Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 202
Filtrar
1.
J Chem Inf Model ; 63(15): 4924-4933, 2023 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-37466559

RESUMEN

The development of drugs targeting somatostatin receptor 2 (SSTR2), generally overexpressed in neuroendocrine tumors, is focus of intense research. A few molecules in conjugation with radionuclides are in clinical use for both diagnostic and therapeutic purposes. These radiopharmaceuticals are composed of a somatostatin analogue biovector conjugated to a chelator moiety bearing the radionuclide. To date, despite valuable efforts, a detailed molecular-level description of the interaction of radiopharmaceuticals in complex with SSTR2 has not yet been accomplished. Therefore, in this work, we carefully analyzed the key dynamical features and detailed molecular interactions of SSTR2 in complex with six radiopharmaceutical compounds selected among the few already in use (64Cu/68Ga-DOTATATE, 68Ga-DOTATOC, 64Cu-SARTATE) and some in clinical development (68Ga-DOTANOC, 64Cu-TETATATE). Through molecular dynamics simulations and exploiting recently available structures of SSTR2, we explored the influence of the different portions of the compounds (peptide, radionuclide, and chelator) in the interaction with the receptor. We identified the most stable binding modes and found distinct interaction patterns characterizing the six compounds. We thus unveiled detailed molecular interactions crucial for the recognition of this class of radiopharmaceuticals. The microscopically well-founded analysis presented in this study provides guidelines for the design of new potent ligands targeting SSTR2.


Asunto(s)
Radiofármacos , Receptores de Somatostatina , Radiofármacos/química , Simulación de Dinámica Molecular , Receptores de Somatostatina/antagonistas & inhibidores , Receptores de Somatostatina/química , Animales , Ratones , Diseño de Fármacos , Terapia Molecular Dirigida
2.
Prog Mol Biol Transl Sci ; 196: 43-57, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36813365

RESUMEN

Somatostatin (SRIF) is a neuropeptide that acts as an important regulator of both endocrine and exocrine secretion and modulates neurotransmission in the central nervous system (CNS). SRIF also regulates cell proliferation in normal tissues and tumors. The physiological actions of SRIF are mediated by a family of five G protein-coupled receptors, called somatostatin receptor (SST) SST1, SST2, SST3, SST4, SST5. These five receptors share similar molecular structure and signaling pathways but they display marked differences in their anatomical distribution, subcellular localization and intracellular trafficking. The SST subtypes are widely distributed in the CNS and peripheral nervous system, in many endocrine glands and tumors, particularly of neuroendocrine origin. In this review, we focus on the agonist-dependent internalization and recycling of the different SST subtypes in vivo in the CNS, peripheral organs and tumors. We also discuss the physiological, pathophysiological and potential therapeutic effects of the intracellular trafficking of SST subtypes.


Asunto(s)
Neoplasias , Receptores de Somatostatina , Humanos , Receptores de Somatostatina/química , Receptores de Somatostatina/metabolismo , Encéfalo/metabolismo
3.
Nat Chem Biol ; 19(2): 133-140, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36138141

RESUMEN

G protein-coupled receptors (GPCRs) modulate every aspect of physiological functions mainly through activating heterotrimeric G proteins. A majority of GPCRs promiscuously couple to multiple G protein subtypes. Here we validate that in addition to the well-known Gi/o pathway, somatostatin receptor 2 and 5 (SSTR2 and SSTR5) couple to the Gq/11 pathway and show that smaller ligands preferentially activate the Gi/o pathway. We further determined cryo-electron microscopy structures of the SSTR2‒Go and SSTR2‒Gq complexes bound to octreotide and SST-14. Structural and functional analysis revealed that G protein selectivity of SSTRs is not only determined by structural elements in the receptor-G protein interface, but also by the conformation of the agonist-binding pocket. Accordingly, smaller ligands fail to stabilize a broader agonist-binding pocket of SSTRs that is required for efficient Gq/11 coupling but not Gi/o coupling. Our studies facilitate the design of drugs with selective G protein signaling to improve therapeutic efficacy.


Asunto(s)
Receptores de Somatostatina , Somatostatina , Receptores de Somatostatina/química , Receptores de Somatostatina/metabolismo , Somatostatina/metabolismo , Microscopía por Crioelectrón , Transducción de Señal , Proteínas de Unión al GTP/metabolismo
4.
Cells ; 11(2)2022 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-35053382

RESUMEN

Somatostatin receptors (SSTRs) are G protein-coupled receptors (GPCRs) known to regulate exocrine secretion, neurotransmission, and inhibit endogenous cell proliferation. SSTR subtypes (SSTR1-SSTR5) exhibit homo- or heterodimerization with unique signaling characteristics. Melanocortin receptor accessory protein 1 (MRAP1) functions as an allosteric modulator of melanocortin receptors and some other GPCRs. In this study, we investigated the differential interaction of MRAP1 and SSTRs and examined the pharmacological modulation of MRAP1 on mouse SSTR2/SSTR3 and SSTR2/SSTR5 heterodimerization in vitro. Our results show that the mouse SSTR2 forms heterodimers with SSTR3 and SSTR5 and that MRAP1 selectively interacts with SSTR3 and SSTR5 but not SSTR2. The interactive binding sites of SSTR2/SSTR3 or SSTR2/SSTR5 with MRAP1 locate on SSTR3 and SSTR5 but not SSTR2. The binding sites of MRAP1 to SSTR3 are extensive, while the ones of SSTR5 are restricted on transmembrane region six and seven. The heterodimerization of mouse SSTR2, SSTR3, and SSTR5 can be modulated by binding protein in addition to an agonist. Upregulation of extracellular signal-regulated kinases phosphorylation, p27Kip1, and increased cell growth inhibition with the co-expression of SSTR2/SSTR3 or SSTR2/SSTR5 with MRAP1 suggest a regulatory effect of MRAP1 on anti-proliferative response of two SSTR heterodimers. Taken together, these results provide a new insight of MRAP1 on the maintenance and regulation of mouse SSTR dimers which might be helpful to better understand the molecular mechanism involving SSTRs in tumor biology or other human disorders.


Asunto(s)
Proteínas de la Membrana/metabolismo , Receptores de Somatostatina/metabolismo , Secuencia de Aminoácidos , Animales , Apoptosis , Células HEK293 , Humanos , Sistema de Señalización de MAP Quinasas , Proteínas de la Membrana/química , Ratones , Unión Proteica , Multimerización de Proteína , Empalme del ARN/genética , Receptores de Somatostatina/química , Homología de Secuencia de Aminoácido
5.
Proteins ; 90(3): 704-719, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34661304

RESUMEN

In the past two decades, the structural biology studies on G-protein coupled receptors (GPCRs) are on the rise. Understanding the relation between the structure and function of GPCRs is important as they play a huge role in various signaling mechanisms in a eukaryotic cell. Somatostatin receptor 3 (SSTR3), one of the GPCRs, is one such important receptor which oversees different cellular processes including cell-to-cell signaling. However, the information available regarding the structural features of SSTR3 responsible for their bioactivity is scarce. In this study, we report a structural understanding of SSTR3-ligand binding that could be helpful in demystifying the structural complexities related to functioning of the receptor. An integrated protocol consisting of different computational structural biology tools including protein structure prediction via comparative modeling, binding site characterization, three-dimensional quantitative structure-activity relationship based on comparative molecular field analysis and comparative molecular similarity indices analysis, density functional theory, and molecular dynamics simulations were performed. Different understandings from the simulation of SSTR3-ligand complexes, mainly the conditions that are favorable for the formation of lowest bioactive state of SSTR3 ligands are reported. In addition to that, we report the important physicochemical descriptors of SSTR3 ligands that could significantly influence their bioactivity. The results of the study could be helpful in developing novel SSTR3 ligands (both agonists and antagonists) with high potency and receptor selectivity.


Asunto(s)
Aminas/química , Lípidos/química , Receptores de Somatostatina/química , Sitios de Unión , Bases de Datos de Compuestos Químicos , Teoría Funcional de la Densidad , Diseño de Fármacos , Humanos , Ligandos , Simulación de Dinámica Molecular , Unión Proteica , Conformación Proteica , Relación Estructura-Actividad Cuantitativa
6.
Int J Biol Macromol ; 185: 562-571, 2021 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-34216658

RESUMEN

The exopolysaccharide preparation of Bacillus amyloliquefaciens amy-1 (EPS) regulates glycemic levels and promotes glucagon-like peptide 1 (GLP-1) secretion in vivo and in vitro. This study aimed to identify the molecular mechanism underlying EPS-induced GLP-1 secretion. HEK293T cells stably expressing human Gα-gustducin were used as a heterologous system for expressing the genes of human bitter taste receptor (T2R) 10, 14, 30, 38 (PAV), 38 (AVI), 43, and 46, which were expressed as recombinant proteins with an N-terminal tag composed of a Lucy peptide and a human somatostatin receptor subtype 3 fragment for membrane targeting and a C-terminal red fluorescent protein for expression monitoring. EPS induced a dose-dependent calcium response from the human NCI-H716 enteroendocrine cell line revealed by fluorescent calcium imaging, but inhibitors of the G protein-coupled receptor pathway suppressed the response. EPS activated heterologously expressed T2R14 and T2R38 (PAV). shRNAs of T2R14 effectively inhibited EPS-induced calcium response and GLP-1 secretion in NCI-H716 cells, suggesting the involvement of T2R14 in these effects. The involvement of T2R38 was not characterized because NCI-H716 cells express T2R38 (AVI). In conclusion, the activation of T2Rs mediates EPS-induced GLP-1 secretion from enteroendocrine cells, and T2R14 is a critical target activated by EPS in these cells.


Asunto(s)
Bacillus amyloliquefaciens/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Polisacáridos Bacterianos/farmacología , Receptores Acoplados a Proteínas G/genética , Calcio/metabolismo , Línea Celular , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Somatostatina/química , Receptores de Somatostatina/genética
7.
Front Endocrinol (Lausanne) ; 12: 646210, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33790868

RESUMEN

Epithelial-mesenchymal transition (EMT) is a dynamic process by which epithelial cells loss their phenotype and acquire mesenchymal traits, including increased migratory and invasive capacities. EMT is involved in physiological processes, such as embryogenesis and wound healing, and in pathological processes such as cancer, playing a pivotal role in tumor progression and metastasis. Pituitary tumors, although typically benign, can be locally invasive. Different studies have shown the association of EMT with increased tumor size and invasion in pituitary tumors, and in particular with a poor response to Somatostatin Receptor Ligands (SRLs) treatment in GH-producing pituitary tumors, the main cause of acromegaly. This review will summarize the current knowledge regarding EMT and SRLs resistance in acromegaly and, based on this relation, will suggest new biomarkers and possible therapies to SRLs resistant tumors.


Asunto(s)
Acromegalia/tratamiento farmacológico , Acromegalia/patología , Resistencia a Medicamentos , Neoplasias de las Glándulas Endocrinas/tratamiento farmacológico , Transición Epitelial-Mesenquimal/efectos de los fármacos , Adenoma Hipofisario Secretor de Hormona del Crecimiento/tratamiento farmacológico , Ligandos , Receptores de Somatostatina/química , Acromegalia/etiología , Biomarcadores/metabolismo , Cadherinas/biosíntesis , Citoesqueleto/efectos de los fármacos , Humanos , Fenotipo , Neoplasias Hipofisarias/tratamiento farmacológico , Somatostatina/metabolismo
8.
Sci Rep ; 11(1): 7677, 2021 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-33828200

RESUMEN

Somatostatin receptor 2 (SSTR2) is a G-protein coupled receptor (GPCR) that controls numerous cellular processes including cell-to-cell signaling. In this study, we report how the lipid and ligand molecules influence the conformational dynamics of the membrane-bound SSTR2. Molecular simulations of different holo and apoenzyme complexes of SSTR2 in the presence and absence of a lipid bilayer were performed, observed, and correlated with previously reported studies. We identified the important SSTR2 residues that take part in the formation of the SSTR2-ligand complex. On analyzing the molecular simulation trajectories, we identified that the residue D3.32 is crucial in determining the bioactive conformation of SSTR2 ligands in the binding site. Based on the results, we suggest that designing a novel SSTR2 ligand with an H-bond donor group at the R1 position, and hydrophobic groups at R2 and R3 might have higher activity and SSTR2-selectivity. We analyzed the simulated systems to identify other important structural features involved in SSTR2-ligand binding and to observe the different conformational changes that occur in the protein after the ligand binding. Additionally, we studied the conformational dynamics of N- and C-terminal regions of SSTR2 in the presence and absence of the lipid bilayer. Both the systems were compared to understand the influence of lipid molecules in the formation of secondary structural domains by these extracellular regions. The comparative study revealed that the secondary structural elements formed by C-terminal residues in presence of lipid molecules is crucial for the functioning of SSTR2. Our study results highlight the structural complexities involved in the functioning of SSTR upon binding with the ligands in the presence and absence of lipid bilayer, which is essential for designing novel drug targets.


Asunto(s)
Modelos Moleculares , Receptores de Somatostatina/química , Enlace de Hidrógeno , Ligandos , Membrana Dobles de Lípidos/química , Conformación Proteica
9.
Biochem Soc Trans ; 49(1): 79-91, 2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33599752

RESUMEN

Primary cilia are hair-like projections of the cell membrane supported by an inner microtubule scaffold, the axoneme, which polymerizes out of a membrane-docked centriole at the ciliary base. By working as specialized signaling compartments, primary cilia provide an optimal environment for many G protein-coupled receptors (GPCRs) and their effectors to efficiently transmit their signals to the rest of the cell. For this to occur, however, all necessary receptors and signal transducers must first accumulate at the ciliary membrane. Serotonin receptor 6 (HTR6) and Somatostatin receptor 3 (SSTR3) are two GPCRs whose signaling in brain neuronal cilia affects cognition and is implicated in psychiatric, neurodegenerative, and oncologic diseases. Over a decade ago, the third intracellular loops (IC3s) of HTR6 and SSTR3 were shown to contain ciliary localization sequences (CLSs) that, when grafted onto non-ciliary GPCRs, could drive their ciliary accumulation. Nevertheless, these CLSs were dispensable for ciliary targeting of HTR6 and SSTR3, suggesting the presence of additional CLSs, which we have recently identified in their C-terminal tails. Herein, we review the discovery and mapping of these CLSs, as well as the state of the art regarding how these CLSs may orchestrate ciliary accumulation of these GPCRs by controlling when and where they interact with the ciliary entry and exit machinery via adaptors such as TULP3, RABL2 and the BBSome.


Asunto(s)
Cilios/metabolismo , Receptores de Serotonina/metabolismo , Receptores de Somatostatina/metabolismo , Animales , Humanos , Dominios y Motivos de Interacción de Proteínas/fisiología , Señales de Clasificación de Proteína/genética , Transporte de Proteínas/fisiología , Receptores de Serotonina/química , Receptores de Serotonina/genética , Receptores de Somatostatina/química , Receptores de Somatostatina/genética
10.
Chembiochem ; 22(7): 1307-1315, 2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33238069

RESUMEN

Multimodal imaging probes have attracted the interest of ongoing research, for example, for the surgical removal of tumors. Modular synthesis approaches allow the construction of hybrid probes consisting of a radiotracer, a fluorophore and a targeting unit. We present the synthesis of a new asymmetric bifunctional cyanine dye that can be used as a structural and functional linker for the construction of such hybrid probes. 68 Ga-DOTATATE, a well-characterized radiopeptide targeting the overexpressed somatostatin receptor subtype 2 (SSTR2) in neuroendocrine tumors, was labeled with our cyanine dye, thus providing additional information along with the data obtained from the radiotracer. We tested the SSTR2-targeting and imaging properties of the resulting probe 68 Ga-DOTA-ICC-TATE in vitro and in a tumor xenograft mouse model. Despite the close proximity between dye and pharmacophore, we observed a high binding affinity towards SSTR2 as well as elevated uptake in SSTR2-overexpressing tumors in the positron emission tomography (PET) scan and histological examination.


Asunto(s)
Carbocianinas/química , Colorantes Fluorescentes/química , Receptores de Somatostatina/metabolismo , Somatostatina/química , Animales , Línea Celular Tumoral , Colorantes Fluorescentes/síntesis química , Humanos , Ratones , Ratones Desnudos , Tumores Neuroendocrinos/diagnóstico por imagen , Tumores Neuroendocrinos/metabolismo , Octreótido/análogos & derivados , Octreótido/química , Compuestos Organometálicos/química , Péptidos/química , Péptidos/metabolismo , Tomografía de Emisión de Positrones , Radiofármacos/química , Radiofármacos/metabolismo , Receptores de Somatostatina/química , Trasplante Heterólogo
11.
Clin Cancer Res ; 27(5): 1399-1409, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33355247

RESUMEN

PURPOSE: The goal of this study was to analyze the role of somatostatin receptor type 2 (SSTR2) as a molecular target for the imaging and treatment of thyroid cancer through analysis of SSTR2 expression and its epigenetic modulation and testing tumor uptake of different radiolabeled SSTR2 analogues. EXPERIMENTAL DESIGN: We analyzed SSTR2 expression by immunostaining of 92 thyroid cancer tissue samples and quantified standard uptake values (SUVmax) of SSTR2 analogue, 68Ga-DOTA-TATE, by PET/CT imaging in 25 patients with metastatic thyroid cancer. We utilized human thyroid cancer cell lines characterized by differential SSTR2 expression (TT, BCPAP, and FTC133) and rat pancreatic cell line (AR42J) with intrinsically high SSTR2 expression for functional in vitro studies. SSTR2-high (AR42J) and SSTR2-low (FTC133) xenograft mouse models were used to test the uptake of radiolabeled SSTR2 analogues and their therapeutic efficacy in vivo. RESULTS: Thyroid cancer had a higher SSTR2 expression than normal thyroid. Hurthle cell thyroid cancer was characterized by the highest 68Ga-DOTA-TATE uptake [median SUVmax, 16.5 (7.9-29)] than other types of thyroid cancers. In vivo studies demonstrated that radiolabeled DOTA-EB-TATE is characterized by significantly higher tumor uptake than DOTA-TATE (P < 0.001) and DOTA-JR11 (P < 0.001). Treatment with 177Lu-DOTA-EB-TATE extended survival and reduced tumor size in a mouse model characterized by high somatostatin (SST) analogues uptake (SUVmax, 15.16 ± 4.34), but had no effects in a model with low SST analogues uptake (SUVmax, 4.8 ± 0.27). CONCLUSIONS: A novel SST analogue, 177Lu-DOTA-EB-TATE, has the potential to be translated from bench to bedside for the targeted therapy of patients characterized by high uptake of SST analogues in metastatic lesions.


Asunto(s)
Radiofármacos/administración & dosificación , Receptores de Somatostatina/metabolismo , Somatostatina/análogos & derivados , Neoplasias de la Tiroides/tratamiento farmacológico , Adulto , Animales , Apoptosis , Proliferación Celular , Femenino , Humanos , Ratones , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Persona de Mediana Edad , Metástasis de la Neoplasia , Tomografía Computarizada por Tomografía de Emisión de Positrones , Pronóstico , Radiofármacos/metabolismo , Receptores de Somatostatina/química , Somatostatina/administración & dosificación , Somatostatina/metabolismo , Neoplasias de la Tiroides/diagnóstico por imagen , Neoplasias de la Tiroides/metabolismo , Neoplasias de la Tiroides/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Life Sci Alliance ; 4(3)2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33372037

RESUMEN

G protein-coupled receptors (GPCRs) are the most common pharmacological target in human clinical practice. To perform their functions, many GPCRs must accumulate inside primary cilia, microtubule-based plasma membrane protrusions working as cellular antennae. Nevertheless, the molecular mechanisms underlying GPCR ciliary targeting remain poorly understood. Serotonin receptor 6 (HTR6) and somatostatin receptor 3 (SSTR3) are two brain-enriched ciliary GPCRs involved in cognition and pathologies such as Alzheimer's disease and cancer. Although the third intracellular loops (IC3) of HTR6 and SSTR3 suffice to target non-ciliary GPCRs to cilia, these IC3s are dispensable for ciliary targeting of HTR6 and SSTR3 themselves, suggesting these GPCRs contain additional ciliary targeting sequences (CTSs). Herein, we discover and characterize novel CTSs in HTR6 and SSTR3 C-terminal tails (CT). These CT-CTSs (CTS2) act redundantly with IC3-CTSs (CTS1), each being sufficient for ciliary targeting. In HTR6, RKQ and LPG motifs are critical for CTS1 and CTS2 function, respectively, whereas in SSTR3 these roles are mostly fulfilled by AP[AS]CQ motifs in IC3 and juxtamembrane residues in CT. Furthermore, we shed light on how these CTSs promote ciliary targeting by modulating binding to ciliary trafficking adapters TULP3 and RABL2.


Asunto(s)
Membrana Celular/metabolismo , Cilios/metabolismo , Receptores de Serotonina/química , Receptores de Serotonina/metabolismo , Receptores de Somatostatina/química , Receptores de Somatostatina/metabolismo , Transducción de Señal/genética , Secuencia de Aminoácidos , Animales , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Dominios y Motivos de Interacción de Proteínas , Transporte de Proteínas/genética , Transfección
13.
Neuropharmacology ; 178: 108198, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32739276

RESUMEN

We provided strong proof of concept evidence that somatostatin mediates potent analgesic and anti-inflammatory actions via its receptor subtype 4 (sst4) located both at the periphery and the central nervous system. Therefore, sst4 agonists are promising novel drug candidates for neuropathic pain and neurogenic inflammation, but rational drug design was not possible due to the lack of knowledge about its 3-dimensional structure. We modeled the sst4 receptor structure, described its agonist binding properties, and characterized the binding of our novel small molecule sst4 agonists (4-phenetylamino-7H-pyrrolo[2,3-d]pyrimidine derivatives) using an in silico platform. In addition to the in silico binding data, somatostatin displacement by Compound 1 was demonstrated in the competitive binding assay on sst4-expressing cells. In vivo effects were investigated in rat models of neurogenic inflammation and chronic traumatic neuropathic pain. We defined high- and low-affinity binding pockets of sst4 for our ligands, binding of the highest affinity compounds were similar to that of the reference ligand J-2156. We showed potent G-protein activation with the highest potency of 10 nM EC50 value and highest efficacy of 342%. Oral administration of 100 µg/kg of 5 compounds significantly inhibited acute neurogenic plasma protein extravasation in the paw skin by 40-60%, one candidate abolished and 3 others diminished sciatic nerve-ligation induced neuropathic hyperalgesia by 28-62%. The in silico predictions on sst4-ligands were tested in biological systems. Low oral dose of our novel agonists inhibit neurogenic inflammation and neuropathic pain, which opens promising drug developmental perspectives for these unmet medical need conditions.


Asunto(s)
Analgésicos/farmacología , Antiinflamatorios/farmacología , Receptores de Somatostatina/agonistas , Secuencia de Aminoácidos , Analgésicos/química , Analgésicos/metabolismo , Animales , Antiinflamatorios/química , Antiinflamatorios/metabolismo , Células CHO , Cricetinae , Cricetulus , Relación Dosis-Respuesta a Droga , Femenino , Células HEK293 , Humanos , Masculino , Dimensión del Dolor/efectos de los fármacos , Dimensión del Dolor/métodos , Unión Proteica/fisiología , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Ratas , Ratas Wistar , Receptores de Somatostatina/química , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo
14.
Trends Pharmacol Sci ; 40(10): 790-800, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31519459

RESUMEN

There are over 420 human solute carrier (SLC) transporters from 65 families that are expressed ubiquitously in the body. The SLCs mediate the movement of ions, drugs, and metabolites across membranes and their dysfunction has been associated with a variety of diseases, such as diabetes, cancer, and central nervous system (CNS) disorders. Thus, SLCs are emerging as important targets for therapeutic intervention. Recent technological advances in experimental and computational biology allow better characterization of SLC pharmacology. Here we describe recent approaches to modulate SLC transporter function, with an emphasis on the use of computational approaches and computer-aided drug design (CADD) to study nutrient transporters. Finally, we discuss future perspectives in the rational design of SLC drugs.


Asunto(s)
Diseño de Fármacos , Proteínas Transportadoras de Solutos/química , Sistema de Transporte de Aminoácidos ASC/química , Sistema de Transporte de Aminoácidos ASC/metabolismo , Humanos , Ligandos , Antígenos de Histocompatibilidad Menor/química , Antígenos de Histocompatibilidad Menor/metabolismo , Modelos Moleculares , Receptores de Somatostatina/química , Receptores de Somatostatina/metabolismo , Proteínas Transportadoras de Solutos/metabolismo , Relación Estructura-Actividad
15.
ChemMedChem ; 14(19): 1727-1734, 2019 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-31403246

RESUMEN

Targeted drug delivery (TDD) is an efficient strategy for cancer treatment. However, the real-time monitoring of drug delivery is still challenging because of a pronounced lack of TDD systems capable of providing a near-infrared (NIR) fluorescence signal for the detection of drug-release events. Herein, a new TDD system, comprising a turn-on NIR fluorescent reporter attached to an anticancer drug and targeting peptide, is reported. This system provides both TDD and NIR fluorescence monitoring of drug-release events in target tissue. In this TDD system, a new carboxy-derivatized xanthene-cyanine (XCy) dye is attached to an anticancer drug, chlorambucil (CLB), through a hydrolytically cleavable ester linker and coupled to a targeting peptide, octreotide amide (OCTA), which is specific to somatostatin receptors SSTR-2 and STTR-5 overexpressed on many tumor cells. This OCTA-G-XCy-CLB (G: γ-aminobutyric acid) conjugate exhibits no detectable fluorescence, whereas, upon the hydrolytic cleavage of the ester linker, a bright NIR fluorescence appears at λ≈710 nm; this signals release of the drug. Real-time TDD monitoring is demonstrated for the example of the human pancreatic cancer cell line overexpressing SSTR-2 and STTR-5, in comparison with the noncancerous Chinese hamster ovary cell line, which contains a reduced number of these receptors.


Asunto(s)
Carbocianinas/química , Clorambucilo/química , Portadores de Fármacos/química , Fluorenos/química , Octreótido/metabolismo , Resinas Sintéticas/química , Xantenos/química , Aminobutiratos/química , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Células CHO , Carbocianinas/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Clorambucilo/farmacología , Cricetulus , Portadores de Fármacos/metabolismo , Colorantes Fluorescentes/química , Colorantes Fluorescentes/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Terapia Molecular Dirigida/métodos , Receptores de Somatostatina/química , Receptores de Somatostatina/genética , Xantenos/metabolismo
16.
Int J Mol Sci ; 20(16)2019 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-31412614

RESUMEN

Endogenous somatostatin shows anti-secretory effects in both physiological and pathological settings, as well as inhibitory activity on cell growth. Since somatostatin is not suitable for clinical practice, researchers developed synthetic somatostatin receptor ligands (SRLs) to overcome this limitation. Currently, SRLs represent pivotal tools in the treatment algorithm of neuroendocrine tumors (NETs). Octreotide and lanreotide are the first-generation SRLs developed and show a preferential binding affinity to somatostatin receptor (SST) subtype 2, while pasireotide, which is a second-generation SRL, has high affinity for multiple SSTs (SST5 > SST2 > SST3 > SST1). A number of studies demonstrated that first-generation and second-generation SRLs show distinct functional properties, besides the mere receptor affinity. Therefore, the aim of the present review is to critically review the current evidence on the biological effects of SRLs in pituitary adenomas and neuroendocrine tumors, by mainly focusing on the differences between first-generation and second-generation ligands.


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Tumores Neuroendocrinos/tratamiento farmacológico , Tumores Neuroendocrinos/metabolismo , Receptores de Somatostatina/metabolismo , Animales , Estudios Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Humanos , Ligandos , Tumores Neuroendocrinos/etiología , Tumores Neuroendocrinos/patología , Neoplasias Hipofisarias/tratamiento farmacológico , Neoplasias Hipofisarias/etiología , Neoplasias Hipofisarias/metabolismo , Neoplasias Hipofisarias/patología , Unión Proteica , Multimerización de Proteína , Receptores de Somatostatina/química , Transducción de Señal , Resultado del Tratamiento
17.
Nanoscale ; 11(30): 14400-14409, 2019 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-31334537

RESUMEN

Somatostatin receptor subtype 2 (SSTR2) is highly expressed in pulmonary neuroendocrine tumors, which account for approximately 25% of all lung cancers including small-cell lung cancer (SCLC). It is possible to establish SCLC-specific imaging agents for multimodal imaging to obtain tumor integrity information. Herein, we constructed novel multifunctional organic melanin nanoparticles (MNPs) as a carrier and surface-loaded somatostatin analog octreotide to produce a human small-cell lung cancer-targeted nanoprobe OCT-PEG-MNPs. MNPs have an excellent photoacoustic imaging (PAI) function and can be directly chelated with the magnetic resonance contrast agent Mn2+, and N-bromo succinimide (NBS) can be used as an oxidant to label the nanoparticles with the long half-life radionuclide 124I by an electrophilic substitution reaction. Therefore, (124I, Mn) OCT-PEG-MNPs can not only be used for PAI but also be used for positron emission tomography (PET) and magnetic resonance imaging (MRI). The NCI-H69 SCLC tumor xenograft model with high SSTR2 expression was constructed to evaluate the multimodal imaging ability of (124I, Mn) OCT-PEG-MNPs. This nanoprobe showed good imaging abilities in PAI, MRI and PET. The PA images showed that the photoacoustic signal in the NCI-H69 tumor site gradually increased with time, and the NCI-H69 xenograft showed a clear increase in the T1-weighted signal intensity after injection of Mn-OCT-PEG-MNPs at 24 h compared to that in the prescan. MicroPET and biodistribution studies showed that the uptake of NCI-H69 tumors (8.03 ± 0.37% ID g-1) was significantly higher than that in the control A549 model (3.35 ± 0.54% ID g-1) after injection of (124I, Mn) OCT-PEG-MNPs at 24 h. The (124I, Mn) OCT-PEG-MNPs were successfully applied to multimodal imaging in a small-cell lung cancer model with high SSTR2 expression. This nanoprobe may be considered for clinical trials since it combines the numerous advantages of organic nanoparticles.


Asunto(s)
Carcinoma de Células Pequeñas/diagnóstico , Neoplasias Pulmonares/diagnóstico , Melaninas/química , Nanopartículas/química , Receptores de Somatostatina/química , Animales , Carcinoma de Células Pequeñas/diagnóstico por imagen , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Radioisótopos de Yodo/química , Neoplasias Pulmonares/diagnóstico por imagen , Imagen por Resonancia Magnética , Manganeso/química , Melaninas/metabolismo , Ratones , Ratones Desnudos , Nanopartículas/toxicidad , Tomografía de Emisión de Positrones , Radiofármacos/química , Receptores de Somatostatina/metabolismo , Trasplante Heterólogo
18.
Amino Acids ; 51(9): 1247-1257, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31350614

RESUMEN

Somatostatin (SST) is an endogenous cyclic tetradecapeptide hormone that exerts multiple biological activities via a family of five receptors. BIM-23052 (DC-23-99) D-Phe-Phe-Phe-D-Trp-Lys-Thr-Phe-Thr-NH2 is a linear SST analog with established in vitro GH-inhibitory activity and high affinity to sstr5, sstr3 and sstr2. The different SSTR subtypes are expressed in different tissues and in some tumor cells. Based on this finding, a series of new analogs of BIM-23052 with expected antitumor activity have been synthesized. The Thr at position 6 in BIM-23052 was replaced by the conformationally hindered Tle, Aib, Ac5c and Ac6c of the new analogs. The peptides were synthesized by standard solid-phase peptide chemistry methods, Fmoc strategy. The cytotoxic effects of the compounds were tested in vitro against a panel of tumor cell lines: HT-29, MDA-MB-23, Hep-G2, HeLa and the normal human diploid cell line Lep-3. All five somatostatin receptor subtypes were modeled and docking was performed to determine the binding affinity of the analogs. The new peptides exhibited different concentration-dependent antiproliferative effect on the tumor cell lines after 24 h of treatment. The compound 3B (Aib6) demonstrated the most pronounced antiproliferative effects on HepG-2 cells with the IC50 = 0.01349 nM. Docking confirmed that all compounds bind well to SST receptors with preference to sstr3 and sstr5, which is most probably the reason for the observed biological effects.


Asunto(s)
Aminoácidos/química , Antineoplásicos/química , Somatostatina/análogos & derivados , Aminoácidos Cíclicos/química , Ácidos Aminoisobutíricos/química , Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Línea Celular Tumoral , Técnicas de Química Sintética , Ácidos Ciclohexanocarboxílicos/química , Cicloleucina/química , Células HT29 , Células HeLa , Humanos , Simulación del Acoplamiento Molecular , Péptidos/síntesis química , Péptidos/farmacología , Receptores de Somatostatina/química , Somatostatina/química , Somatostatina/farmacología , Relación Estructura-Actividad
19.
Endocr Relat Cancer ; 26(6): 585-599, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30939452

RESUMEN

Control of symptoms related to hormonal hypersecretion by functioning neuroendocrine tumors (NETs) is challenging. New therapeutic options are required. Since novel in vitro tumor models seem to better mimic the tumor in vivo conditions, we aimed to study the effect of somatostatin and dopamine receptor agonists (octreotide and cabergoline, respectively) and novel somatostatin-dopamine chimeric multi-receptor drugs (BIM-065, BIM-23A760) using 2D (monolayer) and 3D (spheroids) cultures. Dose-response studies in 2D and 3D human pancreatic NET cell cultures (BON-1 and QGP-1) were performed under serum-containing and serum-deprived conditions. Cell proliferation, somatostatin and dopamine receptor expression (SSTs and D2R), apoptosis, lactate dehydrogenase, as well as serotonin and chromogranin A (CgA) release were assessed. The following results were obtained. 3D cultures of BON-1/QGP-1 allowed better cell survival than 2D cultures in serum-deprived conditions. SSTs and D2R mRNA levels were higher in the 3D model vs 2D model. Octreotide/cabergoline/BIM-065/BIM-23A760 treatment did not affect cell growth or spheroid size. In BON-1 2D-cultures, only BIM-23A760 significantly inhibited CgA release -this effect being more pronounced in 3D cultures. In BON-1 2D cultures, cabergoline/BIM-065/BIM-23A760 treatment decreased serotonin release (maximal effect up to 40%), being this effect again more potent in 3D cultures (up to 67% inhibition; with BIM-23A760 having the most potent effects). In QGP-1, cabergoline/BIM-065 treatment decreased serotonin release only in the 3D model. In conclusion, cultures of NET 3D spheroids represent a promising method for evaluating cell proliferation and secretion in NET cell-line models. Compared to 2D models, 3D models grow relatively serum independent. In 3D model, SST-D2R multi-receptor targeting drugs inhibit CgA and serotonin secretion, but not NET cell growth.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Proliferación Celular/efectos de los fármacos , Cromogranina A/metabolismo , Tumores Neuroendocrinos/patología , Receptores Dopaminérgicos/metabolismo , Receptores de Somatostatina/metabolismo , Serotonina/metabolismo , Antineoplásicos Hormonales/farmacología , Cabergolina/farmacología , Dopamina/análogos & derivados , Dopamina/farmacología , Agonistas de Dopamina/farmacología , Humanos , Tumores Neuroendocrinos/tratamiento farmacológico , Tumores Neuroendocrinos/metabolismo , Octreótido/farmacología , Receptores Dopaminérgicos/química , Receptores de Somatostatina/química , Somatostatina/análogos & derivados , Somatostatina/farmacología , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...