Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 680
Filtrar
1.
Nature ; 623(7988): 820-827, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37938771

RESUMEN

The majority of oncogenic drivers are intracellular proteins, constraining their immunotherapeutic targeting to mutated peptides (neoantigens) presented by individual human leukocyte antigen (HLA) allotypes1. However, most cancers have a modest mutational burden that is insufficient for generating responses using neoantigen-based therapies2,3. Neuroblastoma is a paediatric cancer that harbours few mutations and is instead driven by epigenetically deregulated transcriptional networks4. Here we show that the neuroblastoma immunopeptidome is enriched with peptides derived from proteins essential for tumorigenesis. We focused on targeting the unmutated peptide QYNPIRTTF discovered on HLA-A*24:02, which is derived from the neuroblastoma-dependency gene and master transcriptional regulator PHOX2B. To target QYNPIRTTF, we developed peptide-centric chimeric antigen receptors (PC-CARs) through a counter panning strategy using predicted potentially cross-reactive peptides. We further proposed that PC-CARs can recognize peptides on additional HLA allotypes when presenting a similar overall molecular surface. Informed by our computational modelling results, we show that PHOX2B PC-CARs also recognize QYNPIRTTF presented by HLA-A*23:01, the most common non-A2 allele in people with African ancestry. Finally, we demonstrate potent and specific killing of neuroblastoma cells expressing these HLAs in vitro and complete tumour regression in mice. These data suggest that PC-CARs have the potential to expand the pool of immunotherapeutic targets to include non-immunogenic intracellular oncoproteins and allow targeting through additional HLA allotypes in a clinical setting.


Asunto(s)
Antígenos de Neoplasias , Neuroblastoma , Proteínas Oncogénicas , Péptidos , Receptores Quiméricos de Antígenos , Animales , Humanos , Ratones , África/etnología , Alelos , Secuencia de Aminoácidos , Carcinogénesis , Reacciones Cruzadas , Antígenos HLA-A/química , Antígenos HLA-A/inmunología , Neuroblastoma/genética , Neuroblastoma/inmunología , Neuroblastoma/terapia , Proteínas Oncogénicas/antagonistas & inhibidores , Proteínas Oncogénicas/inmunología , Péptidos/antagonistas & inhibidores , Péptidos/química , Péptidos/inmunología , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/uso terapéutico
2.
Biochem Soc Trans ; 51(3): 925-936, 2023 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-37293994

RESUMEN

The E3 ligase beta-transducin repeat-containing protein (ßTrCP) is an essential component of the ubiquitin-proteasome system that is responsible for the maintenance of cellular protein levels in human cells. Key target substrates for degradation include inhibitor of nuclear factor kappa B, programmed cell death protein 4 and forkhead box protein O3, alongside the transcription factor nuclear factor erythroid-2-related factor 2 (NRF2) that is responsible for cellular protection against oxidative damage. The tumour suppressive nature of many of its substrates and the overexpression of ßTrCP observed in various cancers support a potential therapeutic role for inhibitors in the treatment of cancer. A small molecule substituted pyrazolone, GS143, and the natural product erioflorin have been identified as inhibitors of ßTrCP and protect its targets from proteasomal degradation. Modified peptides based on the sequences of native substrates have also been reported with KD values in the nanomolar range. This review describes the current status of inhibitors of this E3 ligase. The scope for further inhibitor design and the development of PROTAC and molecular glue-type structures is explored in the context of ßTrCP as an example of WD40 domain-containing proteins that are gaining attention as drug targets.


Asunto(s)
Factor 2 Relacionado con NF-E2 , Proteínas con Repetición de beta-Transducina , Humanos , Proteínas con Repetición de beta-Transducina/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Péptidos/antagonistas & inhibidores , Péptidos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales
3.
J Microbiol ; 60(7): 727-734, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35614378

RESUMEN

Three major proteases, elastase B (LasB), protease IV (PIV), and elastase A (LasA) expressed in Pseudomonas aeruginosa play important roles in infections and pathogeneses. These are activated by a proteolytic cascade initiated by the activation of LasB. In this study, we investigated whether LasB could be inhibited using its propeptide (LasBpp). Although LasA and PIV were inhibited by their propeptides, LasB was not inhibited by purified LasBpp because LasB degraded LasBpp. To address this problem, mutant LasBpp variants were constructed to obtain a mutant LasBpp resistant to LasB degradation. A C-terminal deletion series of LasBpp was tested in vivo, and two positive candidates, T2 and T2-1, were selected. However, both caused growth retardation and were unstably expressed in vivo. Since deleting the C-terminal end of LasBpp significantly affected its stable expression, substitution mutations were introduced at the two amino acids near the truncation site of T2-1. The resulting mutants, LasBppE172D, LasBppG173A, and LasBppE172DG173A, significantly diminished LasB activity when overexpressed in vivo and were stably expressed in MW1, a quorum sensing mutant that does not produce LasB. In vitro analysis showed that purified LasBppE172DG173A inhibited LasB activity to a small extent. Summarizing, C-terminal modification of LasBpp profoundly affected the stable expression of LasBpp, and little enhanced the ability of LasBpp to resist degradation by LasB.


Asunto(s)
Metaloendopeptidasas , Pseudomonas aeruginosa , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Metaloendopeptidasas/química , Metaloendopeptidasas/genética , Metaloendopeptidasas/metabolismo , Elastasa Pancreática/genética , Elastasa Pancreática/metabolismo , Péptidos/antagonistas & inhibidores , Péptidos/metabolismo , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo , Percepción de Quorum/genética
4.
Behav Brain Res ; 416: 113537, 2022 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-34416299

RESUMEN

Cocaine use disorder is a serious, chronic and relapsing disease of the nervous system, for which effective treatments do not yet exist. Recently, the role of the N-methyl-d-aspartate (NMDA) receptor subunit GluN2B has been highlighted in cocaine abstinence followed by extinction training. Since the GluN2B subunit is stabilized at synaptic level by the interaction with its scaffolding protein PSD95, in this study we aimed at investigating efficacy of Tat-NR2B9c peptide, a PSD95 inhibitor, which disrupts the interaction of PSD95 with GluN2B, in the attenuation of cocaine seeking-behavior or cue-induced reinstatement. We found that Tat-NR2B9c, administered intravenously, attenuated the reinstatement of active lever presses induced by a priming dose of cocaine or by drug-associated conditioned stimuli. At the same time, the GluN2B/PSD95 complex levels were decreased in the ventral hippocampus of rats that previously self-administered cocaine injected with Tat-NR2B9c during cocaine- or cue-induced reinstatement. In conclusion, we here provide the first evidence showing that the disruption of the GluN2B/PSD95 complexes during cocaine abstinence followed by extinction training may represent a useful strategy to reduce reinstatement of cocaine-seeking behavior.


Asunto(s)
Cocaína/farmacología , Comportamiento de Búsqueda de Drogas , Extinción Psicológica/fisiología , Péptidos/antagonistas & inhibidores , Autoadministración , Administración Intravenosa , Animales , Conducta Animal/efectos de los fármacos , Condicionamiento Clásico/efectos de los fármacos , Señales (Psicología) , Homólogo 4 de la Proteína Discs Large/metabolismo , Masculino , Péptidos/administración & dosificación , Ratas , Ratas Sprague-Dawley
5.
Molecules ; 26(4)2021 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-33670371

RESUMEN

Metastasis is the major cause of death in colorectal cancer and it has been proven that inhibiting an interaction between adenomatous polyposis coli (APC) and Rho guanine nucleotide exchange factor 4 (Asef) efficaciously restrain metastasis. However, current inhibitors cannot achieve a satisfying effect in vivo and need to be optimized. In the present study, we applied molecular dynamics (MD) simulations and extensive analyses to apo and holo APC systems in order to reveal the inhibitor mechanism in detail and provide insights into optimization. MD simulations suggested that apo APC takes on a broad array of conformations and inhibitors stabilize conformation selectively. Representative structures in trajectories show specific APC-ligand interactions, explaining the different binding process. The stability and dynamic properties of systems elucidate the inherent factors of the conformation selection mechanism. Binding free energy analysis quantitatively confirms key interface residues and guide optimization. This study elucidates the conformation selection mechanism in APC-Asef inhibition and provides insights into peptide-based drug design.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/antagonistas & inhibidores , Neoplasias Colorrectales/tratamiento farmacológico , Péptidos/química , Proteína de la Poliposis Adenomatosa del Colon/química , Proteína de la Poliposis Adenomatosa del Colon/genética , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Ligandos , Simulación de Dinámica Molecular , Metástasis de la Neoplasia , Péptidos/antagonistas & inhibidores , Unión Proteica/efectos de los fármacos , Factores de Intercambio de Guanina Nucleótido Rho/antagonistas & inhibidores , Factores de Intercambio de Guanina Nucleótido Rho/química , Factores de Intercambio de Guanina Nucleótido Rho/genética
6.
Mol Pharm ; 18(2): 610-626, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-32584043

RESUMEN

Polyglutamine (polyQ) diseases, such as Huntington's disease and several types of spinocerebellar ataxias, are dominantly inherited progressive neurodegenerative disorders and characterized by the presence of expanded CAG trinucleotide repeats in the respective disease locus of the patient genomes. Patients with polyQ diseases currently need to rely on symptom-relieving treatments because disease-modifying therapeutic interventions remain scarce. Many disease-modifying therapeutic agents are now under clinical testing for treating polyQ diseases, but their delivery to the brain is often too invasive (e.g., intracranial injection) or inefficient, owing to in vivo degradation and clearance by physiological barriers (e.g., oral and intravenous administration). Nanoparticles provide a feasible solution for improving drug delivery to the brain, as evidenced by an increasing number of preclinical studies that document the efficacy of nanomedicines for polyQ diseases over the past 5-6 years. In this review, we present the pathogenic mechanisms of polyQ diseases, the common animal models of polyQ diseases for evaluating the efficacy of nanomedicines, and the common administration routes for delivering nanoparticles to the brain. Next, we summarize the recent preclinical applications of nanomedicines for treating polyQ diseases and improving neurological conditions in vivo, placing emphasis on antisense oligonucleotides, small peptide inhibitors, and small molecules as the disease-modifying agents. We conclude with our perspectives of the burgeoning field of "nanomedicines for polyQ diseases", including the use of inorganic nanoparticles and potential drugs as next-generation nanomedicines, development of higher-order animal models of polyQ diseases, and importance of "brain-nano" interactions.


Asunto(s)
Portadores de Fármacos/química , Enfermedad de Huntington/tratamiento farmacológico , Nanopartículas/química , Fármacos Neuroprotectores/administración & dosificación , Péptidos/antagonistas & inhibidores , Ataxias Espinocerebelosas/tratamiento farmacológico , Administración Intranasal , Administración Oral , Animales , Animales Modificados Genéticamente , Disponibilidad Biológica , Barrera Hematoencefálica/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/patología , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Sitios Genéticos/genética , Humanos , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Inyecciones Intraperitoneales , Inyecciones Intravenosas , Inyecciones Intraventriculares , Inyecciones Espinales , Fármacos Neuroprotectores/farmacocinética , Oligonucleótidos Antisentido/administración & dosificación , Oligonucleótidos Antisentido/farmacocinética , Péptidos/genética , Péptidos/metabolismo , Permeabilidad , Médula Espinal/efectos de los fármacos , Médula Espinal/patología , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/patología , Distribución Tisular , Expansión de Repetición de Trinucleótido
7.
Dalton Trans ; 49(47): 17076-17092, 2020 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-33179675

RESUMEN

Protein phosphorylation is a key event in the signalling pathways that control most cell functions, and its deregulation is observed in many human pathologies, including inflammatory, neurodegenerative and autoimmune diseases and cancer. Compounds able to bind phosphoproteins can potentially be used as analytical tools for investigating phosphorylation-based cell signalling and/or as inhibitors of a particular signalling pathway. Metal complexes are arguably the most important class of receptors for the recognition of phosphate-containing molecules. In the last two decades the phosphate-binding ability of metal complexes has been explored for the binding and/or sensing of phosphorylated peptides and proteins. Among those we will focus this review on mono- and dinuclear copper(ii) and zinc(ii) complexes of varied ligand architectures used as binders of phosphorylated peptides and proteins and as sensors of phosphorylation reactions with fluorescence or other techniques in real-time. The cumulative information of strong and selective associations of the indicated receptors allowed selecting some of them for phosphoprotein/peptide enrichment and staining procedures, in vitro monitoring of kinase/phosphatase activity and disruption of phosphorylation-dependent protein-protein interactions. A perspective on the advance of this important area on the frontier between chemistry and biology is presented.


Asunto(s)
Complejos de Coordinación/farmacología , Cobre/farmacología , Péptidos/antagonistas & inhibidores , Fosfoproteínas/antagonistas & inhibidores , Zinc/farmacología , Complejos de Coordinación/química , Cobre/química , Humanos , Estructura Molecular , Péptidos/metabolismo , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Zinc/química
8.
J Neurosci ; 40(49): 9364-9371, 2020 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-33122390

RESUMEN

Mechanosensitivity is a well-known feature of astrocytes, however, its underlying mechanisms and functional significance remain unclear. There is evidence that astrocytes are acutely sensitive to decreases in cerebral perfusion pressure and may function as intracranial baroreceptors, tuned to monitor brain blood flow. This study investigated the mechanosensory signaling in brainstem astrocytes, as these cells reside alongside the cardiovascular control circuits and mediate increases in blood pressure and heart rate induced by falls in brain perfusion. It was found that mechanical stimulation-evoked Ca2+ responses in astrocytes of the rat brainstem were blocked by (1) antagonists of connexin channels, connexin 43 (Cx43) blocking peptide Gap26, or Cx43 gene knock-down; (2) antagonists of TRPV4 channels; (3) antagonist of P2Y1 receptors for ATP; and (4) inhibitors of phospholipase C or IP3 receptors. Proximity ligation assay demonstrated interaction between TRPV4 and Cx43 channels in astrocytes. Dye loading experiments showed that mechanical stimulation increased open probability of carboxyfluorescein-permeable membrane channels. These data suggest that mechanosensory Ca2+ responses in astrocytes are mediated by interaction between TRPV4 and Cx43 channels, leading to Cx43-mediated release of ATP which propagates/amplifies Ca2+ signals via P2Y1 receptors and Ca2+ recruitment from the intracellular stores. In astrocyte-specific Cx43 knock-out mice the magnitude of heart rate responses to acute increases in intracranial pressure was not affected by Cx43 deficiency. However, these animals displayed lower heart rates at different levels of cerebral perfusion, supporting the hypothesis of connexin hemichannel-mediated release of signaling molecules by astrocytes having an excitatory action on the CNS sympathetic control circuits.SIGNIFICANCE STATEMENT There is evidence suggesting that astrocytes may function as intracranial baroreceptors that play an important role in the control of systemic and cerebral circulation. To function as intracranial baroreceptors, astrocytes must possess a specialized membrane mechanism that makes them exquisitely sensitive to mechanical stimuli. This study shows that opening of connexin 43 (Cx43) hemichannels leading to the release of ATP is the key central event underlying mechanosensory Ca2+ responses in astrocytes. This astroglial mechanism plays an important role in the autonomic control of heart rate. These data add to the growing body of evidence suggesting that astrocytes function as versatile surveyors of the CNS metabolic milieu, tuned to detect conditions of potential metabolic threat, such as hypoxia, hypercapnia, and reduced perfusion.


Asunto(s)
Astrocitos/fisiología , Mecanotransducción Celular/fisiología , Adenosina Trifosfato/metabolismo , Animales , Presión Sanguínea/efectos de los fármacos , Tronco Encefálico/citología , Tronco Encefálico/efectos de los fármacos , Tronco Encefálico/fisiología , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Circulación Cerebrovascular/fisiología , Conexina 43/antagonistas & inhibidores , Conexina 43/genética , Femenino , Frecuencia Cardíaca/fisiología , Masculino , Mecanotransducción Celular/efectos de los fármacos , Ratones , Ratones Noqueados , Péptidos/antagonistas & inhibidores , Péptidos/genética , Estimulación Física , Ratas , Receptores Purinérgicos P2Y1/efectos de los fármacos , Canales Catiónicos TRPV/antagonistas & inhibidores , Canales Catiónicos TRPV/genética
9.
Int J Biol Macromol ; 163: 2374-2391, 2020 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-32961180

RESUMEN

Cataract, the major cause of blindness worldwide occurs due to the misfolding and aggregation of the protein crystallin, which constitute a major portion of the lens protein. Other than the whole protein crystallin, the peptide sequences generated from crystallin as a result of covalent protein damage have also been shown to possess and foster protein aggregation, which can be established as crystallin aggregation models. Thus, the disaggregation or inhibition of these protein aggregates could be a viable approach to combat cataract and preserve lens proteostasis. Herein, we tried to explore the disruption as well as inhibition of the intact α-crystallin protein and α-crystallin derived model peptide aggregates by l-3,4-dihydroxyphenylalanine (levodopa) coated gold (Au) nano/micro-roses as modulators. Thioflavin T fluorescence enhancement assay, and electron microscopic analysis were being employed to probe the anti-aggregation behavior of the Au nano/micro-roses towards the aggregating α-crystallin peptides/protein. Further, computational studies were performed to reveal the nature of molecular interactions between the levodopa molecule and the α-crystallin derived model peptides. Interestingly, both levodopa coated Au nano/micro-roses were found to be capable of inhibiting as well as preventing the aggregation of the intact α-crystallin protein and other model peptides derived from it.


Asunto(s)
Anisotropía , Nanopartículas del Metal/química , Péptidos/química , alfa-Cristalinas/química , Oro/química , Levodopa/farmacología , Péptidos/antagonistas & inhibidores , Agregación Patológica de Proteínas/genética , alfa-Cristalinas/genética
10.
J Autoimmun ; 115: 102543, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32951964

RESUMEN

Over the past four decades, the number of people with Type 1 Diabetes (T1D) has increased by 4% per year, making it an important public health challenge. Currently, no curative therapy exists for T1D and the only available treatment is insulin replacement. HLA-DQ8 has been shown to present antigenic islet peptides driving the activation of CD4+ T-cells in T1D patients. Specifically, the insulin peptide InsB:9-23 activates self-reactive CD4+ T-cells, causing pancreatic beta cell destruction. The aim of the current study was to identify retro-inverso-d-amino acid based peptides (RI-D-peptides) that can suppress T-cell activation by blocking the presentation of InsB:9-23 peptide within HLA-DQ8 pocket. We identified a RI-D-peptide (RI-EXT) that inhibited InsB:9-23 binding to recombinant HLA-DQ8 molecule, as well as its binding to DQ8 expressed on human B-cells. RI-EXT prevented T-cell activation in a cellular antigen presentation assay containing human DQ8 cells loaded with InsB:9-23 peptide and murine T-cells expressing a human T-cell receptor specific for the InsB:9-23-DQ8 complex. Moreover, RI-EXT blocked T-cell activation by InsB:9-23 in a humanized DQ8 mice both ex vivo and in vivo, as shown by decreased production of IL-2 and IFN-γ and reduced lymphocyte proliferation. Interestingly, RI-EXT also blocked lymphocyte activation and proliferation by InsB:9-23 in PBMCs isolated from recent onset DQ8-T1D patients. In summary, we discovered a RI-D-peptide that blocks InsB:9-23 binding to HLA-DQ8 and its presentation to T-cells in T1D. These findings set the stage for using our approach as a novel therapy for patients with T1D and potentially other autoimmune diseases.


Asunto(s)
Linfocitos T CD4-Positivos/efectos de los fármacos , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Antígenos HLA-DQ/metabolismo , Células Secretoras de Insulina/inmunología , Péptidos/antagonistas & inhibidores , Animales , Presentación de Antígeno/efectos de los fármacos , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Línea Celular , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Femenino , Antígenos HLA-DQ/inmunología , Antígenos HLA-DQ/aislamiento & purificación , Humanos , Células Secretoras de Insulina/metabolismo , Activación de Linfocitos/efectos de los fármacos , Masculino , Ratones , Ratones Transgénicos , Simulación de Dinámica Molecular , Péptidos/metabolismo , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo
11.
Oxid Med Cell Longev ; 2020: 3129497, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32377295

RESUMEN

Spinocerebellar ataxia type 17 (SCA17) is caused by a CAG/CAA expansion mutation encoding an expanded polyglutamine (polyQ) tract in TATA-box binding protein (TBP), a general transcription initiation factor. Suppression of cAMP-responsive element binding protein- (CREB-) dependent transcription, impaired nuclear factor erythroid 2-related factor 2 (NRF2) signaling, and interaction of AMP-activated protein kinase (AMPK) with increased oxidative stress have been implicated to be involved in pathogenic mechanisms of polyQ-mediated diseases. In this study, we demonstrated decreased pCREB and NRF2 and activated AMPK contributing to neurotoxicity in SCA17 SH-SY5Y cells. We also showed that licochalcone A and the related in-house derivative compound 3-benzoyl-5-hydroxy-2H-chromen-2-one (LM-031) exhibited antiaggregation, antioxidative, antiapoptosis, and neuroprotective effects in TBP/Q79-GFP-expressing cell models. LM-031 and licochalcone A exerted neuroprotective effects by upregulating pCREB and its downstream genes, BCL2 and GADD45B, and enhancing NRF2. Furthermore, LM-031, but not licochalcone A, reduced activated AMPKα. Knockdown of CREB and NRF2 and treatment of AICAR (5-aminoimidazole-4-carboxamide 1-ß-D-ribofuranoside), an AMPK activator, attenuated the aggregation-inhibiting and neurite outgrowth promoting effects of LM-031 on TBP/Q79 SH-SY5Y cells. The study results suggest the LM-031 as potential therapeutics for SCA17 and probable other polyQ diseases.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Cromonas/farmacología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Proyección Neuronal/efectos de los fármacos , Péptidos/antagonistas & inhibidores , Ataxias Espinocerebelosas/tratamiento farmacológico , Ataxias Espinocerebelosas/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Chalconas/farmacología , Humanos , Péptidos/metabolismo , Ribonucleótidos/farmacología , Ataxias Espinocerebelosas/patología , Proteína de Unión a TATA-Box/metabolismo
12.
IUBMB Life ; 72(7): 1528-1536, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32320524

RESUMEN

In Huntington's disease, the length of the polyglutamine tract in the mutant protein correlates positively with the formation of aggregates and disease symptoms and severity of the disease. Some disease-modifying factors exist. However, no organized study has been carried out to investigate the effect of polyglutamine length in the mutant protein on the efficacy of a therapeutic strategy. We had shown earlier that the helical peptide arising out of the N-terminal stretch of normal huntingtin is able to inhibit aggregation of a number of proteins, including luciferase, α-synuclein, p53, and Rnq1. In this work, we show that polyglutamine stretches of differing lengths, namely 51Q, 72Q, and 103Q, form a mixture of aggregates at different rates, with the rate increasing in a polyQ length-dependent manner. The helical peptide is able to inhibit the rate of aggregation. The extent of inhibition was different when measuring either total aggregation or only fibrillar aggregates, suggesting that the helical peptide with benign polyQ stretch alters the aggregation landscape of different elongated polyQ lengths differently. Our results suggest that designing a therapeutic approach to inhibit protein aggregation must take note of polyQ length of the protein.


Asunto(s)
Proteína Huntingtina/metabolismo , Enfermedad de Huntington/patología , Proteínas Mutantes/metabolismo , Mutación , Fragmentos de Péptidos/farmacología , Péptidos/química , Agregación Patológica de Proteínas , Humanos , Proteína Huntingtina/genética , Enfermedad de Huntington/metabolismo , Proteínas Mutantes/genética , Péptidos/antagonistas & inhibidores
13.
Oncol Rep ; 43(3): 851-863, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32020213

RESUMEN

Efficient enrichment and transmembrane transport of cytotoxic reagents are considered to be effective strategies to increase the efficiency and selectivity of antitumor drugs targeting solid tumors. In the present study, a recombinant protein ABD­LDP­Ec consisting of the albumin­binding domain (ABD), the apoprotein (LDP) of lidamycin (LDM) and an EGFR­targeting oligopeptide (Ec) was prepared by DNA recombination and bacterial fermentation, and was integrated with the enediyne chromophore (AE) of lidamycin to generate its enediyne­integrated analogue ABD­LDP­Ec­AE. ABD­LDP­Ec exhibited high binding capacity to both albumin and EGFR­positive pancreatic cancer cells, and was internalized into the cytoplasm through receptor­mediated endocytosis and albumin­driven macropinocytosis of K­ras mutant cells. In animal experiments, ABD­LDP­Ec demonstrated notable selective distribution in pancreatic carcinoma xenografts by passive targeting of albumin captured in the blood and was retained in the tumor for 48 h. ABD­LDP­Ec and ABD­LDP­Ec­AE exhibited inhibitory activity in cell proliferation and AsPC­1 xenograft growth, and ABD­LDP­Ec­AE increased the tumor growth inhibition rate by 20% compared with natural LDM. The results indicated that the introduction of ABD­based multi­functional drug delivery may be an effective approach to improve the efficacy of antitumor drugs, especially for K­ras mutant cancers.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Péptidos/antagonistas & inhibidores , Proteínas Proto-Oncogénicas p21(ras)/genética , Albúminas/química , Albúminas/genética , Aminoglicósidos/química , Aminoglicósidos/genética , Aminoglicósidos/farmacología , Animales , Antineoplásicos/química , Línea Celular Tumoral , Enediinos/química , Enediinos/farmacología , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Xenoinjertos , Humanos , Ratones , Mutación/genética , Oligopéptidos/química , Oligopéptidos/genética , Oligopéptidos/farmacología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Péptidos/genética , Unión Proteica/efectos de los fármacos , Dominios Proteicos/genética , Proteínas Recombinantes/efectos de los fármacos , Proteínas Recombinantes/genética , Ensayos Antitumor por Modelo de Xenoinjerto
14.
J Autoimmun ; 108: 102402, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31980336

RESUMEN

We have previously identified a signature HLA-DR3 pocket variant, designated HLA-DRß1-Arg74 that confers a high risk for Graves' Disease (GD). In view of the key role of HLA-DRß1-Arg74 in triggering GD we hypothesized that thyroid-stimulating hormone receptor (TSHR) peptides that bind to the HLA-DRß1-Arg74 pocket with high affinity represent key pathogenic TSHR peptides triggering GD, and that blocking their presentation to CD4+ T-cells can be used as a novel therapeutic approach in GD. There were several previous attempts to identify the major pathogenic TSHR peptide utilizing different methodologies, however the results were inconsistent and inconclusive. Therefore, the aim of our study was to use TSHR peptide binding affinity to HLA-DRß1-Arg74 as a method to identify the key pathogenic TSHR peptides that trigger GD. Using virtual screening and ELISA and cellular binding assays we identified 2 TSHR peptides that bound with high affinity to HLA-DRß1-Arg74 - TSHR.132 and TSHR.197. Peptide immunization studies in humanized DR3 mice showed that only TSHR.132, but not TSHR.197, induced autoreactive T-cell proliferation and cytokine responses. Next, we induced experimental autoimmune Graves' disease (EAGD) in a novel BALB/c-DR3 humanized mouse model we created and confirmed TSHR.132 as a major DRß1-Arg74 binding peptide triggering GD in our mouse model. Furthermore, we demonstrated that Cepharanthine, a compound we have previously identified as DRß1-Arg74 blocker, could block the presentation and T-cell responses to TSHR.132 in the EAGD model.


Asunto(s)
Presentación de Antígeno/efectos de los fármacos , Presentación de Antígeno/inmunología , Bencilisoquinolinas/farmacología , Antígeno HLA-DR3/inmunología , Péptidos/antagonistas & inhibidores , Péptidos/inmunología , Receptores de Tirotropina/inmunología , Secuencia de Aminoácidos , Animales , Bencilisoquinolinas/química , Mapeo Epitopo/métodos , Epítopos de Linfocito T/inmunología , Citometría de Flujo , Enfermedad de Graves/diagnóstico , Enfermedad de Graves/tratamiento farmacológico , Enfermedad de Graves/inmunología , Antígeno HLA-DR3/genética , Humanos , Inmunohistoquímica , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Ratones Transgénicos , Modelos Moleculares , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/inmunología , Péptidos/química , Unión Proteica , Receptores de Tirotropina/química , Relación Estructura-Actividad , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/metabolismo
15.
Biochemistry ; 59(3): 290-296, 2020 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-31702899

RESUMEN

Amyloid-ß (Aß) oligomers are well-known toxic molecular species associated with Alzheimer's disease. Recent discoveries of the ability of amyloid fibril surfaces to convert soluble proteins into toxic oligomers suggested that these surfaces could serve as therapeutic targets for intervention. We have shown previously that a short helical peptide could be a key structural motif that can specifically recognize the K16-E22 region of the Aß40 fibril surface with an affinity at the level of several micromolar. Here, we demonstrate that in-tether chiral center-induced helical stabilized peptides could also recognize the fibril surfaces, effectively inhibiting the surface-mediated oligomerization of Aß40. Moreover, through extensive computational sampling, we observed two distinct ways in which the peptide inhibitors recognize the fibril surface. Apart from a binding mode that, in accord with the original design, involves hydrophobic side chains at the binding interface, we observed much more frequently another binding mode in which the hydrophobic staple interacts directly with the fibril surface. The affinity of the peptides for the fibril surface could be adjusted by tuning the hydrophobicity of the staple. The best candidate investigated here exhibits a submicromolar affinity (∼0.75 µM). Collectively, this work opens an avenue for the rational design of candidate drugs with stapled peptides for amyloid-related disease.


Asunto(s)
Péptidos beta-Amiloides/antagonistas & inhibidores , Amiloide/efectos de los fármacos , Péptidos/química , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Amiloide/química , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/uso terapéutico , Humanos , Interacciones Hidrofóbicas e Hidrofílicas/efectos de los fármacos , Simulación de Dinámica Molecular , Fragmentos de Péptidos/química , Péptidos/antagonistas & inhibidores , Péptidos/uso terapéutico , Multimerización de Proteína/efectos de los fármacos
16.
Nanoscale ; 12(1): 115-129, 2020 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-31773115

RESUMEN

Real-time imaging of single virus particles allows the visualization of subtle dynamic events of virus-host interaction. During the human immunodeficiency virus (HIV) infection of resting CD4 T lymphocytes, overcoming cortical actin restriction is an essential step, but the dynamic process and mechanism remain to be characterized. Herein, by using quantum dot (QD) encapsulated fluorescent viral particles and single-virus tracking, we explored detailed scenarios of HIV dynamic entry and crossing the cortical actin barrier. The fine-scale temporal and spatial processes of single HIV virion interaction with the cortical actin were studied in depth during virus entry via plasma membrane fusion. Individual HIV virions modulate the subtle rearrangement of the cortical actin barrier to open a door to facilitate viral entry. The actin-binding protein, α-actinin, was found to be critical for actin dynamics during HIV entry. An α-actinin-derived peptide, actin-binding site 1 peptide (ABS1p), was developed to block HIV infection. Our findings reveal an α-actinin-mediated dynamic cortical actin rearrangement for HIV entry, and identify an antiviral target as well as a corresponding peptide inhibitor based on HIV interaction with the actin cytoskeleton.


Asunto(s)
Actinas/metabolismo , VIH-1/fisiología , Citoesqueleto de Actina/efectos de los fármacos , Antivirales/química , Antivirales/farmacología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/virología , Colorantes Fluorescentes/química , Infecciones por VIH/patología , Infecciones por VIH/virología , VIH-1/química , Humanos , Microscopía Fluorescente , Péptidos/antagonistas & inhibidores , Péptidos/síntesis química , Péptidos/metabolismo , Puntos Cuánticos/química , Imagen de Lapso de Tiempo , Virión/química , Virión/fisiología , Internalización del Virus/efectos de los fármacos
17.
Elife ; 82019 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-31774395

RESUMEN

In area CA1 of the hippocampus, the selection of place cells to represent a new environment is biased towards neurons with higher excitability. However, different environments are represented by orthogonal cell ensembles, suggesting that regulatory mechanisms exist. Activity-dependent plasticity of intrinsic excitability, as observed in vitro, is an attractive candidate. Here, using whole-cell patch-clamp recordings of CA1 pyramidal neurons in anesthetized rats, we have examined how inducing theta-bursts of action potentials affects their intrinsic excitability over time. We observed a long-lasting, homeostatic depression of intrinsic excitability which commenced within minutes, and, in contrast to in vitro observations, was not mediated by dendritic Ih. Instead, it was attenuated by the Kv1.1 channel blocker dendrotoxin K, suggesting an axonal origin. Analysis of place cells' out-of-field firing in mice navigating in virtual reality further revealed an experience-dependent reduction consistent with decreased excitability. We propose that this mechanism could reduce memory interference.


Asunto(s)
Región CA1 Hipocampal/fisiología , Homeostasis/fisiología , Canal de Potasio Kv.1.1/metabolismo , Plasticidad Neuronal/fisiología , Células Piramidales/fisiología , Potenciales de Acción/fisiología , Animales , Axones/metabolismo , Quelantes del Calcio/farmacología , Dendritas/fisiología , Electrofisiología , Hipocampo/fisiología , Canal de Potasio Kv.1.1/efectos de los fármacos , Masculino , Ratones , Neuronas/fisiología , Técnicas de Placa-Clamp , Péptidos/antagonistas & inhibidores , Ratas , Ratas Wistar
18.
J Am Heart Assoc ; 8(22): e013673, 2019 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-31698979

RESUMEN

Background We recently discovered a small endogenous peptide, peptide Lv, with the ability to activate vascular endothelial growth factor receptor 2 and its downstream signaling. As vascular endothelial growth factor through vascular endothelial growth factor receptor 2 contributes to normal development, vasodilation, angiogenesis, and pathogenesis of various diseases, we investigated the role of peptide Lv in vasodilation and developmental and pathological angiogenesis in this study. Methods and Results The endothelial cell proliferation, migration, and 3-dimensional sprouting assays were used to test the abilities of peptide Lv in angiogenesis in vitro. The chick chorioallantoic membranes and early postnatal mice were used to examine its impact on developmental angiogenesis. The oxygen-induced retinopathy and laser-induced choroidal neovascularization mouse models were used for in vivo pathological angiogenesis. The isolated porcine retinal and coronary arterioles were used for vasodilation assays. Peptide Lv elicited angiogenesis in vitro and in vivo. Peptide Lv and vascular endothelial growth factor acted synergistically in promoting endothelial cell proliferation. Peptide Lv-elicited vasodilation was not completely dependent on nitric oxide, indicating that peptide Lv had vascular endothelial growth factor receptor 2/nitric oxide-independent targets. An antibody against peptide Lv, anti-Lv, dampened vascular endothelial growth factor-elicited endothelial proliferation and laser-induced vascular leakage and choroidal neovascularization. While the pathological angiogenesis in mouse eyes with oxygen-induced retinopathy was enhanced by exogenous peptide Lv, anti-Lv dampened this process. Furthermore, deletion of peptide Lv in mice significantly decreased pathological neovascularization compared with their wild-type littermates. Conclusions These results demonstrate that peptide Lv plays a significant role in pathological angiogenesis but may be less critical during development. Peptide Lv is involved in pathological angiogenesis through vascular endothelial growth factor receptor 2-dependent and -independent pathways. As anti-Lv dampened the pathological angiogenesis in the eye, anti-Lv may have a therapeutic potential to treat pathological angiogenesis.


Asunto(s)
Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Membrana Corioalantoides/efectos de los fármacos , Neovascularización Patológica/genética , Péptidos/genética , Péptidos/farmacología , Vasos Retinianos/efectos de los fármacos , Animales , Arteriolas/efectos de los fármacos , Ensayos de Migración Celular , Proliferación Celular/genética , Embrión de Pollo , Membrana Corioalantoides/irrigación sanguínea , Neovascularización Coroidal/genética , Neovascularización Coroidal/metabolismo , Vasos Coronarios/efectos de los fármacos , Retinopatía Diabética/genética , Retinopatía Diabética/metabolismo , Modelos Animales de Enfermedad , Perros , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Noqueados , Neovascularización Patológica/metabolismo , Péptidos/antagonistas & inhibidores , Péptidos/metabolismo , Arteria Retiniana/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sus scrofa , Porcinos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
19.
Sci Rep ; 9(1): 16872, 2019 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-31728006

RESUMEN

CAG repeats RNA causes various fatal neurodegenerative diseases exemplified by Huntington's disease (HD) and several spinocerebellar ataxias (SCAs). Although there are differences in the pathogenic mechanisms, these diseases share the common cause, i.e., expansion of CAG repeats. The shared cause of these diseases raises the possibility for the exploiting the common target as a potential therapeutic approach. Oligonucleotide-based therapeutics are designed earlier with the help of the base pairing rule but are not very promiscuous, considering the nonspecific stimulation of the immune system and the poor cellular delivery. Therefore, small molecules-based therapeutics are preferred for targeting the repeats expansion disorders. Here, we have used the chemical similarity search approach to discern the small molecules that selectively target toxic CAG RNA. The lead compounds showed the specificity towards AA mismatch in biophysical studies including CD, ITC, and NMR spectroscopy and thus aided to forestall the polyQ mediated pathogenicity. Furthermore, the lead compounds also explicitly alleviate the polyQ mediated toxicity in HD cell models and patient-derived cells. These findings suggest that the lead compound could act as a chemical probe for AA mismatch containing RNA as well as plays a neuroprotective role in fatal neurodegenerative diseases like HD and SCAs.


Asunto(s)
Fibroblastos/efectos de los fármacos , Flavonoides/farmacología , Fármacos Neuroprotectores/farmacología , Péptidos/antagonistas & inhibidores , ARN/química , Bibliotecas de Moléculas Pequeñas/farmacología , Benzotiazoles/química , Bioensayo , Supervivencia Celular/efectos de los fármacos , Descubrimiento de Drogas , Fibroblastos/metabolismo , Fibroblastos/patología , Flavonoides/química , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Simulación del Acoplamiento Molecular , Fármacos Neuroprotectores/química , Conformación de Ácido Nucleico , Péptidos/química , Péptidos/metabolismo , Cultivo Primario de Células , Agregado de Proteínas/efectos de los fármacos , ARN/genética , ARN/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Expansión de Repetición de Trinucleótido/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...