Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
2.
Phys Chem Chem Phys ; 16(12): 5855-65, 2014 Mar 28.
Article in English | MEDLINE | ID: mdl-24549246

ABSTRACT

The rational design of supraparticle assemblies requires a detailed understanding of directed assembly processes. The stability of dispersions of nanoscale materials, like single-walled carbon nanotubes (SWCNTs), is still not fully understood, nor are the mechanisms of aggregation and assembly. A detailed balance of attractive van der Waals type interactions with various repulsive barrier mechanisms is needed to control the assembly of industrially viable and functional hybrid-nanoscale supraparticles. We report a detailed study of SWCNT dispersion stability and aggregation kinetics as a function of the nature of the coagulant used in various solvent systems. We explore three classes of coagulants that vary in charge, size, shape, solvation energy, and the ability to bind to the SWCNTs. We use these kinetic data to assess the tube-solvent-coagulant-tube interactions. We compare the relative contributions from two types of repulsive barriers. We find that tube-mediated structured solvent around the SWCNTs does not sufficiently describe our measured kinetic data. A DLVO type, electrical double layer repulsion is used to rationalize our observations. The data presented in this paper require a more detailed theoretical understanding of the physico-chemical environment near nanoparticle surfaces such as aggregating SWCNTs.


Subject(s)
Coagulants/chemistry , Nanotubes, Carbon/chemistry , Diffusion , Kinetics , Solvents/chemistry
3.
Proc Natl Acad Sci U S A ; 109(15): 5750-5, 2012 Apr 10.
Article in English | MEDLINE | ID: mdl-22447776

ABSTRACT

Decoupling of transcription and translation during postmeiotic germ cell differentiation is critical for successful spermatogenesis. Here we establish that the interaction between microRNAs and actin-associated protein Arpc5 sets the stage for an elaborate translational control mechanism by facilitating the sequestration of germ cell mRNAs into translationally inert ribonucleoprotein particles until they are later translated. Our studies reveal that loss of microRNA-dependent regulation of Arpc5, which controls the distribution of germ cell mRNAs between translationally active and inactive pools, results in abnormal round spermatid differentiation and impaired fertility. Interestingly, Arpc5 functions as a broadly acting translational suppressor, as it inhibits translation initiation by blocking 80S formation and facilitates the transport of mRNAs to chromatoid/P bodies. These findings identify a unique role for actin-associated proteins in translational regulation, and suggest that mRNA-specific and general translational control mechanisms work in tandem to regulate critical germ cell differentiation events and diverse somatic cell functions.


Subject(s)
Actin-Related Protein 2-3 Complex/metabolism , Cell Differentiation/genetics , MicroRNAs/metabolism , Spermatozoa/metabolism , Spermatozoa/pathology , Animals , Base Sequence , Chromatin/metabolism , Enzyme Activation , Gene Expression Regulation , Haploidy , HeLa Cells , Humans , Male , Meiosis/genetics , Mice , MicroRNAs/genetics , Molecular Sequence Data , Protamines/metabolism , Protein Binding , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproduction , Ribonuclease III/metabolism , Ribosomes/metabolism , Sperm Head/metabolism , Sperm Head/pathology , Sperm Head/ultrastructure , Testis/abnormalities , Testis/pathology , Testis/ultrastructure
4.
Mol Cell ; 45(6): 826-35, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22387028

ABSTRACT

Although expression of the mammalian RNA-binding protein HuD was considered to be restricted to neurons, we report that HuD is present in pancreatic ß cells, where its levels are controlled by the insulin receptor pathway. We found that HuD associated with a 22-nucleotide segment of the 5' untranslated region (UTR) of preproinsulin (Ins2) mRNA. Modulating HuD abundance did not alter Ins2 mRNA levels, but HuD overexpression decreased Ins2 mRNA translation and insulin production, and conversely, HuD silencing enhanced Ins2 mRNA translation and insulin production. Following treatment with glucose, HuD rapidly dissociated from Ins2 mRNA and enabled insulin biosynthesis. Importantly, HuD-knockout mice displayed higher insulin levels in pancreatic islets, while HuD-overexpressing mice exhibited lower insulin levels in islets and in plasma. In sum, our results identify HuD as a pivotal regulator of insulin translation in pancreatic ß cells.


Subject(s)
ELAV Proteins/metabolism , Insulin/genetics , Insulin/metabolism , Protein Biosynthesis , 5' Untranslated Regions , Animals , ELAV Proteins/genetics , ELAV-Like Protein 4 , Glucose/metabolism , Humans , Insulin-Secreting Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein Precursors/genetics
5.
PLoS One ; 7(12): e50736, 2012.
Article in English | MEDLINE | ID: mdl-23284645

ABSTRACT

BACKGROUND: The receptor for advanced glycation end products (RAGE) on the cell surface transmits inflammatory signals. A member of the immunoglobulin superfamily, RAGE possesses the V, C1, and C2 ectodomains that collectively constitute the receptor's extracellular structure. However, the molecular mechanism of RAGE biogenesis remains unclear, impeding efforts to control RAGE signaling through cellular regulation. METHODOLOGY AND RESULT: We used co-immunoprecipitation and crossing-linking to study RAGE oligomerization and found that RAGE forms dimer-based oligomers. Via non-reducing SDS-polyacrylamide gel electrophoresis and mutagenesis, we found that cysteines 259 and 301 within the C2 domain form intermolecular disulfide bonds. Using a modified tripartite split GFP complementation strategy and confocal microscopy, we also found that RAGE dimerization occurs in the endoplasmic reticulum (ER), and that RAGE mutant molecules without the double disulfide bridges are unstable, and are subjected to the ER-associated degradation. CONCLUSION: Disulfide bond-mediated RAGE dimerization in the ER is the critical step of RAGE biogenesis. Without formation of intermolecular disulfide bonds in the C2 region, RAGE fails to reach cell surface. SIGNIFICANCE: This is the first report of RAGE intermolecular disulfide bond.


Subject(s)
Disulfides/chemistry , Protein Multimerization , Receptors, Immunologic/biosynthesis , Receptors, Immunologic/chemistry , Animals , CHO Cells , Cricetinae , Cricetulus , Cysteine/chemistry , Endoplasmic Reticulum/metabolism , Gene Expression Regulation , Gene Knockout Techniques , Glycosylation , HeLa Cells , Humans , Mice , Protein Structure, Quaternary , Protein Structure, Tertiary , Receptor for Advanced Glycation End Products , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Signal Transduction
6.
Mol Cell Biol ; 31(20): 4219-31, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21859890

ABSTRACT

The RNA-binding protein (RBP) nucleolin promotes the expression of several proliferative proteins. Nucleolin levels are high in cancer cells, but the mechanisms that control nucleolin expression are unknown. Here, we show that nucleolin abundance is controlled posttranscriptionally via factors that associate with its 3' untranslated region (3'UTR). The RBP HuR was found to interact with the nucleolin (NCL) 3'UTR and specifically promoted nucleolin translation without affecting nucleolin mRNA levels. In human cervical carcinoma HeLa cells, analysis of a traceable NCL 3'UTR bearing MS2 RNA hairpins revealed that NCL RNA was mobilized to processing bodies (PBs) after silencing HuR, suggesting that the repression of nucleolin translation may occur in PBs. Immunoprecipitation of MS2-tagged NCL 3'UTR was used to screen for endogenous repressors of nucleolin synthesis. This search identified miR-494 as a microRNA that potently inhibited nucleolin expression, enhanced NCL mRNA association with argonaute-containing complexes, and induced NCL RNA transport to PBs. Importantly, miR-494 and HuR functionally competed for modulation of nucleolin expression. Moreover, the promotion of cell growth previously attributed to HuR was due in part to the HuR-elicited increase in nucleolin expression. Our collective findings indicate that nucleolin expression is positively regulated by HuR and negatively regulated via competition with miR-494.


Subject(s)
3' Untranslated Regions , Antigens, Surface/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Neoplasms/genetics , Phosphoproteins/genetics , RNA-Binding Proteins/genetics , ELAV Proteins , ELAV-Like Protein 1 , HeLa Cells , Humans , RNA, Messenger/genetics , Nucleolin
7.
Mol Cell Biol ; 31(18): 3790-801, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21768308

ABSTRACT

The cellular abundance of topoisomerase IIα (TOP2A) critically maintains DNA topology after replication and determines the efficacy of TOP2 inhibitors in chemotherapy. Here, we report that the RNA-binding protein HuR, commonly overexpressed in cancers, binds to the TOP2A 3'-untranslated region (3'UTR) and increases TOP2A translation. Reducing HuR levels triggered the recruitment of TOP2A transcripts to RNA-induced silencing complex (RISC) components and to cytoplasmic processing bodies. Using a novel MS2-tagged RNA precipitation method, we identified microRNA miR-548c-3p as a mediator of these effects and further uncovered that the interaction of miR-548c-3p with the TOP2A 3'UTR repressed TOP2A translation by antagonizing the action of HuR. Lowering TOP2A by silencing HuR or by overexpressing miR-548c-3p selectively decreased DNA damage after treatment with the chemotherapeutic agent doxorubicin. In sum, HuR enhances TOP2A translation by competing with miR-548c-3p; their combined actions control TOP2A expression levels and determine the effectiveness of doxorubicin.


Subject(s)
Antigens, Neoplasm/genetics , Antigens, Surface/metabolism , DNA Topoisomerases, Type II/genetics , DNA-Binding Proteins/genetics , Doxorubicin/pharmacology , RNA-Binding Proteins/metabolism , 3' Untranslated Regions , Antibiotics, Antineoplastic/pharmacology , Antigens, Neoplasm/biosynthesis , Antigens, Surface/genetics , DNA/metabolism , DNA Damage/drug effects , DNA Topoisomerases, Type II/biosynthesis , DNA-Binding Proteins/biosynthesis , ELAV Proteins , ELAV-Like Protein 1 , Gene Expression Regulation , HeLa Cells , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Poly-ADP-Ribose Binding Proteins , Protein Biosynthesis , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/biosynthesis , RNA-Binding Proteins/genetics , RNA-Induced Silencing Complex/metabolism
8.
Mol Cell Biol ; 30(17): 4197-210, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20584986

ABSTRACT

Neuronal development and plasticity are maintained by tightly regulated gene expression programs. Here, we report that the developmentally regulated microRNA miR-375 affects dendrite formation and maintenance. miR-375 overexpression in mouse hippocampus potently reduced dendrite density. We identified the predominantly neuronal RNA-binding protein HuD as a key effector of miR-375 influence on dendrite maintenance. Heterologous reporter analysis verified that miR-375 repressed HuD expression through a specific, evolutionarily conserved site on the HuD 3' untranslated region. miR-375 overexpression lowered both HuD mRNA stability and translation and recapitulated the effects of HuD silencing, which reduced the levels of target proteins with key functions in neuronal signaling and cytoskeleton organization (N-cadherin, PSD-95, RhoA, NCAM1, and integrin alpha1). Moreover, the increase in neurite outgrowth after brain-derived neurotrophic factor (BDNF) treatment was diminished by miR-375 overexpression; this effect was rescued by reexpression of miR-375-refractory HuD. Our findings indicate that miR-375 modulates neuronal HuD expression and function, in turn affecting dendrite abundance.


Subject(s)
Dendrites/metabolism , ELAV Proteins/genetics , MicroRNAs/metabolism , Neurogenesis , Animals , Cell Line , Dendrites/ultrastructure , ELAV Proteins/metabolism , ELAV-Like Protein 4 , Gene Expression Regulation , Hippocampus/cytology , Humans , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Neurites/metabolism , Neurites/ultrastructure , RNA Stability , RNA, Messenger/genetics
9.
Nat Struct Mol Biol ; 17(6): 732-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20473314

ABSTRACT

Amyloid precursor protein (APP) regulates neuronal synapse function, and its cleavage product Abeta is linked to Alzheimer's disease. Here, we present evidence that the RNA-binding proteins (RBPs) heterogeneous nuclear ribonucleoprotein (hnRNP) C and fragile X mental retardation protein (FMRP) associate with the same APP mRNA coding region element, and they influence APP translation competitively and in opposite directions. Silencing hnRNP C increased FMRP binding to APP mRNA and repressed APP translation, whereas silencing FMRP enhanced hnRNP C binding and promoted translation. Repression of APP translation was linked to colocalization of FMRP and tagged APP RNA within processing bodies; this colocalization was abrogated by hnRNP C overexpression or FMRP silencing. Our findings indicate that FMRP represses translation by recruiting APP mRNA to processing bodies, whereas hnRNP C promotes APP translation by displacing FMRP, thereby relieving the translational block.


Subject(s)
Amyloid beta-Protein Precursor/biosynthesis , Amyloid beta-Protein Precursor/genetics , Fragile X Mental Retardation Protein/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group C/metabolism , Neurons/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , 3' Untranslated Regions , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Base Sequence , Binding, Competitive , Cell Line , Cytoplasmic Structures/metabolism , Fragile X Mental Retardation Protein/genetics , Genes, Reporter , Green Fluorescent Proteins/genetics , Humans , In Vitro Techniques , Mice , Mice, Knockout , Models, Biological , Molecular Sequence Data , Protein Biosynthesis , RNA, Small Interfering/genetics , Recombinant Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...