Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros

Banco de datos
Tipo del documento
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.
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
3.
J Biol Chem ; 289(50): 35015-28, 2014 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-25342747

RESUMEN

The broadly neutralizing antibodies HIV 2F5 and 4E10, which bind to overlapping epitopes in the membrane-proximal external region of the fusion protein gp41, have been proposed to use a two-step mechanism for neutralization; first, they bind and preconcentrate at the viral membrane through their long, hydrophobic CDRH3 loops, and second, they form a high affinity complex with the protein epitope. Accordingly, mutagenesis of the CDRH3 can abolish their neutralizing activity, with no change in the affinity for the peptide epitope. We show here that we can mimic this mechanism by conjugating a cholesterol group outside of the paratope of an antibody. Cholesterol-conjugated antibodies bind to lipid raft domains on the membrane, and because of this enrichment, they show increased antiviral potency. In particular, we find that cholesterol conjugation (i) rescues the antiviral activity of CDRH3-mutated 2F5, (ii) increases the antiviral activity of WT 2F5, (iii) potentiates the non-membrane-binding HIV antibody D5 10-100-fold (depending on the virus strain), and (iv) increases synergy between 2F5 and D5. Conjugation can be made at several positions, including variable and constant domains. Cholesterol conjugation therefore appears to be a general strategy to boost the potency of antiviral antibodies, and, because membrane affinity is engineered outside of the antibody paratope, it can complement affinity maturation strategies.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Anticuerpos Neutralizantes/metabolismo , Colesterol/metabolismo , Anticuerpos Anti-VIH/inmunología , Anticuerpos Anti-VIH/metabolismo , VIH-1/inmunología , Anticuerpos Neutralizantes/inmunología , Membrana Celular/metabolismo , Células HEK293 , Humanos , Pruebas de Neutralización
4.
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
5.
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
6.
J Antimicrob Chemother ; 69(5): 1286-97, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24464268

RESUMEN

OBJECTIVES: The aim of the present work was to evaluate the interaction of two new HIV fusion inhibitors {HIVP3 [C34-polyethylene glycol (PEG)4-cholesterol] and HIVP4 [(C34-PEG4)2-cholesterol]} with membrane model systems and human blood cells in order to clarify where and how the fusion inhibitors locate, allowing us to understand their mechanism of action at the molecular level, and which strategies may be followed to increase efficacy. METHODS: Lipid vesicles with defined compositions were used for peptide partition and localization studies, based on the intrinsic fluorescence of HIVP3 and HIVP4. Lipid monolayers were employed in surface pressure studies. Finally, human erythrocytes and peripheral blood mononuclear cells (PBMCs) isolated from blood samples were used in dipole potential assays. RESULTS: Membrane partition, dipole potential and surface pressure assays indicate that the new fusion inhibitors interact preferentially with cholesterol-rich liquid-ordered membranes, mimicking biological membrane microdomains known as lipid rafts. HIVP3 and HIVP4 are able to interact with human erythrocytes and PBMCs to a similar degree as a previously described simpler drug with monomeric C34 and lacking the PEG spacer, C34-cholesterol. However, the pocket-binding domain (PBD) of both HIVP3 and HIVP4 is more exposed to the aqueous environment than in C34-cholesterol. CONCLUSIONS: The present data allow us to conclude that more efficient blocking of HIV entry results from the synergism between the membranotropic behaviour and the enhanced exposure of the PBD.


Asunto(s)
Proteína gp41 de Envoltorio del VIH/farmacología , Inhibidores de Fusión de VIH/farmacología , Fragmentos de Péptidos/farmacología , Membrana Celular/metabolismo , Eritrocitos/metabolismo , Humanos , Leucocitos Mononucleares/metabolismo , Unión Proteica
7.
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
8.
J Pept Sci ; 20(1): 7-19, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24222478

RESUMEN

Neuromedin U (NMU) is an endogenous peptide implicated in the regulation of feeding, energy homeostasis, and glycemic control, which is being considered for the therapy of obesity and diabetes. A key liability of NMU as a therapeutic is its very short half-life in vivo. We show here that conjugation of NMU to human serum albumin (HSA) yields a compound with long circulatory half-life, which maintains full potency at both the peripheral and central NMU receptors. Initial attempts to conjugate NMU via the prevalent strategy of reacting a maleimide derivative of the peptide with the free thiol of Cys34 of HSA met with limited success, because the resulting conjugate was unstable in vivo. Use of a haloacetyl derivative of the peptide led instead to the formation of a metabolically stable conjugate. HSA-NMU displayed long-lasting, potent anorectic, and glucose-normalizing activity. When compared side by side with a previously described PEG conjugate, HSA-NMU proved superior on a molar basis. Collectively, our results reinforce the notion that NMU-based therapeutics are promising candidates for the treatment of obesity and diabetes.


Asunto(s)
Fármacos Antiobesidad/síntesis química , Hipoglucemiantes/síntesis química , Neuropéptidos/síntesis química , Neuropéptidos/farmacología , Polietilenglicoles/farmacología , Albúmina Sérica/síntesis química , Animales , Fármacos Antiobesidad/farmacocinética , Fármacos Antiobesidad/farmacología , Glucemia , Línea Celular , Evaluación Preclínica de Medicamentos , Humanos , Hipoglucemiantes/farmacocinética , Hipoglucemiantes/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Neuropéptidos/farmacocinética , Polietilenglicoles/farmacocinética , Receptores de Neurotransmisores/agonistas , Albúmina Sérica/farmacocinética , Albúmina Sérica/farmacología , Albúmina Sérica Humana , Pérdida de Peso/efectos de los fármacos
9.
Bioorg Med Chem ; 21(22): 7064-73, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24094437

RESUMEN

Peptide agonists of the glucagon-like peptide 1 (GLP-1) receptor (GLP1R) are rapidly gaining favor as antidiabetic agents, since in addition to increasing glucose-dependent insulin secretion, they also cause weight loss. Oxyntomodulin (OXM), a natural peptide with sequence homology to both glucagon and GLP-1, has glucose-lowering activity in rodents and anorectic activity in rodents and humans, but its clinical utility is limited by a short circulatory half-life due to rapid renal clearance and degradation by dipeptidyl peptidase IV (DPP-IV). Here, we describe the development of a novel DPP-IV-resistant, long-acting GLP1R agonist, based on derivatization of a suitably chosen OXM analog with high molecular weight polyethylene glycol (PEG) ('PEGylation'). PEG-OXM exerts an anti-hyperglycemic effect in diet-induced obese (DIO) mice in a glucose-dependent manner, with a maximally efficacious dose of 0.1mg/kg, and reduces food intake and body weight with a minimally efficacious dose of 1mg/kg. If this pharmacology is recapitulated in patients with type 2 diabetes, these results indicate PEG-OXM as a potential novel once-weekly GLP-1 mimetic with both glucose-lowering activity and weight loss efficacy.


Asunto(s)
Depresores del Apetito/química , Hipoglucemiantes/química , Oxintomodulina/química , Polietilenglicoles/química , Receptores de Glucagón/agonistas , Animales , Depresores del Apetito/síntesis química , Depresores del Apetito/farmacocinética , Peso Corporal/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Receptor del Péptido 1 Similar al Glucagón , Prueba de Tolerancia a la Glucosa , Semivida , Hipoglucemiantes/síntesis química , Hipoglucemiantes/farmacocinética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Primates , Receptores de Glucagón/metabolismo
10.
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
11.
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
12.
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
13.
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
14.
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
15.
Bioorg Med Chem ; 20(15): 4751-9, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22771182

RESUMEN

Neuromedin U (NMU) is an endogenous peptide, whose role in the regulation of feeding and energy homeostasis is well documented. Two NMU receptors have been identified: NMUR1, expressed primarily in the periphery, and NMUR2, expressed predominantly in the brain. We recently demonstrated that acute peripheral administration of NMU exerts potent but acute anorectic activity and can improve glucose homeostasis, with both actions mediated by NMUR1. Here, we describe the development of a metabolically stable analog of NMU, based on derivatization of the native peptide with high molecular weight poly(ethylene) glycol (PEG) ('PEGylation'). PEG size, site of attachment, and conjugation chemistry were optimized, to yield an analog which displays robust and long-lasting anorectic activity and significant glucose-lowering activity in vivo. Studies in NMU receptor-deficient mice showed that PEG-NMU displays an expanded pharmacological profile, with the ability to engage NMUR2 in addition to NMUR1. In light of these data, PEGylated derivatives of NMU represent promising candidates for the treatment of obesity and diabetes.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Neuropéptidos/farmacología , Obesidad/tratamiento farmacológico , Polietilenglicoles/química , Receptores de Neurotransmisores/agonistas , Animales , Relación Dosis-Respuesta a Droga , Prueba de Tolerancia a la Glucosa , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuropéptidos/administración & dosificación , Neuropéptidos/síntesis química , Polietilenglicoles/administración & dosificación , Polietilenglicoles/síntesis química , Polietilenglicoles/farmacología , Receptores de Neurotransmisores/deficiencia , Relación Estructura-Actividad
16.
Proc Natl Acad Sci U S A ; 106(14): 5801-6, 2009 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-19297617

RESUMEN

Peptides derived from the heptad repeat 2 (HR2) region of the HIV fusogenic protein gp41 are potent inhibitors of viral infection, and one of them, enfuvirtide, is used for the treatment of therapy-experienced AIDS patients. The mechanism of action of these peptides is binding to a critical intermediate along the virus-cell fusion pathway, and accordingly, increasing the affinity for the intermediate yields more potent inhibitors. We took a different approach, namely to increase the potency of the HR2 peptide inhibitor C34 by targeting it to the cell compartment where fusion occurs, and we show here that a simple, yet powerful way to accomplish this is attachment of a cholesterol group. C34 derivatized with cholesterol (C34-Chol) shows dramatically increased antiviral potency on a panel of primary isolates, with IC(90) values 15- to 300-fold lower than enfuvirtide and the second-generation inhibitor T1249, making C34-Chol the most potent HIV fusion inhibitor to date. Consistent with its anticipated mechanism of action, the antiviral activity of C34-Chol is unusually persistent: washing target cells after incubation with C34-Chol, but before triggering fusion, increases IC(50) only 7-fold, relative to a 400-fold increase observed for C34. Moreover, derivatization with cholesterol extends the half-life of the peptide in vivo. In the mouse, s.c. administration of 3.5 mg/kg C34-Chol yields a plasma concentration 24 h after injection >300-fold higher than the measured IC(90) values. Because the fusion machinery targeted by C34-Chol is similar in several other enveloped viruses, we believe that these findings may be of general utility.


Asunto(s)
Colesterol/farmacocinética , Sistemas de Liberación de Medicamentos/métodos , Inhibidores de Fusión de VIH/farmacocinética , Animales , Colesterol/química , Relación Dosis-Respuesta a Droga , Inhibidores de Fusión de VIH/síntesis química , Infecciones por VIH/tratamiento farmacológico , Semivida , Células HeLa , Humanos , Concentración 50 Inhibidora , Ratones , Relación Estructura-Actividad
17.
ACS Med Chem Lett ; 13(8): 1248-1254, 2022 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-35978682

RESUMEN

Peptide-based analogues of the gut-derived incretin hormone, glucagon-like peptide 1 (GLP1), stimulate insulin secretion in a glucose-dependent manner. Currently marketed GLP1 receptor (GLP1R) agonists are safe and effective in the management of Type 2 diabetes but often offer only modest weight loss. This has prompted the search for safe and effective alternatives to enhance the weight loss component of these treatments. We have demonstrated that concomitant activation GLP1R and the glucagon receptor (GCGR) can improve glucose metabolism and provide superior weight loss when compared to selective GLP1R agonism in preclinical species. This paper will highlight chemistry structure-activity relationship optimization and summarize in vivo efficacy studies toward the discovery of a once daily balanced dual agonist 12 (MK-1462), which was advanced into clinical trials.

18.
J Virol ; 84(13): 6760-8, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20357085

RESUMEN

The fusion of enveloped viruses with the host cell is driven by specialized fusion proteins to initiate infection. The "class I" fusion proteins harbor two regions, typically two heptad repeat (HR) domains, which are central to the complex conformational changes leading to fusion: the first heptad repeat (HRN) is adjacent to the fusion peptide, while the second (HRC) immediately precedes the transmembrane domain. Peptides derived from the HR regions can inhibit fusion, and one HR peptide, T20 (enfuvirtide), is in clinical use for HIV-1. For paramyxoviruses, the activities of two membrane proteins, the receptor-binding protein (hemagglutinin-neuraminidase [HN] or G) and the fusion protein (F), initiate viral entry. The binding of HN or G to its receptor on a target cell triggers the activation of F, which then inserts into the target cell and mediates the membrane fusion that initiates infection. We have shown that for paramyxoviruses, the inhibitory efficacy of HR peptides is inversely proportional to the rate of F activation. For HIV-1, the antiviral potency of an HRC-derived peptide can be dramatically increased by targeting it to the membrane microdomains where fusion occurs, via the addition of a cholesterol group. We report here that for three paramyxoviruses-human parainfluenza virus type 3 (HPIV3), a major cause of lower respiratory tract diseases in infants, and the emerging zoonotic viruses Hendra virus (HeV) and Nipah virus (NiV), which cause lethal central nervous system diseases-the addition of cholesterol to a paramyxovirus HRC-derived peptide increased antiviral potency by 2 log units. Our data suggest that this enhanced activity is indeed the result of the targeting of the peptide to the plasma membrane, where fusion occurs. The cholesterol-tagged peptides on the cell surface create a protective antiviral shield, target the F protein directly at its site of action, and expand the potential utility of inhibitory peptides for paramyxoviruses.


Asunto(s)
Antivirales/farmacología , Virus Hendra/fisiología , Virus Nipah/fisiología , Virus de la Parainfluenza 3 Humana/fisiología , Internalización del Virus/efectos de los fármacos , Animales , Antivirales/química , Línea Celular , Chlorocebus aethiops , Virus Hendra/efectos de los fármacos , Humanos , Virus Nipah/efectos de los fármacos , Virus de la Parainfluenza 3 Humana/efectos de los fármacos
19.
Nat Struct Mol Biol ; 13(8): 740-7, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16862157

RESUMEN

Elicitation of potent and broadly neutralizing antibodies is an important goal in designing an effective human immunodeficiency virus-1 (HIV-1) vaccine. The HIV-1 gp41 inner-core trimer represents a functionally and structurally conserved target for therapeutics. Here we report the 2.0-A-resolution crystal structure of the complex between the antigen-binding fragment of D5, an HIV-1 cross-neutralizing antibody, and 5-helix, a gp41 inner-core mimetic. Both binding and neutralization depend on residues in the D5 CDR H2 loop protruding into the conserved gp41 hydrophobic pocket, as well as a large pocket in D5 surrounding core gp41 residues. Kinetic analysis of D5 mutants with perturbed D5-gp41 interactions suggests that D5 persistence at the fusion intermediate is crucial for neutralization. Thus, our data validate the gp41 N-peptide trimer fusion intermediate as a target for neutralizing antibodies and provide a template for identification of more potent and broadly neutralizing molecules.


Asunto(s)
Anticuerpos Monoclonales/química , Proteína gp41 de Envoltorio del VIH/química , Proteína gp41 de Envoltorio del VIH/inmunología , VIH-1/inmunología , Anticuerpos Monoclonales/metabolismo , Células Cultivadas , Cristalografía por Rayos X , Proteína gp41 de Envoltorio del VIH/metabolismo , VIH-1/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Leucina/química , Modelos Moleculares , Mutación , Pruebas de Neutralización , Conformación Proteica , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Triptófano/química
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA