Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Nat Commun ; 15(1): 3729, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702330

ABSTRACT

The unique virus-cell interaction in Epstein-Barr virus (EBV)-associated malignancies implies targeting the viral latent-lytic switch is a promising therapeutic strategy. However, the lack of specific and efficient therapeutic agents to induce lytic cycle in these cancers is a major challenge facing clinical implementation. We develop a synthetic transcriptional activator that specifically activates endogenous BZLF1 and efficiently induces lytic reactivation in EBV-positive cancer cells. A lipid nanoparticle encapsulating nucleoside-modified mRNA which encodes a BZLF1-specific transcriptional activator (mTZ3-LNP) is synthesized for EBV-targeted therapy. Compared with conventional chemical inducers, mTZ3-LNP more efficiently activates EBV lytic gene expression in EBV-associated epithelial cancers. Here we show the potency and safety of treatment with mTZ3-LNP to suppress tumor growth in EBV-positive cancer models. The combination of mTZ3-LNP and ganciclovir yields highly selective cytotoxic effects of mRNA-based lytic induction therapy against EBV-positive tumor cells, indicating the potential of mRNA nanomedicine in the treatment of EBV-associated epithelial cancers.


Subject(s)
Epstein-Barr Virus Infections , Herpesvirus 4, Human , Liposomes , Nanoparticles , Trans-Activators , Humans , Herpesvirus 4, Human/genetics , Trans-Activators/metabolism , Trans-Activators/genetics , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/drug therapy , Animals , Nanoparticles/chemistry , Cell Line, Tumor , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Virus Activation/drug effects , Xenograft Model Antitumor Assays , Gene Expression Regulation, Viral/drug effects , Mice, Nude , Female
2.
Nat Commun ; 13(1): 1871, 2022 04 06.
Article in English | MEDLINE | ID: mdl-35387989

ABSTRACT

Three-dimensional (3D) structures of the genome are dynamic, heterogeneous and functionally important. Live cell imaging has become the leading method for chromatin dynamics tracking. However, existing CRISPR- and TALE-based genomic labeling techniques have been hampered by laborious protocols and are ineffective in labeling non-repetitive sequences. Here, we report a versatile CRISPR/Casilio-based imaging method that allows for a nonrepetitive genomic locus to be labeled using one guide RNA. We construct Casilio dual-color probes to visualize the dynamic interactions of DNA elements in single live cells in the presence or absence of the cohesin subunit RAD21. Using a three-color palette, we track the dynamic 3D locations of multiple reference points along a chromatin loop. Casilio imaging reveals intercellular heterogeneity and interallelic asynchrony in chromatin interaction dynamics, underscoring the importance of studying genome structures in 4D.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , RNA, Guide, Kinetoplastida , CRISPR-Cas Systems/genetics , Chromatin/genetics , Chromosomes , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Genomics , RNA, Guide, Kinetoplastida/genetics
3.
ACS Synth Biol ; 11(1): 116-124, 2022 01 21.
Article in English | MEDLINE | ID: mdl-34931802

ABSTRACT

Here we describe TALE.Sense, a versatile platform for sensing DNA sequences in live mammalian cells enabling programmable generation of a customable response that discerns cells containing specified sequence targets. The platform is based on the programmable DNA binding of transcription activator-like effector (TALE) coupled to conditional intein-reconstitution producing a trans-spliced ON-switch for a response circuit. TALE.Sense shows higher efficiency and dynamic range when compared to the reported zinc-finger based DNA-sensor in detecting same DNA sequences. Swapping transcriptional activation modules and introducing SunTag-based amplification loops to TALE.Sense circuits augment detection efficiency of the DNA sensor. The TALE.Sense platform shows versatility when applied to a range of target sites, indicating its suitability for applications to identify live cell variants with anticipated DNA sequences. TALE.Sense could be integrated with other cellular or synthetic circuits by using specified DNA sequences as control-switches, thus expanding the scope in connecting inducible modules for synthetic biology.


Subject(s)
DNA , Transcription Activator-Like Effectors , Animals , DNA/genetics , DNA/metabolism , Inteins , Mammals/genetics , Synthetic Biology , Transcription Activator-Like Effectors/genetics , Transcription Activator-Like Effectors/metabolism , Zinc Fingers/genetics
4.
Genome Biol ; 22(1): 295, 2021 10 18.
Article in English | MEDLINE | ID: mdl-34663425

ABSTRACT

BACKGROUND: Nanopore long-read sequencing technology greatly expands the capacity of long-range, single-molecule DNA-modification detection. A growing number of analytical tools have been developed to detect DNA methylation from nanopore sequencing reads. Here, we assess the performance of different methylation-calling tools to provide a systematic evaluation to guide researchers performing human epigenome-wide studies. RESULTS: We compare seven analytic tools for detecting DNA methylation from nanopore long-read sequencing data generated from human natural DNA at a whole-genome scale. We evaluate the per-read and per-site performance of CpG methylation prediction across different genomic contexts, CpG site coverage, and computational resources consumed by each tool. The seven tools exhibit different performances across the evaluation criteria. We show that the methylation prediction at regions with discordant DNA methylation patterns, intergenic regions, low CG density regions, and repetitive regions show room for improvement across all tools. Furthermore, we demonstrate that 5hmC levels at least partly contribute to the discrepancy between bisulfite and nanopore sequencing. Lastly, we provide an online DNA methylation database ( https://nanome.jax.org ) to display the DNA methylation levels detected by nanopore sequencing and bisulfite sequencing data across different genomic contexts. CONCLUSIONS: Our study is the first systematic benchmark of computational methods for detection of mammalian whole-genome DNA modifications in nanopore sequencing. We provide a broad foundation for cross-platform standardization and an evaluation of analytical tools designed for genome-scale modified base detection using nanopore sequencing.


Subject(s)
DNA Methylation , Epigenome , Nanopore Sequencing , Software , 5-Methylcytosine/analysis , CpG Islands , Genome, Human , Humans
5.
Nat Commun ; 10(1): 4296, 2019 09 20.
Article in English | MEDLINE | ID: mdl-31541098

ABSTRACT

Here we develop a methylation editing toolbox, Casilio-ME, that enables not only RNA-guided methylcytosine editing by targeting TET1 to genomic sites, but also by co-delivering TET1 and protein factors that couple methylcytosine oxidation to DNA repair activities, and/or promote TET1 to achieve enhanced activation of methylation-silenced genes. Delivery of TET1 activity by Casilio-ME1 robustly alters the CpG methylation landscape of promoter regions and activates methylation-silenced genes. We augment Casilio-ME1 to simultaneously deliver the TET1-catalytic domain and GADD45A (Casilio-ME2) or NEIL2 (Casilio-ME3) to streamline removal of oxidized cytosine intermediates to enhance activation of targeted genes. Using two-in-one effectors or modular effectors, Casilio-ME2 and Casilio-ME3 remarkably boost gene activation and methylcytosine demethylation of targeted loci. We expand the toolbox to enable a stable and expression-inducible system for broader application of the Casilio-ME platforms. This work establishes a platform for editing DNA methylation to enable research investigations interrogating DNA methylomes.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , DNA Demethylation , DNA Repair , RNA, Guide, Kinetoplastida/metabolism , 5-Methylcytosine/metabolism , CRISPR-Cas Systems , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , DNA Glycosylases/metabolism , DNA Methylation , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Gene Editing , HEK293 Cells , Humans , Mixed Function Oxygenases/genetics , Oxidation-Reduction , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Sequence Analysis, RNA
7.
Biosci Rep ; 35(2)2015 Apr 28.
Article in English | MEDLINE | ID: mdl-25772301

ABSTRACT

Accumulating evidence indicates that RNA metabolism components assemble into supramolecular cellular structures to mediate functional compartmentalization within the cytoplasmic membrane of the bacterial cell. This cellular compartmentalization could play important roles in the processes of RNA degradation and maturation. These components include Hfq, the RNA chaperone protein, which is involved in the post-transcriptional control of protein synthesis mainly by the virtue of its interactions with several small regulatory ncRNAs (sRNA). The Escherichia coli Hfq is structurally organized into two domains. An N-terminal domain that folds as strongly bent ß-sheets within individual protomers to assemble into a typical toroidal hexameric ring. A C-terminal flexible domain that encompasses approximately one-third of the protein seems intrinsically unstructured. RNA-binding function of Hfq mainly lies within its N-terminal core, whereas the function of the flexible domain remains controversial and largely unknown. In the present study, we demonstrate that the Hfq-C-terminal region (CTR) has an intrinsic property to self-assemble into long amyloid-like fibrillar structures in vitro. We show that normal localization of Hfq within membrane-associated coiled structures in vivo requires this C-terminal domain. This finding establishes for the first time a function for the hitherto puzzling CTR, with a plausible central role in RNA transactions.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli/chemistry , Host Factor 1 Protein/chemistry , RNA, Bacterial/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Host Factor 1 Protein/genetics , Host Factor 1 Protein/metabolism , Protein Binding , Protein Structure, Quaternary , Protein Structure, Tertiary , RNA, Bacterial/genetics , RNA, Bacterial/metabolism
8.
Methods Mol Biol ; 1259: 87-101, 2015.
Article in English | MEDLINE | ID: mdl-25579581

ABSTRACT

The ability to study the localization and organization of proteins within the tiny cells of bacteria, such as Escherichia coli, has paved the way for a new and exciting era of prokaryotic cellular biology. Previously unrecognized levels of spatiotemporal and supramolecular organization of proteins have been revealed within the prokaryotic cell that had long been assumed as a "bag of enzymes." Immunofluorescence (IF) microscopy, which involves cellular immunostaining of native proteins with fluorescently labeled antibodies, is relatively laborious and requires cell fixation and highly specific antibodies. However, IF microscopy allows localization studies of native proteins expressed to their normal cellular levels, as opposed to labeling proteins with large fluorescent tag that can alter protein abundance dependent on changes in mRNAs and/or proteins stability, or whose detection can require overexpression of labeled proteins. In addition, when antibodies against native proteins are not available or lack specificity, epitope tags such as hemagglutinin (HA) or Flag can be used to label chromosomally expressed proteins. The short Flag- and HA-tag, eight or nine amino acids, are unlikely to interfere with the localization or function of the proteins. We describe and discuss here the use of fluorescence microscopy for determination of cellular organization of protein components of the E. coli RNA processing and degradation machinery. We present examples of cellular organization patterns visualized by light microscopy, either by IF microscopy of native and epitope-tagged proteins in fixed cells, or by fluorescence labeling of the proteins in live cells.


Subject(s)
Escherichia coli/metabolism , RNA, Bacterial/metabolism , Escherichia coli Proteins/metabolism
9.
Integr Biol (Camb) ; 7(1): 128-41, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25407044

ABSTRACT

The bacterial actin-homolog MreB is a key player in bacterial cell-wall biosynthesis and is required for the maintenance of the rod-like morphology of Escherichia coli. However, how MreB cellular levels are adjusted to growth conditions is poorly understood. Here, we show that DsrA, an E. coli small noncoding RNA (sRNA), is involved in the post-transcriptional regulation of mreB. DsrA is required for the downregulation of MreB cellular concentration during environmentally induced slow growth-rates, mainly growth at low temperature and during the stationary phase. DsrA interacts in an Hfq-dependent manner with the 5' region of mreB mRNA, which contains signals for translation initiation and thereby affects mreB translation and stability. Moreover, as DsrA is also involved in the regulation of two transcriptional regulators, σ(S) and the nucleoid associated protein H-NS, which negatively regulate mreB transcription, it also indirectly contributes to mreB transcriptional down-regulation. By using quantitative analyses, our results evidence the complexity of this regulation and the tangled interplay between transcriptional and post-transcriptional control. As transcription factors and sRNA-mediated post-transcriptional regulators use different timescales, we propose that the sRNA pathway helps to adapt to changes in temperature, but also indirectly mediates long-term regulation of MreB concentration. The tight regulation and fine-tuning of mreB gene expression in response to cellular stresses is discussed in regard to the effect of the MreB protein on cell elongation.


Subject(s)
Escherichia coli Proteins/genetics , Escherichia coli/cytology , Escherichia coli/genetics , Gene Expression Regulation, Bacterial/genetics , Models, Genetic , RNA, Small Untranslated/genetics , Actins/genetics , Cell Size , Computer Simulation , Stress, Physiological/genetics , Transcriptional Activation/genetics
10.
Biosci Rep ; 34(6): e00166, 2014 Dec 23.
Article in English | MEDLINE | ID: mdl-25299745

ABSTRACT

Multiprotein complexes that carry out RNA degradation and processing functions are found in cells from all domains of life. In Escherichia coli, the RNA degradosome, a four-protein complex, is required for normal RNA degradation and processing. In addition to the degradosome complex, the cell contains other ribonucleases that also play important roles in RNA processing and/or degradation. Whether the other ribonucleases are associated with the degradosome or function independently is not known. In the present work, IP (immunoprecipitation) studies from cell extracts showed that the major hydrolytic exoribonuclease RNase II is associated with the known degradosome components RNaseE (endoribonuclease E), RhlB (RNA helicase B), PNPase (polynucleotide phosphorylase) and Eno (enolase). Further evidence for the RNase II-degradosome association came from the binding of RNase II to purified RNaseE in far western affinity blot experiments. Formation of the RNase II-degradosome complex required the degradosomal proteins RhlB and PNPase as well as a C-terminal domain of RNaseE that contains binding sites for the other degradosomal proteins. This shows that the RNase II is a component of the RNA degradosome complex, a previously unrecognized association that is likely to play a role in coupling and coordinating the multiple elements of the RNA degradation pathways.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Exoribonucleases/metabolism , RNA, Bacterial/metabolism , Binding Sites/genetics , Blotting, Far-Western , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Endoribonucleases/genetics , Endoribonucleases/metabolism , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Exoribonucleases/genetics , Immunoblotting , Immunoprecipitation , Models, Biological , Polyribonucleotide Nucleotidyltransferase/genetics , Polyribonucleotide Nucleotidyltransferase/metabolism , Protein Binding , RNA, Bacterial/genetics
11.
Biochem J ; 458(1): 11-22, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24266791

ABSTRACT

Bacterial RNA processing and degradation involves the co-ordinated action of a large number of RNases, RNA helicases and other proteins. It is not known how this functional network is organized within the cell nor how it is co-ordinated or regulated. In the present study, we show that multiple components of the RNA degradation and processing network of Escherichia coli are localized within extended cellular structures that appear to coil around the periphery of the cell. These include Orn, Hfq, PAP I, RNase III, RppH, RraA and RraB in addition to the previously reported proteins RNase II and RNaseE. Double-label localization studies of several of the proteins showed co-localization of the proteins within the observed structures. Assembly of the proteins into the structures was independent of the MreBCD or MinCDE cytoskeletal systems, RNA synthesis, or nucleoid positioning within the cell. Our results indicate that the components of the RNA processing and degradation network are compartmentalized within the cell rather than diffusely distributed in the cytoplasm. This sequestration provides the cell with a possible mechanism to control access to RNA substrates and to functionally co-ordinate the multiple players of the RNA processing and degradation pathways.


Subject(s)
Cell Compartmentation , Escherichia coli/genetics , RNA Processing, Post-Transcriptional , RNA, Bacterial/metabolism , Blotting, Western , Proteolysis
12.
J Biol Chem ; 288(10): 7241-51, 2013 Mar 08.
Article in English | MEDLINE | ID: mdl-23344958

ABSTRACT

The subcellular localization of the exoribonuclease RNase II is not known despite the advanced biochemical characterization of the enzyme. Here we report that RNase II is organized into cellular structures that appear to coil around the Escherichia coli cell periphery and that RNase II is associated with the cytoplasmic membrane by its amino-terminal amphipathic helix. The helix also acts as an autonomous transplantable membrane binding domain capable of directing normally cytoplasmic proteins to the membrane. Assembly of the organized cellular structures of RNase II required the RNase II amphipathic membrane binding domain. Co-immunoprecipitation of the protein from cell extracts indicated that RNase II interacts with itself. The RNase II self-interaction and the ability of the protein to assemble into organized cellular structures required the membrane binding domain. The ability of RNase II to maintain cell viability in the absence of the exoribonuclease polynucleotide phosphorylase was markedly diminished when the RNase II cellular structures were lost due to changes in the amphipathicity of the amino-terminal helix, suggesting that membrane association and assembly of RNase II into organized cellular structures play an important role in the normal function of the protein within the bacterial cell.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli/enzymology , Exoribonucleases/chemistry , Protein Structure, Secondary , Amino Acid Sequence , Binding Sites/genetics , Blotting, Western , Cell Membrane/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Exoribonucleases/genetics , Exoribonucleases/metabolism , Hydrophobic and Hydrophilic Interactions , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microbial Viability , Microscopy, Fluorescence , Models, Molecular , Mutation , Protein Binding , Protein Structure, Tertiary
13.
J Bacteriol ; 192(12): 3222-6, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20382767

ABSTRACT

The Escherichia coli RNA degradosome proteins are organized into a helical cytoskeletal-like structure within the cell. Here we describe the ATP-dependent assembly of the RhlB component of the degradosome into polymeric filamentous structures in vitro, which suggests that extended polymers of RhlB are likely to comprise a basic core element of the degradosome cytoskeletal structures.


Subject(s)
Cytoskeleton/metabolism , DEAD-box RNA Helicases/metabolism , Escherichia coli Proteins/metabolism , Adenosine Triphosphate/metabolism , DEAD-box RNA Helicases/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Gene Expression Regulation, Bacterial/physiology , Immunohistochemistry , Protein Transport
14.
Mol Microbiol ; 70(4): 780-2, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18990179

ABSTRACT

Ribonuclease E (RNase E) is a component of the Escherichia coli RNA degradosome, a multiprotein complex that also includes RNA helicase B (RhlB), polynucleotide phosphorylase (PNPase) and enolase. The degradosome plays a key role in RNA processing and degradation. The degradosomal proteins are organized as a cytoskeletal-like structure within the cell that has been thought to be associated with the cytoplasmic membrane. The article by Khemici et al. in the current issue of Molecular Microbiology reports that RNase E can directly interact with membrane phospholipids in vitro. The RNase E-membrane interaction is likely to play an important role in the membrane association of the degradosome system. These findings shed light on important but largely unexplored aspects of cellular structure and function, including the organization of the RNA processing machinery of the cell and of bacterial cytoskeletal elements in general.


Subject(s)
Endoribonucleases/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/genetics , Multienzyme Complexes/metabolism , Polyribonucleotide Nucleotidyltransferase/metabolism , RNA Helicases/metabolism , Cell Membrane/metabolism , Endoribonucleases/genetics , Escherichia coli/enzymology , Escherichia coli/ultrastructure , Escherichia coli Proteins/genetics , Multienzyme Complexes/genetics , Phospholipids/metabolism , Polyribonucleotide Nucleotidyltransferase/genetics , Protein Binding , RNA Helicases/genetics
15.
J Biol Chem ; 283(20): 13850-5, 2008 May 16.
Article in English | MEDLINE | ID: mdl-18337249

ABSTRACT

The RNA degradosome of Escherichia coli is a multiprotein complex that plays an essential role in normal RNA processing and decay. It was recently shown that the major degradosome constituents are organized in a coiled cytoskeletal-like structure that extends along the length of the cell. Here we show that the endoribonuclease E (RNaseE) and RNA helicase B (RhlB) components of the degradosome can each independently form coiled structures in the absence of the other degradosome proteins. In contrast, the cytoskeletal organization of the other degradosome proteins required the presence of the RNaseE or RhlB coiled elements. Although the RNaseE and RhlB structures were equally competent to support the helical organization of polynucleotide phosphorylase, the cytoskeletal-like organization of enolase occurred only in the presence of the RNaseE coiled structure. The results indicate that the RNA degradosome proteins are components of the bacterial cytoskeleton rather than existing as randomly distributed multiprotein complexes within the cell and suggest a model for the cellular organization of the components within the helical degradosomal structure.


Subject(s)
Cytoskeleton/metabolism , DEAD-box RNA Helicases/metabolism , Endoribonucleases/metabolism , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial , Multienzyme Complexes/metabolism , Polyribonucleotide Nucleotidyltransferase/metabolism , RNA Helicases/metabolism , Bacterial Proteins/metabolism , Escherichia coli/metabolism , Genotype , Luminescent Proteins/metabolism , Models, Biological , Molecular Conformation , Protein Conformation , Protein Structure, Tertiary , RNA/metabolism , RNA, Bacterial
16.
Proc Natl Acad Sci U S A ; 104(5): 1667-72, 2007 Jan 30.
Article in English | MEDLINE | ID: mdl-17242352

ABSTRACT

RNaseE is the main component of the RNA degradosome of Escherichia coli, which plays an essential role in RNA processing and decay. Localization studies showed that RNaseE and the other known degradosome components (RNA helicase B, polynucleotide phosphorylase, and enolase) are organized as helical filamentous structures that coil around the length of the cell. These resemble the helical structures formed by the MreB and MinD cytoskeletal proteins. Formation of the RNaseE cytoskeletal-like structure requires an internal domain of the protein that does not include the domains required for any of its known interactions or the minimal domain required for endonuclease activity. We conclude that the constituents of the RNA degradosome are components of the E. coli cytoskeleton, either assembled as a primary cytoskeletal structure or secondarily associated with another underlying cytoskeletal element. This suggests a previously unrecognized role for the bacterial cytoskeleton, providing a mechanism to compartmentalize proteins that act on cytoplasmic components, as exemplified by the RNA processing and degradative activities of the degradosome, to regulate their access to important cellular substrates.


Subject(s)
Cytoskeleton/metabolism , Endoribonucleases/physiology , Escherichia coli Proteins/chemistry , Escherichia coli/metabolism , RNA, Bacterial/chemistry , RNA/chemistry , Adenosine Triphosphatases/metabolism , Cytoplasm/metabolism , Endoribonucleases/metabolism , Escherichia coli Proteins/metabolism , Gene Library , Phenotype , Protein Structure, Tertiary , RNA Helicases/chemistry , RNA Stability , Two-Hybrid System Techniques
17.
J Bacteriol ; 188(8): 2993-3001, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16585760

ABSTRACT

Division site placement in Escherichia coli involves interactions of the MinD protein with MinC and MinE and with other MinD molecules to form membrane-associated polymeric structures. In this work, as part of a study of these interactions, we established that heterologous membrane-associated proteins such as MinD can be targeted to the yeast nuclear membrane, dependent only on the presence of a membrane-binding domain and a nuclear targeting sequence. Targeting to the nuclear membrane was equally effective using the intrinsic MinD membrane-targeting domain or the completely unrelated membrane-targeting domain of cytochrome b(5). The chimeric proteins differing in their membrane-targeting sequences were then used to establish the roles of membrane association and specificity of the membrane anchor in MinD interactions, using the yeast two-hybrid system. The chimeric proteins were also used to show that the membrane association of MinD and MinE in E. coli cells had no specificity for the membrane anchor, whereas formation of MinDE polar zones and MinE rings required the presence of the native MinD membrane-targeting sequence.


Subject(s)
Adenosine Triphosphatases/metabolism , Cell Membrane/metabolism , Escherichia coli Proteins/metabolism , Protein Transport/physiology , Adenosine Triphosphatases/genetics , Bacterial Proteins/analysis , Bacterial Proteins/genetics , Cell Cycle Proteins/metabolism , Cytochromes b5/genetics , Diagnosis, Differential , Escherichia coli/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Genes, Reporter , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Microscopy, Fluorescence , Models, Biological , Models, Molecular , Nuclear Envelope/chemistry , Protein Binding , Protein Sorting Signals/genetics , Protein Structure, Tertiary/genetics , Protein Structure, Tertiary/physiology , Protein Transport/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Staining and Labeling/methods , Two-Hybrid System Techniques , Yeasts/chemistry , Yeasts/genetics , Yeasts/metabolism
18.
Nat Rev Microbiol ; 3(12): 959-68, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16322744

ABSTRACT

The site of cell division in bacterial cells is placed with high fidelity at a designated position, usually the midpoint of the cell. In normal cell division in Escherichia coli this is accomplished by the action of the Min proteins, which maintain a high concentration of a septation inhibitor near the ends of the cell, and a low concentration at midcell. This leaves the midcell site as the only available location for formation of the division septum. In other species, such as Bacillus subtilis, this general paradigm is maintained, although some of the proteins differ and the mechanisms used to localize the proteins vary. A second mechanism of negative regulation, the nucleoid-occlusion system, prevents septa forming over nucleoids. This system functions in Gram-negative and Gram-positive bacteria, and is especially important in cells that lack the Min system or in cells in which nucleoid replication or segregation are defective. Here, we review the latest findings on these two systems.


Subject(s)
Cell Division/physiology , Escherichia coli/cytology , Adenosine Triphosphatases/physiology , Bacillus subtilis/cytology , Bacterial Proteins/physiology , Cell Cycle Proteins , Chloroplasts/ultrastructure , Cytoskeletal Proteins/physiology , DNA-Binding Proteins/physiology , Escherichia coli Proteins/physiology , Membrane Proteins/physiology , Spores, Bacterial/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...