Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Res Sq ; 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38699331

ABSTRACT

HIV-1 eradication strategies require complete reactivation of HIV-1 latent cells by Latency Reversing Agents (LRA). Current methods lack effectiveness due to incomplete proviral reactivation. We employed a single-molecule RNA-FISH (smRNA-FISH) and FISH-Quant analysis and found that proviral reactivation is highly variable from cell-to-cell, stochastic, and occurs in bursts and waves, with different kinetics in response to diverse LRAs. Approximately 1-5% of latent cells exhibited stochastic reactivation without LRAs. Through single-cell RNA-seq analysis, we identified NR4A3 and cMYC as extrinsic factors associated with stochastic HIV-1 reactivation. Concomitant with HIV-1 reactivation cMYC was downregulated and NR4A3 was upregulated in both latent cell lines and primary CD4+ T-cells from aviremic patients. By inhibiting cMYC using SN-38, an active metabolite of irinotecan, we induced NR4A3 and HIV-1 expression. Our results suggest that inherent stochasticity in proviral reactivation contributes to cell-to-cell variability, which could potentially be modulated by drugs targeting cMYC and NR4A3.

2.
Methods Mol Biol ; 2807: 45-59, 2024.
Article in English | MEDLINE | ID: mdl-38743220

ABSTRACT

Latent HIV-1 reservoirs are a major obstacle to the eradication of HIV-1. Several cure strategies have been proposed to eliminate latent reservoirs. One of the key strategies involves the reactivation of latent HIV-1 from cells using latency-reversing agents. However, currently it is unclear whether any of the latency-reversing agents are able to completely reactivate HIV-1 provirus transcription in all latent cells. An understanding of the reactivation of HIV-1 provirus at single-cell single-molecule level is necessary to fully comprehend the reactivation of HIV-1 in the reservoirs. Furthermore, since reactivable viruses in the pool of latent reservoirs are rare, combining single-cell imaging techniques with the ability to visualize a large number of reactivated single cells that express both viral RNA and proteins in a pool of uninfected and non-reactivated cells will provide unprecedented information about cell-to-cell variability in reactivation. Here, we describe the single-cell single-molecule RNA-FISH (smRNA-FISH) method to visualize HIV-1 gag RNA combined with the immunofluorescence (IF) method to detect Gag protein to characterize the reactivated cells. This method allows the visualization of subcellular localization of RNA and proteins before and after reactivation and facilitates absolute quantitation of the number of transcripts per cell using FISH-quant. In addition, we describe a high-speed and high-resolution scanning (HSHRS) fluorescence microscopy imaging method to visualize rare and reactivated cells in a pool of non-reactivated cells with high efficiency.


Subject(s)
Fluorescent Antibody Technique , HIV-1 , In Situ Hybridization, Fluorescence , RNA, Viral , Single Molecule Imaging , Single-Cell Analysis , Virus Activation , Virus Latency , HIV-1/physiology , HIV-1/genetics , Humans , In Situ Hybridization, Fluorescence/methods , RNA, Viral/genetics , Single-Cell Analysis/methods , Single Molecule Imaging/methods , Fluorescent Antibody Technique/methods , HIV Infections/virology , Proviruses/genetics
3.
bioRxiv ; 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38746329

ABSTRACT

The liver acts as a master regulator of metabolic homeostasis in part by performing gluconeogenesis. This process is dysregulated in type 2 diabetes, leading to elevated hepatic glucose output. The parenchymal cells of the liver (hepatocytes) are heterogeneous, existing on an axis between the portal triad and the central vein, and perform distinct functions depending on location in the lobule. Here, using single cell analysis of hepatocytes across the liver lobule, we demonstrate that gluconeogenic gene expression ( Pck1 and G6pc ) is relatively low in the fed state and gradually increases first in the periportal hepatocytes during the initial fasting period. As the time of fasting progresses, pericentral hepatocyte gluconeogenic gene expression increases, and following entry into the starvation state, the pericentral hepatocytes show similar gluconeogenic gene expression to the periportal hepatocytes. Similarly, pyruvate-dependent gluconeogenic activity is approximately 10-fold higher in the periportal hepatocytes during the initial fasting state but only 1.5-fold higher in the starvation state. In parallel, starvation suppresses canonical beta-catenin signaling and modulates expression of pericentral and periportal glutamine synthetase and glutaminase, resulting in an enhanced pericentral glutamine-dependent gluconeogenesis. These findings demonstrate that hepatocyte gluconeogenic gene expression and gluconeogenic activity are highly spatially and temporally plastic across the liver lobule, underscoring the critical importance of using well-defined feeding and fasting conditions to define the basis of hepatic insulin resistance and glucose production.

4.
Viruses ; 16(2)2024 02 07.
Article in English | MEDLINE | ID: mdl-38400039

ABSTRACT

SARS-CoV-2 infection remains a global burden. Despite intensive research, the mechanism and dynamics of early viral replication are not completely understood, such as the kinetics of the formation of genomic RNA (gRNA), sub-genomic RNA (sgRNA), and replication centers/organelles (ROs). We employed single-molecule RNA-fluorescence in situ hybridization (smRNA-FISH) to simultaneously detect viral gRNA and sgRNA and immunofluorescence to detect nsp3 protein, a marker for the formation of RO, and carried out a time-course analysis. We found that single molecules of gRNA are visible within the cytoplasm at 30 min post infection (p.i.). Starting from 2 h p.i., most of the viral RNA existed in clusters/speckles, some of which were surrounded by single molecules of sgRNA. These speckles associated with nsp3 protein starting at 3 h p.i., indicating that these were precursors to ROs. Furthermore, RNA replication was asynchronous, as cells with RNA at all stages of replication were found at any given time point. Our probes detected the SARS-CoV-2 variants of concern, and also suggested that the BA.1 strain exhibited a slower rate of replication kinetics than the WA1 strain. Our results provide insights into the kinetics of SARS-CoV-2 early post-entry events, which will facilitate identification of new therapeutic targets for early-stage replication to combat COVID-19.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , COVID-19/metabolism , RNA Replication , In Situ Hybridization, Fluorescence/methods , Reactive Oxygen Species/metabolism , Subgenomic RNA , RNA, Guide, CRISPR-Cas Systems , Fluorescent Antibody Technique , Proteins/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism
5.
bioRxiv ; 2022 Dec 12.
Article in English | MEDLINE | ID: mdl-36561180

ABSTRACT

SARS-CoV-2 infection has caused a major global burden. Despite intensive research, the mechanism and dynamics of early viral replication are not completely understood including the kinetics of formation of plus stranded genomic and subgenomic RNAs (gRNA and sgRNA) starting from the RNA from the first virus that enters the cell. We employed single-molecule RNA-fluorescence in situ hybridization (smRNA-FISH) to simultaneously detect viral gRNA and sgRNA in infected cells and carried out a time course analysis to determine the kinetics of their replication. We visualized the single molecules of gRNA within the cytoplasm of infected cells 30 minutes post-infection and detected the co-expression of gRNA and sgRNA within two hours post-infection. Furthermore, we observed the formation of a replication organelle (RO) from a single RNA, which led to the formation of multiple ROs within the same cells. Single molecule analysis indicated that while gRNA resided in the center of these ROs, the sgRNAs were found to radiate and migrate out of these structures. Our results also indicated that after the initial delay, there was a rapid but asynchronous replication, and the gRNA and sgRNAs dispersed throughout the cell within 4-5 hours post-infection forming multiple ROs that filled the entire cytoplasm. These results provide insight into the kinetics of early post-entry events of SARS-CoV-2 and the formation of RO, which will help to understand the molecular events associated with viral infection and facilitate the identification of new therapeutic targets that can curb the virus at a very early stage of replication to combat COVID-19. Author Summary: SARS-CoV-2 infection continues to be a global burden. Soon after the entry, SARS-CoV-2 replicates by an elaborate process, producing genomic and subgenomic RNAs (gRNA and sgRNAs) within specialized structures called replication organelles (RO). Many questions including the timing of multiplication of gRNA and sgRNA, the generation, subcellular localization, and function of the ROs, and the mechanism of vRNA synthesis within ROs is not completely understood. Here, we have developed probes and methods to simultaneously detect the viral gRNA and a sgRNA at single cell single molecule resolution and have employed a method to scan thousands of cells to visualize the early kinetics of gRNA and sgRNA synthesis soon after the viral entry into the cell. Our results reveal that the replication is asynchronous and ROs are rapidly formed from a single RNA that enters the cell within 2 hours, which multiply to fill the entire cell cytoplasm within ~4 hours after infection. Furthermore, our studies provide a first glimpse of the gRNA and sgRNA synthesis within ROs at single molecule resolution. Our studies may facilitate the development of drugs that inhibit the virus at the earliest possible stages of replication to minimize the pathogenic impact of viral infection.

6.
Exp Eye Res ; 207: 108564, 2021 06.
Article in English | MEDLINE | ID: mdl-33894228

ABSTRACT

Cellular differentiation is marked by temporally and spatially regulated gene expression. The ocular lens is one of the most powerful mammalian model system since it is composed from only two cell subtypes, called lens epithelial and fiber cells. Lens epithelial cells differentiate into fiber cells through a series of spatially and temporally orchestrated processes, including massive production of crystallins, cellular elongation and the coordinated degradation of nuclei and other organelles. Studies of transcriptional and posttranscriptional gene regulatory mechanisms in lens provide a wide range of opportunities to understand global molecular mechanisms of gene expression as steady-state levels of crystallin mRNAs reach very high levels comparable to globin genes in erythrocytes. Importantly, dysregulation of crystallin gene expression results in lens structural abnormalities and cataracts. The mRNA life cycle is comprised of multiple stages, including transcription, splicing, nuclear export into cytoplasm, stabilization, localization, translation and ultimate decay. In recent years, development of modern mRNA detection methods with single molecule and single cell resolution enabled transformative studies to visualize the mRNA life cycle to generate novel insights into the sequential regulatory mechanisms of gene expression during embryogenesis. This review is focused on recent major advancements in studies of transcriptional bursting in differentiating lens fiber cells, analysis of nascent mRNA expression from bi-directional promoters, transient nuclear accumulation of specific mRNAs, condensation of chromatin prior lens fiber cell denucleation, and outlines future studies to probe the interactions of individual mRNAs with specific RNA-binding proteins (RBPs) in the cytoplasm and regulation of translation and mRNA decay.


Subject(s)
Crystallins/genetics , Gene Expression Regulation/physiology , Lens, Crystalline/metabolism , Transcription, Genetic , Animals , Cell Differentiation , Humans , RNA, Messenger/genetics , RNA-Binding Proteins/metabolism , Transcriptional Activation
7.
Nat Commun ; 10(1): 4440, 2019 09 30.
Article in English | MEDLINE | ID: mdl-31570709

ABSTRACT

The IGF2 mRNA-binding proteins (ZBP1/IMP1, IMP2, IMP3) are highly conserved post-transcriptional regulators of RNA stability, localization and translation. They play important roles in cell migration, neural development, metabolism and cancer cell survival. The knockout phenotypes of individual IMP proteins suggest that each family member regulates a unique pool of RNAs, yet evidence and an underlying mechanism for this is lacking. Here, we combine systematic evolution of ligands by exponential enrichment (SELEX) and NMR spectroscopy to demonstrate that the major RNA-binding domains of the two most distantly related IMPs (ZBP1 and IMP2) bind to different consensus sequences and regulate targets consistent with their knockout phenotypes and roles in disease. We find that the targeting specificity of each IMP is determined by few amino acids in their variable loops. As variable loops often differ amongst KH domain paralogs, we hypothesize that this is a general mechanism for evolving specificity and regulation of the transcriptome.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , RNA/metabolism , Animals , Base Sequence , Crystallography, X-Ray , DNA-Binding Proteins/genetics , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Ligands , Magnetic Resonance Spectroscopy , Mice , Models, Molecular , Mutation , Protein Binding , Protein Domains , RNA Stability , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Ribonucleoproteins, Small Nucleolar , SELEX Aptamer Technique , Transcriptome
8.
Proc Natl Acad Sci U S A ; 116(26): 12863-12872, 2019 06 25.
Article in English | MEDLINE | ID: mdl-31189591

ABSTRACT

The molecular function and fate of mRNAs are controlled by RNA-binding proteins (RBPs). Identification of the interacting proteome of a specific mRNA in vivo remains very challenging, however. Based on the widely used technique of RNA tagging with MS2 aptamers for RNA visualization, we developed a RNA proximity biotinylation (RNA-BioID) technique by tethering biotin ligase (BirA*) via MS2 coat protein at the 3' UTR of endogenous MS2-tagged ß-actin mRNA in mouse embryonic fibroblasts. We demonstrate the dynamics of the ß-actin mRNA interactome by characterizing its changes on serum-induced localization of the mRNA. Apart from the previously known interactors, we identified more than 60 additional ß-actin-associated RBPs by RNA-BioID. Among these, the KH domain-containing protein FUBP3/MARTA2 has been shown to be required for ß-actin mRNA localization. We found that FUBP3 binds to the 3' UTR of ß-actin mRNA and is essential for ß-actin mRNA localization, but does not interact with the characterized ß-actin zipcode element. RNA-BioID provides a tool for identifying new mRNA interactors and studying the dynamic view of the interacting proteome of endogenous mRNAs in space and time.


Subject(s)
Actins/genetics , RNA Processing, Post-Transcriptional , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , 3' Untranslated Regions , Actins/metabolism , Animals , Binding Sites , Biotinylation/methods , Cell Line , Mice , Protein Binding , RNA, Messenger/chemistry , RNA-Binding Proteins/chemistry
9.
Article in English | MEDLINE | ID: mdl-32086331

ABSTRACT

The fate of an RNA, from its localization, translation, and ultimate decay, is dictated by interactions with RNA binding proteins (RBPs). ß-actin mRNA has functioned as the classic example of RNA localization in eukaryotic cells. Studies of ß-actin mRNA over the past three decades have allowed understanding of how RBPs, such as ZBP1 (IGF2BP1), can control both RNA localization and translational status. Here, we summarize studies of ß-actin mRNA and focus on how ZBP1 serves as a model for understanding interactions between RNA and their binding protein(s). Central to the study of RNA and RBPs were technological developments that occurred along the way. We conclude with a future outlook highlighting new technologies that may be used to address still unanswered questions about RBP-mediated regulation of mRNA during its life cycle, within the cell.

10.
Curr Opin Cell Biol ; 45: 38-46, 2017 04.
Article in English | MEDLINE | ID: mdl-28258033

ABSTRACT

Regulation of gene expression is key determinant to cell structure and function. RNA localization, where specific mRNAs are transported to subcellular regions and then translated, is highly conserved in eukaryotes ranging from yeast to extremely specialized and polarized cells such as neurons. Messenger RNA and associated proteins (mRNP) move from the site of transcription in the nucleus to their final destination in the cytoplasm both passively through diffusion and actively via directed transport. Dysfunction of RNA localization, transport and translation machinery can lead to pathology. Single-molecule live-cell imaging techniques have revealed unique features of this journey with unprecedented resolution. In this review, we highlight key recent findings that have been made using these approaches and possible implications for spatial control of gene function.


Subject(s)
Active Transport, Cell Nucleus , Gene Expression Regulation , Ribonucleoproteins/metabolism , Animals , Cell Nucleus/metabolism , Cytoplasm/metabolism , Humans , RNA Transport , RNA, Messenger/metabolism , Ribonucleoproteins/analysis
11.
Proc Natl Acad Sci U S A ; 114(10): E1875-E1884, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28223507

ABSTRACT

RNA-protein interactions are essential for proper gene expression regulation, particularly in neurons with unique spatial constraints. Currently, these interactions are defined biochemically, but a method is needed to evaluate them quantitatively within morphological context. Colocalization of two-color labels using wide-field microscopy is a method to infer these interactions. However, because of chromatic aberrations in the objective lens, this approach lacks the resolution to determine whether two molecules are physically in contact or simply nearby by chance. Here, we developed a robust super registration methodology that corrected the chromatic aberration across the entire image field to within 10 nm, which is capable of determining whether two molecules are physically interacting or simply in proximity by random chance. We applied this approach to image single-molecule FISH in combination with immunofluorescence (smFISH-IF) and determined whether the association between an mRNA and binding protein(s) within a neuron was significant or accidental. We evaluated several mRNA-binding proteins identified from RNA pulldown assays to determine which of these exhibit bona fide interactions. Surprisingly, many known mRNA-binding proteins did not bind the mRNA in situ, indicating that adventitious interactions are significant using existing technology. This method provides an ability to evaluate two-color registration compatible with the scale of molecular interactions.


Subject(s)
Neurons/chemistry , Proteins/isolation & purification , RNA, Messenger/isolation & purification , RNA-Binding Proteins/isolation & purification , Fluorescent Dyes/chemistry , Gene Expression Regulation/genetics , In Situ Hybridization, Fluorescence/methods , Neurons/ultrastructure , Proteins/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Single Molecule Imaging/methods
12.
Science ; 352(6292): 1430-5, 2016 Jun 17.
Article in English | MEDLINE | ID: mdl-27313041

ABSTRACT

Translation is the fundamental biological process converting mRNA information into proteins. Single-molecule imaging in live cells has illuminated the dynamics of RNA transcription; however, it is not yet applicable to translation. Here, we report single-molecule imaging of nascent peptides (SINAPS) to assess translation in live cells. The approach provides direct readout of initiation, elongation, and location of translation. We show that mRNAs coding for endoplasmic reticulum (ER) proteins are translated when they encounter the ER membrane. Single-molecule fluorescence recovery after photobleaching provides direct measurement of elongation speed (5 amino acids per second). In primary neurons, mRNAs are translated in proximal dendrites but repressed in distal dendrites and display "bursting" translation. This technology provides a tool with which to address the spatiotemporal translation mechanism of single mRNAs in living cells.


Subject(s)
Fluorescence Recovery After Photobleaching/methods , Molecular Imaging/methods , Neurons/metabolism , Protein Biosynthesis , RNA, Messenger/metabolism , Animals , Cell Line, Tumor , Dendrites/metabolism , Endoplasmic Reticulum/metabolism , Humans , Mice , Ribosomes/metabolism
13.
J Biol Chem ; 288(28): 20361-8, 2013 Jul 12.
Article in English | MEDLINE | ID: mdl-23720759

ABSTRACT

Cells have evolved to regulate the asymmetric distribution of specific mRNA targets to institute spatial and temporal control over gene expression. Over the last few decades, evidence has mounted as to the importance of localization elements in the mRNA sequence and their respective RNA-binding proteins. Live imaging methodologies have shown mechanistic details of this phenomenon. In this minireview, we focus on the advanced biochemical and cell imaging techniques used to tweeze out the finer aspects of mechanisms of mRNA movement.


Subject(s)
Cytoplasm/metabolism , RNA Precursors/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Animals , Cytoplasm/genetics , Humans , In Situ Hybridization , Microscopy, Fluorescence , Models, Genetic , RNA Precursors/genetics , RNA Transport , RNA, Messenger/genetics , RNA-Binding Proteins/genetics
14.
Nature ; 495(7439): 121-5, 2013 Mar 07.
Article in English | MEDLINE | ID: mdl-23434754

ABSTRACT

More than half of mammalian genes generate multiple messenger RNA isoforms that differ in their 3' untranslated regions (3' UTRs) and therefore in regulatory sequences, often associated with cell proliferation and cancer; however, the mechanisms coordinating alternative 3'-UTR processing for specific mRNA populations remain poorly defined. Here we report that the cytoplasmic polyadenylation element binding protein 1 (CPEB1), an RNA-binding protein that regulates mRNA translation, also controls alternative 3'-UTR processing. CPEB1 shuttles to the nucleus, where it co-localizes with splicing factors and mediates shortening of hundreds of mRNA 3' UTRs, thereby modulating their translation efficiency in the cytoplasm. CPEB1-mediated 3'-UTR shortening correlates with cell proliferation and tumorigenesis. CPEB1 binding to pre-mRNAs not only directs the use of alternative polyadenylation sites, but also changes alternative splicing by preventing U2AF65 recruitment. Our results reveal a novel function of CPEB1 in mediating alternative 3'-UTR processing, which is coordinated with regulation of mRNA translation, through its dual nuclear and cytoplasmic functions.


Subject(s)
3' Untranslated Regions/genetics , Alternative Splicing/genetics , Protein Biosynthesis/genetics , Transcription Factors/metabolism , mRNA Cleavage and Polyadenylation Factors/metabolism , Cell Cycle Proteins/genetics , Cell Nucleus/metabolism , Cell Proliferation , Cell Transformation, Neoplastic , Cytoplasm/metabolism , HeLa Cells , Humans , Models, Genetic , Nuclear Proteins/metabolism , Poly A/genetics , Poly A/metabolism , Poly-ADP-Ribose Binding Proteins , Polyadenylation/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribonucleoproteins/metabolism , Splicing Factor U2AF
15.
Nat Med ; 18(1): 83-90, 2011 Dec 04.
Article in English | MEDLINE | ID: mdl-22138752

ABSTRACT

Malignant transformation, invasion and angiogenesis rely on the coordinated reprogramming of gene expression in the cells from which the tumor originated. Although deregulated gene expression has been extensively studied at genomic and epigenetic scales, the contribution of the regulation of mRNA-specific translation to this reprogramming is not well understood. Here we show that cytoplasmic polyadenylation element binding protein 4 (CPEB4), an RNA binding protein that mediates meiotic mRNA cytoplasmic polyadenylation and translation, is overexpressed in pancreatic ductal adenocarcinomas and glioblastomas, where it supports tumor growth, vascularization and invasion. We also show that, in pancreatic tumors, the pro-oncogenic functions of CPEB4 originate in the translational activation of mRNAs that are silenced in normal tissue, including the mRNA of tissue plasminogen activator, a key contributor to pancreatic ductal adenocarcinoma malignancy. Taken together, our results document a key role for post-transcriptional gene regulation in tumor development and describe a detailed mechanism for gene expression reprogramming underlying malignant tumor progression.


Subject(s)
Adenocarcinoma/pathology , Glioblastoma/pathology , Pancreatic Ducts/pathology , Pancreatic Neoplasms/pathology , Protein Biosynthesis/genetics , RNA-Binding Proteins/metabolism , Adenocarcinoma/blood supply , Adenocarcinoma/genetics , Animals , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/blood supply , Glioblastoma/genetics , Humans , Male , Mice , Mice, Nude , Neoplasm Invasiveness/genetics , Neovascularization, Pathologic/genetics , Pancreatic Ducts/blood supply , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/genetics , Polyadenylation , RNA, Small Interfering/genetics , RNA-Binding Proteins/genetics
16.
Nat Cell Biol ; 10(7): 858-65, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18536713

ABSTRACT

Meiotic progression requires the translational activation of stored maternal mRNAs, such as those encoding cyclin B1 or mos. The translation of these mRNAs is regulated by the cytoplasmic polyadenylation element (CPE) present in their 3'UTRs, which recruits the CPE-binding protein CPEB. This RNA-binding protein not only dictates the timing and extent of translational activation by cytoplasmic polyadenylation but also participates, together with the translational repressor Maskin, in the transport and localization, in a quiescent state, of its targets to subcellular locations where their translation will take place. During the early development of Xenopus laevis, CPEB localizes at the animal pole of oocytes and later on at embryonic spindles and centrosomes. Disruption of embryonic CPEB-mediated translational regulation results in abnormalities in the mitotic apparatus and inhibits embryonic mitosis. Here we show that spindle-localized translational activation of CPE-regulated mRNAs, encoding for proteins with a known function in spindle assembly and chromosome segregation, is essential for completion of the first meiotic division and for chromosome segregation in Xenopus oocytes.


Subject(s)
Chromosome Segregation , Gene Expression Regulation, Developmental , Meiosis/physiology , Oocytes , Regulatory Sequences, Nucleic Acid , Spindle Apparatus/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cyclin B/genetics , Cyclin B/metabolism , Cyclin B1 , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Kinesins/genetics , Kinesins/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oocytes/cytology , Oocytes/physiology , Phosphoproteins/genetics , Phosphoproteins/metabolism , Polyadenylation , Protein Biosynthesis , Proto-Oncogene Proteins c-mos/genetics , Proto-Oncogene Proteins c-mos/metabolism , Xenopus Proteins/genetics , Xenopus Proteins/metabolism , Xenopus laevis/embryology
SELECTION OF CITATIONS
SEARCH DETAIL
...