Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Mol Microbiol ; 2023 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-37972006

RESUMEN

Streptococcus mutans is commonly associated with dental caries and the ability to form biofilms is essential for its pathogenicity. We recently identified the Pgf glycosylation machinery of S. mutans, responsible for the post-translational modification of the surface-associated adhesins Cnm and WapA. Since the four-gene pgf operon (pgfS-pgfM1-pgfE-pgfM2) is part of the S. mutans core genome, we hypothesized that the scope of the Pgf system goes beyond Cnm and WapA glycosylation. In silico analyses and tunicamycin sensitivity assays suggested a functional overlap between the Pgf machinery and the rhamnose-glucose polysaccharide synthesis pathway. Phenotypic characterization of pgf mutants (ΔpgfS, ΔpgfE, ΔpgfM1, ΔpgfM2, and Δpgf) revealed that the Pgf system is important for biofilm formation, surface charge, membrane stability, and survival in human saliva. Moreover, deletion of the entire pgf operon (Δpgf strain) resulted in significantly impaired colonization in a rat oral colonization model. Using Cnm as a model, we showed that Cnm is heavily modified with N-acetyl hexosamines but it becomes heavily phosphorylated with the inactivation of the PgfS glycosyltransferase, suggesting a crosstalk between these two post-translational modification mechanisms. Our results revealed that the Pgf machinery contributes to multiple aspects of S. mutans pathobiology that may go beyond Cnm and WapA glycosylation.

2.
Glycobiology ; 33(3): 245-259, 2023 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-36637425

RESUMEN

Streptococcus mutans is a key pathogen associated with dental caries and is often implicated in infective endocarditis. This organism forms robust biofilms on tooth surfaces and can use collagen-binding proteins (CBPs) to efficiently colonize collagenous substrates, including dentin and heart valves. One of the best characterized CBPs of S. mutans is Cnm, which contributes to adhesion and invasion of oral epithelial and heart endothelial cells. These virulence properties were subsequently linked to post-translational modification (PTM) of the Cnm threonine-rich repeat region by the Pgf glycosylation machinery, which consists of 4 enzymes: PgfS, PgfM1, PgfE, and PgfM2. Inactivation of the S. mutans pgf genes leads to decreased collagen binding, reduced invasion of human coronary artery endothelial cells, and attenuated virulence in the Galleria mellonella invertebrate model. The present study aimed to better understand Cnm glycosylation and characterize the predicted 4-epimerase, PgfE. Using a truncated Cnm variant containing only 2 threonine-rich repeats, mass spectrometric analysis revealed extensive glycosylation with HexNAc2. Compositional analysis, complemented with lectin blotting, identified the HexNAc2 moieties as GlcNAc and GalNAc. Comparison of PgfE with the other S. mutans 4-epimerase GalE through structural modeling, nuclear magnetic resonance, and capillary electrophoresis demonstrated that GalE is a UDP-Glc-4-epimerase, while PgfE is a GlcNAc-4-epimerase. While PgfE exclusively participates in protein O-glycosylation, we found that GalE affects galactose metabolism and cell division. This study further emphasizes the importance of O-linked protein glycosylation and carbohydrate metabolism in S. mutans and identifies the PTM modifications of the key CBP, Cnm.


Asunto(s)
Adhesinas Bacterianas , Caries Dental , Humanos , Glicosilación , Adhesinas Bacterianas/genética , Streptococcus mutans/genética , Streptococcus mutans/metabolismo , Adhesión Bacteriana/fisiología , Racemasas y Epimerasas/genética , Racemasas y Epimerasas/metabolismo , Células Endoteliales/metabolismo , Proteínas Portadoras/genética , Colágeno/genética , División Celular
3.
Appl Environ Microbiol ; 87(21): e0114921, 2021 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-34406827

RESUMEN

The cnm gene, coding for the glycosylated collagen- and laminin-binding surface adhesin Cnm, is found in the genomes of approximately 20% of Streptococcus mutans clinical isolates and is associated with systemic infections and increased caries risk. Other surface-associated collagen-binding proteins of S. mutans, such as P1 and WapA, have been demonstrated to form an amyloid quaternary structure with functional implications within biofilms. In silico analysis predicted that the ß-sheet-rich N-terminal collagen-binding domain (CBD) of Cnm has a propensity for amyloid aggregation, whereas the threonine-rich C-terminal domain was predicted to be disorganized. In this study, thioflavin-T fluorescence and electron microscopy were used to show that Cnm forms amyloids in either its native glycosylated or recombinant nonglycosylated form and that the CBD of Cnm is the main amyloidogenic unit of Cnm. We then performed a series of in vitro, ex vivo, and in vivo assays to characterize the amylogenic properties of Cnm. In addition, Congo red birefringence indicated that Cnm is a major amyloidogenic protein of S. mutans biofilms. Competitive binding assays using collagen-coated microtiter plates and dental roots, a substrate rich in collagen, revealed that Cnm monomers inhibit S. mutans binding to collagenous substrates, whereas Cnm amyloid aggregates lose this property. Thus, while Cnm contributes to recognition and initial binding of S. mutans to collagen-rich surfaces, amyloid formation by Cnm might act as a negative regulatory mechanism to modulate collagen-binding activity within S. mutans biofilms and warrants further investigation. IMPORTANCE Streptococcus mutans is a keystone pathogen that promotes caries by acidifying the dental biofilm milieu. The collagen- and laminin-binding glycoprotein Cnm is a virulence factor of S. mutans. Expression of Cnm by S. mutans is hypothesized to contribute to niche expansion, allowing colonization of multiple sites in the body, including collagen-rich surfaces such as dentin and heart valves. Here, we suggest that Cnm function might be modulated by its aggregation status. As a monomer, its primary function is to promote attachment to collagenous substrates via its collagen-binding domain (CBD). However, in later stages of biofilm maturation, the same CBD of Cnm could self-assemble into amyloid fibrils, losing the ability to bind to collagen and likely becoming a component of the biofilm matrix. Our findings shed light on the role of functional amyloids in S. mutans pathobiology and ecology.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Amiloide , Proteínas Amiloidogénicas/metabolismo , Proteínas Portadoras/metabolismo , Colágeno/metabolismo , Streptococcus mutans , Amiloide/metabolismo , Streptococcus mutans/genética
4.
Plant Physiol ; 171(1): 71-81, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26960732

RESUMEN

Symbiosis receptor kinase (SYMRK) is indispensable for activation of root nodule symbiosis (RNS) at both epidermal and cortical levels and is functionally conserved in legumes. Previously, we reported SYMRK to be phosphorylated on "gatekeeper" Tyr both in vitro as well as in planta. Since gatekeeper phosphorylation was not necessary for activity, the significance remained elusive. Herein, we show that substituting gatekeeper with nonphosphorylatable residues like Phe or Ala significantly affected autophosphorylation on selected targets on activation segment/αEF and ß3-αC loop of SYMRK. In addition, the same gatekeeper mutants failed to restore proper symbiotic features in a symrk null mutant where rhizobial invasion of the epidermis and nodule organogenesis was unaffected but rhizobia remain restricted to the epidermis in infection threads migrating parallel to the longitudinal axis of the root, resulting in extensive infection patches at the nodule apex. Thus, gatekeeper phosphorylation is critical for synchronizing epidermal/cortical responses in RNS.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Plantas/metabolismo , Raíces de Plantas/metabolismo , Proteínas Quinasas/metabolismo , Nódulos de las Raíces de las Plantas/metabolismo , Simbiosis , Tirosina/metabolismo , Secuencia de Aminoácidos , Proteínas Portadoras/genética , Fabaceae/metabolismo , Regulación de la Expresión Génica de las Plantas , Genes de Plantas , Mutagénesis , Mutación , Fenotipo , Ácidos Fosfoaminos/análisis , Fosforilación , Epidermis de la Planta , Proteínas de Plantas/genética , Nodulación de la Raíz de la Planta , Raíces de Plantas/microbiología , Proteínas Quinasas/genética , Rhizobium/fisiología , Nódulos de las Raíces de las Plantas/enzimología , Nódulos de las Raíces de las Plantas/genética
5.
Mol Pharm ; 13(10): 3578-3589, 2016 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-27589087

RESUMEN

The continuous rise of antimicrobial resistance and the dearth of new antibiotics in the clinical pipeline raise an urgent call for the development of potent antimicrobial agents. Cationic chitosan derivatives, N-(2-hydroxypropyl)-3-trimethylammonium chitosan chlorides (HTCC), have been widely studied as potent antibacterial agents. However, their systemic structure-activity relationship, activity toward drug-resistant bacteria and fungi, and mode of action are very rare. Moreover, toxicity and efficacy of these polymers under in vivo conditions are yet to be established. Herein, we investigated antibacterial and antifungal efficacies of the HTCC polymers against multidrug resistant bacteria including clinical isolates and pathogenic fungi, studied their mechanism of action, and evaluated cytotoxic and antimicrobial activities in vitro and in vivo. The polymers were found to be active against both bacteria and fungi (MIC = 125-250 µg/mL) and displayed rapid microbicidal kinetics, killing pathogens within 60-120 min. Moreover, the polymers were shown to target both bacterial and fungal cell membrane leading to membrane disruption and found to be effective in hindering bacterial resistance development. Importantly, very low toxicity toward human erythrocytes (HC50 = >10000 µg/mL) and embryo kidney cells were observed for the cationic polymers in vitro. Further, no inflammation toward skin tissue was observed in vivo for the most active polymer even at 200 mg/kg when applied on the mice skin. In a murine model of superficial skin infection, the polymer showed significant reduction of methicillin-resistant Staphylococcus aureus (MRSA) burden (3.2 log MRSA reduction at 100 mg/kg) with no to minimal inflammation. Taken together, these selectively active polymers show promise to be used as potent antimicrobial agents in topical and other infections.


Asunto(s)
Antiinfecciosos/uso terapéutico , Quitosano/análogos & derivados , Compuestos de Amonio Cuaternario/química , Compuestos de Amonio Cuaternario/uso terapéutico , Animales , Antiinfecciosos/administración & dosificación , Antiinfecciosos/efectos adversos , Supervivencia Celular/efectos de los fármacos , Quitosano/efectos adversos , Quitosano/química , Quitosano/uso terapéutico , Farmacorresistencia Fúngica , Farmacorresistencia Bacteriana Múltiple , Femenino , Humanos , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Ratones , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana , Compuestos de Amonio Cuaternario/efectos adversos , Infecciones Cutáneas Estafilocócicas/tratamiento farmacológico , Relación Estructura-Actividad
6.
Biomacromolecules ; 17(9): 3094-102, 2016 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-27442617

RESUMEN

Cationic-amphiphilic antibacterial polymers with optimal amphiphilicity generally target the bacterial membranes instead of mammalian membranes. To date, this balance has been achieved by varying the cationic charge or side chain hydrophobicity in a variety of cationic-amphiphilic polymers. Optimal hydrophobicity of cationic-amphiphilic polymers has been considered as the governing factor for potent antibacterial activity yet minimal mammalian cell toxicity. However, the concomitant role of hydrogen bonding and hydrophobicity with constant cationic charge in the interactions of antibacterial polymers with bacterial membranes is not understood. Also, degradable polymers that result in nontoxic degradation byproducts offer promise as safe antibacterial agents. Here we show that amide- and ester (degradable)-bearing cationic-amphiphilic polymers with tunable side chain hydrophobicity can modulate antibacterial activity and cytotoxicity. Our results suggest that an amide polymer can be a potent antibacterial agent with lower hydrophobicity whereas the corresponding ester polymer needs a relatively higher hydrophobicity to be as effective as its amide counterpart. Our studies reveal that at higher hydrophobicities both amide and ester polymers have similar profiles of membrane-active antibacterial activity and mammalian cell toxicity. On the contrary, at lower hydrophobicities, amide and ester polymers are less cytotoxic, but the former have potent antibacterial and membrane activity compared to the latter. Incorporation of amide and ester moieties made these polymers side chain degradable, with amide polymers being more stable than the ester polymers. Further, the polymers are less toxic, and their degradation byproducts are nontoxic to mice. More importantly, the optimized amide polymer reduces the bacterial burden of burn wound infections in mice models. Our design introduces a new strategy of interplay between the hydrophobic and hydrogen bonding interactions keeping constant cationic charge density for developing potent membrane-active antibacterial polymers with minimal toxicity to mammalian cells.


Asunto(s)
Acinetobacter baumannii/efectos de los fármacos , Antibacterianos/farmacología , Cationes/química , Polímeros/química , Polímeros/farmacología , Animales , Antibacterianos/química , Supervivencia Celular/efectos de los fármacos , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Ratones , Células Tumorales Cultivadas
7.
Angew Chem Int Ed Engl ; 55(27): 7836-40, 2016 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-27010764

RESUMEN

Vancomycin, the drug of last resort for Gram-positive bacterial infections, has also been rendered ineffective by the emergence of resistance in such bacteria. To combat the threat of vancomycin-resistant bacteria (VRB), we report the development of a dipicolyl-vancomycin conjugate (Dipi-van), which leads to enhanced inhibition of cell-wall biosynthesis in VRB and displays in vitro activity that is more than two orders of magnitude higher than that of vancomycin. Conjugation of the dipicolyl moiety, which is a zinc-binding ligand, endowed the parent drug with the ability to bind to pyrophosphate groups of cell-wall lipids while maintaining the inherent binding affinity for pentapeptide termini of cell-wall precursors. Furthermore, no detectable resistance was observed after several serial passages, and the compound reduced the bacterial burden by a factor of 5 logs at 12 mg kg(-1) in a murine model of VRB kidney infection. The findings presented in this report stress the potential of our strategy to combat VRB infections.


Asunto(s)
Antibacterianos/química , Difosfatos/química , Vancomicina/análogos & derivados , Animales , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Diaminas/química , Eritrocitos/citología , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Infecciones por Bacterias Grampositivas/tratamiento farmacológico , Infecciones por Bacterias Grampositivas/microbiología , Infecciones por Bacterias Grampositivas/veterinaria , Humanos , Ratones , Pruebas de Sensibilidad Microbiana , Vancomicina/farmacología , Vancomicina/uso terapéutico , Resistencia a la Vancomicina/efectos de los fármacos , Zinc/química
8.
Bioconjug Chem ; 26(12): 2442-53, 2015 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-26452096

RESUMEN

The emergence of bacterial resistance and biofilm associated infections has created a challenging situation in global health. In this present state of affairs where conventional antibiotics are falling short of being able to provide a solution to these problems, development of novel antibacterial compounds possessing the twin prowess of antibacterial and antibiofilm efficacy is imperative. Herein, we report a library of amino acid tunable lipidated norspermidine conjugates that were prepared by conjugating both amino acids and fatty acids with the amine functionalities of norspermidine through amide bond formation. These lipidated conjugates displayed potent antibacterial activity against various planktonic Gram-positive and Gram-negative bacteria including drug-resistant superbugs such as methicillin-resistant Staphylococcus aureus, vancomycin-resistant Enterococcus faecium, and ß-lactam-resistant Klebsiella pneumoniae. This class of nontoxic and fast-acting antibacterial molecules (capable of killing bacteria within 15 min) did not allow bacteria to develop resistance against them after several passages. Most importantly, an optimized compound in the series was also capable of killing metabolically inactive persisters and stationary phase bacteria. Additionally, this compound was capable of disrupting the preformed biofilms of S. aureus and E. coli. Therefore, this class of antibacterial conjugates have potential in tackling the challenging situation posed by both bacterial resistance as well as drug tolerance due to biofilm formation.


Asunto(s)
Antibacterianos/química , Antibacterianos/farmacología , Bacterias/efectos de los fármacos , Biopelículas/efectos de los fármacos , Espermidina/análogos & derivados , Aminoácidos/química , Aminoácidos/farmacología , Bacterias/crecimiento & desarrollo , Infecciones Bacterianas/tratamiento farmacológico , Biopelículas/crecimiento & desarrollo , Hemólisis/efectos de los fármacos , Humanos , Espermidina/química , Espermidina/farmacología
9.
Angew Chem Int Ed Engl ; 54(46): 13644-9, 2015 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-26473305

RESUMEN

Resistance to glycopeptide antibiotics, the drugs of choice for life-threatening bacterial infections, is on the rise. In order to counter the threat of glycopeptide-resistant bacteria, we report development of a new class of semi-synthetic glycopeptide antibiotics, which not only target the bacterial membrane but also display enhanced inhibition of cell-wall biosynthesis through increased binding affinity to their target peptides. The combined effect of these two mechanisms resulted in improved in vitro activity of two to three orders of magnitude over vancomycin and no propensity to trigger drug resistance in bacteria. In murine model of kidney infection, the optimized compound was able to bring bacterial burden down by about 6 logs at 12 mg kg(-1) with no observed toxicity. The results furnished in this report emphasize the potential of this class of compounds as future antibiotics for drug-resistant Gram-positive infections.


Asunto(s)
Antibacterianos/farmacología , Bacterias/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Pared Celular/efectos de los fármacos , Pared Celular/metabolismo , Glicopéptidos/farmacología , Resistencia a la Vancomicina/efectos de los fármacos , Animales , Antibacterianos/síntesis química , Antibacterianos/química , Bacterias/citología , Membrana Celular/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Glicopéptidos/síntesis química , Glicopéptidos/química , Ratones , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Relación Estructura-Actividad , Vancomicina/farmacología
10.
ACS Chem Biol ; 15(4): 884-889, 2020 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-32195571

RESUMEN

Vancomycin is a standard drug for the treatment of multidrug-resistant Gram-positive bacterial infections. Albeit, development of resistance (VRE, VRSA) and its inefficacy against persistent infections is a demerit. It is also intrinsically inactive against Gram-negative bacteria. Herein, we report a vancomycin derivative, VanQAmC10, that addresses these challenges. VanQAmC10 was rapidly bactericidal against carbapenem-resistant A. baumannii (6 log10 CFU/mL reduction in 6 h), disrupted A. baumannii biofilms, and eradicated their stationary phase cells. In MRSA infected macrophages, the compound reduced the bacterial burden by 1.3 log10 CFU/mL while vancomycin exhibited a static effect. Further investigation indicated that the compound, unlike vancomycin, promoted the intracellular degradative mechanism, autophagy, in mammalian cells, which may have contributed to its intracellular activity. The findings of the work provide new perspectives on the field of glycopeptide antibiotics.


Asunto(s)
Acinetobacter baumannii/efectos de los fármacos , Antibacterianos/farmacología , Autofagia/efectos de los fármacos , Vancomicina/análogos & derivados , Vancomicina/farmacología , Acinetobacter baumannii/fisiología , Animales , Antibacterianos/síntesis química , Antibacterianos/toxicidad , Biopelículas/efectos de los fármacos , Femenino , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana , Vancomicina/toxicidad , Resistencia betalactámica/efectos de los fármacos
11.
ACS Appl Mater Interfaces ; 11(37): 33559-33572, 2019 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-31424189

RESUMEN

An optimum hydrophilic/hydrophobic balance has been recognized as a crucial parameter in designing cationic polymers that mimic antimicrobial peptides (AMPs). To date, this balance was achieved either by hydrophilicity variation through altering the nature and the number of cationic charges or by hydrophobicity modulation through incorporation of alkyl groups of different chain lengths. However, how the hydrophobicity variation through AMPs' building blocks-amino acids-influences the antibacterial efficacy of AMP-mimicking cationic polymers has rarely been explored. Toward this goal, herein we report a class of amino acid conjugated polymers (ACPs) with tunable antibacterial activity through a simple post-polymer-functionalization strategy. Our polymeric design comprised a permanent cationic charge in every repeating unit, whereby the hydrophobicity was tuned through incorporation of different amino acids. Our results revealed that the amino acid alteration has a strong influence on antibacterial efficacy. Upon increasing the amino acid side-chain hydrophobicity, both the antibacterial activity (against broad spectrum of bacteria) and toxicity increased. However, the distinct feature of this class of polymers was their good activity against Acinetobacter baumannii-the top most critical pathogen according to WHO, which has created an alarming situation worldwide, causing the majority of infections in humans. A nontoxic (no hemolysis even at 1000 µg/mL) ACP including a glycine residue (ACP-1 (Gly)) showed very good activity (MIC = 8-16 µg/mL) against both drug-sensitive and drug-resistant strains of A. baumannii, including clinical isolates. This polymer not only was rapidly bactericidal against growing planktonic A. baumannii but also killed nondividing stationary-phase cells instantaneously (<2 min). Moreover, it eradicated the established biofilm formed by drug-resistant A. baumannii clinical isolates. No propensity for bacterial resistance development against this polymer was seen even after 14 continuous passages. Taken together, the results highlight that hydrophobicity modulation through incorporation of amino acids in cationic polymers will provide a significant opportunity in designing new ACPs with potent antibacterial activity and minimum toxicity toward mammalian cells. More importantly, the excellent anti-A. baumannii efficacy of the optimized lead polymer indicates its immense potential for being developed as therapeutic agent.


Asunto(s)
Acinetobacter baumannii/fisiología , Antibacterianos , Biopelículas/efectos de los fármacos , Materiales Biomiméticos , Farmacorresistencia Bacteriana/efectos de los fármacos , Polímeros , Aminoácidos/química , Aminoácidos/farmacología , Antibacterianos/química , Antibacterianos/farmacología , Biopelículas/crecimiento & desarrollo , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacología , Células HEK293 , Humanos , Polímeros/química , Polímeros/farmacología
12.
J Med Chem ; 62(7): 3184-3205, 2019 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-30404451

RESUMEN

Vancomycin, a natural glycopeptide antibiotic, was used as the antibiotic of last resort for the treatment of multidrug-resistant Gram-positive bacterial infections. However, almost 30 years after its use, resistance to vancomycin was first reported in 1986 in France. This became a major health concern, and alternative treatment strategies were urgently needed. New classes of molecules, including semisynthetic antibacterial compounds and newer generations of the previously used antibiotics, were developed. Semisynthetic derivatives of vancomycin with enhanced binding affinity, membrane disruption ability, and lipid binding properties have exhibited promising results against both Gram-positive and Gram-negative bacteria. Various successful approaches developed to overcome the acquired resistance in Gram-positive bacteria, intrinsic resistance in Gram-negative bacteria, and other forms of noninherited resistance to vancomycin have been discussed in this Perspective.


Asunto(s)
Antibacterianos/farmacología , Farmacorresistencia Bacteriana/efectos de los fármacos , Antibacterianos/química , Antibacterianos/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Pared Celular/efectos de los fármacos , Pared Celular/metabolismo , Glicopéptidos/química , Glicopéptidos/metabolismo , Glicopéptidos/farmacología , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Gramnegativas/genética , Bacterias Grampositivas/efectos de los fármacos , Bacterias Grampositivas/genética , Familia de Multigenes , Vancomicina/farmacología
13.
Artículo en Inglés | MEDLINE | ID: mdl-29993814

RESUMEN

Massively parallel sequencing technique, introduced by NGS technology, has resulted in an exponential growth of sequencing data, with greatly reduced cost and increased throughput. This huge explosion of data has introduced new challenges in regard to its storage, integration, processing and analyses. In this paper, we have proposed a novel distributed model under Map-Reduce paradigm to address the NGS big data problem. The architecture of the model involves Map-Reduce based modularized approach involving 3 different phases that support various analytical pipelines. The first phase will generate detailed base level information of various individual genomes, by granulating the alignment data. The other 2 phases independently process this base level information in parallel. One of these 2 phases will provide an integrated DNA profile of multiple individuals, whereas the other phase will generate contigs with similar features in an individual. Each of these 2 phases will generate a repository of genomic information that will facilitate other analytical pipelines. A simulated and real experimental prototypes has been provided as results to show the effectiveness of the model and its superiority over a few existing popular models and tools. A detailed description of the scope of applications of this model is also included in this article.

14.
Chem Commun (Camb) ; 54(39): 4943-4946, 2018 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-29700513

RESUMEN

Here we report the design of membrane-active peptidomimetic molecules with a tunable arrangement of hydrophobic and polar groups. In spite of having the same chemical composition, the effective hydrophobicities of the compounds were different as a consequence of their chemical structure and conformational properties. The compound with lower effective hydrophobicity demonstrated antibacterial activity that was highly selective towards bacteria over mammalian cells. This study, highlighting the role in membrane selectivity of the specific arrangement of the different moieties in the molecular structure, provides useful indications for developing non-toxic antibacterial agents.


Asunto(s)
Antibacterianos/farmacología , Peptidomiméticos/farmacología , Tensoactivos/farmacología , Animales , Antibacterianos/química , Antibacterianos/toxicidad , Línea Celular Transformada , Escherichia coli/efectos de los fármacos , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Liposomas/química , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Ratones , Pruebas de Sensibilidad Microbiana , Conformación Molecular , Simulación de Dinámica Molecular , Estructura Molecular , Peptidomiméticos/química , Peptidomiméticos/toxicidad , Pseudomonas aeruginosa/efectos de los fármacos , Tensoactivos/química , Tensoactivos/toxicidad
15.
ACS Infect Dis ; 4(7): 1093-1101, 2018 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-29726673

RESUMEN

New Delhi metallo-ß-lactamase-1 (NDM-1) is the major contributor to the emergence of carbapenem resistance in Gram-negative pathogens (GNPs) and has caused many clinically available ß-lactam antibiotics to become obsolete. A clinically approved inhibitor of metallo-ß-lactamase (MBL) that could restore the activity of carbapenems against resistant GNPs has not yet been found, making NDM-1 a serious threat to human health. Here, we have rationally developed an inhibitor for the NDM-1 enzyme, which has the ability to penetrate the outer membrane of GNPs and inactivate the enzyme by depleting the metal ion (Zn2+) from the active site. The inhibitor reinstated the activity of meropenem against NDM-1 producing clinical isolates of GNPs like Klebsiella pneumoniae and Escherichia coli. Further, the inhibitor efficiently restored meropenem activity against NDM-1 producing K. pneumoniae in a murine sepsis infection model. These findings demonstrate that a combination of the present inhibitor and meropenem has high potential to be translated clinically to combat carbapenem-resistant GNPs.


Asunto(s)
Antibacterianos/farmacología , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Gramnegativas/metabolismo , Meropenem/farmacología , Vancomicina/farmacología , beta-Lactamasas/metabolismo , Animales , Permeabilidad de la Membrana Celular , Sinergismo Farmacológico , Activación Enzimática/efectos de los fármacos , Femenino , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Bacterias Gramnegativas/genética , Infecciones por Klebsiella/tratamiento farmacológico , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/genética , Klebsiella pneumoniae/metabolismo , Ratones , Estructura Molecular , Vancomicina/análogos & derivados , Vancomicina/síntesis química , Resistencia betalactámica , beta-Lactamasas/genética
16.
Chem Commun (Camb) ; 53(60): 8427-8430, 2017 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-28702588

RESUMEN

l-Lysines were conjugated to lipidated biphenyls using simple synthetic chemistry to obtain selective membrane-active antibacterial agents that inhibit cell-wall biosynthesis. The most selective compound bore promising activity against biofilm-related infections and intracellular bacteria, and also suppressed the stimulation of TNF-α induced by lipoteichoic acid. Belligerent to resistance development, it was active in a murine model of MRSA infection.


Asunto(s)
Antibacterianos/farmacología , Antiinflamatorios no Esteroideos/farmacología , Biopelículas/efectos de los fármacos , Compuestos de Bifenilo/farmacología , Lípidos/farmacología , Lisina/farmacología , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Antibacterianos/síntesis química , Antibacterianos/química , Antiinflamatorios no Esteroideos/síntesis química , Antiinflamatorios no Esteroideos/química , Compuestos de Bifenilo/química , Relación Dosis-Respuesta a Droga , Lípidos/química , Lisina/química , Estructura Molecular
17.
PLoS One ; 12(8): e0183263, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28837596

RESUMEN

Chronic bacterial biofilms place a massive burden on healthcare due to the presence of antibiotic-tolerant dormant bacteria. Some of the conventional antibiotics such as erythromycin, vancomycin, linezolid, rifampicin etc. are inherently ineffective against Gram-negative bacteria, particularly in their biofilms. Here, we report membrane-active macromolecules that kill slow dividing stationary-phase and antibiotic tolerant cells of Gram-negative bacteria. More importantly, these molecules potentiate antibiotics (erythromycin and rifampicin) to biofilms of Gram-negative bacteria. These molecules eliminate planktonic bacteria that are liberated after dispersion of biofilms (dispersed cells). The membrane-active mechanism of these molecules forms the key for potentiating the established antibiotics. Further, we demonstrate that the combination of macromolecules and antibiotics significantly reduces bacterial burden in mouse burn and surgical wound infection models caused by Acinetobacter baumannii and Carbapenemase producing Klebsiella pneumoniae (KPC) clinical isolate respectively. Colistin, a well-known antibiotic targeting the lipopolysaccharide (LPS) of Gram-negative bacteria fails to kill antibiotic tolerant cells and dispersed cells (from biofilms) and bacteria develop resistance to it. On the contrary, these macromolecules prevent or delay the development of bacterial resistance to known antibiotics. Our findings emphasize the potential of targeting the bacterial membrane in antibiotic potentiation for disruption of biofilms and suggest a promising strategy towards developing therapies for topical treatment of Gram-negative infections.


Asunto(s)
Antibacterianos/farmacología , Bacterias Gramnegativas/efectos de los fármacos , Animales , Biopelículas , Recuento de Colonia Microbiana , Sinergismo Farmacológico , Bacterias Gramnegativas/aislamiento & purificación , Ratones , Pruebas de Sensibilidad Microbiana
18.
J Glob Antimicrob Resist ; 5: 71-4, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27436471

RESUMEN

Staphylococcus aureus is a facultative intracellular pathogen and there are limited options for the treatment of severe intracellular bacterial infections. The membrane-active glycopeptide antibiotic Van-QC8 is a permanent positively charged lipophilic vancomycin analogue that demonstrates high activity against clinically relevant drug-resistant Gram-positive bacteria both in vitro and in vivo. In this study, the intracellular activity of Van-QC8 was evaluated against meticillin-resistant S. aureus (MRSA) infection in RAW macrophages. Furthermore, the mechanism of intracellular uptake of Van-QC8 was investigated. Van-QC8 showed time- and concentration-dependent bactericidal activity against intracellular MRSA. Van-QC8 displayed significantly higher intracellular activity compared with vancomycin and linezolid. Cellular uptake of Van-QC8 was found to be through clathrin-dependent and -independent and caveolin-dependent and -independent endocytic pathways. The findings of this study suggest that Van-QC8 could be translated clinically for the treatment of intracellular infections due to MRSA.


Asunto(s)
Antibacterianos/farmacología , Glicopéptidos/farmacología , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Vancomicina/análogos & derivados , Animales , Ratones , Células RAW 264.7
19.
J Med Chem ; 59(23): 10750-10762, 2016 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-27809517

RESUMEN

More than 80% of the bacterial infections are associated with biofilm formation. To combat infections, amphiphilic small molecules have been developed as promising antibiofilm agents. However, cytotoxicity of such molecules still remains a major problem. Herein we demonstrate a concept in which antibacterial versus cytotoxic activities of cationic small molecules are tuned by spatial positioning of hydrophobic moieties while keeping positive charges constant. Compared to the molecules with more pendent hydrophobicity from positive centers (MIC = 1-4 µg/mL and HC50 = 60-65 µg/mL), molecules with more confined hydrophobicity between two centers show similar antibacterial activity but significantly less toxicity toward human erythrocytes (MIC = 1-4 µg/mL and HC50 = 805-1242 µg/mL). Notably, the optimized molecule is shown to be nontoxic toward human cells (HEK 293) at a concentration at which it eradicates established bacterial biofilms. The molecule is also shown to eradicate preformed bacterial biofilm in vivo in a murine model of superficial skin infection.


Asunto(s)
Antibacterianos/farmacología , Bacterias/efectos de los fármacos , Biopelículas/efectos de los fármacos , Enfermedades Cutáneas Bacterianas/tratamiento farmacológico , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Antibacterianos/síntesis química , Antibacterianos/química , Cationes/síntesis química , Cationes/química , Cationes/farmacología , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Enfermedades Cutáneas Bacterianas/microbiología , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad
20.
ChemMedChem ; 11(21): 2367-2371, 2016 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-27731959

RESUMEN

In the global effort to thwart antimicrobial resistance, lipopeptides are an important class of antimicrobial agents, especially against Gram-negative infections. In an attempt to circumvent their synthetic complexities, we designed simple membrane-active agents involving only one amino acid and two lipid tails. Herein we show that the use of two short lipid tails instead of a single long one significantly increases selective antibacterial activity. This study yielded several selective antibacterial compounds, and investigations into the properties of this compound class were conducted with the most active compound. Fluorescence spectroscopic studies revealed the capacity of the representative compound to cause depolarization and permeabilization of bacterial cell membranes. This membrane-active nature of the compound imparts superior activity against persister cells, biofilms, and planktonic cells. Topical application of the compound decreased bacterial burden in mice inflicted with burn-infections caused by Acinetobacter baumannii. We anticipate that the design principles described herein will direct the development of several antimicrobial agents of clinical importance.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA