Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Nat Commun ; 15(1): 4687, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38824166

RESUMEN

Ligand-induced activation of G protein-coupled receptors (GPCRs) can initiate signaling through multiple distinct pathways with differing biological and physiological outcomes. There is intense interest in understanding how variation in GPCR ligand structure can be used to promote pathway selective signaling ("biased agonism") with the goal of promoting desirable responses and avoiding deleterious side effects. Here we present an approach in which a conventional peptide ligand for the type 1 parathyroid hormone receptor (PTHR1) is converted from an agonist which induces signaling through all relevant pathways to a compound that is highly selective for a single pathway. This is achieved not through variation in the core structure of the agonist, but rather by linking it to a nanobody tethering agent that binds with high affinity to a separate site on the receptor not involved in signal transduction. The resulting conjugate represents the most biased agonist of PTHR1 reported to date. This approach holds promise for facile generation of pathway selective ligands for other GPCRs.


Asunto(s)
Receptor de Hormona Paratiroídea Tipo 1 , Receptores Acoplados a Proteínas G , Transducción de Señal , Anticuerpos de Dominio Único , Ligandos , Humanos , Receptor de Hormona Paratiroídea Tipo 1/metabolismo , Receptor de Hormona Paratiroídea Tipo 1/agonistas , Anticuerpos de Dominio Único/metabolismo , Anticuerpos de Dominio Único/farmacología , Células HEK293 , Transducción de Señal/efectos de los fármacos , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Unión Proteica , Animales , Péptidos/química , Péptidos/farmacología , Péptidos/metabolismo
2.
Curr Opin Chem Biol ; 80: 102443, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38503199

RESUMEN

New applications for biomolecules demand novel approaches for their synthesis and modification. Traditional methods for modifying proteins and cells using non-specific labeling chemistry are insufficiently precise to rigorously interrogate the mechanistic biological and physiological questions at the forefront of biomedical science. Site-specific catalytic modification of proteins promises to meet these challenges. Here, we describe recent applications of the enzyme sortase A in facilitating precise biomolecule labeling. We focus on describing new chemistries to broaden the scope of sortase-mediated labeling (sortagging), the development of new probes for imaging via enzymatic labeling, and the modulation of biological systems using probes and reactions mediated by sortase.


Asunto(s)
Aminoaciltransferasas , Proteínas Bacterianas , Cisteína Endopeptidasas , Aminoaciltransferasas/metabolismo , Cisteína Endopeptidasas/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/química , Humanos , Coloración y Etiquetado/métodos , Proteínas/metabolismo , Proteínas/química , Animales
3.
Protein Sci ; 33(2): e4866, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38088474

RESUMEN

Antibodies have proven highly valuable for therapeutic development; however, they are typically poor candidates for applications that require activation of G protein-coupled receptors (GPCRs), the largest collection of targets for clinically approved drugs. Nanobodies (Nbs), the smallest antibody fragments retaining full antigen-binding capacity, have emerged as promising tools for pharmacologic applications, including GPCR modulation. Past work has shown that conjugation of Nbs with ligands can provide GPCR agonists that exhibit improved activity and selectivity compared to their parent ligands. The neurokinin-1 receptor (NK1R), a GPCR targeted for the treatment of pain, is activated by peptide agonists such as Substance P (SP) and neurokinin A (NKA), which induce signaling through multiple pathways (Gs , Gq and ß-arrestin). In this study, we investigated whether conjugating NK1R ligands with Nbs that bind to a separate location on the receptor would provide chimeric compounds with distinctive signaling properties. We employed sortase A-mediated ligation to generate several conjugates consisting of Nbs linked to NK1R ligands. Many of these conjugates exhibited divergent and unexpected signaling properties and transcriptional outputs. For example, some Nb-NKA conjugates showed enhanced receptor binding capacity, high potency partial agonism, prolonged cAMP production, and an increase in transcriptional output associated with Gs signaling; whereas other conjugates were virtually inactive. Nanobody conjugation caused only minor alterations in ligand-induced upstream Gq signaling with unexpected enhancements in transcriptional (downstream) responses. Our findings underscore the potential of nanobody conjugation for providing compounds with advantageous properties such as biased agonism, prolonged duration of action, and enhanced transcriptional responses. These compounds hold promise not only for facilitating fundamental research on GPCR signal transduction mechanisms but also for the development of more potent and enduring therapeutics.


Asunto(s)
Receptores Acoplados a Proteínas G , Transducción de Señal , Ligandos , Receptores Acoplados a Proteínas G/metabolismo , beta-Arrestinas/metabolismo , Péptidos/metabolismo
4.
Commun Biol ; 6(1): 599, 2023 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-37268817

RESUMEN

The parathyroid hormone receptor type 1 (PTH1R) is a G protein-coupled receptor that plays key roles in regulating calcium homeostasis and skeletal development via binding the ligands, PTH and PTH-related protein (PTHrP), respectively. Eiken syndrome is a rare disease of delayed bone mineralization caused by homozygous PTH1R mutations. Of the three mutations identified so far, R485X, truncates the PTH1R C-terminal tail, while E35K and Y134S alter residues in the receptor's amino-terminal extracellular domain. Here, using a variety of cell-based assays, we show that R485X increases the receptor's basal rate of cAMP signaling and decreases its capacity to recruit ß-arrestin2 upon ligand stimulation. The E35K and Y134S mutations each weaken the binding of PTHrP leading to impaired ß-arrestin2 recruitment and desensitization of cAMP signaling response to PTHrP but not PTH. Our findings support a critical role for interaction with ß-arrestin in the mechanism by which the PTH1R regulates bone formation.


Asunto(s)
Proteína Relacionada con la Hormona Paratiroidea , Receptor de Hormona Paratiroídea Tipo 1 , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Receptor de Hormona Paratiroídea Tipo 1/genética , Receptor de Hormona Paratiroídea Tipo 1/química , Receptor de Hormona Paratiroídea Tipo 1/metabolismo , Hormona Paratiroidea/metabolismo , Transducción de Señal/fisiología , Receptores Acoplados a Proteínas G
5.
J Nucl Med ; 64(7): 1017-1023, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36997331

RESUMEN

Primary liver cancer is the third leading cause of cancer-related deaths, and its incidence and mortality are increasing worldwide. Hepatocellular carcinoma (HCC) accounts for 80% of primary liver cancer cases. Glypican-3 (GPC3) is a heparan sulfate proteoglycan that histopathologically defines HCC and represents an attractive tumor-selective marker for radiopharmaceutical imaging and therapy for this disease. Single-domain antibodies are a promising scaffold for imaging because of their favorable pharmacokinetic properties, good tumor penetration, and renal clearance. Although conventional lysine-directed bioconjugation can be used to yield conjugates for radiolabeling full-length antibodies, this stochastic approach risks negatively affecting target binding of the smaller single-domain antibodies. To address this challenge, site-specific approaches have been explored. Here, we used conventional and sortase-based site-specific conjugation methods to engineer GPC3-specific human single-domain antibody (HN3) PET probes. Methods: Bifunctional deferoxamine (DFO) isothiocyanate was used to synthesize native HN3 (nHN3)-DFO. Site-specifically modified HN3 (ssHN3)-DFO was engineered using sortase-mediated conjugation of triglycine-DFO chelator and HN3 containing an LPETG C-terminal tag. Both conjugates were radiolabeled with 89Zr, and their binding affinity in vitro and target engagement of GPC3-positive (GPC3+) tumors in vivo were determined. Results: Both 89Zr-ssHN3 and 89Zr-nHN3 displayed nanomolar affinity for GPC3 in vitro. Biodistribution and PET/CT image analysis in mice bearing isogenic A431 and A431-GPC3+ xenografts, as well as in HepG2 liver cancer xenografts, showed that both conjugates specifically identify GPC3+ tumors. 89Zr-ssHN3 exhibited more favorable biodistribution and pharmacokinetic properties, including higher tumor uptake and lower liver accumulation. Comparative PET/CT studies on mice imaged with both 18F-FDG and 89Zr-ssHN3 showed more consistent tumor accumulation for the single-domain antibody conjugate, further establishing its potential for PET imaging. Conclusion: 89Zr-ssHN3 showed clear advantages in tumor uptake and tumor-to-liver signal ratio over the conventionally modified 89Zr-nHN3 in xenograft models. Our results establish the potential of HN3-based single-domain antibody probes for GPC3-directed PET imaging of liver cancers.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Anticuerpos de Dominio Único , Humanos , Animales , Ratones , Neoplasias Hepáticas/diagnóstico por imagen , Carcinoma Hepatocelular/diagnóstico por imagen , Radioisótopos/química , Glipicanos/química , Tomografía Computarizada por Tomografía de Emisión de Positrones , Anticuerpos Monoclonales/química , Distribución Tisular , Línea Celular Tumoral , Tomografía de Emisión de Positrones/métodos , Circonio/química
6.
Bioconjug Chem ; 33(10): 1867-1875, 2022 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-36107739

RESUMEN

Synthetic molecules that form a covalent bond upon binding to a targeted biomolecule (proximity-induced reactivity) are the subject of intense biomedical interest for the unique pharmacological properties imparted by irreversible binding. However, off-target covalent labeling and the lack of molecules with sufficient specificity limit more widespread applications. We describe the first example of a cross-linking platform that uses a synthetic peptide epitope and a single domain antibody (or nanobody) pair to form a covalent linkage rapidly and specifically. The rate of the cross-linking reaction between peptide and nanobody is faster than most other biocompatible cross-linking reactions, and it can be used to label live cells expressing receptor-nanobody fusions. The rapid kinetics of this system allowed us to probe the consequences on signaling for ligand cross-linking to the A2A-adenosine receptor. Our method may be generally useful to site-specifically link synthetic molecules to receptors on mammalian cell surfaces.


Asunto(s)
Proteínas de la Membrana , Anticuerpos de Dominio Único , Animales , Ligandos , Epítopos , Péptidos/química , Mamíferos
7.
ACS Chem Biol ; 17(8): 2296-2303, 2022 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-35930411

RESUMEN

Peptide epitope tags offer a valuable means for detection and manipulation of protein targets for which high quality detection reagents are not available. Most commonly used epitope tags are bound by conventional, full-size antibodies (Abs). The complex architecture of Abs complicates their application in protein engineering and intracellular applications. To address these shortcomings, single domain antibodies (nanobodies, Nbs) that recognize short peptide epitopes have become increasingly prized. Here, we characterize the interaction between a Nb (Nb6E) and a 14-mer peptide epitope. We identify residues in the peptide epitope essential for high affinity binding. Using this information in combination with computational modeling we propose a mode of interaction between Nb6E and this epitope. We apply this nanobody-epitope pair to augment the potency of a ligand at an engineered adenosine A2A receptor. This characterization of the nanobody-epitope pair opens the door to diverse applications including mechanistic studies of the G protein-coupled receptor function.


Asunto(s)
Anticuerpos de Dominio Único , Anticuerpos , Epítopos/química , Péptidos/química , Ingeniería de Proteínas , Anticuerpos de Dominio Único/química
8.
J Immunol ; 207(5): 1468-1477, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34408009

RESUMEN

Immuno-positron emission tomography (PET), a noninvasive imaging modality, can provide a dynamic approach for longitudinal assessment of cell populations of interest. Transformation of mAbs into single-chain variable fragment (scFv)-based PET imaging agents would allow noninvasive tracking in vivo of a wide range of possible targets. We used sortase-mediated enzymatic labeling in combination with PEGylation to develop an anti-mouse CD4 scFv-based PET imaging agent constructed from an anti-mouse CD4 mAb. This anti-CD4 scFv can monitor the in vivo distribution of CD4+ T cells by immuno-PET. We tracked CD4+ and CD8+ T cells in wild-type mice, in immunodeficient recipients reconstituted with monoclonal populations of OT-II and OT-I T cells, and in a B16 melanoma model. Anti-CD4 and -CD8 immuno-PET showed that the persistence of both CD4+ and CD8+ T cells transferred into immunodeficient mice improved when recipients were immunized with OVA in CFA. In tumor-bearing animals, infiltration of both CD4+ and CD8+ T cells increased as the tumor grew. The approach described in this study should be readily applicable to convert clinically useful Abs into the corresponding scFv PET imaging agents.


Asunto(s)
Antígenos CD4/inmunología , Linfocitos T CD4-Positivos/inmunología , Inmunoterapia/métodos , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma/terapia , Monitorización Inmunológica/métodos , Neoplasias Cutáneas/terapia , Animales , Anticuerpos Monoclonales/metabolismo , Diagnóstico por Imagen , Femenino , Memoria Inmunológica , Melanoma Experimental , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Tomografía de Emisión de Positrones , Anticuerpos de Cadena Única/metabolismo
9.
J Am Chem Soc ; 143(15): 5958-5966, 2021 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-33825470

RESUMEN

The lower respiratory tract infections affecting children worldwide are in large part caused by the parainfluenza viruses (HPIVs), particularly HPIV3, along with human metapneumovirus and respiratory syncytial virus, enveloped negative-strand RNA viruses. There are no vaccines for these important human pathogens, and existing treatments have limited or no efficacy. Infection by HPIV is initiated by viral glycoprotein-mediated fusion between viral and host cell membranes. A viral fusion protein (F), once activated in proximity to a target cell, undergoes a series of conformational changes that first extend the trimer subunits to allow insertion of the hydrophobic domains into the target cell membrane and then refold the trimer into a stable postfusion state, driving the merger of the viral and host cell membranes. Lipopeptides derived from the C-terminal heptad repeat (HRC) domain of HPIV3 F inhibit infection by interfering with the structural transitions of the trimeric F assembly. Clinical application of this strategy, however, requires improving the in vivo stability of antiviral peptides. We show that the HRC peptide backbone can be modified via partial replacement of α-amino acid residues with ß-amino acid residues to generate α/ß-peptides that retain antiviral activity but are poor protease substrates. Relative to a conventional α-lipopeptide, our best α/ß-lipopeptide exhibits improved persistence in vivo and improved anti-HPIV3 antiviral activity in animals.


Asunto(s)
Lipopéptidos/farmacología , Virus de la Parainfluenza 3 Humana/efectos de los fármacos , Infecciones del Sistema Respiratorio/patología , Secuencia de Aminoácidos , Aminoácidos/química , Aminoácidos/metabolismo , Animales , Antivirales/química , Antivirales/metabolismo , Antivirales/farmacología , Línea Celular , Colesterol/química , Diseño de Fármacos , Humanos , Lipopéptidos/química , Lipopéptidos/metabolismo , Virus de la Parainfluenza 3 Humana/aislamiento & purificación , Multimerización de Proteína , Ratas , Infecciones del Sistema Respiratorio/virología , Distribución Tisular , Temperatura de Transición , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/metabolismo , Internalización del Virus/efectos de los fármacos
10.
Nat Methods ; 17(10): 1025-1032, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32929269

RESUMEN

The immune system's ability to recognize peptides on major histocompatibility molecules contributes to the eradication of cancers and pathogens. Tracking these responses in vivo could help evaluate the efficacy of immune interventions and improve mechanistic understanding of immune responses. For this purpose, we employ synTacs, which are dimeric major histocompatibility molecule scaffolds of defined composition. SynTacs, when labeled with positron-emitting isotopes, can noninvasively image antigen-specific CD8+ T cells in vivo. Using radiolabeled synTacs loaded with the appropriate peptides, we imaged human papillomavirus-specific CD8+ T cells by positron emission tomography in mice bearing human papillomavirus-positive tumors, as well as influenza A virus-specific CD8+ T cells in the lungs of influenza A virus-infected mice. It is thus possible to visualize antigen-specific CD8+ T-cell populations in vivo, which may serve prognostic and diagnostic roles.


Asunto(s)
Linfocitos T CD8-positivos/fisiología , Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/virología , Papillomaviridae/inmunología , Tomografía de Emisión de Positrones/métodos , Animales , Antígenos , Clonación Molecular , Epítopos/genética , Epítopos/metabolismo , Femenino , Regulación de la Expresión Génica/inmunología , Antígenos de Histocompatibilidad Clase I/clasificación , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Inmunoglobulina G/clasificación , Inmunoglobulina G/inmunología , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/inmunología
11.
Nat Commun ; 11(1): 2087, 2020 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-32350260

RESUMEN

Antibodies conjugated to bioactive compounds allow targeted delivery of therapeutics to cell types of choice based on that antibody's specificity. Here we develop a new type of conjugate that consists of a nanobody and a peptidic ligand for a G protein-coupled receptor (GPCR), fused via their C-termini. We address activation of parathyroid hormone receptor-1 (PTHR1) and improve the signaling activity and specificity of otherwise poorly active N-terminal peptide fragments of PTH by conjugating them to nanobodies (VHHs) that recognize PTHR1. These C-to-C conjugates show biological activity superior to that of the parent fragment peptide in vitro. In an exploratory experiment in mice, a VHH-PTH peptide conjugate showed biological activity, whereas the corresponding free peptide did not. The lead conjugate also possesses selectivity for PTHR1 superior to that of PTH(1-34). This design approach, dubbed "conjugation of ligands and antibodies for membrane proteins" (CLAMP), can yield ligands with high potency and specificity.


Asunto(s)
Receptores Acoplados a Proteínas G/metabolismo , Anticuerpos de Dominio Único/metabolismo , Secuencia de Aminoácidos , Animales , Femenino , Células HEK293 , Humanos , Ligandos , Ratones , Modelos Moleculares , Hormona Paratiroidea/metabolismo , Péptidos/química , Péptidos/metabolismo , Unión Proteica , Receptor de Hormona Paratiroídea Tipo 1/metabolismo
12.
Mol Immunol ; 114: 513-523, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31518855

RESUMEN

A substantial fraction of eukaryotic proteins is folded and modified in the endoplasmic reticulum (ER) prior to export and secretion. Proteins that enter the ER but fail to fold correctly must be degraded, mostly in a process termed ER-associated degradation (ERAD). Both protein folding in the ER and ERAD are essential for proper immune function. Several E2 and E3 enzymes localize to the ER and are essential for various aspects of ERAD, but their functions and regulation are incompletely understood. Here we identify and characterize single domain antibody fragments derived from the variable domain of alpaca heavy chain-only antibodies (VHHs or nanobodies) that bind to the ER-localized E2 UBC6e, an enzyme implicated in ERAD. One such VHH, VHH05 recognizes a 14 residue stretch and enhances the rate of E1-catalyzed ubiquitin E2 loading in vitroand interferes with phosphorylation of UBC6e in response to cell stress. Identification of the peptide epitope recognized by VHH05 places it outside the E2 catalytic core, close to the position of activation-induced phosphorylation on Ser184. Our data thus suggests a site involved in allosteric regulation of UBC6e's activity. This VHH should be useful not only to dissect the participation of UBC6e in ERAD and in response to cell stress, but also as a high affinity epitope tag-specific reagent of more general utility.


Asunto(s)
Epítopos/inmunología , Péptidos/inmunología , Anticuerpos de Dominio Único/inmunología , Enzimas Ubiquitina-Conjugadoras/inmunología , Anticuerpos/inmunología , Línea Celular Tumoral , Células Cultivadas , Degradación Asociada con el Retículo Endoplásmico/inmunología , Células HeLa , Humanos , Células K562 , Fosforilación/inmunología , Ubiquitina/inmunología , Ubiquitina-Proteína Ligasas/inmunología
13.
J Immunol ; 203(6): 1619-1628, 2019 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-31391235

RESUMEN

Proteins are composed of α-amino acid residues. This consistency in backbone structure likely serves an important role in the display of an enormous diversity of peptides by class II MHC (MHC-II) products, which make contacts with main chain atoms of their peptide cargo. Peptides that contain residues with an extra carbon in the backbone (derived from ß-amino acids) have biological properties that differ starkly from those of their conventional counterparts. How changes in the structure of the peptide backbone affect the loading of peptides onto MHC-II or recognition of the resulting complexes by TCRs has not been widely explored. We prepared a library of analogues of MHC-II-binding peptides derived from OVA, in which at least one α-amino acid residue was replaced with a homologous ß-amino acid residue. The latter contain an extra methylene unit in the peptide backbone but retain the original side chain. We show that several of these α/ß-peptides retain the ability to bind tightly to MHC-II, activate TCR signaling, and induce responses from T cells in mice. One α/ß-peptide exhibited enhanced stability in the presence of an endosomal protease relative to the index peptide. Conjugation of this backbone-modified peptide to a camelid single-domain Ab fragment specific for MHC-II enhanced its biological activity. Our results suggest that backbone modification offers a method to modulate MHC binding and selectivity, T cell stimulatory capacity, and susceptibility to processing by proteases such as those found within endosomes where Ag processing occurs.


Asunto(s)
Aminoácidos/metabolismo , Antígenos de Histocompatibilidad Clase II/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión/fisiología , Línea Celular Tumoral , Genes MHC Clase II/fisiología , Ligandos , Ratones , Fragmentos de Péptidos/metabolismo , Unión Proteica/fisiología , Receptores de Antígenos de Linfocitos T/metabolismo , Relación Estructura-Actividad , Linfocitos T/metabolismo
14.
ACS Chem Biol ; 14(8): 1836-1844, 2019 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-31348637

RESUMEN

Commonly used methods to monitor internalization of cell surface structures involve application of fluorescently or otherwise labeled antibodies against the target of interest. Genetic modification of the protein of interest, for example through creation of fusions with fluorescent or enzymatically active protein domains, is another approach to follow trafficking behavior. The former approach requires indirect methods, such as multiple rounds of cell staining, to distinguish between a target that remains surface-disposed and an internalized and/or recycled species. The latter approach necessitates the creation of fusions whose behavior may not accurately reflect that of their unmodified counterparts. Here, we report a method for the characterization of protein internalization in real time through sortase-mediated, site-specific labeling of single-domain antibodies or viral proteins with a newly developed, cathepsin-sensitive quenched-fluorophore probe. Quenched probes of this type have been used to measure enzyme activity in complex environments and for different cell types, but not as a sensor of protein movement into living cells. This approach allows a quantitative assessment of the movement of proteins into protease-containing endosomes in real time in living cells. We demonstrate considerable variation in the rate of endosomal delivery for different cell surface receptors. We were also able to characterize the kinetics of influenza virus delivery to cathepsin-positive compartments, showing highly coordinated arrival in endosomal compartments. This approach should be useful for identifying proteins expressed on cells of interest for targeted endosomal delivery of payloads, such as antibody-drug conjugates or antigens that require processing.


Asunto(s)
Colorantes Fluorescentes/química , Virus de la Influenza A/fisiología , Proteínas de la Membrana/metabolismo , Péptidos/química , Rodaminas/química , Aminoaciltransferasas/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Línea Celular Tumoral , Cisteína Endopeptidasas/metabolismo , Perros , Endosomas/metabolismo , Células de Riñón Canino Madin Darby , Ratones , Microscopía Confocal/métodos , Microscopía Fluorescente/métodos , Transporte de Proteínas , Internalización del Virus
15.
Proc Natl Acad Sci U S A ; 115(49): 12383-12388, 2018 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-30442659

RESUMEN

Human parathyroid hormone (PTH) and N-terminal fragments thereof activate two receptors, hPTHR1 and hPTHR2, which share ∼51% sequence similarity. A peptide comprising the first 34 residues of PTH is fully active at both receptors and is used to treat osteoporosis. We have used this system to explore the hypothesis that backbone modification of a promiscuous peptidic agonist can provide novel receptor-selective agonists. We tested this hypothesis by preparing a set of variants of PTH(1-34)-NH2 that contained a single ß-amino-acid residue replacement at each of the first eight positions. These homologs, each containing one additional backbone methylene unit relative to PTH(1-34)-NH2 itself, displayed a wide range of potencies in cell-based assays for PTHR1 or PTHR2 activation. The ß-scan series allowed us to identify two homologs, each containing two αâ†’ß replacements, that were highly selective, one for PTHR1 and the other for PTHR2. These findings suggest that backbone modification of peptides may provide a general strategy for achieving activation selectivity among polypeptide-modulated receptors, and that success requires consideration of both ß2- and ß3-residues, which differ in terms of side-chain location.


Asunto(s)
Hormona Paratiroidea/química , Péptidos/síntesis química , Péptidos/farmacología , Receptor de Hormona Paratiroídea Tipo 1/agonistas , Receptor de Hormona Paratiroídea Tipo 2/agonistas , Secuencia de Aminoácidos , Células HEK293 , Humanos , Unión Proteica , Conformación Proteica
16.
Cancer Immunol Res ; 6(7): 870-880, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29792298

RESUMEN

High-risk human papillomavirus-associated cancers express viral oncoproteins (e.g., E6 and E7) that induce and maintain the malignant phenotype. The viral origin of these proteins makes them attractive targets for development of a therapeutic vaccine. Camelid-derived single-domain antibody fragments (nanobodies or VHHs) that recognize cell surface proteins on antigen-presenting cells (APC) can serve as targeted delivery vehicles for antigens attached to them. Such VHHs were shown to induce CD4+ and CD8+ T-cell responses against model antigens conjugated to them via sortase, but antitumor responses had not yet been investigated. Here, we tested the ability of an anti-CD11b VHH (VHHCD11b) to target APCs and serve as the basis for a therapeutic vaccine to induce CD8+ T-cell responses against HPV+ tumors. Mice immunized with VHHCD11b conjugated to an H-2Db-restricted immunodominant E7 epitope (E749-57) had more E7-specific CD8+ T cells compared with those immunized with E749-57 peptide alone. These CD8+ T cells acted prophylactically and conferred protection against a subsequent challenge with HPV E7-expressing tumor cells. In a therapeutic setting, VHHCD11b-E749-57 vaccination resulted in greater numbers of CD8+ tumor-infiltrating lymphocytes compared with mice receiving E749-57 peptide alone in HPV+ tumor-bearing mice, as measured by in vivo noninvasive VHH-based immune-positron emission tomography (immunoPET), which correlated with tumor regression and survival outcome. Together, these results demonstrate that VHHs can serve as a therapeutic cancer vaccine platform for HPV-induced cancers. Cancer Immunol Res; 6(7); 870-80. ©2018 AACR.


Asunto(s)
Antígenos/inmunología , Neoplasias/etiología , Papillomaviridae/inmunología , Infecciones por Papillomavirus/complicaciones , Infecciones por Papillomavirus/inmunología , Anticuerpos de Dominio Único/inmunología , Animales , Biomarcadores , Vacunas contra el Cáncer/inmunología , Modelos Animales de Enfermedad , Femenino , Inmunización , Inmunoterapia , Ratones , Ratones Noqueados , Modelos Biológicos , Neoplasias/diagnóstico , Neoplasias/metabolismo , Neoplasias/terapia , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/inmunología , Infecciones por Papillomavirus/virología , Tomografía de Emisión de Positrones , Unión Proteica
17.
J Med Chem ; 60(21): 8816-8833, 2017 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-29064243

RESUMEN

The parathyroid hormone receptor 1 (PTHR1) is a member of the B-family of GPCRs; these receptors are activated by long polypeptide hormones and constitute targets of drug development efforts. Parathyroid hormone (PTH, 84 residues) and PTH-related protein (PTHrP, 141 residues) are natural agonists of PTHR1, and an N-terminal fragment of PTH, PTH(1-34), is used clinically to treat osteoporosis. Conventional peptides in the 20-40-mer length range are rapidly degraded by proteases, which may limit their biomedical utility. We have used the PTHR1-ligand system to explore the impact of broadly distributed replacement of α-amino acid residues with ß-amino acid residues on susceptibility to proteolysis and agonist activity. This effort led us to identify new PTHR1 agonists that contain α → ß replacements throughout their sequences, manifest potent agonist activity in cellular assays, and display remarkable resistance to proteolysis, in cases remaining active after extended exposure to simulated gastric fluid. The strategy we have employed suggests a path toward identifying protease-resistant agonists of other B-family GPCRs.


Asunto(s)
Proteolisis/efectos de los fármacos , Receptor de Hormona Paratiroídea Tipo 1/agonistas , Aminoácidos/química , Aminoácidos/farmacología , Humanos , Ligandos , Hormona Paratiroidea/farmacología , Proteína Relacionada con la Hormona Paratiroidea/farmacología , Inhibidores de Proteasas/síntesis química
18.
Biosens Bioelectron ; 89(Pt 2): 789-794, 2017 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-27816596

RESUMEN

Current techniques to characterize leukocyte subgroups in blood require long sample preparation times and sizable sample volumes. A simplified method for leukocyte characterization using smaller blood volumes would thus be useful in diagnostic settings. Here we describe a flow system comprised of two functionalized graphene oxide (GO) surfaces that allow the capture of distinct leukocyte populations from small volumes blood using camelid single-domain antibodyfragments (VHHs) as capture agents. We used site-specifically labeled leukocytes to detect and identify cells exposed to fungal challenge. Combining the chemical and optical properties of GO with the versatility of the VHH scaffold in the context of a flow system provides a quick and efficient method for the capture and characterization of functional leukocytes.


Asunto(s)
Anticuerpos Inmovilizados/química , Candida albicans/aislamiento & purificación , Separación Celular/métodos , Grafito/química , Leucocitos/citología , Leucocitos/microbiología , Anticuerpos de Dominio Único/química , Aminoaciltransferasas/análisis , Animales , Proteínas Bacterianas/análisis , Candidiasis/sangre , Cisteína Endopeptidasas/análisis , Femenino , Citometría de Flujo , Ratones Endogámicos C57BL , Ratones Transgénicos , Nanoestructuras/química
19.
ACS Chem Biol ; 10(3): 844-54, 2015 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-25559929

RESUMEN

Oligomers that contain both α- and ß-amino acid residues, or "α/ß-peptides", have emerged as promising mimics of signal-bearing polypeptides that can inhibit or augment natural protein-protein interactions. α/ß-Peptides that contain a sufficient proportion of ß residues evenly distributed along the sequence can be highly resistant to enzymatic degradation, which is favorable with regard to in vivo applications. Little is known, however, about recognition of α/ß-peptides by the immune system. Prior studies have focused almost entirely on examples that contain a single ß residue; such α/ß-peptides frequently retain the immunological profile of the analogous α-peptide. We have conducted α-peptide vs α/ß-peptide comparisons involving higher ß residue content, focusing on molecules with αααß and ααßαααß backbone repeat patterns. Among analogues of an 18-mer derived from the Bim BH3 domain and an 8-mer derived from secreted phospholipase-2 (sPLA2), we find that recognition by antibodies raised against the prototype α-peptide is suppressed by periodic α → ß replacements. Complementary studies reveal that antibodies raised against Bim BH3- or sPLA2-derived α/ß-peptides fail to recognize prototype α-peptides displaying identical side chain repertoires. Because polypeptides containing d-α-amino acid residues are of growing interest for biomedical applications, we included the enantiomer of the sPLA2-derived α-peptide in these studies; this d-peptide is fully competent as a hapten, but the resulting antibodies do not cross react with the enantiomeric peptide. Among analogues of the 9-mer CD8(+) T-cell viral epitope GP33, we observe that periodic α → ß replacements suppress participation in the MHC I + peptide + T-cell receptor ternary complexes that activate cytotoxic T-lymphocytes, due in part to disruption of MHC binding.


Asunto(s)
Anticuerpos/química , Antígenos Virales/química , Proteínas Reguladoras de la Apoptosis/química , Epítopos/química , Proteínas de la Membrana/química , Oligopéptidos/química , Fosfolipasas A2/química , Proteínas Proto-Oncogénicas/química , Secuencia de Aminoácidos , Animales , Anticuerpos/inmunología , Anticuerpos/aislamiento & purificación , Especificidad de Anticuerpos , Antígenos Virales/inmunología , Proteínas Reguladoras de la Apoptosis/inmunología , Proteína 11 Similar a Bcl2 , Sitios de Unión , Pollos , Ensayo de Inmunoadsorción Enzimática , Epítopos/inmunología , Humanos , Cinética , Proteínas de la Membrana/inmunología , Modelos Moleculares , Datos de Secuencia Molecular , Oligopéptidos/síntesis química , Oligopéptidos/inmunología , Fosfolipasas A2/inmunología , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas/inmunología , Receptores de Antígenos de Linfocitos T/química , Receptores de Antígenos de Linfocitos T/inmunología , Estereoisomerismo , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/inmunología
20.
Nat Biotechnol ; 32(7): 653-5, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24929976

RESUMEN

Systematic modification of the backbone of bioactive polypeptides through ß-amino acid residue incorporation could provide a strategy for generating molecules with improved drug properties, but such alterations can result in lower receptor affinity and potency. Using an agonist of parathyroid hormone receptor-1 (PTHR1), a G protein-coupled receptor in the B-family, we present an approach for αâ†’ß residue replacement that enables both high activity and improved pharmacokinetic properties in vivo.


Asunto(s)
Hormonas Peptídicas/química , Hormonas Peptídicas/farmacocinética , Receptor de Hormona Paratiroídea Tipo 1/antagonistas & inhibidores , Sustitución de Aminoácidos , Aminoácidos/química , Aminoácidos/farmacocinética , Animales , Masculino , Tasa de Depuración Metabólica , Ratones Endogámicos C57BL , Hormonas Peptídicas/sangre , Relación Estructura-Actividad , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA