Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.027
Filtrar
1.
PLoS One ; 19(4): e0300453, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38683783

RESUMEN

The activity-regulated cytoskeleton-associated protein (Arc) is a complex regulator of synaptic plasticity in glutamatergic neurons. Understanding its molecular function is key to elucidate the neurobiology of memory and learning, stress regulation, and multiple neurological and psychiatric diseases. The recent development of anti-Arc nanobodies has promoted the characterization of the molecular structure and function of Arc. This study aimed to validate two anti-Arc nanobodies, E5 and H11, as selective modulators of the human Arc N-lobe (Arc-NL), a domain that mediates several molecular functions of Arc through its peptide ligand binding site. The structural characteristics of recombinant Arc-NL-nanobody complexes were solved at atomic resolution using X-ray crystallography. Both anti-Arc nanobodies bind specifically to the multi-peptide binding site of Arc-NL. Isothermal titration calorimetry showed that the Arc-NL-nanobody interactions occur at nanomolar affinity, and that the nanobodies can displace a TARPγ2-derived peptide from the binding site. Thus, both anti-Arc-NL nanobodies could be used as competitive inhibitors of endogenous Arc ligands. Differences in the CDR3 loops between the two nanobodies indicate that the spectrum of short linear motifs recognized by the Arc-NL should be expanded. We provide a robust biochemical background to support the use of anti-Arc nanobodies in attempts to target Arc-dependent synaptic plasticity. Function-blocking anti-Arc nanobodies could eventually help unravel the complex neurobiology of synaptic plasticity and allow to develop diagnostic and treatment tools.


Asunto(s)
Proteínas del Citoesqueleto , Proteínas del Tejido Nervioso , Anticuerpos de Dominio Único , Humanos , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/inmunología , Anticuerpos de Dominio Único/metabolismo , Sitios de Unión , Proteínas del Citoesqueleto/metabolismo , Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/inmunología , Ligandos , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/inmunología , Cristalografía por Rayos X , Unión Proteica , Modelos Moleculares , Secuencia de Aminoácidos
2.
Elife ; 132024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38639993

RESUMEN

In the Firmicutes phylum, GpsB is a membrane associated protein that coordinates peptidoglycan synthesis with cell growth and division. Although GpsB has been studied in several bacteria, the structure, function, and interactome of Staphylococcus aureus GpsB is largely uncharacterized. To address this knowledge gap, we solved the crystal structure of the N-terminal domain of S. aureus GpsB, which adopts an atypical, asymmetric dimer, and demonstrates major conformational flexibility that can be mapped to a hinge region formed by a three-residue insertion exclusive to Staphylococci. When this three-residue insertion is excised, its thermal stability increases, and the mutant no longer produces a previously reported lethal phenotype when overexpressed in Bacillus subtilis. In S. aureus, we show that these hinge mutants are less functional and speculate that the conformational flexibility imparted by the hinge region may serve as a dynamic switch to fine-tune the function of the GpsB complex and/or to promote interaction with its various partners. Furthermore, we provide the first biochemical, biophysical, and crystallographic evidence that the N-terminal domain of GpsB binds not only PBP4, but also FtsZ, through a conserved recognition motif located on their C-termini, thus coupling peptidoglycan synthesis to cell division. Taken together, the unique structure of S. aureus GpsB and its direct interaction with FtsZ/PBP4 provide deeper insight into the central role of GpsB in S. aureus cell division.


Asunto(s)
Proteínas Bacterianas , Proteínas del Citoesqueleto , Unión Proteica , Conformación Proteica , Staphylococcus aureus , Staphylococcus aureus/metabolismo , Staphylococcus aureus/genética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/química , Proteínas del Citoesqueleto/metabolismo , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/química , Cristalografía por Rayos X , Proteínas de Unión a las Penicilinas/metabolismo , Proteínas de Unión a las Penicilinas/genética , Proteínas de Unión a las Penicilinas/química , Modelos Moleculares
3.
Int J Biol Macromol ; 259(Pt 2): 129255, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38199552

RESUMEN

Several harmful bacteria have evolved resistance to conventional antibiotics due to their extensive usage. FtsZ, a principal bacterial cell division protein, is considered as an important drug target to combat resistance. We identified a caffeoyl anilide derivative, (E)-N-(4-(3-(3,4-dihydroxyphenyl)acryloyl)phenyl)-1-adamantylamide (compound 11) as a new antimicrobial agent targeting FtsZ. Compound 11 caused cell elongation in Mycobacterium smegmatis, Bacillus subtilis, and Escherichia coli cells, indicating that it inhibits cell partitioning. Compound 11 inhibited the assembly of Mycobacterium smegmatis FtsZ (MsFtsZ), forming short and thin filaments in vitro. Interestingly, the compound increased the rate of GTP hydrolysis of MsFtsZ. Compound 11 also impeded the assembly of Mycobacterium tuberculosis FtsZ. Fluorescence and absorption spectroscopic analysis suggested that compound 11 binds to MsFtsZ and produces conformational changes in FtsZ. The docking analysis indicated that the compound binds at the interdomain cleft of MsFtsZ. Further, it caused delocalization of the Z-ring in Mycobacterium smegmatis and Bacillus subtilis without affecting DNA segregation. Notably, compound 11 did not inhibit tubulin polymerization, the eukaryotic homolog of FtsZ, suggesting its specificity on bacteria. The evidence indicated that compound 11 exerts its antibacterial effect by impeding FtsZ assembly and has the potential to be developed as a broad-spectrum antimicrobial agent.


Asunto(s)
Antibacterianos , Proteínas del Citoesqueleto , Proteínas del Citoesqueleto/química , Antibacterianos/química , División Celular , Proliferación Celular , Proteínas Bacterianas/química
4.
J Biomol Struct Dyn ; 42(5): 2653-2666, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37158088

RESUMEN

Earlier molecular dynamics studies of the FtsZ protein revealed that the protein has high intrinsic flexibility which the crystal structures cannot reveal. However, the input structure in these simulation studies was based on the available crystal structure data and therefore, the effect of the C-terminal Intrinsically Disordered Region (IDR) of FtsZ could not be observed in any of these studies. Recent investigations have revealed that the C-terminal IDR is crucial for FtsZ assembly in vitro and Z ring formation in vivo. Therefore, in this study, we simulated FtsZ with the IDR. Simulations of the FtsZ monomer in different nucleotide bound forms (without nucleotide, GTP, GDP) were performed. In the conformations of FtsZ monomer with GTP, GTP binds variably with the protein. Such a variable interaction with the monomer has not been observed in any previous simulation studies of FtsZ and not observed in crystal structures. We found that central helix bends towards the C-terminal domain in the GTP bound form, hence, making way for polymerization. A nucleotide dependent shift/rotation of the C-terminal domain was observed in simulation time averaged structures.Communicated by Ramaswamy H. Sarma.


Asunto(s)
Proteínas Bacterianas , Simulación de Dinámica Molecular , Proteínas Bacterianas/química , Proteínas del Citoesqueleto/química , Nucleótidos , Escherichia coli/metabolismo , Guanosina Trifosfato/química
5.
J Vis Exp ; (199)2023 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-37811947

RESUMEN

A main event in bacterial cell division is the septation process, where the protein FtsZ is the key element. FtsZ polymerizes forming a ring-like structure (Z-ring) in the middle of the cell that serves as a scaffold for other division proteins. Super-resolution microscopy in bacterial models Escherichia coli and Bacillus subtilis showed that the Z-ring is discontinuous, while live cell imaging studies demonstrated that FtsZ moves along the ring by a mechanism known as treadmilling. To study the dynamics of FtsZ in vivo, a special cell placement in a vertical position is necessary for imaging the complete structure of the ring in the XY plane. In the case of FtsZ imaging in multicellular cyanobacteria, such as Anabaena sp. PCC7120, maintaining the filaments in a vertical position is challenging because of the size of the cells and the filaments' length. In this article, we describe a method that allows the vertical immobilization of Anabaena sp. PCC 7120 filaments using low melting point agarose and syringes, to record the Z-ring in a mutant that expresses a FtsZ-sfGFP fusion protein. This method is a rapid and inexpensive way to register protein dynamics at the division site using confocal microscopy.


Asunto(s)
Cianobacterias , Microscopía , Microscopía/métodos , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/química , Imagen de Lapso de Tiempo , Proteínas Bacterianas/química , Escherichia coli/genética , Cianobacterias/metabolismo
6.
Nat Commun ; 14(1): 3543, 2023 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-37336883

RESUMEN

PEAK pseudokinases are molecular scaffolds which dimerize to regulate cell migration, morphology, and proliferation, as well as cancer progression. The mechanistic role dimerization plays in PEAK scaffolding remains unclear, as there are no structures of PEAKs in complex with their interactors. Here, we report the cryo-EM structure of dimeric PEAK3 in complex with an endogenous 14-3-3 heterodimer. Our structure reveals an asymmetric binding mode between PEAK3 and 14-3-3 stabilized by one pseudokinase domain and the SHED domain of the PEAK3 dimer. The binding interface contains a canonical phosphosite-dependent primary interaction and a unique secondary interaction not observed in previous structures of 14-3-3/client complexes. Additionally, we show that PKD regulates PEAK3/14-3-3 binding, which when prevented leads to PEAK3 nuclear enrichment and distinct protein-protein interactions. Altogether, our data demonstrate that PEAK3 dimerization forms an unusual secondary interface for 14-3-3 binding, facilitating 14-3-3 regulation of PEAK3 localization and interactome diversity.


Asunto(s)
Proteínas 14-3-3 , Proteínas del Citoesqueleto , Proteínas del Citoesqueleto/química , Proteínas 14-3-3/química , Multimerización de Proteína
7.
Protein Sci ; 32(5): e4638, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37027210

RESUMEN

Palladin is an actin binding protein that is specifically upregulated in metastatic cancer cells but also colocalizes with actin stress fibers in normal cells and is critical for embryonic development as well as wound healing. Of nine isoforms present in humans, only the 90 kDa isoform of palladin, comprising three immunoglobulin (Ig) domains and one proline-rich region, is ubiquitously expressed. Previous work has established that the Ig3 domain of palladin is the minimal binding site for F-actin. In this work, we compare functions of the 90 kDa isoform of palladin to the isolated actin binding domain. To understand the mechanism of action for how palladin can influence actin assembly, we monitored F-actin binding and bundling as well as actin polymerization, depolymerization, and copolymerization. Together, these results demonstrate that there are key differences between the Ig3 domain and full-length palladin in actin binding stoichiometry, polymerization, and interactions with G-actin. Understanding the role of palladin in regulating the actin cytoskeleton may help us develop means to prevent cancer cells from reaching the metastatic stage of cancer progression.


Asunto(s)
Actinas , Proteínas del Citoesqueleto , Humanos , Actinas/análisis , Actinas/química , Actinas/metabolismo , Proteínas del Citoesqueleto/química , Proteínas de Microfilamentos/metabolismo , Citoesqueleto de Actina/química , Isoformas de Proteínas/metabolismo , Fosfoproteínas/química
8.
ACS Chem Biol ; 18(3): 629-642, 2023 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-36854145

RESUMEN

Methicillin-resistant Staphylococcus aureus (MRSA) is a multidrug-resistant (MDR) bacterial pathogen of acute clinical significance. Resistance to current standard-of-care antibiotics, such as vancomycin and linezolid, among nosocomial and community-acquired MRSA clinical isolates is on the rise. This threat to global public health highlights the need to develop new antibiotics for the treatment of MRSA infections. Here, we describe a new benzamide FtsZ inhibitor (TXH9179) with superior antistaphylococcal activity relative to earlier-generation benzamides like PC190723 and TXA707. TXH9179 was found to be 4-fold more potent than TXA707 against a library of 55 methicillin-sensitive S. aureus (MSSA) and MRSA clinical isolates, including MRSA isolates resistant to vancomycin and linezolid. TXH9179 was also associated with a lower frequency of resistance relative to TXA707 in all but one of the MSSA and MRSA isolates examined, with the observed resistance being due to mutations in the ftsZ gene. TXH9179 induced changes in MRSA cell morphology, cell division, and FtsZ localization are fully consistent with its actions as a FtsZ inhibitor. Crystallographic studies demonstrate the direct interaction of TXH9179 with S. aureus FtsZ (SaFtsZ), while delineating the key molecular contacts that drive complex formation. TXH9179 was not associated with any mammalian cytotoxicity, even at a concentration 10-fold greater than that producing antistaphylococcal activity. In serum, the carboxamide prodrug of TXH9179 (TXH1033) is rapidly hydrolyzed to TXH9179 by serum acetylcholinesterases. Significantly, both intravenously and orally administered TXH1033 exhibited enhanced in vivo efficacy relative to the carboxamide prodrug of TXA707 (TXA709) in treating a mouse model of systemic (peritonitis) MRSA infection. Viewed as a whole, our results highlight TXH9179 as a promising new benzamide FtsZ inhibitor worthy of further development.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina , Profármacos , Infecciones Estafilocócicas , Animales , Ratones , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Proteínas Bacterianas/química , Benzamidas/farmacología , Benzamidas/uso terapéutico , Proteínas del Citoesqueleto/química , Linezolid/farmacología , Linezolid/uso terapéutico , Mamíferos , Meticilina/farmacología , Meticilina/uso terapéutico , Pruebas de Sensibilidad Microbiana , Profármacos/farmacología , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus , Vancomicina/farmacología
9.
Adv Biol (Weinh) ; 7(3): e2200172, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36593513

RESUMEN

Mimicking bacterial cell division in well-defined cell-free systems has the potential to elucidate the minimal set of proteins required for cytoskeletal formation, membrane constriction, and final abscission. Membrane-anchored FtsZ polymers are often regarded as a sufficient system to realize this chain of events. By using purified FtsZ and its membrane-binding protein FtsA or the gain-of-function mutant FtsA* expressed in PURE (Protein synthesis Using Reconstituted Elements) from a DNA template, it is shown in this study that cytoskeletal structures are formed, and yield constricted liposomes exhibiting various morphologies. However, the resulting buds remain attached to the parental liposome by a narrow membrane neck. No division events can be monitored even after long-time tracking by fluorescence microscopy, nor when the osmolarity of the external solution is increased. The results provide evidence that reconstituted FtsA-FtsZ proto-rings coating the membrane necks are too stable to enable abscission. The prospect of combining a DNA-encoded FtsZ system with assisting mechanisms to achieve synthetic cell division is discussed.


Asunto(s)
Proteínas Bacterianas , Liposomas , Liposomas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/química , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/metabolismo , Escherichia coli/metabolismo , División Celular/genética
10.
FEBS J ; 290(14): 3527-3532, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36349414

RESUMEN

Self-assembling protein filaments are at the heart of cell function. Among them, tubulin-like proteins are essential for cell division, DNA segregation and cytoskeletal functions across the domains of life. FtsZ and tubulin share their core structures, a characteristic nucleotide-binding pocket and similar protofilament architecture. GTP hydrolysis between consecutive subunits drives their assembly dynamics. Two recent studies provide previously missing, filament atomic structures of bacterial FtsZ and a recently discovered archaeal tubulin in their nucleotide triphosphate-bound states. Both filament structures reveal strikingly conserved interfacial GTPase active sites, with Mg2+ and K+ /Na+ cations and an NxDxxD/E triad of catalytic residues, probably inherited from the common ancestor of FtsZs and tubulins. Moreover, both proteins exhibit nucleotide-regulated subunit association mediated by interfacial water bridges, as well as polymerization-induced structural changes, likely enabling related dynamic assembly mechanisms.


Asunto(s)
GTP Fosfohidrolasas , Tubulina (Proteína) , Tubulina (Proteína)/metabolismo , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/metabolismo , Proteínas del Citoesqueleto/química , Archaea/genética , Archaea/metabolismo , Proteínas Bacterianas/metabolismo , Bacterias/metabolismo , Nucleótidos , Guanosina Trifosfato/metabolismo
11.
Proc Natl Acad Sci U S A ; 119(50): e2208227119, 2022 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-36490318

RESUMEN

The spatiotemporal regulation of cell division is a fundamental issue in cell biology. Bacteria have evolved a variety of different systems to achieve proper division site placement. In many cases, the underlying molecular mechanisms are still incompletely understood. In this study, we investigate the function of the cell division regulator MipZ from Caulobacter crescentus, a P-loop ATPase that inhibits the polymerization of the treadmilling tubulin homolog FtsZ near the cell poles, thereby limiting the assembly of the cytokinetic Z ring to the midcell region. We show that MipZ interacts with FtsZ in both its monomeric and polymeric forms and induces the disassembly of FtsZ polymers in a manner that is not dependent but enhanced by the FtsZ GTPase activity. Using a combination of biochemical and genetic approaches, we then map the MipZ-FtsZ interaction interface. Our results reveal that MipZ employs a patch of surface-exposed hydrophobic residues to interact with the C-terminal region of the FtsZ core domain. In doing so, it sequesters FtsZ monomers and caps the (+)-end of FtsZ polymers, thereby promoting their rapid disassembly. We further show that MipZ influences the conformational dynamics of interacting FtsZ molecules, which could potentially contribute to modulating their assembly kinetics. Together, our findings show that MipZ uses a combination of mechanisms to control FtsZ polymerization, which may be required to robustly regulate the spatiotemporal dynamics of Z ring assembly within the cell.


Asunto(s)
Caulobacter crescentus , Proteínas del Citoesqueleto , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/química , Polímeros , Proteínas Bacterianas/genética , Proteínas Bacterianas/química , Caulobacter crescentus/genética , División Celular
12.
Biophys J ; 121(22): 4325-4341, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36225113

RESUMEN

Desmosomes are large, macromolecular protein assemblies that mechanically couple the intermediate filament cytoskeleton to sites of cadherin-mediated cell adhesion, thereby providing structural integrity to tissues that routinely experience large forces. Proper desmosomal adhesion is necessary for the normal development and maintenance of vertebrate tissues, such as epithelia and cardiac muscle, while dysfunction can lead to severe disease of the heart and skin. Therefore, it is important to understand the relationship between desmosomal adhesion and the architecture of the molecules that form the adhesive interface, the desmosomal cadherins (DCs). However, desmosomes are embedded in two plasma membranes and are linked to the cytoskeletal networks of two cells, imposing extreme difficulty on traditional structural studies of DC architecture, which have yielded conflicting results. Consequently, the relationship between DC architecture and adhesive function remains unclear. To overcome these challenges, we utilized excitation-resolved fluorescence polarization microscopy to quantify the orientational order of the extracellular and intracellular domains of three DC isoforms: desmoglein 2, desmocollin 2, and desmoglein 3. We found that DC ectodomains were significantly more ordered than their cytoplasmic counterparts, indicating a drastic difference in DC architecture between opposing sides of the plasma membrane. This difference was conserved among all DCs tested, suggesting that it may be an important feature of desmosomal architecture. Moreover, our findings suggest that the organization of DC ectodomains is predominantly the result of extracellular adhesive interactions. We employed azimuthal orientation mapping to show that DC ectodomains are arranged with rotational symmetry about the membrane normal. Finally, we performed a series of mathematical simulations to test the feasibility of a recently proposed antiparallel arrangement of DC ectodomains, finding that it is supported by our experimental data. Importantly, the strategies employed here have the potential to elucidate molecular mechanisms for diseases that result from defective desmosome architecture.


Asunto(s)
Proteínas del Citoesqueleto , Desmosomas , Desmosomas/metabolismo , Proteínas del Citoesqueleto/química , Cadherinas/metabolismo , Adhesión Celular/fisiología , Cadherinas Desmosómicas/análisis , Cadherinas Desmosómicas/metabolismo
13.
J Mol Biol ; 434(21): 167817, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36087777

RESUMEN

Bacterial cell division begins with the formation of the Z-ring via polymerization of FtsZ and the localization of Z-ring beneath the inner membrane through membrane anchors. In Mycobacterium tuberculosis (Mtb), SepF is one such membrane anchor, but our understanding of the underlying mechanism is very limited. Here we used molecular dynamics simulations to characterize how SepF itself, a water-soluble protein, tethers to acidic membranes that mimic the Mtb inner membrane. In addition to an amphipathic helix (residues 1-12) at the N-terminus, membrane binding also occurs through two stretches of positively charged residues (Arg27-Arg37 and Arg95-Arg107) in the long linker preceding the FtsZ-binding core domain (residues 128-218). The additional interactions via the disordered linker stabilize the membrane tethering of SepF, and keep the core domain of SepF and hence the attached Z-ring close to the membrane. The resulting membrane proximity of the Z-ring in turn enables its interactions with and thus recruitment of two membrane proteins, FtsW and CrgA, at the late stage of cell division.


Asunto(s)
Proteínas Bacterianas , División Celular , Membrana Celular , Proteínas del Citoesqueleto , Mycobacterium tuberculosis , Proteínas Bacterianas/química , Proteínas del Citoesqueleto/química , Mycobacterium tuberculosis/citología , Mycobacterium tuberculosis/fisiología , Simulación de Dinámica Molecular , Membrana Celular/química , Dominios Proteicos
14.
Proc Natl Acad Sci U S A ; 119(35): e2208457119, 2022 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-35994671

RESUMEN

The nicotinamide adenine dinucleotide hydrolase (NADase) sterile alpha toll/interleukin receptor motif containing-1 (SARM1) acts as a central executioner of programmed axon death and is a possible therapeutic target for neurodegenerative disorders. While orthosteric inhibitors of SARM1 have been described, this multidomain enzyme is also subject to intricate forms of autoregulation, suggesting the potential for allosteric modes of inhibition. Previous studies have identified multiple cysteine residues that support SARM1 activation and catalysis, but which of these cysteines, if any, might be selectively targetable by electrophilic small molecules remains unknown. Here, we describe the chemical proteomic discovery of a series of tryptoline acrylamides that site-specifically and stereoselectively modify cysteine-311 (C311) in the noncatalytic, autoregulatory armadillo repeat (ARM) domain of SARM1. These covalent compounds inhibit the NADase activity of WT-SARM1, but not C311A or C311S SARM1 mutants, show a high degree of proteome-wide selectivity for SARM1_C311 and stereoselectively block vincristine- and vacor-induced neurite degeneration in primary rodent dorsal root ganglion neurons. Our findings describe selective, covalent inhibitors of SARM1 targeting an allosteric cysteine, pointing to a potentially attractive therapeutic strategy for axon degeneration-dependent forms of neurological disease.


Asunto(s)
Proteínas del Dominio Armadillo , Cisteína , Proteínas del Citoesqueleto , Proteínas del Dominio Armadillo/antagonistas & inhibidores , Proteínas del Dominio Armadillo/química , Proteínas del Dominio Armadillo/genética , Axones , Proteínas del Citoesqueleto/antagonistas & inhibidores , Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/genética , Homeostasis , NAD+ Nucleosidasa , Proteómica
15.
Curr Opin Chem Biol ; 69: 102176, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35780654

RESUMEN

During axon degeneration, NAD+ levels are largely controlled by two enzymes: nicotinamide mononucleotide adenylyltransferase 2 (NMNAT2) and sterile alpha and toll interleukin motif containing protein 1 (SARM1). NMNAT2, which catalyzes the formation of NAD+ from NMN and ATP, is actively degraded leading to decreased NAD+ levels. SARM1 activity further decreases the concentration of NAD+ by catalyzing its hydrolysis to form nicotinamide and a mixture of ADPR and cADPR. Notably, SARM1 knockout mice show decreased neurodegeneration in animal models of axon degeneration, highlighting the therapeutic potential of targeting this novel NAD+ hydrolase. This review discusses recent advances in the SARM1 field, including SARM1 structure, regulation, and catalysis as well as the identification of the first SARM1 inhibitors.


Asunto(s)
Proteínas del Dominio Armadillo , Nicotinamida-Nucleótido Adenililtransferasa , Animales , Proteínas del Dominio Armadillo/química , Proteínas del Dominio Armadillo/genética , Proteínas del Dominio Armadillo/metabolismo , Axones/metabolismo , Biología , Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/metabolismo , Ratones , NAD/metabolismo , Nicotinamida-Nucleótido Adenililtransferasa/genética , Nicotinamida-Nucleótido Adenililtransferasa/metabolismo
16.
J Phys Chem B ; 126(28): 5219-5230, 2022 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-35799411

RESUMEN

In many species of Gram-positive bacteria, SepF participated in the membrane tethering of FtsZ Z-ring during bacteria division. However, atomic-level details of interaction between SepF and FtsZ in an assembled state are lacking. Here, by combining solid-state NMR (SSNMR) with biochemical analyses, the interaction of Bacillus subtilis SepF and the C-terminal domain (CTD) of FtsZ was investigated. We obtained near complete chemical shift assignments of SepF and determined the structural model of the SepF monomer. Interaction with FtsZ-CTD caused further packing of SepF rings, and SSNMR experiments revealed the affected residues locating at α1, α2, ß3, and ß4 of SepF. Solution NMR experiments of dimeric SepF constructed by point mutation strategy proved a prerequisite role of α-α interface formation in SepF for FtsZ binding. Overall, our results provide structural insights into the mechanisms of SepF-FtsZ interaction for better understanding the function of SepF in bacteria.


Asunto(s)
Bacillus subtilis , Proteínas del Citoesqueleto , Bacillus subtilis/química , Proteínas Bacterianas/química , División Celular , Proteínas del Citoesqueleto/química , Espectroscopía de Resonancia Magnética
17.
Biochem J ; 479(14): 1543-1558, 2022 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-35789252

RESUMEN

The respiratory pathogen, Streptococcus pneumoniae has acquired multiple-drug resistance over the years. An attractive strategy to combat pneumococcal infection is to target cell division to inhibit the proliferation of S. pneumoniae. This work presents Vitamin K3 as a potential anti-pneumococcal drug that targets FtsZ, the master coordinator of bacterial cell division. Vitamin K3 strongly inhibited S. pneumoniae proliferation with a minimum inhibitory concentration (MIC) and a minimum bactericidal concentration (MBC) of 6 µg/ml. Vitamin K3 disrupted the Z-ring localization in both S. pneumoniae and Bacillus subtilis within 30 min of treatment, while the membrane integrity and nucleoid segregation remain unchanged. Several complementary experiments showed that Vitamin K3 inhibits the assembly of purified S. pneumoniae FtsZ (SpnFtsZ) and induces conformational changes in the protein. Interestingly, Vitamin K3 interfered with GTP binding onto FtsZ and increased the GTPase activity of FtsZ polymers. The intrinsic tryptophan fluorescence of SpnFtsZ revealed that Vitamin K3 delays the nucleation of FtsZ polymers and reduces the rate of polymerization. In the presence of a non-hydrolyzable analog of GTP, Vitamin K3 did not show inhibition of FtsZ polymerization. These results indicated that Vitamin K3 induces conformational changes in FtsZ that increase GTP hydrolysis and thereby, destabilize the FtsZ polymers. Together, our data provide evidence that Vitamin K3 derives its potent anti-pneumococcal activity by inhibiting FtsZ assembly.


Asunto(s)
Streptococcus pneumoniae , Vitamina K 3 , Bacillus subtilis , Proteínas Bacterianas/química , Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/genética , Guanosina Trifosfato/metabolismo , Polímeros/metabolismo , Streptococcus pneumoniae/metabolismo , Vitamina K 3/metabolismo
18.
J Antibiot (Tokyo) ; 75(7): 385-395, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35618784

RESUMEN

FtsZ inhibitors represent a new drug class as no drugs using this mode of action (MOA) have been approved by regulators. 3-alkoxy substituted 2,6-difluorobenzamide scaffold is one of the most studied FtsZ inhibitors among which the most promising anti-MRSA candidate TXA709 is in clinical trial. In this paper, we present the screening and evaluation of a benzamide class that is functionalized at the alkoxy fragment targeting Gram-negative bacteria. The variations in 3-alkoxy substitutions, specifically the hydroxylated alkyl residues to the secondary and stereogenic pseudo-benzylic carbon of their methyleneoxy linker, are particularly active against K. pneumoniae ATCC 10031 in marked contrast to the derivatives related to PC190723, all of which were inactive against Gram-negative bacteria. The two lead molecules TXA6101 and TXY6129 inhibit the polymerization of E. coli FtsZ in a concentration-dependent manner and induce changes in the morphology of E. coli and K. pneumoniae consistent with inhibition of cell division. These classes of compounds, however, were found to be substrates for efflux pumps in Gram-negative bacteria.


Asunto(s)
Proteínas del Citoesqueleto , Escherichia coli , Antibacterianos/química , Antibacterianos/farmacología , Proteínas Bacterianas/química , Benzamidas/química , Benzamidas/farmacología , Proteínas del Citoesqueleto/química , Klebsiella pneumoniae
19.
J Mol Graph Model ; 114: 108189, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35453046

RESUMEN

Bacterial resistance caused by widespread use and abuse of antibiotics is threatening human health, and the development of new antibacterial agents with novel antibacterial targets has become urgent. Filamenting temperature-sensitive mutant Z (FtsZ), as a key protein in bacterial division, has received extensive attention. PC190723 exhibits an outstanding antibacterial activity by producing potent inhibitory ability on FtsZ protein, but its influence on the conformation of FtsZ protein at the molecular level is still unclear. In this study, we explored the effect of PC190723 on the conformation and function of FtsZ protein through molecular dynamics (MD) simulation and post-analysis. The results showed that PC190723 increased the high-affinity conformational stability of FtsZ protein, which disrupts the normal assembly of the Z-ring. In particular, the interactions of residues S8-sheet (VAL260-GLY266) increased in the FtsZPC190723 system, which may be the reason for promotes the formation of protofilament. In brief, the mechanism of PC190723 inhibiting FtsZ protein was explained at the molecular level by MD simulation, which provides new ideas for the identification of new FtsZ inhibitors as antibacterial agents.


Asunto(s)
Proteínas del Citoesqueleto , Simulación de Dinámica Molecular , Antibacterianos/farmacología , Proteínas Bacterianas/química , Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/metabolismo , Humanos , Piridinas , Tiazoles
20.
Mol Cell ; 82(9): 1643-1659.e10, 2022 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-35334231

RESUMEN

The NADase SARM1 (sterile alpha and TIR motif containing 1) is a key executioner of axon degeneration and a therapeutic target for several neurodegenerative conditions. We show that a potent SARM1 inhibitor undergoes base exchange with the nicotinamide moiety of nicotinamide adenine dinucleotide (NAD+) to produce the bona fide inhibitor 1AD. We report structures of SARM1 in complex with 1AD, NAD+ mimetics and the allosteric activator nicotinamide mononucleotide (NMN). NMN binding triggers reorientation of the armadillo repeat (ARM) domains, which disrupts ARM:TIR interactions and leads to formation of a two-stranded TIR domain assembly. The active site spans two molecules in these assemblies, explaining the requirement of TIR domain self-association for NADase activity and axon degeneration. Our results reveal the mechanisms of SARM1 activation and substrate binding, providing rational avenues for the design of new therapeutics targeting SARM1.


Asunto(s)
Proteínas del Dominio Armadillo , NAD , Proteínas del Dominio Armadillo/genética , Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/genética , NAD/metabolismo , NAD+ Nucleosidasa/metabolismo , Dominios Proteicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...