Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 4123, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750014

ABSTRACT

Avian influenza A viruses (IAVs) pose a public health threat, as they are capable of triggering pandemics by crossing species barriers. Replication of avian IAVs in mammalian cells is hindered by species-specific variation in acidic nuclear phosphoprotein 32 (ANP32) proteins, which are essential for viral RNA genome replication. Adaptive mutations enable the IAV RNA polymerase (FluPolA) to surmount this barrier. Here, we present cryo-electron microscopy structures of monomeric and dimeric avian H5N1 FluPolA with human ANP32B. ANP32B interacts with the PA subunit of FluPolA in the monomeric form, at the site used for its docking onto the C-terminal domain of host RNA polymerase II during viral transcription. ANP32B acts as a chaperone, guiding FluPolA towards a ribonucleoprotein-associated FluPolA to form an asymmetric dimer-the replication platform for the viral genome. These findings offer insights into the molecular mechanisms governing IAV genome replication, while enhancing our understanding of the molecular processes underpinning mammalian adaptations in avian-origin FluPolA.


Subject(s)
Cryoelectron Microscopy , Genome, Viral , Influenza A Virus, H5N1 Subtype , Nuclear Proteins , Virus Replication , Humans , Influenza A Virus, H5N1 Subtype/genetics , Virus Replication/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/chemistry , Animals , RNA-Dependent RNA Polymerase/metabolism , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/chemistry , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Viral Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/chemistry , Adaptation, Physiological/genetics , Influenza, Human/virology , RNA, Viral/metabolism , RNA, Viral/genetics , HEK293 Cells , Protein Multimerization , Models, Molecular
2.
PLoS Biol ; 21(11): e3002370, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37943954

ABSTRACT

During influenza A virus infection, the viral RNA polymerase transcribes the viral negative-sense segmented RNA genome and replicates it in a two-step process via complementary RNA within viral ribonucleoprotein (vRNP) complexes. While numerous viral and host factors involved in vRNP functions have been identified, dissecting the roles of individual factors remains challenging due to the complex cellular environment in which vRNP activity has been studied. To overcome this challenge, we reconstituted viral transcription and a full cycle of replication in a test tube using vRNPs isolated from virions and recombinant factors essential for these processes. This novel system uncovers the minimal components required for influenza virus replication and also reveals new roles of regulatory factors in viral replication. Moreover, it sheds light on the molecular interplay underlying the temporal regulation of viral transcription and replication. Our highly robust in vitro system enables systematic functional analysis of factors modulating influenza virus vRNP activity and paves the way for imaging key steps of viral transcription and replication.


Subject(s)
Influenza A virus , Influenza, Human , Orthomyxoviridae , Humans , Influenza A virus/genetics , Influenza, Human/genetics , Viral Proteins/genetics , Viral Proteins/metabolism , Ribonucleoproteins/genetics , Virus Replication/physiology , RNA, Viral/genetics
3.
Nucleic Acids Res ; 50(10): 5713-5725, 2022 06 10.
Article in English | MEDLINE | ID: mdl-35639917

ABSTRACT

The segmented negative-sense RNA genome of influenza A virus is assembled into ribonucleoprotein complexes (RNP) with viral RNA-dependent RNA polymerase and nucleoprotein (NP). It is in the context of these RNPs that the polymerase transcribes and replicates viral RNA (vRNA). Host acidic nuclear phosphoprotein 32 (ANP32) family proteins play an essential role in vRNA replication by mediating the dimerization of the viral polymerase via their N-terminal leucine-rich repeat (LRR) domain. However, whether the C-terminal low-complexity acidic region (LCAR) plays a role in RNA synthesis remains unknown. Here, we report that the LCAR is required for viral genome replication during infection. Specifically, we show that the LCAR directly interacts with NP and this interaction is mutually exclusive with RNA. Furthermore, we show that the replication of a short vRNA-like template that can be replicated in the absence of NP is less sensitive to LCAR truncations compared with the replication of full-length vRNA segments which is NP-dependent. We propose a model in which the LCAR interacts with NP to promote NP recruitment to nascent RNA during influenza virus replication, ensuring the co-replicative assembly of RNA into RNPs.


Subject(s)
Nuclear Proteins , RNA, Viral , Genome, Viral , Nuclear Proteins/metabolism , Nucleocapsid Proteins/genetics , Nucleoproteins/genetics , Nucleoproteins/metabolism , Phosphoproteins/genetics , RNA, Viral/genetics , RNA, Viral/metabolism , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Virus Replication/genetics
4.
J Virol ; 96(5): e0197921, 2022 03 09.
Article in English | MEDLINE | ID: mdl-35019720

ABSTRACT

Influenza A virus (IAV) contains a segmented RNA genome that is transcribed and replicated by the viral RNA polymerase in the cell nucleus. Replicated RNA segments are assembled with viral polymerase and oligomeric nucleoprotein into viral ribonucleoprotein (vRNP) complexes which are exported from the nucleus and transported across the cytoplasm to be packaged into progeny virions. Host GTPase Rab11a associated with recycling endosomes is believed to contribute to this process by mediating the cytoplasmic transport of vRNPs. However, how vRNPs interact with Rab11a remains poorly understood. In this study, we utilized a combination of biochemical, proteomic, and biophysical approaches to characterize the interaction between the viral polymerase and Rab11a. Using pulldown assays, we showed that vRNPs but not complementary RNPs (cRNPs) from infected cell lysates bind to Rab11a. We also showed that the viral polymerase directly interacts with Rab11a and that the C-terminal two-thirds of the PB2 polymerase subunit (PB2-C) comprising the cap-binding, mid-link, 627, and nuclear localization signal (NLS) domains mediate this interaction. Small-angle X-ray scattering (SAXS) experiments confirmed that PB2-C associates with Rab11a in solution forming a compact folded complex with a 1:1 stoichiometry. Furthermore, we demonstrate that the switch I region of Rab11a, which has been shown to be important for binding Rab11 family-interacting proteins (Rab11-FIPs), is also important for PB2-C binding, suggesting that IAV polymerase and Rab11-FIPs compete for the same binding site. Our findings expand our understanding of the interaction between the IAV polymerase and Rab11a in the cytoplasmic transport of vRNPs. IMPORTANCE The influenza virus RNA genome segments are replicated in the cell nucleus and are assembled into viral ribonucleoprotein (vRNP) complexes with viral RNA polymerase and nucleoprotein (NP). Replicated vRNPs need to be exported from the nucleus and trafficked across the cytoplasm to the cell membrane, where virion assembly takes place. The host GTPase Rab11a plays a role in vRNP trafficking. In this study, we showed that the viral polymerase directly interacts with Rab11a mediating the interaction between vRNPs and Rab11a. We mapped this interaction to the C-terminal domains of the PB2 polymerase subunit and the switch I region of Rab11a. Identifying the exact site of Rab11a binding on the viral polymerase could uncover a novel target site for the development of an influenza antiviral drug.


Subject(s)
GTP Phosphohydrolases , Influenza A virus , RNA, Viral , RNA-Dependent RNA Polymerase , Viral Proteins , Virus Replication , GTP Phosphohydrolases/metabolism , Influenza A virus/enzymology , Influenza A virus/genetics , Nucleoproteins/metabolism , Protein Binding , Protein Domains , Protein Transport/genetics , Proteomics , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Ribonucleoproteins/metabolism , Scattering, Small Angle , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication/genetics
5.
Nat Commun ; 13(1): 251, 2022 01 11.
Article in English | MEDLINE | ID: mdl-35017564

ABSTRACT

Influenza A viruses cause seasonal epidemics and global pandemics, representing a considerable burden to healthcare systems. Central to the replication cycle of influenza viruses is the viral RNA-dependent RNA polymerase which transcribes and replicates the viral RNA genome. The polymerase undergoes conformational rearrangements and interacts with viral and host proteins to perform these functions. Here we determine the structure of the 1918 influenza virus polymerase in transcriptase and replicase conformations using cryo-electron microscopy (cryo-EM). We then structurally and functionally characterise the binding of single-domain nanobodies to the polymerase of the 1918 pandemic influenza virus. Combining these functional and structural data we identify five sites on the polymerase which are sensitive to inhibition by nanobodies. We propose that the binding of nanobodies at these sites either prevents the polymerase from assuming particular functional conformations or interactions with viral or host factors. The polymerase is highly conserved across the influenza A subtypes, suggesting these sites as effective targets for potential influenza antiviral development.


Subject(s)
DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/genetics , Orthomyxoviridae/genetics , Pandemics , Single-Domain Antibodies/chemistry , Animals , Cryoelectron Microscopy , Genome, Viral , HEK293 Cells , Humans , Influenza A virus/genetics , Models, Molecular , Protein Binding , Protein Conformation , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase , Sf9 Cells , Single-Domain Antibodies/genetics , Viral Proteins/chemistry , Viral Proteins/genetics
6.
Nucleic Acids Res ; 50(3): 1484-1500, 2022 02 22.
Article in English | MEDLINE | ID: mdl-35037045

ABSTRACT

The SARS-CoV-2 coronavirus is the causal agent of the current global pandemic. SARS-CoV-2 belongs to an order, Nidovirales, with very large RNA genomes. It is proposed that the fidelity of coronavirus (CoV) genome replication is aided by an RNA nuclease complex, comprising the non-structural proteins 14 and 10 (nsp14-nsp10), an attractive target for antiviral inhibition. Our results validate reports that the SARS-CoV-2 nsp14-nsp10 complex has RNase activity. Detailed functional characterization reveals nsp14-nsp10 is a versatile nuclease capable of digesting a wide variety of RNA structures, including those with a blocked 3'-terminus. Consistent with a role in maintaining viral genome integrity during replication, we find that nsp14-nsp10 activity is enhanced by the viral RNA-dependent RNA polymerase complex (RdRp) consisting of nsp12-nsp7-nsp8 (nsp12-7-8) and demonstrate that this stimulation is mediated by nsp8. We propose that the role of nsp14-nsp10 in maintaining replication fidelity goes beyond classical proofreading by purging the nascent replicating RNA strand of a range of potentially replication-terminating aberrations. Using our developed assays, we identify drug and drug-like molecules that inhibit nsp14-nsp10, including the known SARS-CoV-2 major protease (Mpro) inhibitor ebselen and the HIV integrase inhibitor raltegravir, revealing the potential for multifunctional inhibitors in COVID-19 treatment.


Subject(s)
Antiviral Agents/pharmacology , Drug Evaluation, Preclinical , Exoribonucleases/metabolism , Genome, Viral/genetics , Genomic Instability , SARS-CoV-2/enzymology , SARS-CoV-2/genetics , Viral Nonstructural Proteins/metabolism , Viral Regulatory and Accessory Proteins/metabolism , Coronavirus RNA-Dependent RNA Polymerase/metabolism , Exoribonucleases/antagonists & inhibitors , Genome, Viral/drug effects , Genomic Instability/drug effects , Genomic Instability/genetics , HIV Integrase Inhibitors/pharmacology , Isoindoles/pharmacology , Multienzyme Complexes/antagonists & inhibitors , Multienzyme Complexes/metabolism , Organoselenium Compounds/pharmacology , RNA, Viral/biosynthesis , RNA, Viral/genetics , Raltegravir Potassium/pharmacology , SARS-CoV-2/drug effects , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Regulatory and Accessory Proteins/antagonists & inhibitors , Virus Replication/drug effects , Virus Replication/genetics
7.
Nucleic Acids Res ; 49(22): 13019-13030, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34850141

ABSTRACT

SARS-CoV-2 is a positive-sense RNA virus responsible for the Coronavirus Disease 2019 (COVID-19) pandemic, which continues to cause significant morbidity, mortality and economic strain. SARS-CoV-2 can cause severe respiratory disease and death in humans, highlighting the need for effective antiviral therapies. The RNA synthesis machinery of SARS-CoV-2 is an ideal drug target and consists of non-structural protein 12 (nsp12), which is directly responsible for RNA synthesis, and numerous co-factors involved in RNA proofreading and 5' capping of viral RNAs. The formation of the 5' 7-methylguanosine (m7G) cap structure is known to require a guanylyltransferase (GTase) as well as a 5' triphosphatase and methyltransferases; however, the mechanism of SARS-CoV-2 RNA capping remains poorly understood. Here we find that SARS-CoV-2 nsp12 is involved in viral RNA capping as a GTase, carrying out the addition of a GTP nucleotide to the 5' end of viral RNA via a 5' to 5' triphosphate linkage. We further show that the nsp12 NiRAN (nidovirus RdRp-associated nucleotidyltransferase) domain performs this reaction, and can be inhibited by remdesivir triphosphate, the active form of the antiviral drug remdesivir. These findings improve understanding of coronavirus RNA synthesis and highlight a new target for novel or repurposed antiviral drugs against SARS-CoV-2.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Antiviral Agents/pharmacology , Coronavirus RNA-Dependent RNA Polymerase/metabolism , Nucleotidyltransferases/antagonists & inhibitors , RNA, Viral/biosynthesis , SARS-CoV-2/enzymology , Adenosine Triphosphate/pharmacology , Coronavirus RNA-Dependent RNA Polymerase/antagonists & inhibitors , Genome, Viral/genetics , Guanosine/analogs & derivatives , Guanosine/metabolism , Humans , Nucleotidyltransferases/metabolism , RNA Caps/genetics , SARS-CoV-2/genetics , Vaccinia virus/enzymology , Vaccinia virus/metabolism , COVID-19 Drug Treatment
8.
Acta Crystallogr F Struct Biol Commun ; 77(Pt 7): 208-214, 2021 Jul 01.
Article in English | MEDLINE | ID: mdl-34196611

ABSTRACT

Influenza A viruses of the H1N1 and H3N2 subtypes are responsible for seasonal epidemic events. The influenza nucleoprotein (NP) binds to the viral genomic RNA and is essential for its replication. Efforts are under way to produce therapeutics and vaccines targeting the NP. Despite this, no structure of an NP from an H3N2 virus has previously been determined. Here, the structure of the A/Northern Territory/60/1968 (H3N2) influenza virus NP is presented at 2.2 Šresolution. The structure is highly similar to those of the A/WSN/1933 (H1N1) and A/Hong Kong/483/97 (H5N1) NPs. Nonconserved amino acids are widely dispersed both at the sequence and structural levels. A movement of the 73-90 RNA-binding loop is observed to be the key difference between the structure determined here and previous structures. The data presented here increase the understanding of structural conservation amongst influenza NPs and may aid in the design of universal interventions against influenza.


Subject(s)
Influenza A Virus, H3N2 Subtype/chemistry , Influenza A Virus, H3N2 Subtype/genetics , Nucleoproteins/chemistry , Nucleoproteins/genetics , Amino Acid Sequence , Crystallography, X-Ray/methods , Humans , Influenza, Human/genetics , Protein Structure, Secondary , Protein Structure, Tertiary
9.
Nature ; 587(7835): 638-643, 2020 11.
Article in English | MEDLINE | ID: mdl-33208942

ABSTRACT

Aquatic birds represent a vast reservoir from which new pandemic influenza A viruses can emerge1. Influenza viruses contain a negative-sense segmented RNA genome that is transcribed and replicated by the viral heterotrimeric RNA polymerase (FluPol) in the context of viral ribonucleoprotein complexes2,3. RNA polymerases of avian influenza A viruses (FluPolA) replicate viral RNA inefficiently in human cells because of species-specific differences in acidic nuclear phosphoprotein 32 (ANP32), a family of essential host proteins for FluPol activity4. Host-adaptive mutations, particularly a glutamic-acid-to-lysine mutation at amino acid residue 627 (E627K) in the 627 domain of the PB2 subunit, enable avian FluPolA to overcome this restriction and efficiently replicate viral RNA in the presence of human ANP32 proteins. However, the molecular mechanisms of genome replication and the interplay with ANP32 proteins remain largely unknown. Here we report cryo-electron microscopy structures of influenza C virus polymerase (FluPolC) in complex with human and chicken ANP32A. In both structures, two FluPolC molecules form an asymmetric dimer bridged by the N-terminal leucine-rich repeat domain of ANP32A. The C-terminal low-complexity acidic region of ANP32A inserts between the two juxtaposed PB2 627 domains of the asymmetric FluPolA dimer, suggesting a mechanism for how the adaptive PB2(E627K) mutation enables the replication of viral RNA in mammalian hosts. We propose that this complex represents a replication platform for the viral RNA genome, in which one of the FluPol molecules acts as a replicase while the other initiates the assembly of the nascent replication product into a viral ribonucleoprotein complex.


Subject(s)
Cryoelectron Microscopy , Gammainfluenzavirus/enzymology , Host-Pathogen Interactions , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/metabolism , Animals , Chickens/virology , Genome, Viral/genetics , HEK293 Cells , Humans , Gammainfluenzavirus/genetics , Models, Molecular , Nuclear Proteins/ultrastructure , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology , Protein Multimerization , RNA, Viral/biosynthesis , RNA, Viral/genetics , RNA-Binding Proteins/ultrastructure , RNA-Dependent RNA Polymerase/ultrastructure , Sf9 Cells
10.
bioRxiv ; 2020 Jan 06.
Article in English | MEDLINE | ID: mdl-32511388

ABSTRACT

Influenza A virus and coronavirus strains cause a mild to severe respiratory disease that can result in death. Although vaccines exist against circulating influenza A viruses, such vaccines are ineffective against emerging pandemic influenza A viruses. Currently, no vaccine exists against coronavirus infections, including pandemic SARS-CoV-2, the causative agent of the Coronavirus Disease 2019 (COVID-19). To combat these RNA virus infections, alternative antiviral strategies are needed. A key drug target is the viral RNA polymerase, which is responsible for viral RNA synthesis. In January 2020, the World Health Organisation identified enisamium as a candidate therapeutic against SARS-CoV-2. Enisamium is an isonicotinic acid derivative that is an inhibitor of multiple influenza B and A virus strains in cell culture and clinically approved in 11 countries. Here we show using in vitro assays that enisamium and its putative metabolite, VR17-04, inhibit the activity of the influenza virus and the SARS-CoV-2 RNA polymerase. VR17-04 displays similar efficacy against the SARS-CoV-2 RNA polymerase as the nucleotide analogue remdesivir triphosphate. These results suggest that enisamium is a broad-spectrum small molecule inhibitor of RNA virus RNA synthesis, and implicate it as a possible therapeutic option for treating SARS-CoV-2 infection. Unlike remdesivir, enisamium does not require intravenous administration which may be advantageous for the development of COVID-19 treatments outside a hospital setting.

11.
Nature ; 573(7773): 287-290, 2019 09.
Article in English | MEDLINE | ID: mdl-31485076

ABSTRACT

Influenza A viruses are responsible for seasonal epidemics, and pandemics can arise from the transmission of novel zoonotic influenza A viruses to humans1,2. Influenza A viruses contain a segmented negative-sense RNA genome, which is transcribed and replicated by the viral-RNA-dependent RNA polymerase (FluPolA) composed of PB1, PB2 and PA subunits3-5. Although the high-resolution crystal structure of FluPolA of bat influenza A virus has previously been reported6, there are no complete structures available for human and avian FluPolA. Furthermore, the molecular mechanisms of genomic viral RNA (vRNA) replication-which proceeds through a complementary RNA (cRNA) replicative intermediate, and requires oligomerization of the polymerase7-10-remain largely unknown. Here, using crystallography and cryo-electron microscopy, we determine the structures of FluPolA from human influenza A/NT/60/1968 (H3N2) and avian influenza A/duck/Fujian/01/2002 (H5N1) viruses at a resolution of 3.0-4.3 Å, in the presence or absence of a cRNA or vRNA template. In solution, FluPolA forms dimers of heterotrimers through the C-terminal domain of the PA subunit, the thumb subdomain of PB1 and the N1 subdomain of PB2. The cryo-electron microscopy structure of monomeric FluPolA bound to the cRNA template reveals a binding site for the 3' cRNA at the dimer interface. We use a combination of cell-based and in vitro assays to show that the interface of the FluPolA dimer is required for vRNA synthesis during replication of the viral genome. We also show that a nanobody (a single-domain antibody) that interferes with FluPolA dimerization inhibits the synthesis of vRNA and, consequently, inhibits virus replication in infected cells. Our study provides high-resolution structures of medically relevant FluPolA, as well as insights into the replication mechanisms of the viral RNA genome. In addition, our work identifies sites in FluPolA that could be targeted in the development of antiviral drugs.


Subject(s)
Genome, Viral/genetics , Influenza A Virus, H3N2 Subtype/enzymology , Influenza A Virus, H5N1 Subtype/enzymology , Models, Molecular , RNA-Dependent RNA Polymerase/chemistry , Cryoelectron Microscopy , Crystallization , Protein Structure, Tertiary , Single-Domain Antibodies/metabolism , Virus Replication
12.
Nucleic Acids Res ; 45(19): 11043-11055, 2017 Nov 02.
Article in English | MEDLINE | ID: mdl-28977553

ABSTRACT

In prokaryotes, RNA polymerase and ribosomes can bind concurrently to the same RNA transcript, leading to the functional coupling of transcription and translation. The interactions between RNA polymerase and ribosomes are crucial for the coordination of transcription with translation. Here, we report that RNA polymerase directly binds ribosomes and isolated large and small ribosomal subunits. RNA polymerase and ribosomes form a one-to-one complex with a micromolar dissociation constant. The formation of the complex is modulated by the conformational and functional states of RNA polymerase and the ribosome. The binding interface on the large ribosomal subunit is buried by the small subunit during protein synthesis, whereas that on the small subunit remains solvent-accessible. The RNA polymerase binding site on the ribosome includes that of the isolated small ribosomal subunit. This direct interaction between RNA polymerase and ribosomes may contribute to the coupling of transcription to translation.


Subject(s)
DNA-Directed RNA Polymerases/metabolism , Escherichia coli Proteins/metabolism , Protein Biosynthesis , Ribosome Subunits/metabolism , Transcription, Genetic , DNA-Directed RNA Polymerases/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Kinetics , Models, Molecular , Protein Binding , Protein Domains , Ribosome Subunits/chemistry , Ribosome Subunits/genetics
13.
J Biol Chem ; 290(34): 20856-20864, 2015 Aug 21.
Article in English | MEDLINE | ID: mdl-26163516

ABSTRACT

The translational GTPase BipA regulates the expression of virulence and pathogenicity factors in several eubacteria. BipA-dependent expression of virulence factors occurs under starvation conditions, such as encountered during infection of a host. Under these conditions, BipA associates with the small ribosomal subunit. BipA also has a second function to promote the efficiency of late steps in biogenesis of large ribosomal subunits at low temperatures, presumably while bound to the ribosome. During starvation, the cellular concentration of stress alarmone guanosine-3', 5'-bis pyrophosphate (ppGpp) is increased. This increase allows ppGpp to bind to BipA and switch its binding specificity from ribosomes to small ribosomal subunits. A conformational change of BipA upon ppGpp binding could explain the ppGpp regulation of the binding specificity of BipA. Here, we present the structures of the full-length BipA from Escherichia coli in apo, GDP-, and ppGpp-bound forms. The crystal structure and small-angle x-ray scattering data of the protein with bound nucleotides, together with a thermodynamic analysis of the binding of GDP and of ppGpp to BipA, indicate that the ppGpp-bound form of BipA adopts the structure of the GDP form. This suggests furthermore, that the switch in binding preference only occurs when both ppGpp and the small ribosomal subunit are present. This molecular mechanism would allow BipA to interact with both the ribosome and the small ribosomal subunit during stress response.


Subject(s)
Apoproteins/chemistry , Enteropathogenic Escherichia coli/genetics , Enteropathogenic Escherichia coli/pathogenicity , Escherichia coli Proteins/chemistry , GTP Phosphohydrolases/chemistry , Guanosine Diphosphate/chemistry , Phosphoproteins/chemistry , Pyrophosphatases/chemistry , Apoproteins/genetics , Apoproteins/metabolism , Crystallography, X-Ray , Enteropathogenic Escherichia coli/enzymology , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Gene Expression , Guanosine Diphosphate/metabolism , Kinetics , Models, Molecular , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Binding , Protein Structure, Tertiary , Pyrophosphatases/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ribosome Subunits, Small/genetics , Ribosome Subunits, Small/metabolism , Signal Transduction , Stress, Physiological , Thermodynamics , Virulence
14.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 5): 830-7, 2013 May.
Article in English | MEDLINE | ID: mdl-23633592

ABSTRACT

Plant Deg5 and Deg8 are two members of the HtrA proteases, a family of oligomeric serine endopeptidases that are involved in a variety of protein quality-control processes. These two HtrA proteases are located in the thylakoid lumen and participate in high-light stress responses by collaborating with other chloroplast proteins. Deg5 and Deg8 degrade photodamaged D1 protein of the photosystem II reaction centre, allowing its in situ replacement. Here, the crystal structures of Arabidopsis thaliana Deg5 (S266A) and Deg8 (S292A) are reported at 2.6 and 2.0 Å resolution, respectively. The Deg5 trimer contains two calcium ions in a central channel, suggesting a link between photodamage control and calcium ions in chloroplasts. Previous structures of HtrA proteases have indicated that their regulation usually requires C-terminal PDZ domain(s). Deg5 is unique in that it contains no PDZ domain and the trimeric structure of Deg5 (S266A) reveals a novel catalytic triad conformation. A similar triad conformation is observed in the hexameric structure of the single PDZ-domain-containing Deg8 (S292A). These findings suggest a novel activation mechanism for plant HtrA proteases and provide structural clues to their function in light-stress response.


Subject(s)
Arabidopsis Proteins/chemistry , PDZ Domains , Serine Endopeptidases/chemistry , Arabidopsis Proteins/metabolism , Calcium/metabolism , Catalytic Domain , Chloroplasts/metabolism , Crystallography, X-Ray , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/metabolism , Hydrogen Bonding , Light , Models, Molecular , Periplasmic Proteins/chemistry , Periplasmic Proteins/metabolism , Protein Conformation , Serine Endopeptidases/metabolism
15.
Article in English | MEDLINE | ID: mdl-23295491

ABSTRACT

Arabidopsis thaliana Deg8, an ATP-independent serine endopeptidase, is involved in the repair of photosystem II (PSII), specifically the degradation of the photo-damaged PSII reaction centre D1 protein. To understand the molecular mechanism underlying the participation of Deg8 in the degradation of the photo-damaged D1 protein, the structure of Deg8 is needed. Until recently, however, no structure of Deg8 had been solved. In this study, Deg8 from A. thaliana was cloned, overexpressed and purified in Escherichia coli. Crystallization was performed at 277 K using tribasic sodium citrate as the precipitant and the crystals diffracted to 2.0 Šresolution, belonging to space group C2 with unit-cell parameters a = 129.5, b = 124.2, c = 93.3 Å, α = γ = 90, ß = 132.4°. Assuming one trimer in the asymmetric unit, the Matthews coefficient and the solvent content were calculated to be 2.35 Å(3) Da(-1) and 47.6%, respectively.


Subject(s)
Arabidopsis Proteins/chemistry , Arabidopsis Proteins/isolation & purification , Serine Endopeptidases/chemistry , Serine Endopeptidases/isolation & purification , Arabidopsis Proteins/genetics , Base Sequence , Cloning, Molecular , Crystallization/methods , Crystallography, X-Ray , Molecular Sequence Data , Mutation , Protein Conformation , Serine Endopeptidases/genetics
16.
J Biol Chem ; 287(44): 37564-9, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-22961982

ABSTRACT

Eukaryotic organelles have developed elaborate protein quality control systems to ensure their normal activity, among which Deg/HtrA proteases play an essential role. Plant Deg2 protease is a homologue of prokaryotic DegQ/DegP proteases and is located in the chloroplast stroma, where its proteolytic activity is required to maintain the efficiency of photosynthetic machinery during stress. Here, we demonstrate that Deg2 exhibits dual protease-chaperone activities, and we present the hexameric structure of Deg2 complexed with co-purified peptides. The structure shows that Deg2 contains a unique second PDZ domain (PDZ2) following a conventional PDZ domain (PDZ1), with PDZ2 orchestrating the cage assembly of Deg2. We discovered a conserved internal ligand for PDZ2 that mediates hexamer formation and thus locks the protease in the resting state. These findings provide insight into the diverse modes of PDZ domain-mediated regulation of Deg proteases.


Subject(s)
Arabidopsis Proteins/chemistry , Arabidopsis/enzymology , Serine Endopeptidases/chemistry , Caseins/chemistry , Crystallography, X-Ray , Hydrogen Bonding , Hydrogen-Ion Concentration , Ligands , Protein Multimerization , Protein Structure, Quaternary , Protein Structure, Secondary , Protein Structure, Tertiary , Proteolysis
17.
Article in English | MEDLINE | ID: mdl-22750879

ABSTRACT

Arabidopsis thaliana Deg5 is an ATP-independent serine protease which resides on the luminal side of the thylakoid in chloroplasts. Deg5 and another Deg/HtrA-family protease, Deg8, have a synergistic function in the turnover of the D1 protein of photosystem II (PSII), which is prone to damage arising from high light exposure. An inactive mutant of the protein, Deg5(S266A), was overexpressed in Escherichia coli. After purification and crystallization, crystals that diffracted to 2.6 Šresolution were obtained. The crystals belonged to the monoclinic space group C2, with unit-cell parameters a = 109.1, b = 126.0, c = 83.3 Å, ß = 102.9°, and contained three molecules in the asymmetric unit. The calculated Matthews coefficient and solvent content were 3.0 Å(3) Da(-1) and 59.0%, respectively.


Subject(s)
Arabidopsis Proteins/chemistry , Arabidopsis/enzymology , Serine Endopeptidases/chemistry , Arabidopsis Proteins/genetics , Arabidopsis Proteins/isolation & purification , Crystallization , Crystallography, X-Ray , Gene Expression , Serine Endopeptidases/genetics , Serine Endopeptidases/isolation & purification
18.
J Antimicrob Chemother ; 65(8): 1681-7, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20513704

ABSTRACT

OBJECTIVES: T-2307, a novel arylamidine synthesized at Toyama Chemical Co., Ltd, has in vitro and in vivo broad-spectrum activities against pathogenic fungi. T-2307 particularly exhibits potent in vitro and in vivo activity against Candida albicans, suggesting that its uptake might be mediated by a transport system. In this report, we studied the uptake of T-2307 in C. albicans. METHODS: C. albicans cells and rat hepatocytes were exposed to 0.02 microM [(14)C]T-2307. After incubation, the reaction mixture was concentrated and layered on a silicon layer (mixture of silicon oil and liquid paraffin) inside a tube. The tube was then centrifuged to transfer cells into the bottom layer (sodium hydroxide) for solubilization. The bottom layer was neutralized and measured for radioactivity. RESULTS: T-2307 was concentrated from the extracellular medium by C. albicans cells in 10 mM phosphate buffer solution supplemented with 1% glucose by 3200- to 5100-fold. The accumulation was approximately two orders of magnitude greater than that achieved with a rat hepatocyte preparation. T-2307 uptake was sensitive to temperature and extracellular pH, and was reduced in the presence of inhibitors of mitochondrial respiration, oxidative phosphorylation and plasma membrane proton pump, and by an uncoupler. Furthermore, T-2307 uptake was concentration dependent and an Eadie-Hofstee plot suggested the involvement of two transport systems. CONCLUSIONS: The considerably higher concentrations of T-2307 were selectively accumulated in C. albicans via transporter-mediated systems, as compared with the concentrations in rat hepatocytes. This transporter-mediated uptake of T-2307 contributes to its potent anticandidal activity.


Subject(s)
Antifungal Agents/metabolism , Antifungal Agents/pharmacology , Candida albicans/drug effects , Candida albicans/metabolism , Animals , Biological Transport , Cells, Cultured , Hepatocytes/metabolism , Membrane Transport Proteins/metabolism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...