Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25.990
Filtrar
1.
Nat Commun ; 15(1): 3437, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38653755

RESUMO

Phytoalexin sakuranetin functions in resistance against rice blast. However, the mechanisms underlying the effects of sakuranetin remains elusive. Here, we report that rice lines expressing resistance (R) genes were found to contain high levels of sakuranetin, which correlates with attenuated endocytic trafficking of plasma membrane (PM) proteins. Exogenous and endogenous sakuranetin attenuates the endocytosis of various PM proteins and the fungal effector PWL2. Moreover, accumulation of the avirulence protein AvrCO39, resulting from uptake into rice cells by Magnaporthe oryzae, was reduced following treatment with sakuranetin. Pharmacological manipulation of clathrin-mediated endocytic (CME) suggests that this pathway is targeted by sakuranetin. Indeed, attenuation of CME by sakuranetin is sufficient to convey resistance against rice blast. Our data reveals a mechanism of rice against M. oryzae by increasing sakuranetin levels and repressing the CME of pathogen effectors, which is distinct from the action of many R genes that mainly function by modulating transcription.


Assuntos
Ascomicetos , Resistência à Doença , Endocitose , Flavonoides , Oryza , Fitoalexinas , Doenças das Plantas , Proteínas de Plantas , Oryza/microbiologia , Oryza/metabolismo , Oryza/efeitos dos fármacos , Oryza/genética , Doenças das Plantas/microbiologia , Endocitose/efeitos dos fármacos , Resistência à Doença/genética , Resistência à Doença/efeitos dos fármacos , Proteínas de Plantas/metabolismo , Proteínas de Plantas/genética , Sesquiterpenos/farmacologia , Sesquiterpenos/metabolismo , Regulação da Expressão Gênica de Plantas/efeitos dos fármacos , Membrana Celular/metabolismo , Membrana Celular/efeitos dos fármacos , Plantas Geneticamente Modificadas , Proteínas Fúngicas/metabolismo , Proteínas Fúngicas/genética
2.
Nat Commun ; 15(1): 3424, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38654023

RESUMO

Developing unique mechanisms of action are essential to combat the growing issue of antimicrobial resistance. Supramolecular assemblies combining the improved biostability of non-natural compounds with the complex membrane-attacking mechanisms of natural peptides are promising alternatives to conventional antibiotics. However, for such compounds the direct visual insight on antibacterial action is still lacking. Here we employ a design strategy focusing on an inducible assembly mechanism and utilized electron microscopy (EM) to follow the formation of supramolecular structures of lysine-rich heterochiral ß3-peptides, termed lamellin-2K and lamellin-3K, triggered by bacterial cell surface lipopolysaccharides. Combined molecular dynamics simulations, EM and bacterial assays confirmed that the phosphate-induced conformational change on these lamellins led to the formation of striped lamellae capable of incising the cell envelope of Gram-negative bacteria thereby exerting antibacterial activity. Our findings also provide a mechanistic link for membrane-targeting agents depicting the antibiotic mechanism derived from the in-situ formation of active supramolecules.


Assuntos
Antibacterianos , Membrana Celular , Simulação de Dinâmica Molecular , Antibacterianos/farmacologia , Antibacterianos/química , Membrana Celular/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Testes de Sensibilidade Microbiana , Peptídeos/química , Peptídeos/farmacologia , Microscopia Eletrônica , Bactérias Gram-Negativas/efeitos dos fármacos , Escherichia coli/efeitos dos fármacos
3.
J Biol Chem ; 300(3): 105701, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38301897

RESUMO

Fungal keratitis is the foremost cause of corneal infections worldwide, of which Fusariumspp. is the common etiological agent that causes loss of vision and warrants surgical intervention. An increase in resistance to the available drugs along with severe side effects of the existing antifungals demands for new effective antimycotics. Here, we demonstrate that antimicrobial peptide S100A12 directly binds to the phospholipids of the fungal membrane, disrupts the structural integrity, and induces generation of reactive oxygen species in fungus. In addition, it inhibits biofilm formation by Fusariumspp. and exhibits antifungal property against Fusariumspp. both in vitro and in vivo. Taken together, our results delve into specific effect of S100A12 against Fusariumspp. with an aim to investigate new antifungal compounds to combat fungal keratitis.


Assuntos
Antifúngicos , Biofilmes , Membrana Celular , Fusarium , Proteína S100A12 , Antifúngicos/metabolismo , Antifúngicos/farmacologia , Biofilmes/efeitos dos fármacos , Infecções Oculares Fúngicas/microbiologia , Fusarium/efeitos dos fármacos , Ceratite/microbiologia , Proteína S100A12/metabolismo , Proteína S100A12/farmacologia , Humanos , Membrana Celular/efeitos dos fármacos , Fosfolipídeos/metabolismo , Espécies Reativas de Oxigênio/metabolismo
4.
Nature ; 625(7995): 572-577, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38172635

RESUMO

Gram-negative bacteria are extraordinarily difficult to kill because their cytoplasmic membrane is surrounded by an outer membrane that blocks the entry of most antibiotics. The impenetrable nature of the outer membrane is due to the presence of a large, amphipathic glycolipid called lipopolysaccharide (LPS) in its outer leaflet1. Assembly of the outer membrane requires transport of LPS across a protein bridge that spans from the cytoplasmic membrane to the cell surface. Maintaining outer membrane integrity is essential for bacterial cell viability, and its disruption can increase susceptibility to other antibiotics2-6. Thus, inhibitors of the seven lipopolysaccharide transport (Lpt) proteins that form this transenvelope transporter have long been sought. A new class of antibiotics that targets the LPS transport machine in Acinetobacter was recently identified. Here, using structural, biochemical and genetic approaches, we show that these antibiotics trap a substrate-bound conformation of the LPS transporter that stalls this machine. The inhibitors accomplish this by recognizing a composite binding site made up of both the Lpt transporter and its LPS substrate. Collectively, our findings identify an unusual mechanism of lipid transport inhibition, reveal a druggable conformation of the Lpt transporter and provide the foundation for extending this class of antibiotics to other Gram-negative pathogens.


Assuntos
Antibacterianos , Proteínas da Membrana Bacteriana Externa , Lipopolissacarídeos , Proteínas de Membrana Transportadoras , Acinetobacter/química , Acinetobacter/efeitos dos fármacos , Acinetobacter/genética , Antibacterianos/farmacologia , Antibacterianos/metabolismo , Proteínas da Membrana Bacteriana Externa/antagonistas & inibidores , Proteínas da Membrana Bacteriana Externa/química , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/metabolismo , Sítios de Ligação/efeitos dos fármacos , Transporte Biológico/efeitos dos fármacos , Membrana Celular/química , Membrana Celular/efeitos dos fármacos , Membrana Celular/genética , Membrana Celular/metabolismo , Lipopolissacarídeos/metabolismo , Proteínas de Membrana Transportadoras/química , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Viabilidade Microbiana , Conformação Proteica/efeitos dos fármacos , Especificidade por Substrato
5.
J Mol Biol ; 435(11): 167953, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-37330283

RESUMO

Membranes form the first line of defence of bacteria against potentially harmful molecules in the surrounding environment. Understanding the protective properties of these membranes represents an important step towards development of targeted anti-bacterial agents such as sanitizers. Use of propanol, isopropanol and chlorhexidine can significantly decrease the threat imposed by bacteria in the face of growing anti-bacterial resistance via mechanisms that include membrane disruption. Here we have employed molecular dynamics simulations and nuclear magnetic resonance to explore the impact of chlorhexidine and alcohol on the S. aureus cell membrane, as well as the E. coli inner and outer membranes. We identify how sanitizer components partition into these bacterial membranes, and show that chlorhexidine is instrumental in this process.


Assuntos
1-Propanol , 2-Propanol , Antibacterianos , Clorexidina , Escherichia coli , Higienizadores de Mão , Staphylococcus aureus , Antibacterianos/farmacologia , Clorexidina/farmacologia , Escherichia coli/efeitos dos fármacos , Espectroscopia de Ressonância Magnética , Staphylococcus aureus/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , 1-Propanol/farmacologia , 2-Propanol/farmacologia , Higienizadores de Mão/farmacologia
6.
World J Microbiol Biotechnol ; 39(6): 164, 2023 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-37069369

RESUMO

Underestimating fungal infections led to a gap in the development of antifungal medication. However, rising rates of morbidity and mortality with fungal infection have revealed an alarming rise in antifungal resistance also. Due to the eukaryotic properties of fungi and the close evolutionary similarity between fungal cells and human hosts, therapeutic targeting of Candida infections is troublesome, along with the development of resistance. The discovery of new antifungals is so far behind schedule that the antifungal pipeline is nearly empty. Previously, we have reported the activity and susceptibility of Sodium lignosulfonate (LIG) against C. albicans. In this work, we have established the mechanistic actions of LIG's activity. We performed flow cytometric analysis for membrane integrity, ergosterol binding assay, crystal violet assay, and membrane leakage assay to analyze quantitatively that the C. albicans membrane is being disrupted in response to LIG. Electron microscopic analysis with SEM and TEM confirmed changes in Candida cellular morphology and membrane perturbation respectively. These findings indicated that LIG causes cell membrane damage in C. albicans. This knowledge about LIG's mechanism of action against C. albicans could be used to explore it further as a lead antifungal molecule to develop it as a potent candidate for antifungal therapeutics in the future.


Assuntos
Antifúngicos , Candida albicans , Lignina , Lignina/análogos & derivados , Candida albicans/citologia , Candida albicans/efeitos dos fármacos , Antifúngicos/farmacologia , Membrana Celular/química , Membrana Celular/efeitos dos fármacos
7.
Nature ; 613(7945): 729-734, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36450357

RESUMO

Peptidoglycan and almost all surface glycopolymers in bacteria are built in the cytoplasm on the lipid carrier undecaprenyl phosphate (UndP)1-4. These UndP-linked precursors are transported across the membrane and polymerized or directly transferred to surface polymers, lipids or proteins. UndP is then flipped to regenerate the pool of cytoplasmic-facing UndP. The identity of the flippase that catalyses transport has remained unknown. Here, using the antibiotic amphomycin that targets UndP5-7, we identified two broadly conserved protein families that affect UndP recycling. One (UptA) is a member of the DedA superfamily8; the other (PopT) contains the domain DUF368. Genetic, cytological and syntenic analyses indicate that these proteins are UndP transporters. Notably, homologues from Gram-positive and Gram-negative bacteria promote UndP transport in Bacillus subtilis, indicating that recycling activity is broadly conserved among family members. Inhibitors of these flippases could potentiate the activity of antibiotics targeting the cell envelope.


Assuntos
Proteínas de Bactérias , Proteínas de Transporte , Sequência Conservada , Evolução Molecular , Bactérias Gram-Negativas , Bactérias Gram-Positivas , Fosfatos de Poli-Isoprenil , Antibacterianos/farmacologia , Bacillus subtilis/citologia , Bacillus subtilis/efeitos dos fármacos , Bacillus subtilis/genética , Bacillus subtilis/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/classificação , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas de Transporte/química , Proteínas de Transporte/classificação , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Bactérias Gram-Negativas/citologia , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Negativas/genética , Bactérias Gram-Negativas/metabolismo , Bactérias Gram-Positivas/citologia , Bactérias Gram-Positivas/efeitos dos fármacos , Bactérias Gram-Positivas/genética , Bactérias Gram-Positivas/metabolismo , Fosfatos de Poli-Isoprenil/metabolismo , Sintenia , Peptidoglicano/metabolismo , Parede Celular/química , Parede Celular/metabolismo
8.
Bioorg Chem ; 130: 106252, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36379149

RESUMO

We investigated synthetic amino acid-based approach to design short peptide-based antibiotics. Tautomerically restricted, amphiphilic 1-aryl-l-histidines along with hydrophobic tryptophan were utilized to synthesize the designed peptides. l-Trp-l-His(1-biphenyl)-NHBzl (12e, IC50 = 1.91 µg/mL; MIC = 3.46 µg/mL) and l-His[1-(4-n-butylphenyl)]-l-Trp-l-His[1-(4-n-butylphenyl)]-NHBzl (16d, IC50 = 1.36 µg/mL; MIC = 2.46 µg/mL) produced potency against Cryptococcus neoformans. Peptides with moderate antibacterial activities (IC50s = 4.40-8.80 µg/mL) were also identified. The mechanism of action and cellular changes revealed that membrane disruption due to interactions of the positively charged peptides with the negatively charged membrane of the cryptococcal cells result in permeabilization, leading to pore formation. The internal localization of the peptides instigated the interactions with DNA causing fragmentation of the genetic material, which together with membrane disruption led to cell death. Flow cytometric analysis points to cells death by apoptotic pathway. Time kill kinetics and synergistic study confirmed the fungicidal nature and synergism with amphotericin B.


Assuntos
Membrana Celular , Criptococose , Cryptococcus neoformans , Peptídeos , Aminoácidos/metabolismo , Antifúngicos/farmacologia , Antifúngicos/metabolismo , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Criptococose/tratamento farmacológico , Testes de Sensibilidade Microbiana , Peptídeos/farmacologia , Peptídeos/metabolismo
9.
Nature ; 608(7922): 390-396, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35922513

RESUMO

Antibiotics that use novel mechanisms are needed to combat antimicrobial resistance1-3. Teixobactin4 represents a new class of antibiotics with a unique chemical scaffold and lack of detectable resistance. Teixobactin targets lipid II, a precursor of peptidoglycan5. Here we unravel the mechanism of teixobactin at the atomic level using a combination of solid-state NMR, microscopy, in vivo assays and molecular dynamics simulations. The unique enduracididine C-terminal headgroup of teixobactin specifically binds to the pyrophosphate-sugar moiety of lipid II, whereas the N terminus coordinates the pyrophosphate of another lipid II molecule. This configuration favours the formation of a ß-sheet of teixobactins bound to the target, creating a supramolecular fibrillar structure. Specific binding to the conserved pyrophosphate-sugar moiety accounts for the lack of resistance to teixobactin4. The supramolecular structure compromises membrane integrity. Atomic force microscopy and molecular dynamics simulations show that the supramolecular structure displaces phospholipids, thinning the membrane. The long hydrophobic tails of lipid II concentrated within the supramolecular structure apparently contribute to membrane disruption. Teixobactin hijacks lipid II to help destroy the membrane. Known membrane-acting antibiotics also damage human cells, producing undesirable side effects. Teixobactin damages only membranes that contain lipid II, which is absent in eukaryotes, elegantly resolving the toxicity problem. The two-pronged action against cell wall synthesis and cytoplasmic membrane produces a highly effective compound targeting the bacterial cell envelope. Structural knowledge of the mechanism of teixobactin will enable the rational design of improved drug candidates.


Assuntos
Antibacterianos , Bactérias , Membrana Celular , Depsipeptídeos , Viabilidade Microbiana , Antibacterianos/química , Antibacterianos/farmacologia , Bactérias/citologia , Bactérias/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Parede Celular/efeitos dos fármacos , Parede Celular/metabolismo , Depsipeptídeos/química , Depsipeptídeos/farmacologia , Difosfatos/química , Farmacorresistência Bacteriana/efeitos dos fármacos , Humanos , Lipídeos/química , Testes de Sensibilidade Microbiana , Viabilidade Microbiana/efeitos dos fármacos , Microscopia de Força Atômica , Simulação de Dinâmica Molecular , Ressonância Magnética Nuclear Biomolecular , Estrutura Secundária de Proteína , Pirrolidinas/química , Açúcares/química
10.
Elife ; 112022 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-35670565

RESUMO

We report the real-time response of Escherichia coli to lactoferricin-derived antimicrobial peptides (AMPs) on length scales bridging microscopic cell sizes to nanoscopic lipid packing using millisecond time-resolved synchrotron small-angle X-ray scattering. Coupling a multiscale scattering data analysis to biophysical assays for peptide partitioning revealed that the AMPs rapidly permeabilize the cytosolic membrane within less than 3 s-much faster than previously considered. Final intracellular AMP concentrations of ∼80-100 mM suggest an efficient obstruction of physiologically important processes as the primary cause of bacterial killing. On the other hand, damage of the cell envelope and leakage occurred also at sublethal peptide concentrations, thus emerging as a collateral effect of AMP activity that does not kill the bacteria. This implies that the impairment of the membrane barrier is a necessary but not sufficient condition for microbial killing by lactoferricins. The most efficient AMP studied exceeds others in both speed of permeabilizing membranes and lowest intracellular peptide concentration needed to inhibit bacterial growth.


Assuntos
Antibacterianos , Peptídeos Catiônicos Antimicrobianos , Membrana Celular , Escherichia coli , Lactoferrina , Antibacterianos/farmacologia , Peptídeos Catiônicos Antimicrobianos/metabolismo , Peptídeos Catiônicos Antimicrobianos/farmacologia , Bactérias/efeitos dos fármacos , Bactérias/metabolismo , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Escherichia coli/efeitos dos fármacos , Escherichia coli/metabolismo , Espaço Intracelular/química , Espaço Intracelular/microbiologia , Lactoferrina/metabolismo , Lactoferrina/farmacologia , Fatores de Tempo
11.
J Agric Food Chem ; 70(20): 6123-6133, 2022 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-35576531

RESUMO

The present study sought to identify and characterize a novel antimicrobial peptide, named MOp2 from Moringa oleifera seed protein hydrolysates, and elucidate its potential antimicrobial effects on Staphylococcus aureus. MOp2, with the amino acid sequence of His-Val-Leu-Asp-Thr-Pro-Leu-Leu (HVLDTPLL), was characterized as a hydrophobic anionic AMP of the ß-sheet structure. MOp2 exhibited negligible hemolytic activity at 2.0× MIC, suggesting its inhibitory effect on the growth of S. aureus (MIC: 2.204 mM). It maintained more than 90% of antimicrobial activity under 5% salt and about 78% of antimicrobial activity at a high temperature of 115 °C for 30 min. Protease, especially acid protease, reduced its antimicrobial activity to different extents. Moreover, MOp2 caused irreversible membrane damage to S. aureus cells by increasing the membrane permeability, resulting in the release of intracellular nucleotide pools. Additionally, molecular docking revealed that MOp2 could inhibit S. aureus growth by interacting with dihydrofolate reductase and DNA gyrase through hydrogen bonding and hydrophobic interactions. Overall, MOp2 could be a potential novel antimicrobial agent against S. aureus in food processing.


Assuntos
Peptídeos Antimicrobianos , Moringa oleifera , Staphylococcus aureus , Antibacterianos/química , Antibacterianos/farmacologia , Anti-Infecciosos/química , Anti-Infecciosos/farmacologia , Peptídeos Antimicrobianos/química , Peptídeos Antimicrobianos/farmacologia , Membrana Celular/efeitos dos fármacos , Simulação de Acoplamento Molecular , Moringa oleifera/química , Peptídeo Hidrolases/análise , Extratos Vegetais/química , Hidrolisados de Proteína/farmacologia , Sementes/química , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/prevenção & controle , Staphylococcus aureus/metabolismo
12.
Sci Rep ; 12(1): 1987, 2022 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-35132082

RESUMO

In earlier reports, we have shown the antimicrobial activity of a host neuropeptide, alpha-melanocyte stimulating hormone (α-MSH) and its cationic analogues against Staphylococcus aureus. These analogues of α-MSH showed enhanced staphylocidal activity without any significant mammalian cell toxicity. Therefore, here, we explored the antimicrobial activity of α-MSH and its cationic analogues against Escherichia coli. Though the presence of lipopolysaccharide (LPS) in Gram-negative bacteria enables them to resist most conventional antibiotics, encouragingly α-MSH and its four analogues showed killing of both logarithmic and stationary phase E. coli cells in a time, dose and cationicity-dependent manner. In fact, the most cationic analogue, KKK-MSH with a + 5 charge, demonstrated successful eradication of 105 CFU/mL of E. coli cells within 15 min at a concentration as low as 1 µM. BC displacement experiment revealed that cationicity of the peptides was directly related to the killing efficacy of these α-MSH analogues against E. coli cells via initial LPS-binding, leading to rapid disruption of the LPS-outer membrane complex followed by inner bacterial membrane damage and eventual cell death. Here, we propose α-MSH based cationic peptides as promising future agents with broad-spectrum antibacterial efficacy against both Gram-negative and Gram-positive pathogens.


Assuntos
Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Escherichia coli/efeitos dos fármacos , Lipopolissacarídeos/metabolismo , alfa-MSH/análogos & derivados , alfa-MSH/farmacologia , Relação Dose-Resposta a Droga , Farmacorresistência Bacteriana , Escherichia coli/citologia , Escherichia coli/metabolismo , Ligação Proteica , Relação Estrutura-Atividade , alfa-MSH/química , alfa-MSH/metabolismo
13.
PLoS Pathog ; 18(2): e1010307, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35130322

RESUMO

Antibiotic tolerance is an understudied potential contributor to antibiotic treatment failure and the emergence of multidrug-resistant bacteria. The molecular mechanisms governing tolerance remain poorly understood. A prominent type of ß-lactam tolerance relies on the formation of cell wall-deficient spheroplasts, which maintain structural integrity via their outer membrane (OM), an asymmetric lipid bilayer consisting of phospholipids on the inner leaflet and a lipid-linked polysaccharide (lipopolysaccharide, LPS) enriched in the outer monolayer on the cell surface. How a membrane structure like LPS, with its reliance on mere electrostatic interactions to maintain stability, is capable of countering internal turgor pressure is unknown. Here, we have uncovered a novel role for the PhoPQ two-component system in tolerance to the ß-lactam antibiotic meropenem in Enterobacterales. We found that PhoPQ is induced by meropenem treatment and promotes an increase in 4-amino-4-deoxy-L-aminoarabinose [L-Ara4N] modification of lipid A, the membrane anchor of LPS. L-Ara4N modifications likely enhance structural integrity, and consequently tolerance to meropenem, in several Enterobacterales species. Importantly, mutational inactivation of the negative PhoPQ regulator mgrB (commonly selected for during clinical therapy with the last-resort antibiotic colistin, an antimicrobial peptide [AMP]) results in dramatically enhanced tolerance, suggesting that AMPs can collaterally select for meropenem tolerance via stable overactivation of PhoPQ. Lastly, we identify histidine kinase inhibitors (including an FDA-approved drug) that inhibit PhoPQ-dependent LPS modifications and consequently potentiate meropenem to enhance lysis of tolerant cells. In summary, our results suggest that PhoPQ-mediated LPS modifications play a significant role in stabilizing the OM, promoting survival when the primary integrity maintenance structure, the cell wall, is removed.


Assuntos
Proteínas de Bactérias/metabolismo , Carbapenêmicos/farmacologia , Tolerância a Medicamentos , Enterobacter cloacae/efeitos dos fármacos , Enterobacter cloacae/metabolismo , Lipopolissacarídeos/metabolismo , Antibacterianos/farmacologia , Peptídeos Antimicrobianos/farmacologia , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Colistina/farmacologia , Enterobacter cloacae/genética , Regulação da Expressão Gênica , Histidina Quinase/antagonistas & inibidores , Humanos , Lipídeo A/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Testes de Sensibilidade Microbiana
14.
Microbiol Spectr ; 10(1): e0126221, 2022 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-35171048

RESUMO

The emergence of antimicrobial resistance warrants for the development of improved treatment approaches. In this regard, peptide nucleic acids (PNAs) have shown great promise, exhibiting antibiotic properties through the targeting of cellular nucleic acids. We aimed to study the efficacy of PNA as an anti-tuberculosis agent. Since the efficacy of PNA is limited by its low penetration into the cell, we also investigated combinatorial treatments using permeabilizing drugs to improve PNA efficacy. Various concentrations of anti-inhA PNA, permeabilizing drugs, and their combinations were screened against extracellular and intracellular mycobacteria.0.625 to 5 µM anti-inhA PNA was observed to merely inhibit the growth of extracellular M. smegmatis, while low intracellular bacterial load was reduced by 2 or 2.5 log-fold when treated with 2.5 or 5 µM PNA, respectively. Anti-inhA PNA against M. tuberculosis H37Ra exhibited bactericidal properties at 2.5 and 5 µM and enabled a slight reduction in intracellular M. tuberculosis at concentrations from 2.5 to 20 µM. Of the permeabilizing drugs tested, ethambutol showed the most permeabilizing potential and ultimately potentiated anti-inhA PNA to the greatest extent, reducing its efficacious concentration to 1.25 µM against both M. smegmatis and M. tuberculosis. Furthermore, an enhanced clearance of 1.3 log-fold was observed for ethambutol-anti-inhA PNA combinations against intracellular M. tuberculosis. Thus, permeabilizing drug-PNA combinations indeed exhibit improved efficacies. We therefore propose that anti-inhA PNA could improve therapy even when applied in minute doses as an addition to the current anti-tuberculosis drug regimen. IMPORTANCE Peptide nucleic acids have great potential in therapeutics as anti-gene/anti-sense agents. However, their limited uptake in cells has curtailed their widespread application. Through this study, we explore a PNA-drug combinatorial strategy to improve the efficacy of PNAs and reduce their effective concentrations. This work also focuses on improving tuberculosis treatment, which is hindered by the emergence of antimicrobial-resistant strains of Mycobacterium tuberculosis. It is observed that the antibacterial efficacy of anti-inhA PNA is enhanced when it is combined with permeabilizing drugs, particularly ethambutol. This indicates that the addition of even small concentrations of anti-inhA PNA to the current TB regimen could potentiate their therapeutic efficiency. We hypothesize that this system would also overcome isoniazid resistance, since the resistance mutations lie outside the designed anti-inhA PNA target site.


Assuntos
Antituberculosos/farmacologia , Proteínas de Bactérias/genética , Ceftazidima/farmacologia , Colistina/farmacologia , Etambutol/farmacologia , Mycobacterium smegmatis/efeitos dos fármacos , Mycobacterium tuberculosis/efeitos dos fármacos , Oxirredutases/genética , Ácidos Nucleicos Peptídicos/farmacologia , Proteínas de Bactérias/metabolismo , Membrana Celular/efeitos dos fármacos , Permeabilidade da Membrana Celular , Sinergismo Farmacológico , Humanos , Testes de Sensibilidade Microbiana , Mycobacterium smegmatis/genética , Mycobacterium smegmatis/crescimento & desenvolvimento , Mycobacterium smegmatis/metabolismo , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/crescimento & desenvolvimento , Mycobacterium tuberculosis/metabolismo , Oxirredutases/metabolismo , Ácidos Nucleicos Peptídicos/genética , Tuberculose/tratamento farmacológico , Tuberculose/microbiologia
15.
Org Biomol Chem ; 20(9): 1974-1981, 2022 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-35179161

RESUMO

Trehalose-based probes are useful tools that allow the detection of the mycomembrane of mycobacteria through the metabolic labeling approach. Trehalose analogues conjugated to fluorescent probes can be used, and other probes are functionalized with a bioorthogonal chemical reporter for a two-step labeling approach. The synthesis of such trehalose-based probes mainly relies on the desymmetrization of natural trehalose using a large number of regioselective protection-deprotection steps to differentiate the eight hydroxyl groups. Herein, in order to avoid these time-consuming steps, we reinvestigated our previously reported tandem protocol mediated by FeCl3·6H2O, with the aim of modifying the ratio of the products to allow the challenging desymmetrization of the C2-symmetrical disaccharide trehalose. We demonstrate the usefulness of this method in providing easy access to trehalose analogues with a bioorthogonal moiety or a fluorophore in C-2, and also present their use in a one-step and two-step labeling approach, either of which can be used to study the mycomembrane in live mycobacteria.


Assuntos
Antibacterianos/farmacologia , Membrana Celular/efeitos dos fármacos , Cloretos/farmacologia , Corynebacterium/efeitos dos fármacos , Compostos Férricos/farmacologia , Trealose/farmacologia , Antibacterianos/síntese química , Antibacterianos/química , Cloretos/química , Compostos Férricos/química , Testes de Sensibilidade Microbiana , Trealose/síntese química , Trealose/química
16.
Nat Commun ; 13(1): 587, 2022 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-35102167

RESUMO

Developing CAR T cells for acute myeloid leukemia (AML) has been hampered by a paucity of targets that are expressed on AML blasts and not on hematopoietic progenitor cells (HPCs). Here we demonstrate that GRP78 is expressed on the cell surface of primary AML blasts but not HPCs. To target GRP78, we generate T cell expressing a GRP78-specific peptide-based CAR, which show evidence of minimal fratricide post activation/transduction and antigen-dependent T cell differentiation. GRP78-CAR T cells recognize and kill GRP78-positive AML cells without toxicity to HPCs. In vivo, GRP78-CAR T cells have significant anti-AML activity. To prevent antigen-dependent T cell differentiation, we block CAR signaling and GRP78 cell surface expression post activation by using dasatinib during GRP78-CAR T cell manufacturing. This significantly improves their effector function in vitro and in vivo. Thus, targeting cell surface GRP78-positive AML with CAR T cells is feasible, and warrants further active exploration.


Assuntos
Membrana Celular/metabolismo , Chaperona BiP do Retículo Endoplasmático/imunologia , Células-Tronco Hematopoéticas/imunologia , Leucemia Mieloide Aguda/imunologia , Receptores de Antígenos Quiméricos/imunologia , Linfócitos T/imunologia , Animais , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Citocinas/metabolismo , Citotoxicidade Imunológica/efeitos dos fármacos , Dasatinibe/farmacologia , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Humanos , Leucemia Mieloide Aguda/genética , Camundongos Endogâmicos NOD , Camundongos SCID , Linfócitos T/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
17.
J Med Chem ; 65(4): 3046-3065, 2022 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-35133820

RESUMO

Infectious diseases remain significant health concerns worldwide, and resistance is particularly common in patients with tuberculosis caused by Mycobacterium tuberculosis. The development of anti-infectives with novel modes of action may help overcome resistance. In this regard, membrane-active agents, which modulate membrane components essential for the survival of pathogens, present attractive antimicrobial agents. Key advantages of membrane-active compounds include their ability to target slow-growing or dormant bacteria and their favorable pharmacokinetics. Here, we comprehensively review recent advances in the development of membrane-active chemotypes that target mycobacterial membranes and discuss clinically relevant membrane-active antibacterial agents that have shown promise in counteracting bacterial infections. We discuss the relationship between the membrane properties and the synthetic requirements within the chemical scaffold, as well as the limitations of current membrane-active chemotypes. This review will lay the chemical groundwork for the development of membrane-active antituberculosis agents and will foster the discovery of more effective antitubercular agents.


Assuntos
Antituberculosos/farmacologia , Membrana Celular/efeitos dos fármacos , Mycobacterium tuberculosis/efeitos dos fármacos , Animais , Antituberculosos/química , Desenho de Fármacos , Humanos , Lipídeos/química , Mycobacterium tuberculosis/ultraestrutura , Tuberculose/tratamento farmacológico
18.
Int J Mol Sci ; 23(4)2022 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-35216106

RESUMO

Antimicrobial resistance (AMR) is a global health challenge with methicillin resistant Staphylococcus aureus (MRSA), a leading cause of nosocomial infection. In the search for novel antibiotics, marine sponges have become model organisms as they produce diverse bioactive compounds. We investigated and compared the antibacterial potential of 3 bis-indole alkaloids-bromodeoxytopsentin, bromotopsentin and spongotine A-isolated from the Northeastern Atlantic sponge Spongosorites calcicola. Antimicrobial activity was determined by MIC and time-kill assays. The mechanism of action of bis-indoles was assessed using bacterial cytological profiling via fluorescence microscopy. Finally, we investigated the ability of bis-indole alkaloids to decrease the cytotoxicity of pathogens upon co-incubation with HeLa cells through the measurement of mammalian cell lysis. The bis-indoles were bactericidal to clinically relevant Gram-positive pathogens including MRSA and to the Gram-negative gastroenteric pathogen Vibrio parahaemolyticus. Furthermore, the alkaloids were synergistic in combination with conventional antibiotics. Antimicrobial activity of the bis-indole alkaloids was due to rapid disruption and permeabilization of the bacterial cell membrane. Significantly, the bis-indoles reduced pathogen cytotoxicity toward mammalian cells, indicating their ability to prevent bacterial virulence. In conclusion, sponge bis-indole alkaloids are membrane-permeabilizing agents that represent good antibiotic candidates because of their potency against Gram-positive and Gram-negative bacterial pathogens.


Assuntos
Membrana Celular/efeitos dos fármacos , Alcaloides Indólicos/farmacologia , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Poríferos/química , Animais , Antibacterianos/farmacologia , Linhagem Celular Tumoral , Células HeLa , Humanos , Testes de Sensibilidade Microbiana/métodos
19.
Biochem Biophys Res Commun ; 598: 26-31, 2022 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-35151200

RESUMO

Globins are heme proteins such as hemoglobin (Hb), myoglobin (Mb) and neuroglobin (Ngb), playing important roles in biological system. In addition to normal functions, zebrafish Ngb was able to penetrate cell membranes, whereas less was known for other globin members. In this study, to improve the cell-membrane-penetrating activity of globins, we used sperm whale Mb as a model protein and constructed a quadruple mutant of G5K/Q8K/A19K/V21K Mb (termed 4K Mb), by introduction of four positive charges on the protein surface, which was designed according to the amino acid alignment with that of zebrafish Ngb. Spectroscopic and crystallographic studies showed that the four positively charged Lys residues did not affect the protein structure. Cell-membrane-penetrating essay further showed that 4K Mb exhibited enhanced activity compared to that of native Mb. This study provides valuable information for the effect of distribution of charged residues on the protein structure and the cell-membrane-penetrating activity of globins. Therefore, it will guide the design of protein-based biomaterials for biological applications.


Assuntos
Membrana Celular/metabolismo , Mioglobina/química , Mioglobina/metabolismo , Animais , Membrana Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Dicroísmo Circular , Cristalografia por Raios X , Fluoresceína-5-Isotiocianato/química , Humanos , Lisina/química , Células MCF-7 , Mutação , Mioglobina/genética , Mioglobina/farmacocinética , Espectrofotometria Ultravioleta , Cachalote
20.
Microbiol Spectr ; 10(1): e0012722, 2022 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-35196787

RESUMO

The emergence of antifungal resistance, especially to the most widely used azole class of ergosterol biosynthesis inhibitors, makes fungal infections difficult to treat in clinics and agriculture. When exposed to azoles, fungi can make adaptive responses to alleviate azole toxicity and produce azole tolerance. However, except for azole efflux pumps and ergosterol biosynthesis genes, the role of most azole responsive genes in azole resistance is unknown. In this study, STK-17, whose transcription is upregulated by azoles, was characterized as a novel kinase that is required for azole resistance. Deletion or dysfunction of STK-17 led to azole hypersensitivity in Neurospora crassa and to other ergosterol biosynthesis inhibitors such as amorolfine, terbinafine, and amphotericin B, but not fatty acid and ceramide biosynthesis inhibitors. STK-17 was also required for oxidative stress resistance, but this was not connected to azole resistance. RNA-seq results showed that stk-17 deletion affected the basal expression and the response to ketoconazole of some membrane protein genes, indicating functional association of STK-17 with the membrane. Notably, deletion of stk-17 affected the normal response to azoles of erg genes, including the azole target-encoding gene erg11, and erg2, erg6, and erg24, and led to abnormal accumulation of sterols in the presence of azoles. HPLC-MS/MS analysis revealed increased intracellular azole accumulation in the stk-17 mutant, possibly due to enhanced azole influx and reduced azole efflux that was independent of the major efflux pump CDR4. Importantly, STK-17 was widely distributed and functionally conserved among fungi, thus providing a potential antifungal target. IMPORTANCE Antifungal resistance is increasing worldwide, especially to the most widely used azole class of ergosterol biosynthesis inhibitors, making control of fungal infections more challenging. A lot of effort has been expended in elucidating the mechanism of azole resistance and revealing potential antifungal targets. In this study, by analyzing azole-responsive genes in Neurospora crassa, we discovered STK-17, a novel kinase, that is required for azole resistance in several types of fungi. It has a role in regulating membrane homeostasis, responses to azole by ergosterol biosynthesis genes and azole accumulation, thus, deepening our understanding on the mechanism of azole stress response. Additionally, STK-17 is conserved among fungi and plays important roles in fungal development and stress resistance. Kinase inhibitors are broadly used for treating diseases, and our study pinpoints a potential drug target for antifungal development.


Assuntos
Antifúngicos/metabolismo , Azóis/metabolismo , Membrana Celular/metabolismo , Proteínas Fúngicas/metabolismo , Neurospora crassa/enzimologia , Proteínas Quinases/metabolismo , Antifúngicos/farmacologia , Azóis/farmacologia , Membrana Celular/efeitos dos fármacos , Membrana Celular/genética , Farmacorresistência Fúngica , Ergosterol/biossíntese , Proteínas Fúngicas/genética , Regulação Fúngica da Expressão Gênica , Homeostase , Testes de Sensibilidade Microbiana , Neurospora crassa/efeitos dos fármacos , Neurospora crassa/genética , Neurospora crassa/metabolismo , Proteínas Quinases/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...