Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Pharmacol Res ; 207: 107298, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39032840

RESUMEN

Acquired drug resistance is the major cause for disease recurrence in cancer patients, and this is particularly true for patients with metastatic melanoma that carry a BRAF V600E mutation. To address this problem, we investigated cyclic membrane-active peptides as an alternative therapeutic modality to kill drug-tolerant and resistant melanoma cells to avoid acquired drug resistance. We selected two stable cyclic peptides (cTI and cGm), previously shown to have anti-melanoma properties, and compared them with dabrafenib, a drug used to treat cancer patients with the BRAF V600E mutation. The peptides act via a fast membrane-permeabilizing mechanism and kill metastatic melanoma cells that are sensitive, tolerant, or resistant to dabrafenib. Melanoma cells do not become resistant to long-term treatment with cTI, nor do they evolve their lipid membrane composition, as measured by lipidomic and proteomic studies. In vivo studies in mice demonstrated that the combination treatment of cTI and dabrafenib resulted in fewer metastases and improved overall survival. Such cyclic membrane-active peptides are thus well suited as templates to design new anticancer therapeutic strategies.


Asunto(s)
Antineoplásicos , Proliferación Celular , Resistencia a Antineoplásicos , Imidazoles , Melanoma , Oximas , Péptidos Cíclicos , Péptidos Cíclicos/farmacología , Péptidos Cíclicos/uso terapéutico , Animales , Melanoma/tratamiento farmacológico , Melanoma/patología , Humanos , Resistencia a Antineoplásicos/efectos de los fármacos , Línea Celular Tumoral , Imidazoles/farmacología , Imidazoles/uso terapéutico , Proliferación Celular/efectos de los fármacos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Oximas/farmacología , Oximas/uso terapéutico , Péptidos Catiónicos Antimicrobianos/farmacología , Péptidos Catiónicos Antimicrobianos/uso terapéutico , Ratones , Femenino , Proteínas de Unión al ADN
2.
Cell Mol Life Sci ; 79(1): 38, 2021 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-34971427

RESUMEN

Bacteria that occupy an intracellular niche can evade extracellular host immune responses and antimicrobial molecules. In addition to classic intracellular pathogens, other bacteria including uropathogenic Escherichia coli (UPEC) can adopt both extracellular and intracellular lifestyles. UPEC intracellular survival and replication complicates treatment, as many therapeutic molecules do not effectively reach all components of the infection cycle. In this study, we explored cell-penetrating antimicrobial peptides from distinct structural classes as alternative molecules for targeting bacteria. We identified two ß-hairpin peptides from the horseshoe crab, tachyplesin I and polyphemusin I, with broad antimicrobial activity toward a panel of pathogenic and non-pathogenic bacteria in planktonic form. Peptide analogs [I11A]tachyplesin I and [I11S]tachyplesin I maintained activity toward bacteria, but were less toxic to mammalian cells than native tachyplesin I. This important increase in therapeutic window allowed treatment with higher concentrations of [I11A]tachyplesin I and [I11S]tachyplesin I, to significantly reduce intramacrophage survival of UPEC in an in vitro infection model. Mechanistic studies using bacterial cells, model membranes and cell membrane extracts, suggest that tachyplesin I and polyphemusin I peptides kill UPEC by selectively binding and disrupting bacterial cell membranes. Moreover, treatment of UPEC with sublethal peptide concentrations increased zinc toxicity and enhanced innate macrophage antimicrobial pathways. In summary, our combined data show that cell-penetrating peptides are attractive alternatives to traditional small molecule antibiotics for treating UPEC infection, and that optimization of native peptide sequences can deliver effective antimicrobials for targeting bacteria in extracellular and intracellular environments.


Asunto(s)
Antibacterianos/farmacología , Péptidos Catiónicos Antimicrobianos/farmacología , Péptidos Antimicrobianos/farmacología , Bacterias/efectos de los fármacos , Infecciones Bacterianas/tratamiento farmacológico , Proteínas de Unión al ADN/farmacología , Péptidos Cíclicos/farmacología , Animales , Células de la Médula Ósea , Membrana Celular/efectos de los fármacos , Células Cultivadas , Eritrocitos , Cangrejos Herradura/metabolismo , Humanos , Ratones Endogámicos C57BL , Cultivo Primario de Células
3.
J Biol Chem ; 295(32): 10911-10925, 2020 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-32414842

RESUMEN

Cyclotides are plant-derived peptides characterized by an ∼30-amino acid-long cyclic backbone and a cystine knot motif. Cyclotides have diverse bioactivities, and their cytotoxicity has attracted significant attention for its potential anticancer applications. Hybanthus enneaspermus (Linn) F. Muell is a medicinal herb widely used in India as a libido enhancer, and a previous study has reported that it may contain cyclotides. In the current study, we isolated 11 novel cyclotides and 1 known cyclotide (cycloviolacin O2) from H. enneaspermus and used tandem MS to determine their amino acid sequences. We found that among these cyclotides, hyen C comprises a unique sequence in loops 1, 2, 3, 4, and 6 compared with known cyclotides. The most abundant cyclotide in this plant, hyen D, had anticancer activity comparable to that of cycloviolacin O2, one of the most cytotoxic known cyclotides. We also provide mechanistic insights into how these novel cyclotides interact with and permeabilize cell membranes. Results from surface plasmon resonance experiments revealed that hyen D, E, L, and M and cycloviolacin O2 preferentially interact with model lipid membranes that contain phospholipids with phosphatidyl-ethanolamine headgroups. The results of a lactate dehydrogenase assay indicated that exposure to these cyclotides compromises cell membrane integrity. Using live-cell imaging, we show that hyen D induces rapid membrane blebbing and cell necrosis. Cyclotide-membrane interactions correlated with the observed cytotoxicity, suggesting that membrane permeabilization and disintegration underpin cyclotide cytotoxicity. These findings broaden our knowledge on the indigenous Indian herb H. enneaspermus and have uncovered cyclotides with potential anticancer activity.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Ciclotidas/farmacología , Descubrimiento de Drogas , Plantas Medicinales/química , Violaceae/química , Secuencia de Aminoácidos , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/aislamiento & purificación , Línea Celular Tumoral , Ciclotidas/química , Ciclotidas/aislamiento & purificación , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Proteínas de Plantas/química , Proteínas de Plantas/aislamiento & purificación , Proteínas de Plantas/farmacología , Resonancia por Plasmón de Superficie , Espectrometría de Masas en Tándem
4.
J Biol Chem ; 293(23): 9041-9052, 2018 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-29703751

RESUMEN

Gating modifier toxins (GMTs) are venom-derived peptides isolated from spiders and other venomous creatures and modulate activity of disease-relevant voltage-gated ion channels and are therefore being pursued as therapeutic leads. The amphipathic surface profile of GMTs has prompted the proposal that some GMTs simultaneously bind to the cell membrane and voltage-gated ion channels in a trimolecular complex. Here, we examined whether there is a relationship among spider GMT amphipathicity, membrane binding, and potency or selectivity for voltage-gated sodium (NaV) channels. We used NMR spectroscopy and in silico calculations to examine the structures and physicochemical properties of a panel of nine GMTs and deployed surface plasmon resonance to measure GMT affinity for lipids putatively found in proximity to NaV channels. Electrophysiology was used to quantify GMT activity on NaV1.7, an ion channel linked to chronic pain. Selectivity of the peptides was further examined against a panel of NaV channel subtypes. We show that GMTs adsorb to the outer leaflet of anionic lipid bilayers through electrostatic interactions. We did not observe a direct correlation between GMT amphipathicity and affinity for lipid bilayers. Furthermore, GMT-lipid bilayer interactions did not correlate with potency or selectivity for NaVs. We therefore propose that increased membrane binding is unlikely to improve subtype selectivity and that the conserved amphipathic GMT surface profile is an adaptation that facilitates simultaneous modulation of multiple NaVs.


Asunto(s)
Canal de Sodio Activado por Voltaje NAV1.7/metabolismo , Venenos de Araña/farmacología , Toxinas Biológicas/farmacología , Secuencia de Aminoácidos , Animales , Proteínas de Artrópodos/química , Proteínas de Artrópodos/metabolismo , Proteínas de Artrópodos/farmacología , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Células HEK293 , Humanos , Membrana Dobles de Lípidos/metabolismo , Modelos Moleculares , Venenos de Araña/química , Venenos de Araña/metabolismo , Arañas/química , Toxinas Biológicas/química , Toxinas Biológicas/metabolismo , Bloqueadores del Canal de Sodio Activado por Voltaje/química , Bloqueadores del Canal de Sodio Activado por Voltaje/metabolismo , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacología
5.
J Biol Chem ; 293(5): 1536-1549, 2018 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-29255091

RESUMEN

Crotalicidin (Ctn), a cathelicidin-related peptide from the venom of a South American rattlesnake, possesses potent antimicrobial, antitumor, and antifungal properties. Previously, we have shown that its C-terminal fragment, Ctn(15-34), retains the antimicrobial and antitumor activities but is less toxic to healthy cells and has improved serum stability. Here, we investigated the mechanisms of action of Ctn and Ctn(15-34) against Gram-negative bacteria. Both peptides were bactericidal, killing ∼90% of Escherichia coli and Pseudomonas aeruginosa cells within 90-120 and 5-30 min, respectively. Studies of ζ potential at the bacterial cell membrane suggested that both peptides accumulate at and neutralize negative charges on the bacterial surface. Flow cytometry experiments confirmed that both peptides permeabilize the bacterial cell membrane but suggested slightly different mechanisms of action. Ctn(15-34) permeabilized the membrane immediately upon addition to the cells, whereas Ctn had a lag phase before inducing membrane damage and exhibited more complex cell-killing activity, probably because of two different modes of membrane permeabilization. Using surface plasmon resonance and leakage assays with model vesicles, we confirmed that Ctn(15-34) binds to and disrupts lipid membranes and also observed that Ctn(15-34) has a preference for vesicles that mimic bacterial or tumor cell membranes. Atomic force microscopy visualized the effect of these peptides on bacterial cells, and confocal microscopy confirmed their localization on the bacterial surface. Our studies shed light onto the antimicrobial mechanisms of Ctn and Ctn(15-34), suggesting Ctn(15-34) as a promising lead for development as an antibacterial/antitumor agent.


Asunto(s)
Antibacterianos , Permeabilidad de la Membrana Celular/efectos de los fármacos , Membrana Celular , Venenos de Crotálidos , Crotalus , Escherichia coli , Fragmentos de Péptidos , Pseudomonas aeruginosa , Animales , Antibacterianos/química , Antibacterianos/farmacología , Membrana Celular/química , Membrana Celular/metabolismo , Venenos de Crotálidos/química , Venenos de Crotálidos/farmacología , Escherichia coli/química , Escherichia coli/metabolismo , Fragmentos de Péptidos/química , Fragmentos de Péptidos/farmacología , Pseudomonas aeruginosa/química , Pseudomonas aeruginosa/metabolismo , Resonancia por Plasmón de Superficie
6.
J Am Chem Soc ; 141(51): 20460-20469, 2019 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-31765148

RESUMEN

Peptides with pharmaceutical activities are attractive drug leads, and knowledge of their mode-of-action is essential for translation into the clinic. Comparison of native and enantiomeric peptides has long been used as a powerful approach to discriminate membrane- or receptor-mediated modes-of-action on the basis of the assumption that interactions with cell membranes are independent of peptide chirality. Here, we revisit this paradigm with the cyclotide kalata B1, a drug scaffold with intrinsic membrane-binding activity whose enantiomer is less potent than native peptide. To investigate this chirality dependence, we compared peptide-lipid binding using mirror image model membranes. We synthesized phospholipids with non-natural chirality and demonstrate that native kalata B1 binds with higher affinity to phospholipids with chirality found in eukaryotic membranes. This study shows for the first time that the chiral environment of lipid bilayers can modulate the function of membrane-active peptides and challenges the view that peptide-lipid interactions are achiral.


Asunto(s)
Membrana Celular/química , Ciclotidas/química , Fosfolípidos/química , Sitios de Unión , Ciclotidas/síntesis química , Ciclotidas/aislamiento & purificación , Células Eucariotas/citología , Voluntarios Sanos , Humanos , Leucocitos Mononucleares/citología , Modelos Moleculares
7.
Cell Mol Life Sci ; 75(24): 4511-4524, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30109357

RESUMEN

Sea anemone venoms have long been recognized as a rich source of peptides with interesting pharmacological and structural properties, but they still contain many uncharacterized bioactive compounds. Here we report the discovery, three-dimensional structure, activity, tissue localization, and putative function of a novel sea anemone peptide toxin that constitutes a new, sixth type of voltage-gated potassium channel (KV) toxin from sea anemones. Comprised of just 17 residues, κ-actitoxin-Ate1a (Ate1a) is the shortest sea anemone toxin reported to date, and it adopts a novel three-dimensional structure that we have named the Proline-Hinged Asymmetric ß-hairpin (PHAB) fold. Mass spectrometry imaging and bioassays suggest that Ate1a serves a primarily predatory function by immobilising prey, and we show this is achieved through inhibition of Shaker-type KV channels. Ate1a is encoded as a multi-domain precursor protein that yields multiple identical mature peptides, which likely evolved by multiple domain duplication events in an actinioidean ancestor. Despite this ancient evolutionary history, the PHAB-encoding gene family exhibits remarkable sequence conservation in the mature peptide domains. We demonstrate that this conservation is likely due to intra-gene concerted evolution, which has to our knowledge not previously been reported for toxin genes. We propose that the concerted evolution of toxin domains provides a hitherto unrecognised way to circumvent the effects of the costly evolutionary arms race considered to drive toxin gene evolution by ensuring efficient secretion of ecologically important predatory toxins.


Asunto(s)
Venenos de Cnidarios/química , Péptidos/química , Canales de Potasio con Entrada de Voltaje/química , Anémonas de Mar/química , Secuencia de Aminoácidos , Animales , Venenos de Cnidarios/genética , Venenos de Cnidarios/metabolismo , Evolución Molecular , Modelos Moleculares , Péptidos/genética , Péptidos/metabolismo , Canales de Potasio con Entrada de Voltaje/genética , Canales de Potasio con Entrada de Voltaje/metabolismo , Conformación Proteica , Pliegue de Proteína , Anémonas de Mar/genética , Anémonas de Mar/metabolismo , Transcriptoma
8.
Biophys J ; 112(4): 630-642, 2017 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-28256223

RESUMEN

Cyclotides are ultra-stable cyclic disulfide-rich peptides from plants. Their biophysical effects and medically interesting activities are related to their membrane-binding properties, with particularly high affinity for phosphatidylethanolamine lipids. In this study we were interested in understanding the molecular details of cyclotide-membrane interactions, specifically with regard to the spatial orientation of the cyclotide kalata B1 from Oldenlandia affinis when embedded in a lipid bilayer. Our experimental approach was based on the use of solid-state 19F-NMR of oriented bilayers in conjunction with the conformationally restricted amino acid L-3-(trifluoromethyl)bicyclopent-[1.1.1]-1-ylglycine as an orientation-sensitive 19F-NMR probe. Its rigid connection to the kalata B1 backbone scaffold, together with the well-defined structure of the cyclotide, allowed us to calculate the protein alignment in the membrane directly from the orientation-sensitive 19F-NMR signal. The hydrophobic and polar residues on the surface of kalata B1 form well-separated patches, endowing this cyclotide with a pronounced amphipathicity. The peptide orientation, as determined by NMR, showed that this amphipathic structure matches the polar/apolar interface of the lipid bilayer very well. A location in the amphiphilic headgroup region of the bilayer was supported by 15N-NMR of uniformly labeled protein, and confirmed using solid-state 31P- and 2H-NMR. 31P-NMR relaxation data indicated a change in lipid headgroup dynamics induced by kalata B1. Changes in the 2H-NMR order parameter profile of the acyl chains suggest membrane thinning, as typically observed for amphiphilic peptides embedded near the polar/apolar bilayer interface. Furthermore, from the 19F-NMR analysis two important charged residues, E7 and R28, were found to be positioned equatorially. The observed location thus would be favorable for the postulated binding of E7 to phosphatidylethanolamine lipid headgroups. Furthermore, it may be speculated that this pair of side chains could promote oligomerization of kalata B1 through electrostatic intermolecular contacts via their complementary charges.


Asunto(s)
Ciclotidas/química , Ciclotidas/metabolismo , Membrana Dobles de Lípidos/metabolismo , Secuencia de Aminoácidos , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Oldenlandia/metabolismo , Conformación Proteica
9.
J Biol Chem ; 291(33): 17049-65, 2016 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-27311819

RESUMEN

ProTx-II is a disulfide-rich peptide toxin from tarantula venom able to inhibit the human voltage-gated sodium channel 1.7 (hNaV1.7), a channel reported to be involved in nociception, and thus it might have potential as a pain therapeutic. ProTx-II acts by binding to the membrane-embedded voltage sensor domain of hNaV1.7, but the precise peptide channel-binding site and the importance of membrane binding on the inhibitory activity of ProTx-II remain unknown. In this study, we examined the structure and membrane-binding properties of ProTx-II and several analogues using NMR spectroscopy, surface plasmon resonance, fluorescence spectroscopy, and molecular dynamics simulations. Our results show a direct correlation between ProTx-II membrane binding affinity and its potency as an hNaV1.7 channel inhibitor. The data support a model whereby a hydrophobic patch on the ProTx-II surface anchors the molecule at the cell surface in a position that optimizes interaction of the peptide with the binding site on the voltage sensor domain. This is the first study to demonstrate that binding of ProTx-II to the lipid membrane is directly linked to its potency as an hNaV1.7 channel inhibitor.


Asunto(s)
Membrana Dobles de Lípidos/química , Simulación de Dinámica Molecular , Canal de Sodio Activado por Voltaje NAV1.7/química , Venenos de Araña/química , Sitios de Unión , Humanos , Resonancia Magnética Nuclear Biomolecular
10.
J Biol Chem ; 291(22): 11829-42, 2016 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-27026701

RESUMEN

The µO-conotoxins MrVIA, MrVIB, and MfVIA inhibit the voltage-gated sodium channel NaV1.8, a well described target for the treatment of pain; however, little is known about the residues or structural elements that define this activity. In this study, we determined the three-dimensional structure of MfVIA, examined its membrane binding properties, performed alanine-scanning mutagenesis, and identified residues important for its activity at human NaV1.8. A second round of mutations resulted in (E5K,E8K)MfVIA, a double mutant with greater positive surface charge and greater affinity for lipid membranes compared with MfVIA. This analogue had increased potency at NaV1.8 and was analgesic in the mouse formalin assay.


Asunto(s)
Analgésicos/farmacología , Membrana Celular/metabolismo , Conotoxinas/farmacología , Canal de Sodio Activado por Voltaje NAV1.8/metabolismo , Dolor/prevención & control , Secuencia de Aminoácidos , Animales , Conducta Animal/efectos de los fármacos , Cristalografía por Rayos X , Electrofisiología , Células HEK293 , Humanos , Liposomas , Espectroscopía de Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Canal de Sodio Activado por Voltaje NAV1.8/química , Canal de Sodio Activado por Voltaje NAV1.8/genética , Dolor/inducido químicamente , Conformación Proteica , Homología de Secuencia de Aminoácido
11.
Biochim Biophys Acta ; 1858(4): 872-82, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26850736

RESUMEN

Many venom peptides are potent and selective inhibitors of voltage-gated ion channels, including channels that are validated therapeutic targets for treatment of a wide range of human diseases. However, the development of novel venom-peptide-based therapeutics requires an understanding of their mechanism of action. In the case of voltage-gated ion channels, venom peptides act either as pore blockers that bind to the extracellular side of the channel pore or gating modifiers that bind to one or more of the membrane-embedded voltage sensor domains. In the case of gating modifiers, it has been debated whether the peptide must partition into the membrane to reach its binding site. In this study, we used surface plasmon resonance, fluorescence spectroscopy and molecular dynamics to directly compare the lipid-binding properties of two gating modifiers (µ-TRTX-Hd1a and ProTx-I) and two pore blockers (ShK and KIIIA). Only ProTx-I was found to bind to model membranes. Our results provide further evidence that the ability to insert into the lipid bilayer is not a requirement to be a gating modifier. In addition, we characterised the surface of ProTx-I that mediates its interaction with neutral and anionic phospholipid membranes and show that it preferentially interacts with anionic lipids.


Asunto(s)
Membranas/efectos de los fármacos , Péptidos/química , Venenos de Araña/química , Sitios de Unión/efectos de los fármacos , Humanos , Activación del Canal Iónico/efectos de los fármacos , Membranas/química , Péptidos/toxicidad , Venenos de Araña/toxicidad
12.
Biopolymers ; 108(5)2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28459137

RESUMEN

Chlorotoxin (CTX), a disulfide-rich peptide from the scorpion Leiurus quinquestriatus, has several promising biopharmaceutical properties, including preferential affinity for certain cancer cells, high serum stability, and cell penetration. These properties underpin its potential for use as a drug design scaffold, especially for the treatment of cancer; indeed, several analogs of CTX have reached clinical trials. Here, we focus on its ability to internalize into cells-a trait associated with a privileged subclass of peptides called cell-penetrating peptides-and whether it can be improved through conservative substitutions. Mutants of CTX were made using solid-phase peptide synthesis and internalization into human cervical carcinoma (HeLa) cells was monitored by fluorescence and confocal microscopy. CTX_M1 (ie, [K15R/K23R]CTX) and CTX_M2 (ie, [K15R/K23R/Y29W]CTX) mutants showed at least a twofold improvement in uptake compared to CTX. We further showed that these mutants internalize into HeLa cells largely via an energy-dependent mechanism. Importantly, the mutants have high stability, remaining intact in serum for over 24 h; thus, retaining the characteristic stability of their parent peptide. Overall, we have shown that simple conservative substitutions can enhance the cellular uptake of CTX, suggesting that such type of mutations might be useful for improving uptake of other peptide toxins.


Asunto(s)
Venenos de Escorpión/metabolismo , Secuencia de Aminoácidos , Animales , Arginina/química , Membrana Celular/química , Membrana Celular/metabolismo , Disulfuros/química , Espectroscopía de Resonancia por Spin del Electrón , Células HeLa , Humanos , Membrana Dobles de Lípidos/química , Membrana Dobles de Lípidos/metabolismo , Lisina/química , Microscopía Confocal , Mutagénesis , Estabilidad Proteica , Venenos de Escorpión/síntesis química , Venenos de Escorpión/genética , Escorpiones/metabolismo , Alineación de Secuencia
13.
Biopolymers ; 108(1)2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27487329

RESUMEN

Cyclotides are plant-derived host defense peptides displaying exceptional stability due to their cyclic cystine knot comprising three intertwined disulfide bonds and a cyclic backbone. Their six conserved cysteine residues are separated by backbone loops with diverse sequences. Prototypical cyclotides from the Möbius (kalata B1) and trypsin inhibitor (MCoTI-II) subfamilies lack sequence homology with one another, but both are able to penetrate cells, apparently via different mechanisms. To delineate the influence of the sequences of the loops on the structure and cell internalization of these two cyclotide subfamilies, a series of Möbius/trypsin inhibitor loop-chimeras of kalata B1 and MCoTI-II were synthesized, and structurally and functionally characterized. NMR analysis showed that the structural fold of the majority of chimeric peptides was minimally affected by the loop substitutions. Substituting loops 3, 5, or 6 of MCoTI-II into the corresponding loops of kalata B1 attenuated its hemolytic and cytotoxic activities, and greatly reduced its cell-penetrating properties. On the other hand, replacing loops of MCoTI-II with the corresponding loops of kalata B1 did not introduce cytotoxicity into the chimeras. Loops 2, 3, and 4 of MCoTI-II were found to contribute little to cell-penetrating properties. Overall, this study provides valuable insights into the structural basis for the hemolytic, cytotoxic, and cell-penetrating properties of kalata B1 and MCoTI-II, which could be useful for future engineering of cyclotides to carry bioactive epitopes to intracellular targets.


Asunto(s)
Ciclotidas/química , Proteínas de Plantas/química , Secuencia de Aminoácidos , Supervivencia Celular/efectos de los fármacos , Cucurbitaceae/metabolismo , Ciclotidas/síntesis química , Ciclotidas/toxicidad , Eritrocitos/citología , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Células HeLa , Hemólisis/efectos de los fármacos , Humanos , Espectroscopía de Resonancia Magnética , Estructura Terciaria de Proteína
14.
Langmuir ; 33(26): 6630-6637, 2017 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-28605904

RESUMEN

Cyclotides are cyclic disulfide-rich peptides that are chemically and thermally stable and possess pharmaceutical and insecticidal properties. The activities reported for cyclotides correlate with their ability to target phosphatidylethanolamine (PE)-phospholipids and disrupt cell membranes. However, the mechanism by which this disruption occurs remains unclear. In the current study we examine the effect of the prototypic cyclotides, kalata B1 (kB1) and kalata B2 (kB2), on tethered lipid bilayer membranes (tBLMs) using swept frequency electrical impedance spectroscopy. We confirmed that kB1 and kB2 bind to bilayers only if they contain PE-phospholipids. We hypothesize that the increase in membrane conduction and capacitance observed upon addition of kB1 or kB2 is unlikely to result from ion channel like pores but is consistent with the formation of lipidic toroidal pores. This hypothesis is supported by the concentration dependence of effects of kB1 and kB2 being suggestive of a critical micelle concentration event rather than a progressive increase in conduction arising from increased channel insertion. Additionally, conduction behavior is readily reversible when the peptide is rinsed from the bilayer. Our results support a mechanism by which kB1 and kB2 bind to and disrupt PE-containing membranes by decreasing the overall membrane critical packing parameter, as would a surfactant, which then opens or increases the size of existing membrane defects. The cyclotides need not participate directly in the conductive pore but might exert their effect indirectly through altering membrane packing constraints and inducing purely lipidic conductive pores.


Asunto(s)
Tensoactivos/química , Secuencia de Aminoácidos , Ciclotidas , Membrana Dobles de Lípidos
15.
Biochemistry ; 55(2): 396-405, 2016 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-26685975

RESUMEN

The SET protein is a promising drug target in cancer therapy, because of its ability to inhibit the function of the tumor suppressor gene protein phosphatase 2A (PP2A). COG peptides, derived from apolipoprotein E (apoE), are potent antagonists of SET; they induce cytotoxicity in cancer cells upon binding to intracellular SET and modulate the nuclear factor kappa B (NF-κB) signaling pathway. However, the therapeutic potential of COG peptides is limited, because of their poor proteolytic stability and low bioavailability. In this study, the COG peptide, COG1410, was stabilized by grafting it onto the ultrastable cyclic peptide scaffold, Momordica cochinchinensis trypsin inhibitor-II (MCoTI-II). The grafted MCoTI-II peptides were cytotoxic to a cancer cell line and showed high stability in human serum. The most potent grafted MCoTI-II peptide inhibited lipopolysaccharide (LPS)-mediated activation of NF-κB in murine macrophages. Overall, this study demonstrates the application of the MCoTI-II scaffold for the development of stable peptide drugs for cancer therapy.


Asunto(s)
Chaperonas de Histonas/antagonistas & inhibidores , Péptidos Cíclicos/química , Péptidos Cíclicos/farmacología , Factores de Transcripción/antagonistas & inhibidores , Apolipoproteínas E/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ciclotidas/química , Ciclotidas/farmacología , Proteínas de Unión al ADN , Humanos , Imagen por Resonancia Magnética , FN-kappa B/metabolismo , Regiones Promotoras Genéticas/genética , Proteína Fosfatasa 2/metabolismo
16.
J Biol Chem ; 290(32): 19853-62, 2015 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-26100634

RESUMEN

Maculatin 1.1 (Mac1) is an antimicrobial peptide from the skin of Australian tree frogs and is known to possess selectivity toward Gram-positive bacteria. Although Mac1 has membrane disrupting activity, it is not known how Mac1 selectively targets Gram-positive over Gram-negative bacteria. The interaction of Mac1 with Escherichia coli, Staphylococcus aureus, and human red blood cells (hRBC) and with their mimetic model membranes is here reported. The peptide showed a 16-fold greater growth inhibition activity against S. aureus (4 µM) than against E. coli (64 µM) and an intermediate cytotoxicity against hRBC (30 µM). Surprisingly, Sytox Green uptake monitored by flow cytometry showed that Mac1 compromised both bacterial membranes with similar efficiency at ∼20-fold lower concentration than the reported minimum inhibition concentration against S. aureus. Mac1 also reduced the negative potential of S. aureus and E. coli membrane with similar efficacy. Furthermore, liposomes mimicking the cell membrane of S. aureus (POPG/TOCL) and E. coli (POPE/POPG) were lysed at similar concentrations, whereas hRBC-like vesicles (POPC/SM/Chol) remained mostly intact in the presence of Mac1. Remarkably, when POPG/TOCL and POPE/POPG liposomes were co-incubated, Mac1 did not induce leakage from POPE/POPG liposomes, suggesting a preference toward POPG/TOCL membranes that was supported by surface plasma resonance assays. Interestingly, circular dichroism spectroscopy showed a similar helical conformation in the presence of the anionic liposomes but not the hRBC mimics. Overall, the study showed that Mac1 disrupts bacterial membranes in a similar fashion before cell death events and would preferentially target S. aureus over E. coli or hRBC membranes.


Asunto(s)
Proteínas Anfibias/farmacología , Péptidos Catiónicos Antimicrobianos/farmacología , Membrana Celular/efectos de los fármacos , Eritrocitos/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Staphylococcus aureus/efectos de los fármacos , Proteínas Anfibias/aislamiento & purificación , Animales , Péptidos Catiónicos Antimicrobianos/aislamiento & purificación , Anuros/metabolismo , Cardiolipinas/química , Membrana Celular/química , Colesterol/química , Relación Dosis-Respuesta a Droga , Eritrocitos/química , Eritrocitos/citología , Escherichia coli/química , Escherichia coli/citología , Hemólisis/efectos de los fármacos , Humanos , Liposomas/química , Fosfatidilcolinas/química , Fosfatidiletanolaminas/química , Fosfatidilgliceroles/química , Especificidad de la Especie , Esfingomielinas/química , Staphylococcus aureus/química , Staphylococcus aureus/citología
17.
J Am Chem Soc ; 138(17): 5706-13, 2016 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-27064294

RESUMEN

Enantiomeric forms of BTD-2, PG-1, and PM-1 were synthesized to delineate the structure and function of these ß-sheet antimicrobial peptides. Activity and lipid-binding assays confirm that these peptides act via a receptor-independent mechanism involving membrane interaction. The racemic crystal structure of BTD-2 solved at 1.45 Å revealed a novel oligomeric form of ß-sheet antimicrobial peptides within the unit cell: an antiparallel trimer, which we suggest might be related to its membrane-active form. The BTD-2 oligomer extends into a larger supramolecular state that spans the crystal lattice, featuring a steric-zipper motif that is common in structures of amyloid-forming peptides. The supramolecular structure of BTD-2 thus represents a new mode of fibril-like assembly not previously observed for antimicrobial peptides, providing structural evidence linking antimicrobial and amyloid peptides.


Asunto(s)
Amiloide/química , Antiinfecciosos/química , Péptidos/química , Dicroismo Circular , Cristalografía por Rayos X , Conformación Proteica , Resonancia por Plasmón de Superficie
18.
New Phytol ; 210(2): 717-30, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26668107

RESUMEN

Plants have evolved many strategies to protect themselves from attack, including peptide toxins that are ribosomally synthesized and thus adaptable directly by genetic polymorphisms. Certain toxins in Clitoria ternatea (butterfly pea) are cyclic cystine-knot peptides of c. 30 residues, called cyclotides, which have co-opted the plant's albumin-1 gene family for their production. How butterfly pea albumin-1 genes were commandeered and how these cyclotides are utilized in defence remain unclear. The role of cyclotides in host plant ecology and biotechnological applications requires exploration. We characterized the sequence diversity and expression dynamics of precursor and processing proteins implicated in butterfly pea cyclotide biosynthesis by expression profiling through RNA-sequencing (RNA-seq). Peptide-enriched extracts from various organs were tested for activity against insect-like membranes and the model nematode Caenorhabditis elegans. We found that the evolution and deployment of cyclotides involved their diversification to exhibit different chemical properties and expression between organs facing different defensive challenges. Cyclotide-enriched fractions from soil-contacting organs were effective at killing nematodes, whereas similar enriched fractions from aerial organs contained cyclotides that exhibited stronger interactions with insect-like membrane lipids. Cyclotides are employed as versatile and combinatorial mediators of defence in C. ternatea and have specialized to affect different classes of attacking organisms.


Asunto(s)
Evolución Molecular , Genes de Plantas , Péptidos Cíclicos/metabolismo , Plantas/genética , Plantas/inmunología , Secuencia de Aminoácidos , Análisis por Conglomerados , Secuencia Conservada , Regulación de la Expresión Génica de las Plantas , Especificidad de Órganos/genética , Péptidos Cíclicos/biosíntesis , Péptidos Cíclicos/química , Proteínas de Plantas/metabolismo , Dominios Proteicos , Procesamiento Proteico-Postraduccional , Suelo/química , Especificidad de la Especie , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Agua
19.
Biopolymers ; 106(6): 853-863, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27287767

RESUMEN

The transcription factor p53 has a tumor suppressor role in leading damaged cells to apoptosis. Its activity is regulated/inhibited in healthy cells by the proteins MDM2 and MDMX. Overexpression of MDM2 and/or MDMX in cancer cells inactivates p53, facilitating tumor development. A 12-mer dual inhibitor peptide (pDI) was previously reported to be able to target and inhibit MDMX:p53 and MDM2:p53 interactions with nanomolar potency in vitro. With the aim of improving its cellular inhibitory activity, we produced a series of constrained pDI analogs featuring lactam staples that stabilize the bioactive helical conformation and fused them with a cell-penetrating peptide to increase cytosol delivery. We compared pDI and its analogs on their inhibitory potency, toxicity, and ability to enter cancer cells. Overall, the results show that these analogs keep their nanomolar affinity for MDM2 and MDMX and are highly active against cancer cells. © 2016 Wiley Periodicals, Inc. Biopolymers (Pept Sci) 106: 853-863, 2016.


Asunto(s)
Antineoplásicos , Péptidos de Penetración Celular , Sistemas de Liberación de Medicamentos , Complejos Multiproteicos , Proteínas Nucleares , Proteínas Proto-Oncogénicas c-mdm2 , Proteínas Proto-Oncogénicas , Proteína p53 Supresora de Tumor , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Proteínas de Ciclo Celular , Péptidos de Penetración Celular/química , Péptidos de Penetración Celular/farmacocinética , Péptidos de Penetración Celular/farmacología , Células HeLa , Humanos , Complejos Multiproteicos/antagonistas & inhibidores , Complejos Multiproteicos/metabolismo , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/metabolismo
20.
Biochemistry ; 54(31): 4863-76, 2015 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-26174911

RESUMEN

Enterocin NKR-5-3B, one of the multiple bacteriocins produced by Enterococcus faecium NKR-5-3, is a 64-amino acid novel circular bacteriocin that displays broad-spectrum antimicrobial activity. Here we report the identification, characterization, and three-dimensional nuclear magnetic resonance solution structure determination of enterocin NKR-5-3B. Enterocin NKR-5-3B is characterized by four helical segments that enclose a compact hydrophobic core, which together with its circular backbone impart high stability and structural integrity. We also report the corresponding structural gene, enkB, that encodes an 87-amino acid precursor peptide that undergoes a yet to be described enzymatic processing that involves adjacent cleavage and ligation of Leu(24) and Trp(87) to yield the mature (circular) enterocin NKR-5-3B.


Asunto(s)
Bacteriocinas/química , Enterococcus faecium/química , Bacteriocinas/biosíntesis , Bacteriocinas/genética , Enterococcus faecium/genética , Enterococcus faecium/metabolismo , Resonancia Magnética Nuclear Biomolecular , Estructura Terciaria de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA