Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros











Intervalo de año de publicación
1.
FASEB J ; 35(12): e22026, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34818435

RESUMEN

Antibiotic resistance is becoming a severe obstacle in the fight against acute and chronic infectious diseases that accompany most degenerative illnesses from neoplasia to osteo-arthritis and obesity. Currently, the race is on to identify pharmaceutical molecules or combinations of molecules able to prevent or reduce the insurgence and/or progression of infectivity. Attempts to substitute antibiotics with antimicrobial peptides have, thus far, met with little success against multidrug-resistant (MDR) bacterial strains. During the last decade, we designed and studied the activity and features of human ß-defensin analogs, which are salt-resistant, and hence active also under high salt concentrations as, for instance, in cystic fibrosis. Herein, we describe the design, synthesis, and major features of a new 21 aa long molecule, peptide γ2. The latter derives from the γ-core of the ß-defensin natural molecules, a small fragment of these molecules still bearing high antibacterial activity. We found that peptide γ2, which contains only one disulphide bond, recapitulates most of the biological properties of natural human ß-defensins and can also counteract both Gram-positive and Gram-negative MDR bacterial strains and biofilm formation. Moreover, it has great stability in human serum thereby enhancing its antibacterial presence and activity without cytotoxicity in human cells. In conclusion, peptide γ2 is a promising new weapon also in the battle against intractable infectious diseases.


Asunto(s)
Antibacterianos/farmacología , Péptidos Antimicrobianos/farmacología , Bacterias/crecimiento & desarrollo , Biopelículas/crecimiento & desarrollo , beta-Defensinas/química , Bacterias/efectos de los fármacos , Biopelículas/efectos de los fármacos , Farmacorresistencia Bacteriana Múltiple , Humanos , Pruebas de Sensibilidad Microbiana
2.
Antiviral Res ; 171: 104592, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31473342

RESUMEN

Filoviridae currently includes five official and one proposed genera. Genus Ebolavirus includes five established and one proposed ebolavirus species for Bombali virus (BOMV), Bundibugyo virus (BDBV), Ebola virus (EBOV), Reston virus (RESTV), Sudan virus (SUDV) and Taï Forest virus (TAFV), and genus Marburgvirus includes a single species for Marburg virus (MARV) and Ravn virus (RAVV). Ebola virus (EBOV) has emerged as a significant public health concern since the 2013-2016 Ebola Virus Disease outbreak in Western Africa. Currently, there are no therapeutics approved and the need for Ebola-specific therapeutics remains a gap. In search for anti-Ebola therapies we tested the idea of using inhibitory properties of peptides corresponding to the C-terminal heptad-repeat (HR2) domains of class I fusion proteins against EBOV infection. The fusion protein GP2 of EBOV belongs to class I, suggesting that a similar strategy to HIV may be applied to inhibit EBOV infection. The serum half-life of peptides was expanded by cholesterol conjugation to allow daily dosing. The peptides were further constrained to stabilize a helical structure to increase the potency of inhibition. The EC50s of lead peptides were in low micromolar range, as determined by a high-content imaging test of EBOV-infected cells. Lead peptides were tested in an EBOV lethal mouse model and efficacy of the peptides were determined following twice-daily administration of peptides for 9 days. The most potent peptide was able to protect mice from lethal challenge of mouse-adapted Ebola virus. These data show that engineered peptides coupled with cholesterol can inhibit viral production, protect mice against lethal EBOV infection, and may be used to build novel therapeutics against EBOV.


Asunto(s)
Antivirales/farmacología , Ebolavirus/efectos de los fármacos , Marburgvirus/efectos de los fármacos , Péptidos/farmacología , Secuencia de Aminoácidos , Animales , Antivirales/química , Línea Celular , Colesterol/química , Modelos Animales de Enfermedad , Fiebre Hemorrágica Ebola/virología , Enfermedad del Virus de Marburg/virología , Ratones , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Péptidos/química , Conformación Proteica , Relación Estructura-Actividad
3.
Sci Rep ; 9(1): 12169, 2019 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-31434916

RESUMEN

The interaction between the short 88Ser-Arg-Ser-Arg-Tyr92 sequence of the urokinase receptor (uPAR) and the formyl peptide receptor type 1 (FPR1) elicits cell migration. We generated the Ac-(D)-Tyr-(D)-Arg-Aib-(D)-Arg-NH2 (RI-3) peptide which inhibits the uPAR/FPR1 interaction, reducing migration of FPR1 expressing cells toward N-formyl-methionyl-leucyl-phenylalanine (fMLF) and Ser-Arg-Ser-Arg-Tyr (SRSRY) peptides. To understand the structural basis of the RI-3 inhibitory effects, the FPR1/fMLF, FPR1/SRSRY and FPR1/RI-3 complexes were modeled and analyzed, focusing on the binding pocket of FPR1 and the interaction between the amino acids that signal to the FPR1 C-terminal loop. We found that RI-3 shares the same binding site of fMLF and SRSRY on FPR1. However, while fMLF and SRSRY display the same agonist activation signature (i.e. the series of contacts that transmit the conformational transition throughout the complex), translating binding into signaling, RI-3 does not interact with the activation region of FPR1 and hence does not activate signaling. Indeed, fluorescein-conjugated RI-3 prevents either fMLF and SRSRY uptake on FPR1 without triggering FPR1 internalization and cell motility in the absence of any stimulus. Collectively, our data show that RI-3 is a true FPR1 antagonist and suggest a pharmacophore model useful for development of compounds that selectively inhibit the uPAR-triggered, FPR1-mediated cell migration.


Asunto(s)
Péptidos/metabolismo , Receptores de Formil Péptido/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/química , Secuencia de Aminoácidos , Animales , Sitios de Unión , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Células HEK293 , Humanos , Simulación de Dinámica Molecular , Péptidos/química , Péptidos/farmacología , Unión Proteica , Mapas de Interacción de Proteínas , Estructura Terciaria de Proteína , Ratas , Receptores de Formil Péptido/química , Receptores de Formil Péptido/genética , Relación Estructura-Actividad
4.
Sci Rep ; 8(1): 585, 2018 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-29330364

RESUMEN

A promising emerging area for the treatment of obesity and diabetes is combinatorial hormone therapy, where single-molecule peptides are rationally designed to integrate the complementary actions of multiple endogenous metabolically-related hormones. We describe here a proof-of-concept study on developing unimolecular polypharmacy agents through the use of selection methods based on phage-displayed peptide libraries (PDL). Co-agonists of the glucagon (GCG) and GLP-1 receptors were identified from a PDL sequentially selected on GCGR- and GLP1R-overexpressing cells. After two or three rounds of selection, 7.5% of randomly picked clones were GLP1R/GCGR co-agonists, and a further 1.53% were agonists of a single receptor. The phages were sequenced and 35 corresponding peptides were synthesized. 18 peptides were potent co-agonists, 8 of whom showed EC50 ≤ 30 pM on each receptor, comparable to the best rationally designed co-agonists reported in the literature. Based on literature examples, two sequences were engineered to stabilize against dipeptidyl peptidase IV cleavage and prolong the in vivo half-life: the engineered peptides were comparably potent to the parent peptides on both receptors, highlighting the potential use of phage-derived peptides as therapeutic agents. The strategy described here appears of general value for the discovery of optimized polypharmacology paradigms across several metabolically-related hormones.


Asunto(s)
Receptor del Péptido 1 Similar al Glucagón/agonistas , Péptidos/síntesis química , Péptidos/farmacología , Receptores de Glucagón/agonistas , Diabetes Mellitus/tratamiento farmacológico , Dipeptidil Peptidasa 4/metabolismo , Humanos , Obesidad/tratamiento farmacológico , Biblioteca de Péptidos , Péptidos/genética , Polifarmacia , Análisis de Secuencia de ADN
5.
J Exp Clin Cancer Res ; 36(1): 180, 2017 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-29216889

RESUMEN

BACKGROUND: Accumulating evidence demonstrates that the Urokinase Receptor (uPAR) regulates tumor cell migration through its assembly in composite regulatory units with transmembrane receptors, and uPAR88-92 is the minimal sequence required to induce cell motility through the Formyl Peptide Receptor type 1 (FPR1). Both uPAR and FPR1 are involved in melanoma tumor progression, suggesting that they may be targeted for therapeutic purposes. In this study, the role of the uPAR-FPR1 cross-talk to sustain melanoma cell ability to invade extracellular matrix and cross endothelial barriers is investigated. Also, the possibility that inhibition of the uPAR mediated FPR1-dependent signaling may prevent matrix invasion and transendothelial migration of melanoma cells was investigated. METHODS: Expression levels of uPAR and FPR1 were assessed by immunocytochemistry, Western Blot and qRT-PCR. Cell migration was investigated by Boyden chamber and wound-healing assays. Migration and invasion kinetics, trans-endothelial migration and proliferation of melanoma cells were monitored in real time using the xCELLigence technology. The agonist-triggered FPR1 internalization was visualized by confocal microscope. Cell adhesion to endothelium was determined by fluorometer measurement of cell-associated fluorescence or identified on multiple z-series by laser confocal microscopy. The 3D-organotypic models were set up by seeding melanoma cells onto collagen I matrices embedded dermal fibroblasts. Data were analyzed by one-way ANOVA and post-hoc Dunnett t-test for multiple comparisons. RESULTS: We found that the co-expression of uPAR and FPR1 confers to A375 and M14 melanoma cells a clear-cut capability to move towards chemotactic gradients, to cross extracellular matrix and endothelial monolayers. FPR1 activity is required, as cell migration and invasion were abrogated by receptor desensitization. Finally, melanoma cell ability to move toward chemotactic gradients, invade matrigel or fibroblast-embedded collagen matrices and cross endothelial monolayers are prevented by anti-uPAR84-95 antibodies or by the RI-3 peptide which we have previously shown to inhibit the uPAR84-95/FPR1 interaction. CONCLUSIONS: Collectively, our findings identify uPAR and FPR1 as relevant effectors of melanoma cell invasiveness and suggest that inhibitors of the uPAR84-95/FPR1 cross-talk may be useful for the treatment of metastatic melanoma.


Asunto(s)
Melanoma/metabolismo , Receptores de Formil Péptido/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Línea Celular Tumoral , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Humanos , Melanoma/genética , Melanoma/patología , Receptores de Formil Péptido/genética , Transfección , Activador de Plasminógeno de Tipo Uroquinasa/genética
6.
Sci Rep ; 7(1): 1312, 2017 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-28465589

RESUMEN

The development of metastases is a multistep process that requires the activation of physiological and biochemical processes that govern migration, invasion and entry of metastatic cells into blood vessels. The urokinase receptor (uPAR) promotes cell migration by interacting with the Formyl Peptide Receptors (FPRs). Since both uPAR and FPR1 are involved in tumor progression, the uPAR-FPR1 interaction is an attractive therapeutic target. We previously described peptide antagonists of the uPAR-FPR1 interaction that inhibited cell migration and angiogenesis. To develop enzyme-resistant analogues, we applied here the Retro-Inverso (RI) approach, whereby the topology of the side chains is maintained by inverting the sequence of the peptide and the chirality of all residues. Molecular dynamics suggests that peptide RI-3 adopts the turn structure typical of uPAR-FPR1 antagonists. Accordingly, RI-3 is a nanomolar competitor of N-formyl-Met-Leu-Phe for binding to FPR1 and inhibits migration, invasion, trans-endothelial migration of sarcoma cells and VEGF-triggered endothelial tube formation. When sarcoma cells were subcutaneously injected in nude mice, tumor size, intra-tumoral microvessel density, circulating tumor cells and pulmonary metastases were significantly reduced in animals treated daily with 6 mg/Kg RI-3 as compared to animals treated with vehicle only. Thus, RI-3 represents a promising lead for anti-metastatic drugs.


Asunto(s)
Neovascularización Patológica/tratamiento farmacológico , Péptidos/administración & dosificación , Receptores del Activador de Plasminógeno Tipo Uroquinasa/antagonistas & inhibidores , Sarcoma/tratamiento farmacológico , Animales , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Ratones , Simulación de Dinámica Molecular , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Metástasis de la Neoplasia , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Receptores de Formil Péptido/antagonistas & inhibidores , Receptores de Formil Péptido/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Sarcoma/genética , Sarcoma/patología , Ensayos Antitumor por Modelo de Xenoinjerto
7.
J Pept Sci ; 23(4): 303-310, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28078813

RESUMEN

'Privileged scaffolds' are molecular frameworks which have been successfully exploited for small molecule drug discovery. Peptide privileged scaffolds, featuring a strictly conserved multiple-disulfide framework and high variability in the rest of the sequence, display a broad range of biological effects, including antimicrobial and antiviral activity. Unlike small molecules, however, the cost of manufacturing these peptides is high, and their synthesis challenging. We previously described a simplified privileged scaffold corresponding to the γ-core of human ß-defensin-3 (HBD3). The γ-core is a common structural signature found in virtually all host defense peptides (HDPs) stabilized by multiple disulfides, and we showed that for HBD3, it represents the evolutionary starting point of the full-length molecule and, thus, is itself a primordial HDP. Accordingly, we showed that the peptide folded rapidly and was stable in human serum, and displayed many of the biological activities of HBD3. We report here that in addition to the previously reported antibacterial activity on planktonic bacteria, the γ-core peptide is active against biofilm formation and maturation. We also show that it is readily cell penetrant, like HBD3, although with a different mechanism, which is independent from CD98. Overall, the potency of the single-disulfide, 23-amino acid γ-core is comparable with the full-length peptide across the whole spectrum of examined properties, and the peptide is not toxic to human cells. The HBD3 γ-core peptide may therefore represent the first example of an economically viable lead peptide derived from a HDP privileged scaffold. Copyright © 2017 European Peptide Society and John Wiley & Sons, Ltd.


Asunto(s)
Antibacterianos/farmacología , Descubrimiento de Drogas , Péptidos/farmacología , Pseudomonas aeruginosa/efectos de los fármacos , Staphylococcus aureus/efectos de los fármacos , beta-Defensinas/química , Antibacterianos/síntesis química , Antibacterianos/química , Biopelículas/efectos de los fármacos , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Pruebas de Sensibilidad Microbiana , Péptidos/síntesis química , Péptidos/química , Relación Estructura-Actividad
8.
Colloids Surf B Biointerfaces ; 152: 311-316, 2017 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-28131091

RESUMEN

The HIV broadly neutralizing antibody 2F5 targets the transiently exposed epitope in the membrane proximal external region (MPER) of HIV-1 gp41, by a two-step mechanism involving the viral membrane and this viral glycoprotein. It was recently shown that 2F5 conjugation with a cholesterol moiety outside of the antibody paratope substantially increases its antiviral activity. Additionally, the antiviral activity of D5, a human antibody that binds to the N-terminal heptad repeat (NHR) of gp41 and lacks membrane binding, was boosted by the same cholesterol conjugation. In this work, we evaluated the membrane affinity of both antibodies towards membranes of different compositions, using surface plasmon resonance. A correlation was found between membrane affinity and antiviral activity against HIV-1. We propose that the conjugation of cholesterol to 2F5 or D5 allows a higher degree of antibody pre-concentration at the viral membrane. This way, the antibodies become more available to bind efficiently to the gp41 epitope, blocking viral fusion faster than the unconjugated antibody. These results set up a relevant strategy to improve the rational design of therapeutic antibodies against HIV.


Asunto(s)
Anticuerpos Neutralizantes/química , Anticuerpos Neutralizantes/farmacología , Anticuerpos Anti-VIH/química , Anticuerpos Anti-VIH/farmacología , Antivirales/química , Antivirales/farmacología , VIH/efectos de los fármacos , Membranas Artificiales , Pruebas de Neutralización , Resonancia por Plasmón de Superficie
9.
Sci Rep ; 5: 18450, 2015 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-26688341

RESUMEN

Host defence peptides (HDPs) are critical components of innate immunity. Despite their diversity, they share common features including a structural signature, designated "γ-core motif". We reasoned that for each HDPs evolved from an ancestral γ-core, the latter should be the evolutionary starting point of the molecule, i.e. it should represent a structural scaffold for the modular construction of the full-length molecule, and possess biological properties. We explored the γ-core of human ß-defensin 3 (HBD3) and found that it: (a) is the folding nucleus of HBD3; (b) folds rapidly and is stable in human serum; (c) displays antibacterial activity; (d) binds to CD98, which mediates HBD3 internalization in eukaryotic cells; (e) exerts antiviral activity against human immunodeficiency virus and herpes simplex virus; and (f) is not toxic to human cells. These results demonstrate that the γ-core within HBD3 is the ancestral core of the full-length molecule and is a viable HDP per se, since it is endowed with the most important biological features of HBD3. Notably, the small, stable scaffold of the HBD3 γ-core can be exploited to design disease-specific antimicrobial agents.


Asunto(s)
Secuencias de Aminoácidos/genética , Antiinfecciosos/metabolismo , Inmunidad Innata/genética , beta-Defensinas/metabolismo , Antiinfecciosos/uso terapéutico , Antivirales/metabolismo , Antivirales/uso terapéutico , Proteína-1 Reguladora de Fusión/química , Proteína-1 Reguladora de Fusión/metabolismo , VIH-1/efectos de los fármacos , Humanos , Péptidos/química , Péptidos/metabolismo , Unión Proteica , Pliegue de Proteína , Simplexvirus/efectos de los fármacos , beta-Defensinas/química , beta-Defensinas/genética
10.
J Pept Sci ; 21(9): 743-9, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26292842

RESUMEN

Immunoadhesins are engineered proteins combining the constant domain (Fc) of an antibody with a ligand-binding (adhesion) domain. They have significant potential as therapeutic agents, because they maintain the favourable pharmacokinetics of antibodies with an expanded repertoire of ligand-binding domains: proteins, peptides, or small molecules. We have recently reported that the addition of a cholesterol group to two HIV antibodies can dramatically improve their antiviral potency. Cholesterol, which can be conjugated at various positions in the antibody, including the constant (Fc) domain, endows the conjugate with affinity for the membrane lipid rafts, thus increasing its concentration at the site where viral entry occurs. Here, we extend this strategy to an HIV immunoadhesin, combining a cholesterol-conjugated Fc domain with the peptide fusion inhibitor C41. The immunoadhesin C41-Fc-chol displayed high affinity for Human Embryonic Kidney (HEK) 293 cells, and when tested on a panel of HIV-1 strains, it was considerably more potent than the unconjugated C41-Fc construct. Potentiation of antiviral activity was comparable to what was previously observed for the cholesterol-conjugated HIV antibodies. Given the key role of cholesterol in lipid raft formation and viral fusion, we expect that the same strategy should be broadly applicable to enveloped viruses, for many of which it is already known the sequence of a peptide fusion inhibitor similar to C41. Moreover, the sequence of heptad repeat-derived fusion inhibitors can often be predicted from genomic information alone, opening a path to immunoadhesins against emerging viruses.


Asunto(s)
Antivirales/química , Colesterol/química , Péptidos/química , Antivirales/farmacología , Diseño de Fármacos , Células HEK293 , Infecciones por VIH/tratamiento farmacológico , VIH-1/efectos de los fármacos , Humanos , Péptidos/farmacología , Internalización del Virus/efectos de los fármacos
11.
Chem Biol ; 22(2): 217-28, 2015 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-25641165

RESUMEN

Human ß-defensins play a pivotal role in the innate immune response. Although expressed by and acting at epithelial surfaces, little is known about their specific interaction with epithelial structures. Here, we identify the transmembrane protein CD98 as a cell surface receptor involved in the internalization of human ß-defensin 3 (hBD3) in human epithelial A549 cells. CD98 and hBD3 extensively colocalize on the basolateral domain of A549. While verifying their direct binding by fluorescence resonance energy transfer and surface plasmon resonance, we mapped the interaction to CD98 residues 304-414, i.e. to the region known to interact with the proteins of intestinal bacteria during colonic invasion. Treatment of A549 cells with hBD3 dramatically reduces CD98 expression and conversely, knockdown of CD98 expression impairs hBD3 cell surface binding and internalization. Competition for bacterial binding to CD98 and downregulation of CD98 expression may represent novel mechanisms for the antibacterial activity of hBD3.


Asunto(s)
Proteína-1 Reguladora de Fusión/metabolismo , beta-Defensinas/metabolismo , Antibacterianos/síntesis química , Antibacterianos/farmacología , Biotina/química , Línea Celular Tumoral , Células Epiteliales/citología , Células Epiteliales/metabolismo , Escherichia coli/efectos de los fármacos , Transferencia Resonante de Energía de Fluorescencia , Proteína-1 Reguladora de Fusión/antagonistas & inhibidores , Proteína-1 Reguladora de Fusión/genética , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Microscopía Confocal , Simulación de Dinámica Molecular , Unión Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas , Proteómica , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Resonancia por Plasmón de Superficie , beta-Defensinas/química , beta-Defensinas/farmacología
12.
J Pept Sci ; 21(5): 379-86, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25331523

RESUMEN

While it is now possible to identify and genetically fingerprint the causative agents of emerging viral diseases, often with extraordinary speed, suitable therapies cannot be developed with equivalent speed, because drug discovery requires information that goes beyond knowledge of the viral genome. Peptides, however, may represent a special opportunity. For all enveloped viruses, fusion between the viral and the target cell membrane is an obligatory step of the life cycle. Class I fusion proteins harbor regions with a repeating pattern of amino acids, the heptad repeats (HRs), that play a key role in fusion, and HR-derived peptides such as enfuvirtide, in clinical use for HIV, can block the process. Because of their characteristic sequence pattern, HRs are easily identified in the genome by means of computer programs, providing the sequence of candidate peptide inhibitors directly from genomic information. Moreover, a simple chemical modification, the attachment of a cholesterol group, can dramatically increase the antiviral potency of HR-derived inhibitors and simultaneously improve their pharmacokinetics. Further enhancement can be provided by dimerization of the cholesterol-conjugated peptide. The examples reported so far include inhibitors of retroviruses, paramyxoviruses, orthomyxoviruses, henipaviruses, coronaviruses, and filoviruses. For some of these viruses, in vivo efficacy has been demonstrated in suitable animal models. The combination of bioinformatic lead identification and potency/pharmacokinetics improvement provided by cholesterol conjugation may form the basis for a rapid response strategy, where development of an emergency cholesterol-conjugated therapeutic would immediately follow the availability of the genetic information of a new enveloped virus.


Asunto(s)
Colesterol/química , Péptidos/química , Péptidos/farmacocinética , Virosis/tratamiento farmacológico , Animales , Antivirales/química , Antivirales/farmacocinética , Antivirales/farmacología , Biología Computacional/métodos , Diseño de Fármacos , Humanos , Péptidos/farmacología , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/efectos de los fármacos , Proteínas Virales de Fusión/genética
13.
J Virol ; 87(24): 13785-94, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24109233

RESUMEN

Measles virus (MV) infection causes an acute childhood disease that can include infection of the central nervous system and can rarely progress to severe neurological disease for which there is no specific treatment. We generated potent antiviral peptide inhibitors of MV entry and spreading and MV-induced cell fusion. Dimers of MV-specific peptides derived from the C-terminal heptad repeat region of the MV fusion protein, conjugated to cholesterol, efficiently protect SLAM transgenic mice from fatal MV infection. Fusion inhibitors hold promise for the prophylaxis of MV infection in unvaccinated and immunocompromised people, as well as potential for the treatment of grave neurological complications of measles.


Asunto(s)
Antivirales/farmacología , Encéfalo/virología , Virus del Sarampión/efectos de los fármacos , Sarampión/prevención & control , Proteínas Virales de Fusión/antagonistas & inhibidores , Animales , Encéfalo/efectos de los fármacos , Línea Celular , Humanos , Sarampión/tratamiento farmacológico , Sarampión/mortalidad , Sarampión/virología , Virus del Sarampión/genética , Virus del Sarampión/fisiología , Ratones , Ratones Transgénicos , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/metabolismo , Internalización del Virus/efectos de los fármacos
14.
PLoS One ; 7(5): e36833, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22666328

RESUMEN

Fusion between the viral and target cell membranes is an obligatory step for the infectivity of all enveloped virus, and blocking this process is a clinically validated therapeutic strategy.Viral fusion is driven by specialized proteins which, although specific to each virus, act through a common mechanism, the formation of a complex between two heptad repeat (HR) regions. The HR regions are initially separated in an intermediate termed "prehairpin", which bridges the viral and cell membranes, and then fold onto each other to form a 6-helical bundle (6HB), driving the two membranes to fuse. HR-derived peptides can inhibit viral infectivity by binding to the prehairpin intermediate and preventing its transition to the 6HB.The antiviral activity of HR-derived peptides differs considerably among enveloped viruses. For weak inhibitors, potency can be increased by peptide engineering strategies, but sequence-specific optimization is time-consuming. In seeking ways to increase potency without changing the native sequence, we previously reported that attachment to the HR peptide of a cholesterol group ("cholesterol-tagging") dramatically increases its antiviral potency, and simultaneously increases its half-life in vivo. We show here that antiviral potency may be increased by combining cholesterol-tagging with dimerization of the HR-derived sequence, using as examples human parainfluenza virus, Nipah virus, and HIV-1. Together, cholesterol-tagging and dimerization may represent strategies to boost HR peptide potency to levels that in some cases may be compatible with in vivo use, possibly contributing to emergency responses to outbreaks of existing or novel viruses.


Asunto(s)
Antivirales/química , Antivirales/farmacología , Productos Biológicos/química , Diseño de Fármacos , Fragmentos de Péptidos/química , Fragmentos de Péptidos/farmacología , Proteínas Virales de Fusión/química , Secuencia de Aminoácidos , Animales , Antivirales/metabolismo , Colesterol/metabolismo , Cricetinae , Células HeLa , Humanos , Datos de Secuencia Molecular , Fragmentos de Péptidos/metabolismo , Multimerización de Proteína , Estructura Terciaria de Proteína , Virus ARN/efectos de los fármacos , Virus ARN/fisiología , Replicación Viral/efectos de los fármacos
15.
J Virol ; 85(24): 12867-80, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21976650

RESUMEN

During paramyxovirus entry into a host cell, receptor engagement by a specialized binding protein triggers conformational changes in the adjacent fusion protein (F), leading to fusion between the viral and cell membranes. According to the existing paradigm of paramyxovirus membrane fusion, the initial activation of F by the receptor binding protein sets off a spring-loaded mechanism whereby the F protein progresses independently through the subsequent steps in the fusion process, ending in membrane merger. For human parainfluenza virus type 3 (HPIV3), the receptor binding protein (hemagglutinin-neuraminidase [HN]) has three functions: receptor binding, receptor cleaving, and activating F. We report that continuous receptor engagement by HN activates F to advance through the series of structural rearrangements required for fusion. In contrast to the prevailing model, the role of HN-receptor engagement in the fusion process is required beyond an initiating step, i.e., it is still required even after the insertion of the fusion peptide into the target cell membrane, enabling F to mediate membrane merger. We also report that for Nipah virus, whose receptor binding protein has no receptor-cleaving activity, the continuous stimulation of the F protein by a receptor-engaged binding protein is key for fusion. We suggest a general model for paramyxovirus fusion activation in which receptor engagement plays an active role in F activation, and the continued engagement of the receptor binding protein is essential to F protein function until the onset of membrane merger. This model has broad implications for the mechanism of paramyxovirus fusion and for strategies to prevent viral entry.


Asunto(s)
Proteína HN/metabolismo , Virus Nipah/fisiología , Virus de la Parainfluenza 3 Humana/fisiología , Receptores Virales/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Proteínas Virales de Fusión/metabolismo , Internalización del Virus , Línea Celular , Humanos , Modelos Biológicos , Unión Proteica , Proteínas del Envoltorio Viral/química , Proteínas Virales de Fusión/química
16.
J Biol Chem ; 286(49): 42141-42149, 2011 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-21994935

RESUMEN

We previously described fusion-inhibitory peptides that are targeted to the cell membrane by cholesterol conjugation and potently inhibit enveloped viruses that fuse at the cell surface, including HIV, parainfluenza, and henipaviruses. However, for viruses that fuse inside of intracellular compartments, fusion-inhibitory peptides have exhibited very low antiviral activity. We propose that for these viruses, too, membrane targeting via cholesterol conjugation may yield potent compounds. Here we compare the activity of fusion-inhibitory peptides derived from the influenza hemagglutinin (HA) and show that although the unconjugated peptides are inactive, the cholesterol-conjugated compounds are effective inhibitors of infectivity and membrane fusion. We hypothesize that the cholesterol moiety, by localizing the peptides to the target cell membrane, allows the peptides to follow the virus to the intracellular site of fusion. The cholesterol-conjugated peptides trap HA in a transient intermediate state after fusion is triggered but before completion of the refolding steps that drive the merging of the viral and cellular membranes. These results provide proof of concept for an antiviral strategy that is applicable to intracellularly fusing viruses, including known and emerging viral pathogens.


Asunto(s)
Colesterol/química , Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Animales , Línea Celular , Membrana Celular/metabolismo , Membrana Celular/virología , Chlorocebus aethiops , Colesterol/metabolismo , Endosomas/metabolismo , Prueba de Complementación Genética , Hemaglutininas/química , Humanos , Orthomyxoviridae/metabolismo , Péptidos/química , Desnaturalización Proteica , Pliegue de Proteína , Virus ARN/metabolismo , Células Vero
17.
J Virol ; 85(16): 8422-6, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21653662

RESUMEN

We have previously described heterotypic peptides from parainfluenza virus that potently inhibit Nipah virus in vitro but are not efficacious in vivo. In contrast, our second-generation inhibitors, featuring a cholesterol moiety, are also efficacious in vivo. The difference between in vitro and in vivo results led us to investigate the basis for this discrepancy. Here, we compare the activities of the compounds in standard laboratory cells and in cells relevant to the natural tropism of Nipah virus, i.e., primary neurons, and show that while our first-generation inhibitors are poorly active in primary neurons, the cholesterol-conjugated compounds are highly potent. These results highlight the advantage of evaluating antiviral potency in cells relevant to natural host target tissue.


Asunto(s)
Antivirales/farmacología , Colesterol/farmacología , Neuronas/virología , Virus Nipah/efectos de los fármacos , Péptidos/farmacología , Proteínas del Envoltorio Viral/metabolismo , Antivirales/química , Línea Celular , Colesterol/metabolismo , Células Epiteliales/virología , Humanos , Virus Nipah/fisiología
18.
J Pept Sci ; 17(4): 270-80, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21294225

RESUMEN

Obesity is one of the major risk factors for type 2 diabetes, and the development of agents, that can simultaneously achieve glucose control and weight loss, is being actively pursued. Therapies based on peptide mimetics of the gut hormone glucagon-like peptide 1 (GLP-1) are rapidly gaining favor, due to their ability to increase insulin secretion in a strictly glucose-dependent manner, with little or no risk of hypoglycemia, and to their additional benefit of causing a modest, but durable weight loss. Oxyntomodulin (OXM), a 37-amino acid peptide hormone of the glucagon (GCG) family with dual agonistic activity on both the GLP-1 (GLP1R) and the GCG (GCGR) receptors, has been shown to reduce food intake and body weight in humans, with a lower incidence of treatment-associated nausea than GLP-1 mimetics. As for other peptide hormones, its clinical application is limited by the short circulatory half-life, a major component of which is cleavage by the enzyme dipeptidyl peptidase IV (DPP-IV). SAR studies on OXM, described herein, led to the identification of molecules resistant to DPP-IV degradation, with increased potency as compared to the natural hormone. Analogs derivatized with a cholesterol moiety display increased duration of action in vivo. Moreover, we identified a single substitution which can change the OXM pharmacological profile from a dual GLP1R/GCGR agonist to a selective GLP1R agonist. The latter finding enabled studies, described in detail in a separate study (Pocai A, Carrington PE, Adams JR, Wright M, Eiermann G, Zhu L, Du X, Petrov A, Lassman ME, Jiang G, Liu F, Miller C, Tota LM, Zhou G, Zhang X, Sountis MM, Santoprete A, Capitò E, Chicchi GG, Thornberry N, Bianchi E, Pessi A, Marsh DJ, SinhaRoy R. Glucagon-like peptide 1/glucagon receptor dual agonism reverses obesity in mice. Diabetes 2009; 58: 2258-2266), which highlight the potential of GLP1R/GCGR dual agonists as a potentially superior class of therapeutics over the pure GLP1R agonists currently in clinical use.


Asunto(s)
Dipeptidil Peptidasa 4/metabolismo , Oxintomodulina/química , Oxintomodulina/metabolismo , Secuencia de Aminoácidos , Animales , Glucemia/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Humanos , Ratones , Datos de Secuencia Molecular , Estructura Molecular , Obesidad/tratamiento farmacológico , Oxintomodulina/farmacología , Oxintomodulina/uso terapéutico , Péptidos/síntesis química , Péptidos/química , Péptidos/genética , Pérdida de Peso/efectos de los fármacos
19.
PLoS Pathog ; 6(10): e1001168, 2010 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-21060819

RESUMEN

In the paramyxovirus cell entry process, receptor binding triggers conformational changes in the fusion protein (F) leading to viral and cellular membrane fusion. Peptides derived from C-terminal heptad repeat (HRC) regions in F have been shown to inhibit fusion by preventing formation of the fusogenic six-helix bundle. We recently showed that the addition of a cholesterol group to HRC peptides active against Nipah virus targets these peptides to the membrane where fusion occurs, dramatically increasing their antiviral effect. In this work, we report that unlike the untagged HRC peptides, which bind to the postulated extended intermediate state bridging the viral and cell membranes, the cholesterol tagged HRC-derived peptides interact with F before the fusion peptide inserts into the target cell membrane, thus capturing an earlier stage in the F-activation process. Furthermore, we show that cholesterol tagging renders these peptides active in vivo: the cholesterol-tagged peptides cross the blood brain barrier, and effectively prevent and treat in an established animal model what would otherwise be fatal Nipah virus encephalitis. The in vivo efficacy of cholesterol-tagged peptides, and in particular their ability to penetrate the CNS, suggests that they are promising candidates for the prevention or therapy of infection by Nipah and other lethal paramyxoviruses.


Asunto(s)
Colesterol/uso terapéutico , Infecciones por Henipavirus/prevención & control , Virus Nipah/fisiología , Paramyxovirinae/fisiología , Proteínas Virales de Fusión/antagonistas & inhibidores , Internalización del Virus , Secuencias de Aminoácidos/efectos de los fármacos , Secuencias de Aminoácidos/fisiología , Secuencia de Aminoácidos , Animales , Células Cultivadas , Chlorocebus aethiops , Colesterol/química , Colesterol/farmacología , Regulación hacia Abajo , Infecciones por Henipavirus/inmunología , Infecciones por Henipavirus/terapia , Humanos , Modelos Biológicos , Modelos Moleculares , Datos de Secuencia Molecular , Terapia Molecular Dirigida , Virus Nipah/efectos de los fármacos , Virus Nipah/inmunología , Virus Nipah/patogenicidad , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/uso terapéutico , Células Vero , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/metabolismo , Proteínas Virales de Fusión/fisiología
20.
Proc Natl Acad Sci U S A ; 107(23): 10655-60, 2010 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-20483992

RESUMEN

Eliciting a broadly neutralizing polyclonal antibody response against HIV-1 remains a major challenge. One approach to vaccine development is prevention of HIV-1 entry into cells by blocking the fusion of viral and cell membranes. More specifically, our goal is to elicit neutralizing antibodies that target a transient viral entry intermediate (the prehairpin intermediate) formed by the HIV-1 gp41 protein. Because this intermediate is transient, a stable mimetic is required to elicit an immune response. Previously, a series of engineered peptides was used to select a mAb (denoted D5) that binds to the surface of the gp41 prehairpin intermediate, as demonstrated by x-ray crystallographic studies. D5 inhibits the replication of HIV-1 clinical isolates, providing proof-of-principle for this vaccine approach. Here, we describe a series of peptide mimetics of the gp41 prehairpin intermediate designed to permit a systematic analysis of the immune response generated in animals. To improve the chances of detecting weak neutralizing polyclonal responses, two strategies were employed in the initial screening: use of a neutralization-hypersensitive virus and concentration of the IgG fraction from immunized animal sera. This allowed incremental improvements through iterative cycles of design, which led to vaccine candidates capable of generating a polyclonal antibody response, detectable in unfractionated sera, that neutralize tier 1 HIV-1 and simian HIV primary isolates in vitro. Our findings serve as a starting point for the design of more potent immunogens to elicit a broadly neutralizing response against the gp41 prehairpin intermediate.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Materiales Biomiméticos , Anticuerpos Anti-VIH/inmunología , Proteína gp41 de Envoltorio del VIH/inmunología , VIH-1/inmunología , Sueros Inmunes/inmunología , Vacunación , Secuencia de Aminoácidos , Animales , Cobayas , Proteína gp41 de Envoltorio del VIH/química , VIH-1/química , VIH-1/aislamiento & purificación , Datos de Secuencia Molecular , Péptidos/química , Péptidos/inmunología , Conejos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA