Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 982
Filtrar
1.
Biomolecules ; 14(4)2024 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-38672487

RESUMO

Tuberculosis (TB) is the leading global cause of death f rom an infectious bacterial agent. Therefore, limiting its epidemic spread is a pressing global health priority. The chaperone-like protein HtpG of M. tuberculosis (Mtb) is a large dimeric and multi-domain protein with a key role in Mtb pathogenesis and promising antigenic properties. This dual role, likely associated with the ability of Heat Shock proteins to act both intra- and extra-cellularly, makes HtpG highly exploitable both for drug and vaccine development. This review aims to gather the latest updates in HtpG structure and biological function, with HtpG operating in conjunction with a large number of chaperone molecules of Mtb. Altogether, these molecules help Mtb recovery after exposure to host-like stress by assisting the whole path of protein folding rescue, from the solubilisation of aggregated proteins to their refolding. Also, we highlight the role of structural biology in the development of safer and more effective subunit antigens. The larger availability of structural information on Mtb antigens and a better understanding of the host immune response to TB infection will aid the acceleration of TB vaccine development.


Assuntos
Antígenos de Bactérias , Proteínas de Bactérias , Mycobacterium tuberculosis , Vacinas contra a Tuberculose , Fatores de Virulência , Mycobacterium tuberculosis/imunologia , Antígenos de Bactérias/imunologia , Antígenos de Bactérias/química , Fatores de Virulência/imunologia , Fatores de Virulência/química , Humanos , Vacinas contra a Tuberculose/imunologia , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/química , Tuberculose/imunologia , Tuberculose/prevenção & controle , Tuberculose/microbiologia , Animais , Chaperonas Moleculares/imunologia , Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo
2.
J Phys Chem B ; 128(16): 3929-3936, 2024 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-38619541

RESUMO

Yersinia pestis, the causative agent of plague, is capable of evading the human immune system response by recruiting the plasma circulating vitronectin proteins, which act as a shield and avoid its lysis. Vitronectin recruitment is mediated by its interaction with the bacterial transmembrane protein Ail, protruding from the Y. pestis outer membrane. By using all-atom long-scale molecular dynamic simulations of Ail embedded in a realistic model of the bacterial membrane, we have shown that vitronectin forms a stable complex, mediated by interactions between the disordered moieties of the two proteins. The main amino acids driving the complexation have also been evidenced, thus favoring the possible rational design of specific peptides which, by inhibiting vitronectin recruitment, could act as original antibacterial agents.


Assuntos
Proteínas da Membrana Bacteriana Externa , Simulação de Dinâmica Molecular , Vitronectina , Vitronectina/química , Vitronectina/metabolismo , Proteínas da Membrana Bacteriana Externa/química , Proteínas da Membrana Bacteriana Externa/metabolismo , Humanos , Yersinia pestis/química , Yersinia pestis/metabolismo , Fatores de Virulência/química , Fatores de Virulência/metabolismo , Domínios Proteicos , Ligação Proteica
3.
Nature ; 620(7973): 445-452, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37495693

RESUMO

To replicate inside macrophages and cause tuberculosis, Mycobacterium tuberculosis must scavenge a variety of nutrients from the host1,2. The mammalian cell entry (MCE) proteins are important virulence factors in M. tuberculosis1,3, where they are encoded by large gene clusters and have been implicated in the transport of fatty acids4-7 and cholesterol1,4,8 across the impermeable mycobacterial cell envelope. Very little is known about how cargos are transported across this barrier, and it remains unclear how the approximately ten proteins encoded by a mycobacterial mce gene cluster assemble to transport cargo across the cell envelope. Here we report the cryo-electron microscopy (cryo-EM) structure of the endogenous Mce1 lipid-import machine of Mycobacterium smegmatis-a non-pathogenic relative of M. tuberculosis. The structure reveals how the proteins of the Mce1 system assemble to form an elongated ABC transporter complex that is long enough to span the cell envelope. The Mce1 complex is dominated by a curved, needle-like domain that appears to be unrelated to previously described protein structures, and creates a protected hydrophobic pathway for lipid transport across the periplasm. Our structural data revealed the presence of a subunit of the Mce1 complex, which we identified using a combination of cryo-EM and AlphaFold2, and name LucB. Our data lead to a structural model for Mce1-mediated lipid import across the mycobacterial cell envelope.


Assuntos
Proteínas de Bactérias , Microscopia Crioeletrônica , Lipídeos , Proteínas de Membrana Transportadoras , Mycobacterium tuberculosis , Internalização do Vírus , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/ultraestrutura , Proteínas de Membrana Transportadoras/química , Proteínas de Membrana Transportadoras/metabolismo , Proteínas de Membrana Transportadoras/ultraestrutura , Mycobacterium tuberculosis/química , Mycobacterium tuberculosis/metabolismo , Mycobacterium tuberculosis/ultraestrutura , Tuberculose/microbiologia , Fatores de Virulência/química , Fatores de Virulência/metabolismo , Transportadores de Cassetes de Ligação de ATP/química , Transportadores de Cassetes de Ligação de ATP/metabolismo , Transportadores de Cassetes de Ligação de ATP/ultraestrutura , Periplasma/metabolismo , Domínios Proteicos , Interações Hidrofóbicas e Hidrofílicas , Complexos Multiproteicos/química , Complexos Multiproteicos/metabolismo , Complexos Multiproteicos/ultraestrutura
4.
J Biol Chem ; 299(2): 102829, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36581211

RESUMO

Candida albicans is a deadly pathogen responsible for millions of mucosal and systemic infections per year. The pathobiology of C. albicans is largely dependent on the damaging and immunostimulatory properties of the peptide candidalysin (CL), a key virulence factor. When CL forms pores in the plasma membrane of epithelial cells, it activates a response network grounded in activation of the epidermal growth factor receptor. Prior reviews have characterized the resulting CL immune activation schemas but lacked insights into the molecular mechanism of CL membrane damage. We recently demonstrated that CL functions by undergoing a unique self-assembly process; CL forms polymers and loops in aqueous solution prior to inserting and forming pores in cell membranes. This mechanism, the first of its kind to be observed, informs new therapeutic avenues to treat Candida infections. Recently, variants of CL were identified in other Candida species, providing an opportunity to identify the residues that are key for CL to function. In this review, we connect the ability of CL to damage cell membranes to its immunostimulatory properties.


Assuntos
Candida albicans , Proteínas Fúngicas , Fatores de Virulência , Candida albicans/química , Proteínas Fúngicas/química , Fatores de Virulência/química
5.
J Biol Chem ; 299(1): 102761, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36463964

RESUMO

Pathogenic species from the Mycobacterium genus are responsible for a number of adverse health conditions in humans and animals that threaten health security and the economy worldwide. Mycobacteria have up to five specialized secretion systems (ESX-1 to ESX-5) that transport virulence factors across their complex cell envelope to facilitate manipulation of their environment. In pathogenic species, these virulence factors influence the immune system's response and are responsible for membrane disruption and contributing to cell death. While structural details of these secretion systems have been recently described, gaps still remain in the structural understanding of the secretion mechanisms of most substrates. Here, we describe the crystal structure of Mycobacterium tuberculosis ESX-1 secretion-associated substrate EspB bound to its chaperone EspK. We found that EspB interacts with the C-terminal domain of EspK through its helical tip. Furthermore, cryogenic electron microscopy, size exclusion chromatography analysis, and small-angle X-ray scattering experiments show that EspK keeps EspB in its secretion-competent monomeric form and prevents its oligomerization. The structure presented in this study suggests an additional secretion mechanism in ESX-1, analogous to the chaperoning of proline-glutamate (PE)-proline-proline-glutamate (PPE) proteins by EspG, where EspK facilitates the secretion of EspB in Mycobacterium species.


Assuntos
Proteínas da Membrana Bacteriana Externa , Proteínas de Bactérias , Mycobacterium tuberculosis , Fatores de Virulência , Humanos , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Glutamatos/metabolismo , Mycobacterium tuberculosis/metabolismo , Prolina/metabolismo , Fatores de Virulência/química , Fatores de Virulência/metabolismo , Morte Celular , Proteínas da Membrana Bacteriana Externa/química , Proteínas da Membrana Bacteriana Externa/metabolismo , Cristalização , Microscopia Crioeletrônica
6.
J Mol Biol ; 434(23): 167871, 2022 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-36404438

RESUMO

Porphyromonas gingivalis is a gram-negative oral anaerobic pathogen and is one of the key causative agents of periodontitis. P. gingivalis utilises a range of virulence factors, including the cysteine protease RgpB, to drive pathogenesis and these are exported and attached to the cell surface via the type IX secretion system (T9SS). All cargo proteins possess a conserved C-terminal signal domain (CTD) which is recognised by the T9SS, and the outer membrane ß-barrel protein PorV (PG0027/LptO) can interact with cargo proteins as they are exported to the bacterial surface. Using a combination of solution nuclear magnetic resonance (NMR) spectroscopy, biochemical analyses, machine-learning-based modelling and molecular dynamics (MD) simulations, we present a structural model of a PorV:RgpB-CTD complex from P. gingivalis. This is the first structural insight into CTD recognition by the T9SS and shows how the conserved motifs in the CTD are the primary sites that mediate binding. In PorV, interactions with extracellular surface loops are important for binding the CTD, and together these appear to cradle and lock RgpB-CTD in place. This work provides insight into cargo recognition by PorV but may also have important implications for understanding other aspects of type-IX dependent secretion.


Assuntos
Proteínas de Bactérias , Sistemas de Secreção Bacterianos , Proteínas de Membrana , Simulação de Dinâmica Molecular , Porphyromonas gingivalis , Proteínas de Bactérias/química , Proteínas de Membrana/química , Porphyromonas gingivalis/metabolismo , Porphyromonas gingivalis/patogenicidade , Fatores de Virulência/química , Sistemas de Secreção Bacterianos/química , Domínios Proteicos
7.
J Cell Biol ; 221(12)2022 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-36194176

RESUMO

Pore-forming toxins (PFTs) are important virulence factors produced by many pathogenic bacteria. Here, we show that the Vibrio cholerae toxin MakA is a novel cholesterol-binding PFT that induces non-canonical autophagy in a pH-dependent manner. MakA specifically binds to cholesterol on the membrane at pH < 7. Cholesterol-binding leads to oligomerization of MakA on the membrane and pore formation at pH 5.5. Unlike other cholesterol-dependent cytolysins (CDCs) which bind cholesterol through a conserved cholesterol-binding motif (Thr-Leu pair), MakA contains an Ile-Ile pair that is essential for MakA-cholesterol interaction. Following internalization, endosomal acidification triggers MakA pore-assembly followed by ESCRT-mediated membrane repair and V-ATPase-dependent unconventional LC3 lipidation on the damaged endolysosomal membranes. These findings characterize a new cholesterol-binding toxin that forms pores in a pH-dependent manner and reveals the molecular mechanism of host autophagy manipulation.


Assuntos
Autofagia , Proteínas de Bactérias , Colesterol , Citotoxinas , Vibrio cholerae , Fatores de Virulência , Adenosina Trifosfatases/metabolismo , Motivos de Aminoácidos , Autofagia/efeitos dos fármacos , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Colesterol/metabolismo , Citotoxinas/metabolismo , Citotoxinas/farmacologia , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Endossomos/química , Endossomos/metabolismo , Concentração de Íons de Hidrogênio , Lisossomos/química , Lisossomos/metabolismo , Ligação Proteica , Vibrio cholerae/química , Vibrio cholerae/metabolismo , Fatores de Virulência/química , Fatores de Virulência/metabolismo
8.
J Biol Chem ; 298(10): 102441, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36055404

RESUMO

Vibrio cholerae cytolysin (VCC) is a potent membrane-damaging ß-barrel pore-forming toxin. Upon binding to the target membranes, VCC monomers first assemble into oligomeric prepore intermediates and subsequently transform into transmembrane ß-barrel pores. VCC harbors a designated pore-forming motif, which, during oligomeric pore formation, inserts into the membrane and generates a transmembrane ß-barrel scaffold. It remains an enigma how the molecular architecture of the pore-forming motif regulates the VCC pore-formation mechanism. Here, we show that a specific pore-forming motif residue, E289, plays crucial regulatory roles in the pore-formation mechanism of VCC. We find that the mutation of E289A drastically compromises pore-forming activity, without affecting the structural integrity and membrane-binding potential of the toxin monomers. Although our single-particle cryo-EM analysis reveals WT-like oligomeric ß-barrel pore formation by E289A-VCC in the membrane, we demonstrate that the mutant shows severely delayed kinetics in terms of pore-forming ability that can be rescued with elevated temperature conditions. We find that the pore-formation efficacy of E289A-VCC appears to be more profoundly dependent on temperature than that of the WT toxin. Our results suggest that the E289A mutation traps membrane-bound toxin molecules in the prepore-like intermediate state that is hindered from converting into the functional ß-barrel pores by a large energy barrier, thus highlighting the importance of this residue for the pore-formation mechanism of VCC.


Assuntos
Proteínas de Bactérias , Citotoxinas , Proteínas Citotóxicas Formadoras de Poros , Vibrio cholerae , Fatores de Virulência , Membrana Celular/metabolismo , Citotoxinas/química , Citotoxinas/genética , Vibrio cholerae/patogenicidade , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Fatores de Virulência/química , Fatores de Virulência/genética , Proteínas Citotóxicas Formadoras de Poros/química , Proteínas Citotóxicas Formadoras de Poros/genética , Motivos de Aminoácidos , Mutação , Ácido Glutâmico/química , Ácido Glutâmico/genética
9.
J Virol ; 96(17): e0099422, 2022 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-35993736

RESUMO

Wild birds are the reservoir for all avian influenza viruses (AIV). In poultry, the transition from low pathogenic (LP) AIV of H5 and H7 subtypes to highly pathogenic (HP) AIV is accompanied mainly by changing the hemagglutinin (HA) monobasic cleavage site (CS) to a polybasic motif (pCS). Galliformes, including turkeys and chickens, succumb with high morbidity and mortality to HPAIV infections, although turkeys appear more vulnerable than chickens. Surprisingly, the genetic determinants for virulence and pathogenesis of HPAIV in turkeys are largely unknown. Here, we determined the genetic markers for virulence and transmission of HPAIV H7N1 in turkeys, and we explored the host responses in this species compared to those of chickens. We found that recombinant LPAIV H7N1 carrying pCS was avirulent in chickens but exhibited high virulence in turkeys, indicating that virulence determinants vary in these two galliform species. A transcriptome analysis indicated that turkeys mount a different host response than do chickens, particularly from genes involved in RNA metabolism and the immune response. Furthermore, we found that the HA glycosylation at residue 123, acquired by LP viruses shortly after transmission from wild birds and preceding the transition from LP to HP, had a role in virus fitness and virulence in chickens, though it was not a prerequisite for high virulence in turkeys. Together, these findings indicate variable virulence determinants and host responses in two closely related galliformes, turkeys and chickens, after infection with HPAIV H7N1. These results could explain the higher vulnerability to HPAIV of turkeys compared to chickens. IMPORTANCE Infection with HPAIV in chickens and turkeys, two closely related galliform species, results in severe disease and death. Although the presence of a polybasic cleavage site (pCS) in the hemagglutinin of AIV is a major virulence determinant for the transition of LPAIV to HPAIV, there are knowledge gaps on the genetic determinants (including pCS) and the host responses in turkeys compared to chickens. Here, we found that the pCS alone was sufficient for the transformation of a LP H7N1 into a HPAIV in turkeys but not in chickens. We also noticed that turkeys exhibited a different host response to an HPAIV infection, namely, a widespread downregulation of host gene expression associated with protein synthesis and the immune response. These results are important for a better understanding of the evolution of HPAIV from LPAIV and of the different outcomes and the pathomechanisms of HPAIV infections in chickens and turkeys.


Assuntos
Galinhas , Vírus da Influenza A Subtipo H7N1 , Influenza Aviária , Perus , Fatores de Virulência , Virulência , Animais , Galinhas/virologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/química , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Vírus da Influenza A Subtipo H7N1/genética , Vírus da Influenza A Subtipo H7N1/patogenicidade , Influenza Aviária/mortalidade , Influenza Aviária/virologia , Perus/virologia , Virulência/genética , Fatores de Virulência/química , Fatores de Virulência/genética
10.
Molecules ; 27(14)2022 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-35889366

RESUMO

Toll-interleukin receptor (TIR) domains have emerged as critical players involved in innate immune signaling in humans but are also expressed as potential virulence factors within multiple pathogenic bacteria. However, there has been a shortage of structural studies aimed at elucidating atomic resolution details with respect to their interactions, potentially owing to their dynamic nature. Here, we used a combination of biophysical and biochemical studies to reveal the dynamic behavior and functional interactions of a panel of both bacterial TIR-containing proteins and mammalian receptor TIR domains. Regarding dynamics, all three bacterial TIR domains studied here exhibited an inherent exchange that led to severe resonance line-broadening, revealing their intrinsic dynamic nature on the intermediate NMR timescale. In contrast, the three mammalian TIR domains studied here exhibited a range in terms of their dynamic exchange that spans multiple timescales. Functionally, only the bacterial TIR domains were catalytic towards the cleavage of NAD+, despite the conservation of the catalytic nucleophile on human TIR domains. Our development of NMR-based catalytic assays allowed us to further identify differences in product formation for gram-positive versus gram-negative bacterial TIR domains. Differences in oligomeric interactions were also revealed, whereby bacterial TIR domains self-associated solely through their attached coil-coil domains, in contrast to the mammalian TIR domains that formed homodimers and heterodimers through reactive cysteines. Finally, we provide the first atomic-resolution studies of a bacterial coil-coil domain and provide the first atomic model of the TIR domain from a human anti-inflammatory IL-1R8 protein that undergoes a slow inherent exchange.


Assuntos
Bactérias , Fatores de Virulência , Animais , Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Bactérias Gram-Negativas/metabolismo , Humanos , Mamíferos/metabolismo , Transdução de Sinais , Fatores de Virulência/química
11.
Comput Biol Med ; 145: 105517, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35585729

RESUMO

Quorum sensing (QS) is a bacterial communication strategy controlling cells density, biofilm formation, virulence, sporulation, and survival. Since QS is considered a virulence factor in drug-resistant pathogenic bacteria, inhibition of QS can contribute to control the spread of these bacteria. We propose in this study to test in silico, 19 natural compounds for their potential to inhibit QS transcriptional regulators of Pseudomonas aeruginosa (LasR and PqsE) and Chromobacterium violaceum (CviR and CviR'). Molecular docking was performed to explore the binding energies between selected compounds, and QS signaling proteins. Additionally, molecular dynamics (MD) simulations of the complexes protein-ligand were tested to evaluate the stability of the complexs throughout the simulation process. The simulation interaction diagram (SID) was achieved to compute the radius of gyration (rGyr), solvent accessible surface area (SASA), intramolecular HBs, molecular surface area (MolSA), and polar surface area (PSA). Additionally, the physicochemical properties, pharmacokinetics, drug-likeness, and toxicity analysis of the best-selected compounds were determined. Among these compounds, catechin and nakinadine B were identified as potent QS antagonists that showed the best XP GScore and stable interaction during molecular dynamic simulation. Catechin interacts with LasR and CviR' displaying XP GScore -10.969 kcal/mol and -9.936 kcal/mol respectively. Additionally, nakinadine B interacts with PqsE and CviR giving XP GScore -7.442 kcal/mol and -10.34 kcal/mol respectively. RMSD plot analysis showed that both catechin and nakinadine B were stable during 50 ns simulation time with the tested target proteins. The predictive result of toxicity demonstrated that catechin and nakinadine B doesn't induce cytotoxicity, immunotoxicity, carcinogenicity, mutagenicity, hepatotoxicity and were at medium risk for hERG inhibition. Also they were found to be inactive for androgen receptor and aromatase. These results imply that catechin and nakinadine B may be suggested as QS modulators, which may reduce the virulence factors of drug-resistant bacteria.


Assuntos
Catequina , Percepção de Quorum , Antibacterianos/química , Proteínas de Bactérias/química , Biofilmes , Catequina/farmacologia , Resistência a Medicamentos , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Fatores de Virulência/química , Fatores de Virulência/metabolismo , Fatores de Virulência/farmacologia
12.
Proc Natl Acad Sci U S A ; 119(11): e2118002119, 2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-35271389

RESUMO

SignificanceYeiE has been identified as a master virulence factor of Cronobacter sakazakii. In this study, we determined the crystal structures of the regulatory domain of YeiE in complex with its physiological ligand sulfite ion (SO32-). The structure provides the basis for the molecular mechanisms for sulfite sensing and the ligand-dependent conformational changes of the regulatory domain. The genes under the control of YeiE in response to sulfite were investigated to reveal the functional roles of YeiE in the sulfite tolerance of the bacteria. We propose the molecular mechanism underlying the ability of gram-negative pathogens to defend against the innate immune response involving sulfite, thus providing a strategy to control the pathogenesis of bacteria.


Assuntos
Proteínas de Bactérias , Cronobacter sakazakii , Estresse Fisiológico , Sulfitos , Fatores de Transcrição , Fatores de Virulência , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Cronobacter sakazakii/genética , Cronobacter sakazakii/metabolismo , Cronobacter sakazakii/patogenicidade , Cristalização , Ligantes , Domínios Proteicos , Sulfitos/metabolismo , Fatores de Transcrição/química , Fatores de Transcrição/genética , Fatores de Virulência/química , Fatores de Virulência/genética
13.
Front Immunol ; 13: 815775, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35154132

RESUMO

Many species of bacteria produce toxins such as cholesterol-dependent cytolysins that form pores in cell membranes. Membrane pores facilitate infection by releasing nutrients, delivering virulence factors, and causing lytic cell damage - cytolysis. Oxysterols are oxidized forms of cholesterol that regulate cellular cholesterol and alter immune responses to bacteria. Whether oxysterols also influence the protection of cells against pore-forming toxins is unresolved. Here we tested the hypothesis that oxysterols stimulate the intrinsic protection of epithelial cells against damage caused by cholesterol-dependent cytolysins. We treated epithelial cells with oxysterols and then challenged them with the cholesterol-dependent cytolysin, pyolysin. Treating HeLa cells with 27-hydroxycholesterol, 25-hydroxycholesterol, 7α-hydroxycholesterol, or 7ß-hydroxycholesterol reduced pyolysin-induced leakage of lactate dehydrogenase and reduced pyolysin-induced cytolysis. Specifically, treatment with 10 ng/ml 27-hydroxycholesterol for 24 h reduced pyolysin-induced lactate dehydrogenase leakage by 88%, and reduced cytolysis from 74% to 1%. Treating HeLa cells with 27-hydroxycholesterol also reduced pyolysin-induced leakage of potassium ions, prevented mitogen-activated protein kinase cell stress responses, and limited alterations in the cytoskeleton. Furthermore, 27-hydroxycholesterol reduced pyolysin-induced damage in lung and liver epithelial cells, and protected against the cytolysins streptolysin O and Staphylococcus aureus α-hemolysin. Although oxysterols regulate cellular cholesterol by activating liver X receptors, cytoprotection did not depend on liver X receptors or changes in total cellular cholesterol. However, oxysterol cytoprotection was partially dependent on acyl-CoA:cholesterol acyltransferase (ACAT) reducing accessible cholesterol in cell membranes. Collectively, these findings imply that oxysterols stimulate the intrinsic protection of epithelial cells against pore-forming toxins and may help protect tissues against pathogenic bacteria.


Assuntos
Bactérias/química , Proteínas de Bactérias/toxicidade , Toxinas Bacterianas/toxicidade , Proteínas Hemolisinas/toxicidade , Oxisteróis/farmacologia , Fatores de Virulência/toxicidade , Proteínas de Bactérias/química , Toxinas Bacterianas/química , Células Epiteliais/metabolismo , Células HeLa , Proteínas Hemolisinas/química , Humanos , Fatores de Virulência/química
14.
Int J Mol Sci ; 23(3)2022 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-35163540

RESUMO

Cytospora chrysosperma is the main causal agent of poplar canker disease in China, especially in some areas with poor site conditions. Pathogens secrete a large number of effectors to interfere the plant immunity and promote their infection and colonization. Nevertheless, the roles of effectors in C. chrysosperma remain poorly understood. In this study, we identified and functionally characterized a candidate effector CcSp84 from C. chrysosperma, which contained a nuclear localization signal motif at the C-terminal and was highly induced during infection stages. Transient expression of CcSp84 in Nicotiana benthamiana leaves could trigger cell death. Additionally, deletion of CcSp84 significantly reduced fungal virulence to the polar twigs, while no obvious defects were observed in fungal growth and sensitivity to H2O2. Confocal microscopy revealed that CcSp84 labeled with a green fluorescent protein (GFP) was mainly accumulated in the plant nucleus. Further analysis revealed that the plant nucleus localization of CcSp84 was necessary to trigger plant immune responses, including ROS accumulation, callose deposition, and induced expression of jasmonic acid and ethylene defense-related genes. Collectively, our results suggest that CcSp84 is a virulence-related effector, and plant nucleus localization is required for its functions.


Assuntos
Ascomicetos/patogenicidade , Núcleo Celular/metabolismo , Nicotiana/crescimento & desenvolvimento , Fatores de Virulência/química , Fatores de Virulência/metabolismo , Ascomicetos/metabolismo , Vias Biossintéticas , Proteínas Fúngicas/química , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Deleção de Genes , Regulação Fúngica da Expressão Gênica , Regulação da Expressão Gênica de Plantas , Glucanos/metabolismo , Microscopia Confocal , Sinais de Localização Nuclear , Imunidade Vegetal , Folhas de Planta/genética , Folhas de Planta/crescimento & desenvolvimento , Plantas Geneticamente Modificadas/crescimento & desenvolvimento , Domínios Proteicos , Espécies Reativas de Oxigênio , Nicotiana/genética , Nicotiana/metabolismo , Fatores de Virulência/genética
15.
J Am Chem Soc ; 144(4): 1545-1555, 2022 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-35060711

RESUMO

Development of viable therapeutics to effectively combat tier I pneumopathogens such as Yersinia pestis requires a thorough understanding of proteins vital for pathogenicity. The host invasion protein Ail, although indispensable for Yersinia pathogenesis, has evaded detailed characterization, as it is an outer membrane protein with intrinsically low stability and high aggregation propensity. Here, we identify molecular elements of the metastable Ail structure that considerably alter protein-lipid and intraprotein thermodynamics. In addition, we find that four residues Q50, L88, L92, and A94 contribute additively to the lowered stability of Ail, and their conserved substitution is sufficient to re-engineer Ail to Out14, a thermodynamically hyperstable low-aggregation variant with a functional scaffold. Interestingly, Ail also shows two (parallel) folding pathways, which has not yet been reported for ß-barrel membrane proteins. Additionally, we identify the molecular mechanism of enhanced thermodynamic stability of Out14. We show that this enhanced stability of Out14 is due to a favorable change in the nonpolar accessible surface, and the accumulation of a kinetically accelerated off-pathway folding intermediate, which is absent in wild-type Ail. Such engineered hyperstable Ail ß-barrels can be harnessed for targeted drug screening and developing medical countermeasures against Yersiniae. Application of similar strategies will help design effective translational therapeutics to combat biopathogens.


Assuntos
Proteínas da Membrana Bacteriana Externa/química , Fatores de Virulência/química , Yersinia pestis/metabolismo , Sequência de Aminoácidos , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/metabolismo , Cinética , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , Conformação Proteica em Folha beta , Dobramento de Proteína , Estabilidade Proteica , Alinhamento de Sequência , Termodinâmica , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
16.
Appl Biochem Biotechnol ; 194(1): 37-53, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34762267

RESUMO

In the Asian region, Helicobacter pylori infects about 80% populations, which is most leading cause of peptic ulcers, and it is an asymptomatic infection. Studies reported that the particular bacteria carry specific virulence factors that leads to severe complications. These virulence factors can be used as a drug targets to inhibit their growth and pathogenicity. Chronic infection with H. pylori virulence factors are CagA, VacA and HtrA positive strains the risk factor of gastric cancer. In this study, we aimed to study the antagonistic interaction pattern between the potential eight algal peptides against the virulence factors of H. pylori through in silico analysis intended to treat peptic ulcer and prevent the further complications such as cancer. The proteins of virulent factors are docked using C-Docker algorithm and calculated the bind energy of the complexes. The results showed that the peptide derived from a green alga, Tetradesmus sp. are active against the three virulent factors such as cag-A, vac-A, and Htr-A with multiple hydrogen, vdW, electrostatic interactions, and mild π-hydrophobic bindings with the libdock energy score for CagA, VacA and HtrA are 175.625, 158.603 and 89.397 kcal/mol. These primes and the peptide lead to develop a better and potential inhibitors against H. pylori infection.


Assuntos
Proteínas de Algas/química , Proteínas de Bactérias , Clorófitas/química , Helicobacter pylori , Peptídeos/química , Fatores de Virulência , Proteínas de Algas/farmacologia , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/química , Simulação por Computador , Helicobacter pylori/química , Helicobacter pylori/patogenicidade , Peptídeos/farmacologia , Fatores de Virulência/antagonistas & inibidores , Fatores de Virulência/química
17.
Appl Biochem Biotechnol ; 194(1): 354-367, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34843077

RESUMO

Gastric cancer is a pathological condition induced by the bacteria Helicobacter pylori. Targeting the key virulence factors of H. pylori causing gastric cancer is a promising method for treating gastric cancer. Recently, research has been focused on analyzing the adrenergic, cholinergic, and anti-cancer properties of their venom proteins. Testing the anti-cancer activity of the lethal proteins in the venom of P. volitans provides a bioactive compound for cancer treatment. Still, it is also helpful to eliminate the ecological imbalance caused by these fish in the marine environment. This study focuses on an in silico approach using Z-dock to analyze the bioactive prospective of the venom proteins of P. volitans against the essential virulence proteins of H. pylori responsible for inducing cancer. Our in silico docking study using a computational model of the venom proteins and H. pylori proteins has displayed the possible interactions between these proteins. The results revealed that P. volitans hyaluronidase and PV toxin's venom proteins effectively interact with H. pylori proteins Cag A, Cag L, GGT, Cag D, and urease that may be promising proteins in cancer therapy.


Assuntos
Proteínas de Bactérias/química , Proteínas de Peixes/química , Venenos de Peixe/química , Helicobacter pylori/química , Simulação de Acoplamento Molecular , Perciformes , Fatores de Virulência/química , Animais , Humanos , Neoplasias Gástricas
18.
J Virol ; 96(1): e0141921, 2022 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-34668772

RESUMO

African swine fever (ASF) is currently causing a major pandemic affecting the swine industry and protein availability from Central Europe to East and South Asia. No commercial vaccines are available, making disease control dependent on the elimination of affected animals. Here, we show that the deletion of the African swine fever virus (ASFV) E184L gene from the highly virulent ASFV Georgia 2010 (ASFV-G) isolate produces a reduction in virus virulence during the infection in swine. Of domestic pigs intramuscularly inoculated with a recombinant virus lacking the E184L gene (ASFV-G-ΔE184L), 40% experienced a significantly (5 days) delayed presentation of clinical disease and, overall, had a 60% rate of survival compared to animals inoculated with the virulent parental ASFV-G. Importantly, all animals surviving ASFV-G-ΔE184L infection developed a strong antibody response and were protected when challenged with ASFV-G. As expected, a pool of sera from ASFV-G-ΔE184L-inoculated animals lacked any detectable antibody response to peptides partially representing the E184L protein, while sera from animals inoculated with an efficacious vaccine candidate, ASFV-G-ΔMGF, strongly recognize the same set of peptides. These results support the potential use of the E184L deletion for the development of vaccines able to differentiate infected from vaccinated animals (DIVA). Therefore, it is shown here that the E184L gene is a novel ASFV determinant of virulence that can potentially be used to increase safety in preexisting vaccine candidates, as well as to provide them with DIVA capabilities. To our knowledge, E184L is the first ASFV gene product experimentally shown to be a functional DIVA antigenic marker. IMPORTANCE No commercial vaccines are available to prevent African swine fever (ASF). The ASF pandemic caused by the ASF virus Georgia 2010 (ASFV-G) strain is seriously affecting pork production in a contiguous geographical area from Central Europe to East Asia. The only effective experimental vaccines are viruses attenuated by deleting ASFV genes associated with virus virulence. Therefore, identification of such genes is of critical importance for vaccine development. Here, we report the discovery of a novel determinant of ASFV virulence, the E184L gene. Deletion of the E184L gene from the ASFV-G genome (ASFV-G-ΔE184L) produced a reduction in virus virulence, and importantly, animals surviving infection with ASFV-G-ΔE184L were protected from developing ASF after challenge with the virulent parental virus ASFV-G. Importantly, the virus protein encoded by E184L is highly immunogenic, making a virus lacking this gene a vaccine candidate that allows the differentiation of infected from vaccinated animals (DIVA). Here, we show that unlike what is observed in animals inoculated with the vaccine candidate ASFV-G-ΔMGF, ASFV-G-ΔE184L-inoculated animals do not mount a E184L-specific antibody response, indicating the feasibility of using the E184L deletion as the antigenic marker for the development of a DIVA vaccine in ASFV.


Assuntos
Vírus da Febre Suína Africana/genética , Febre Suína Africana/virologia , Interações Hospedeiro-Patógeno , Deleção de Sequência , Proteínas Virais/genética , Fatores de Virulência/genética , Febre Suína Africana/diagnóstico , Vírus da Febre Suína Africana/classificação , Sequência de Aminoácidos , Animais , Temperatura Corporal , Sequência Conservada , Regulação Viral da Expressão Gênica , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/virologia , Filogenia , Suínos , Proteínas Virais/química , Proteínas Virais/metabolismo , Viremia , Virulência , Fatores de Virulência/química , Fatores de Virulência/metabolismo , Replicação Viral
19.
Molecules ; 26(22)2021 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-34833903

RESUMO

Multi-drug resistance (MDR) bacterial pathogens pose a threat to global health and warrant the discovery of new therapeutic molecules, particularly those that can neutralize their virulence and stop the evolution of new resistant mechanisms. The superbug nosocomial pathogen, Pseudomonas aeruginosa, uses a multiple virulence factor regulator (MvfR) to regulate the expression of multiple virulence proteins during acute and persistent infections. The present study targeted MvfR with the intention of designing novel anti-virulent compounds, which will function in two ways: first, they will block the virulence and pathogenesis P. aeruginosa by disrupting the quorum-sensing network of the bacteria, and second, they will stop the evolution of new resistant mechanisms. A structure-based virtual screening (SBVS) method was used to screen druglike compounds from the Asinex antibacterial library (~5968 molecules) and the comprehensive marine natural products database (CMNPD) (~32 thousand compounds), against the ligand-binding domain (LBD) of MvfR, to identify molecules that show high binding potential for the relevant pocket. In this way, two compounds were identified: Top-1 (4-((carbamoyloxy)methyl)-10,10-dihydroxy-2,6-diiminiodecahydropyrrolo[1,2-c]purin-9-yl sulfate) and Top-2 (10,10-dihydroxy-2,6-diiminio-4-(((sulfonatocarbamoyl)oxy)methyl)decahydropyrrolo[1,2-c]purin-9-yl sulfate), in contrast to the co-crystallized M64 control. Both of the screened leads were found to show deep pocket binding and interactions with several key residues through a network of hydrophobic and hydrophilic interactions. The docking results were validated by a long run of 200 ns of molecular dynamics simulation and MM-PB/GBSA binding free energies. All of these analyses confirmed the presence of strong complex formation and rigorous intermolecular interactions. An additional analysis of normal mode entropy and a WaterSwap assay were also performed to complement the aforementioned studies. Lastly, the compounds were found to show an acceptable range of pharmacokinetic properties, making both compounds potential candidates for further experimental studies to decipher their real biological potency.


Assuntos
Antibacterianos/farmacologia , Pseudomonas aeruginosa/patogenicidade , Fatores de Virulência/antagonistas & inibidores , Antibacterianos/química , Antibacterianos/farmacocinética , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/química , Proteínas de Bactérias/fisiologia , Sítios de Ligação , Bases de Dados de Produtos Farmacêuticos , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos , Farmacorresistência Bacteriana Múltipla , Humanos , Ligação de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Ligantes , Testes de Sensibilidade Microbiana , Simulação de Dinâmica Molecular , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/fisiologia , Bibliotecas de Moléculas Pequenas , Interface Usuário-Computador , Fatores de Virulência/química , Fatores de Virulência/fisiologia
20.
ACS Chem Biol ; 16(12): 2776-2786, 2021 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-34767712

RESUMO

Nonribosomal peptide synthetases (NRPSs) are typically multimodular enzymes that assemble amino acids or carboxylic acids into complex natural products. Here, we characterize a monomodular NRPS, PvfC, encoded by the Pseudomonas virulence factor (pvf) gene cluster that is essential for virulence and signaling in different bacterial species. PvfC exhibits a unique adenylation-thiolation-reductase (ATR) domain architecture that is understudied in bacteria. We show that the activity of PvfC is essential in the production of seven leucine-derived heterocyclic natural products, including two pyrazines, a pyrazinone, and a rare disubstituted imidazole, as well as three pyrazine N-oxides that require an additional N-oxygenation step. Mechanistic studies reveal that PvfC, without a canonical peptide-forming domain, makes a dipeptide aldehyde intermediate en route to both the pyrazinone and imidazole. Our work identifies a novel biosynthetic route for the production of pyrazinones, an emerging class of signaling molecules and virulence factors. Our discovery also showcases the ability of monomodular NRPSs to generate amino acid- and dipeptide-aldehydes that lead to diverse natural products. The diversity-prone biosynthesis by the pvf-encoded enzymes sets the stage for further understanding the functions of pvf in bacterial cell-to-cell signaling.


Assuntos
Produtos Biológicos/química , Peptídeo Sintases/metabolismo , Fatores de Virulência/química , Aldeídos/química , Aminoácidos/química , Ácidos Carboxílicos/química , Óxidos N-Cíclicos/química , Dipeptídeos/química , Imidazóis/química , Pseudomonas/enzimologia , Pirazinas/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...