuclear proteins to the region -514 to -475 of the promoter. Such region contains a pair of Serum Responsive Elements (SRE), an Ets-Binding Site (EBS) and a Glucocorticoid Responsive Element (GRE). In this study, we show that Parp-1, HnRNP-U, CBF-A, BUB-3, Kif4, APEX-1 and Ifi204 bind these regulatory elements of the apoD promoter. Specific binding of HnRNP-U and Parp-1 was confirmed by Electrophoretic Mobility Shift Assay (EMSA). In a biotin pull-down assay, Kif4 and BUB-3 bind preferentially the SRE1 and the EBS-GRE sites, respectively, while APEX-1 seems recruited indirectly to these elements. We found that the mRNA expression of some of these binding factors is upregulated in growth-arrested cells and that these proteins also transactivate the apoD promoter. In agreement with these results, mutants of APEX-1 and of Parp-1 defective for their DNA-binding and catalytic activities could not transactivate the promoter. The knockdown of Parp-1 and HnRNP-U and the use of specific inhibitors of MEK1/2 and of Parp-1 also inhibited the induction of apoD gene expression. Moreover, ERK1/2 was found activated in a biphasic manner post serum-starvation and the inhibition of Parp-1 causes a sustained activation of ERK2 but not ERK1 for up to 2h. Altogether, these findings demonstrate the importance of Parp-1, APEX-1 and ERK1/2 catalytic activities in the growth arrest-induced apoD gene expression.


Subject(s)
Apolipoproteins D/genetics , Cell Growth Processes/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/physiology , Glycoproteins/genetics , Membrane Transport Proteins/genetics , Mitogen-Activated Protein Kinase 1/physiology , Poly(ADP-ribose) Polymerases/physiology , Promoter Regions, Genetic , Animals , Catalysis , Cell Cycle/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Enzyme Activation , Gene Expression Regulation , Humans , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 3/physiology , NIH 3T3 Cells , Nuclear Proteins/metabolism , Nuclear Proteins/physiology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Transcription Factors/metabolism , Transcription Factors/physiology
10.
Stem Cells ; 27(3): 489-97, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19074414

ABSTRACT

Mesenchymal stromal cell (MSC) markers are expressed on brain tumor-initiating cells involved in the development of hypoxic glioblastoma. Given that MSCs can survive hypoxia and that the glucose-6-phosphate transporter (G6PT) provides metabolic control that contributes to MSC mobilization and survival, we investigated the effects of low oxygen (1.2% O(2)) exposure on G6PT gene expression. We found that MSCs significantly expressed G6PT and the glucose-6-phosphatase catalytic subunit beta, whereas expression of the glucose-6-phosphatase catalytic subunit alpha and the islet-specific glucose-6-phosphatase catalytic subunit-related protein was low to undetectable. Analysis of the G6PT promoter sequence revealed potential binding sites for hypoxia inducible factor (HIF)-1alpha and for the aryl hydrocarbon receptor (AhR) and its dimerization partner, the AhR nuclear translocator (ARNT), AhR:ARNT. In agreement with this, hypoxia and the hypoxia mimetic cobalt chloride induced the expression of G6PT, vascular endothelial growth factor (VEGF), and HIF-1alpha. Gene silencing of HIF-1alpha prevented G6PT and VEGF induction in hypoxic MSCs whereas generation of cells stably expressing HIF-1alpha resulted in increased endogenous G6PT gene expression. A semisynthetic analog of the polyketide mumbaistatin, a potent G6PT inhibitor, specifically reduced MSC-HIF-1alpha cell survival. Collectively, our data suggest that G6PT may account for the metabolic flexibility that enables MSCs to survive under conditions characterized by hypoxia and could be specifically targeted within developing tumors.


Subject(s)
Anthraquinones/pharmacology , Gene Expression/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Mesenchymal Stem Cells/metabolism , Monosaccharide Transport Proteins/physiology , Stromal Cells/drug effects , Stromal Cells/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Hypoxia/drug effects , Cell Movement , Cells, Cultured , Cobalt/pharmacology , Electrophoretic Mobility Shift Assay , Female , Glucose-6-Phosphatase/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunoblotting , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mice , Mice, Inbred C57BL , Monosaccharide Transport Proteins/antagonists & inhibitors , Monosaccharide Transport Proteins/genetics , Necrosis/genetics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/cytology
11.
Biochim Biophys Acta ; 1773(6): 954-69, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17477983

ABSTRACT

Apolipoprotein D is a lipocalin, primarily associated with high density lipoproteins in human plasma. Its expression is induced in several pathological and stressful conditions including growth arrest suggesting that it could act as a nonspecific stress protein. A survey of cellular stresses shows those causing an extended growth arrest, as hydrogen peroxide and UV light increase apoD expression. Alternatively, lipopolysaccharide (LPS), a pro-inflammatory agonist showed a time- and dose-dependent effect on apoD expression that correlates with an increase in proliferation. At the promoter level, NF-kB, AP-1 and APRE-3 proved to be the elements implicated in the LPS response. Colocalization of apoDh-GFP fusion constructs with DNA and Golgi markers, immunocytochemistry of the endogenous protein and cell fractionation showed that both serum starvation and LPS treatment caused a displacement of apoD localization. In normal conditions, apoD is mainly perinuclear but it accumulates in cytoplasm and nucleus under these stress conditions. Since nuclear apoD appears derived from the secreted protein, it may act as an extracellular ligand transporter as well as a transcriptional regulator depending on its location. This role of apoD inside the cell is not only dependent of endogenous apoD but may also be provided by exogenous apoD entering the cell.


Subject(s)
Apolipoproteins D/biosynthesis , Cell Nucleus/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Glycoproteins/biosynthesis , Hydrogen Peroxide/pharmacology , Membrane Transport Proteins/biosynthesis , Oxidants/pharmacology , Ultraviolet Rays , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/radiation effects , Animals , Apolipoproteins D/genetics , Cell Nucleus/genetics , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Glycoproteins/genetics , Golgi Apparatus/genetics , Golgi Apparatus/metabolism , HeLa Cells , Humans , Lipopolysaccharides/pharmacology , Membrane Transport Proteins/genetics , Mice , NIH 3T3 Cells , Stress, Physiological/genetics , Stress, Physiological/metabolism , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...