Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 453
Filtrar
1.
Biomed Pharmacother ; 179: 117389, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39243426

RESUMEN

An important goal in the opioid field is to discover effective analgesic drugs with minimal side effects. MCRT demonstrated potent antinociceptive effects with limited side effects, making it a promising candidate. However, its pharmacological properties and how it minimizes side effects remain unknown. Various mouse pain and opioid side effect models were used to evaluate the antinociceptive properties and safety at the spinal level. The targets of MCRT were identified through cAMP measurement, isolated tissue assays, and pharmacological experiments. Immunofluorescence was employed to visualize protein expression. MCRT displayed distinct antinociceptive effects between acute and chronic inflammatory pain models due to its multifunctional properties at the µ opioid receptor (MOR), µ-δ heterodimer (MDOR), and neuropeptide FF receptor 2 (NPFFR2). Activation of NPFFR2 reduced MOR-mediated antinociception, leading to bell-shaped response curves in acute pain models. However, activation of MDOR produced more effective antinociception in chronic inflammatory pain models. MCRT showed limited tolerance and opioid-induced hyperalgesia in both acute and chronic pain models and did not develop cross-tolerance to morphine. Additionally, MCRT did not exhibit addictive properties, gastrointestinal inhibition, and effects on motor coordination. Mechanistically, peripheral chronic inflammation or repeated administration of morphine and MCRT induced an increase in MDOR in the spinal cord. Chronic administration of MCRT had no apparent effect on microglial activation in the spinal cord. These findings suggest that MCRT is a versatile compound that provides potent antinociception with minimal opioid-related side effects. MDOR could be a promising target for managing chronic inflammatory pain and addressing the opioid crisis.


Asunto(s)
Analgésicos Opioides , Dolor Crónico , Modelos Animales de Enfermedad , Inflamación , Inyecciones Espinales , Receptores Opioides mu , Animales , Dolor Crónico/tratamiento farmacológico , Receptores Opioides mu/metabolismo , Ratones , Masculino , Inflamación/tratamiento farmacológico , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/efectos adversos , Analgésicos Opioides/farmacología , Receptores de Neuropéptido/metabolismo , Receptores de Neuropéptido/antagonistas & inhibidores , Receptores Opioides delta/metabolismo , Ratones Endogámicos C57BL , Analgésicos/farmacología , Analgésicos/administración & dosificación , Morfina/administración & dosificación , Morfina/farmacología , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Hiperalgesia/tratamiento farmacológico , Humanos , Oligopéptidos/administración & dosificación , Oligopéptidos/farmacología
2.
J Chem Inf Model ; 64(18): 7056-7067, 2024 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-39207455

RESUMEN

The growth hormone-releasing hormone receptor (GHRHR) belongs to Class B1 of G protein-coupled receptors (GPCRs). Class B1 GPCR peptides such, as growth hormone-releasing hormone (GHRH), have been proposed to bind in a two-step model, where first the C-terminal region of the peptide interacts with the extracellular domain of the receptor and, subsequently, the N-terminus interacts with the seven transmembrane domain of the receptor, resulting in activation. The GHRHR has recently been highlighted as a promising drug target toward several types of cancer and has been shown to be overexpressed in prostate, breast, pancreatic, and ovarian cancer. Indeed, peptide GHRHR antagonists have displayed promising results in many cancer models. However, no nonpeptide GHRHR-targeting compounds have yet been identified. We have utilized several computational tools to target GHRHR and identify potential small-molecule compounds directed at this receptor. These compounds were validated in vitro using a cyclic adenosine monophosphate (cAMP) ELISA to measure activity at the GHRHR. In vitro results suggest that several of the novel small-molecule compounds could inhibit GHRH-induced cAMP accumulation. Preliminary analysis of the specificity/selectivity of one of the most effective hit compounds indicated that the effect seen was via inhibition of the GHRHR. We therefore report the first nonpeptide antagonists of GHRHR and propose a structural basis for inhibition induced by the compounds, which may assist in the future design of lead GHRHR compounds for treating disorders attributed to dysregulated/aberrant GHRHR signaling.


Asunto(s)
Receptores de Neuropéptido , Receptores de Hormona Reguladora de Hormona Hipofisaria , Humanos , Receptores de Hormona Reguladora de Hormona Hipofisaria/metabolismo , Receptores de Hormona Reguladora de Hormona Hipofisaria/antagonistas & inhibidores , Receptores de Neuropéptido/antagonistas & inhibidores , Receptores de Neuropéptido/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Bibliotecas de Moléculas Pequeñas/química , Simulación del Acoplamiento Molecular , AMP Cíclico/metabolismo , Descubrimiento de Drogas
3.
Anal Chem ; 96(31): 12927-12935, 2024 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-39041225

RESUMEN

Mas-related G protein-coupled receptor X2 (MrgprX2) plays a crucial role in anaphylactoid reactions and allergic diseases. Some antagonists with reasonable potency and selectivity have been reported. Cell membrane chromatography (CMC) is effective for discovering ligands. Protein-tag-based CMC models (e.g., SNAP tags and HALO tags) have enhanced performance but also increased nonspecific adsorption of small molecules. The Avi tag, a short peptide sequence, binds biotin specifically via BirA catalysis. Our study showed that 2-iminobiotin (IB) can be a BirA substrate, enabling the development of a new cell membrane stationary phase (CMSP) based on the chemical properties (modifying carboxyl silica gel and specifically labeling the Avi tag) of IB. First, we constructed the MrgprX2-Avi-tag HEK293T cell line. Next, we synthesized IB-modified silica gel (SiO2-IB) stepwise. Finally, we immobilized Avi-tagged MrgprX2 cell membranes on SiO2-IB under BirA catalysis. We characterized the developed CMSP and used it to establish a MrgprX2-Avi-tag/CMC-HPLC/MS two-dimensional screening platform, successfully screening vitexicarpin fromViticis Fructus extract via a 2D/CMC platform. In vitro and in vivo experiments confirmed that vitexicarpin targets the MrgprX2 receptor, demonstrating antiallergic effects. Our IB-Avi tag-based CMC approach effectively decreased nonspecific adsorption of the screening materials. The Avi-tag-based 2D/CMC platform is suitable for screening potential drug candidates.


Asunto(s)
Membrana Celular , Receptores Acoplados a Proteínas G , Humanos , Receptores Acoplados a Proteínas G/metabolismo , Células HEK293 , Membrana Celular/metabolismo , Animales , Receptores de Neuropéptido/metabolismo , Receptores de Neuropéptido/antagonistas & inhibidores , Ratones , Cromatografía Líquida de Alta Presión , Proteínas del Tejido Nervioso
4.
Front Immunol ; 15: 1406438, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38817611

RESUMEN

Introduction: Atopic dermatitis (AD) is a chronic inflammatory skin disorder characterised by itching, erythema, and epidermal barrier dysfunction. The pathogenesis of AD is complex and multifactorial; however,mast cell (MC) activation has been reported to be one of the crucial mechanisms in the pathogenesis of AD. The MC receptor Mas related G protein-coupled receptor-X2 (MRGPRX2) has been identified as a prominent alternative receptor to the IgE receptor in causing MC activation and the subsequent release of inflammatory mediators. The current study aimed to evaluate the therapeutic effect of a novel small molecule MRGPRX2 antagonist GE1111 in AD using in vitro and in vivo approaches. Methods: We developed an in vitro cell culture disease model by using LAD-2 MC, HaCaT keratinocytes and RAW 264.7 macrophage cell lines. We challenged keratinocytes and macrophage cells with CST-14 treated MC supernatant in the presence and absence of GE1111 and measured the expression of tight junction protein claudin 1, inflammatory cytokines and macrophage phagocytosis activity through immunohistochemistry, western blotting, RT-qPCR and fluorescence imaging techniques. In addition to this, we developed a DFNB-induced AD model in mice and evaluated the protective effect and underlying mechanism of GE1111. Results and Discussion: Our in vitro findings demonstrated a potential therapeutic effect of GE1111, which inhibits the expression of TSLP, IL-13, MCP-1, TNF-a, and IL-1ß in MC and keratinocytes. In addition to this, GE1111 was able to preserve the expression of claudin 1 in keratinocytes and the phagocytotic activity of macrophage cells. The in vivo results demonstrated that GE1111 treatment significantly reduced phenotypic changes associated with AD (skin thickening, scaling, erythema and epidermal thickness). Furthermore, immunohistochemical analysis demonstrated that GE1111 treatment preserved the expression of the tight junction protein Involucrin and reduced the expression of the inflammatory mediator periostin in the mouse model of AD. These findings were supported by gene and protein expression analysis, where GE1111 treatment reduced the expression of TSLP, IL-13, and IL-1ß, as well as downstream signalling pathways of MRGPRX2 in AD skin lesions. In conclusion, our findings provide compelling in vitro and in vivo evidence supporting the contribution of MRGPRX2-MC interaction with keratinocytes and macrophages in the pathogenesis of AD.


Asunto(s)
Citocinas , Dermatitis Atópica , Receptores Acoplados a Proteínas G , Receptores de Neuropéptido , Piel , Animales , Humanos , Ratones , Citocinas/metabolismo , Dermatitis Atópica/tratamiento farmacológico , Dermatitis Atópica/inmunología , Modelos Animales de Enfermedad , Células HaCaT , Mediadores de Inflamación/metabolismo , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Mastocitos/efectos de los fármacos , Mastocitos/inmunología , Mastocitos/metabolismo , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/metabolismo , Células RAW 264.7 , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropéptido/antagonistas & inhibidores , Receptores de Neuropéptido/metabolismo , Piel/efectos de los fármacos , Piel/inmunología , Piel/patología
5.
J Sep Sci ; 47(11): e2300924, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38819784

RESUMEN

Mas-related G protein-coupled receptor X2 (MrgprX2) is acknowledged as a mast cell-specific receptor, playing a crucial role in orchestrating anaphylactoid responses through mast cell degranulation. It holds promise as a target for regulating allergic and inflammatory diseases mediated by mast cells. Polygonum cuspidatum (PC) has shown notable anti-anaphylactoid effects, while its pharmacologically active components remain unclear. In this study, we successfully utilized MrgprX2 high-expressing cell membrane chromatography (CMC), in conjunction with liquid chromatography-mass spectrometry (LC-MS), to identify active anti-anaphylactoid components in PC. Our study pinpointed polydatin, resveratrol, and emodin-8-O-ß-d-glucoside as potential anti-anaphylactoid compounds in PC. Their anti-anaphylactoid activities were evaluated through ß-aminohexosidase and histamine release assays, demonstrating a concentration-dependent inhibition for both ß-aminohexosidase and histamine release. This approach, integrating MrgprX2 high-expression CMC with LC-MS, proves effective in screening potential anti-anaphylactoid ingredients in natural herbal medicines. The findings from this study illuminated the anti-anaphylactoid properties of specific components in PC and provided an efficient method for the drug development of natural products.


Asunto(s)
Fallopia japonica , Receptores Acoplados a Proteínas G , Receptores de Neuropéptido , Receptores Acoplados a Proteínas G/metabolismo , Fallopia japonica/química , Receptores de Neuropéptido/metabolismo , Receptores de Neuropéptido/antagonistas & inhibidores , Humanos , Espectrometría de Masas , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Membrana Celular/química , Cromatografía Liquida , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Mastocitos/efectos de los fármacos , Mastocitos/metabolismo , Extractos Vegetales/farmacología , Extractos Vegetales/química , Glucósidos/farmacología , Glucósidos/química , Glucósidos/análisis , Estructura Molecular , Cromatografía Líquida con Espectrometría de Masas
6.
J Med Chem ; 67(9): 7603-7619, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38687204

RESUMEN

The design of bifunctional compounds is a promising approach toward the development of strong analgesics with reduced side effects. We here report the optimization of the previously published lead peptide KGFF09, which contains opioid receptor agonist and neuropeptide FF receptor antagonist pharmacophores and is shown to induce potent antinociception and reduced side effects. We evaluated the novel hybrid peptides for their in vitro activity at MOP, NPFFR1, and NPFFR2 and selected four of them (DP08/14/32/50) for assessment of their acute antinociceptive activity in mice. We further selected DP32 and DP50 and observed that their antinociceptive activity is mostly peripherally mediated; they produced no respiratory depression, no hyperalgesia, significantly less tolerance, and strongly attenuated withdrawal syndrome, as compared to morphine and the recently FDA-approved TRV130. Overall, these data suggest that MOP agonist/NPFF receptor antagonist hybrids might represent an interesting strategy to develop novel analgesics with reduced side effects.


Asunto(s)
Receptores de Neuropéptido , Receptores Opioides mu , Animales , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inhibidores , Receptores Opioides mu/metabolismo , Ratones , Receptores de Neuropéptido/agonistas , Receptores de Neuropéptido/antagonistas & inhibidores , Receptores de Neuropéptido/metabolismo , Masculino , Analgésicos/farmacología , Analgésicos/química , Analgésicos/uso terapéutico , Analgésicos/síntesis química , Humanos , Relación Estructura-Actividad , Analgésicos Opioides/farmacología , Analgésicos Opioides/química
7.
Bioorg Med Chem Lett ; 56: 128485, 2022 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-34861349

RESUMEN

Mas-related G protein-coupled receptor X2 (MRGPRX2) mediates the itch response in neurons and is involved in atopic dermatitis (AD)-associated inflammation and itch. Potent and MRGPRX2-selective ligands are essential to an understanding of the detailed function of the receptor and to develop new therapeutic agents for its related diseases. (+)-TAN-67 (1), the enantiomer of the δ-opioid receptor (DOR) selective ligand (-)-TAN-67 (1), has been reported to activate MRGPRX2, although (+)-1 also interacts with DOR, which prevents investigators from interrogating the function of MRGPRX2. Here, we have succeeded in developing a novel unnatural morphinan compound (+)-2a by a transformation based on the structure of (+)-1, which removes the DOR binding affinity. (+)-2a activated both human MRGPRX2 and the mouse orthologue Mrgprb2 in in vitro experiments and induced itch-like behaviors in mice to the same extent as (+)-1. The (+)-2a-induced itch response in mice was suppressed by administration of the tripeptide QWF, an MRGPRX2/Mrgprb2 antagonist, or the antipruritic drug nalfurafine. Together, (+)-2a serves as a useful tool to elucidate the itch-related function/action of MRGPRX2 and its mouse orthologue Mrgprb2.


Asunto(s)
Conducta Animal/efectos de los fármacos , Desarrollo de Medicamentos , Morfinanos/efectos adversos , Proteínas del Tejido Nervioso/metabolismo , Prurito/inducido químicamente , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropéptido/metabolismo , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Humanos , Ligandos , Ratones , Estructura Molecular , Morfinanos/síntesis química , Morfinanos/química , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores de Neuropéptido/antagonistas & inhibidores , Receptores Opioides delta , Relación Estructura-Actividad
8.
Nature ; 600(7887): 170-175, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34789874

RESUMEN

The MRGPRX family of receptors (MRGPRX1-4) is a family of mas-related G-protein-coupled receptors that have evolved relatively recently1. Of these, MRGPRX2 and MRGPRX4 are key physiological and pathological mediators of itch and related mast cell-mediated hypersensitivity reactions2-5. MRGPRX2 couples to both Gi and Gq in mast cells6. Here we describe agonist-stabilized structures of MRGPRX2 coupled to Gi1 and Gq in ternary complexes with the endogenous peptide cortistatin-14 and with a synthetic agonist probe, respectively, and the development of potent antagonist probes for MRGPRX2. We also describe a specific MRGPRX4 agonist and the structure of this agonist in a complex with MRGPRX4 and Gq. Together, these findings should accelerate the structure-guided discovery of therapeutic agents for pain, itch and mast cell-mediated hypersensitivity.


Asunto(s)
Microscopía por Crioelectrón , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/química , Prurito/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/química , Receptores de Neuropéptido/antagonistas & inhibidores , Receptores de Neuropéptido/química , Agonismo Inverso de Drogas , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/química , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/ultraestructura , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/química , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/ultraestructura , Humanos , Modelos Moleculares , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/ultraestructura , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/ultraestructura , Receptores de Neuropéptido/metabolismo , Receptores de Neuropéptido/ultraestructura
9.
Biomed Pharmacother ; 142: 111951, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34333290

RESUMEN

Tissue expansion is widely used to obtain new skin tissue for repairing defects in the clinical practice of plastic surgery. One major complication can be dermal thinning during expansion, which usually leads to skin rupture. Collagen synthesis can determine dermal thickness and can be influenced by macrophage polarization during expansion. The aim of the study was to test whether pigment epithelium-derived factor (PEDF) could be a modulator of collagen synthesis in fibroblasts by regulating macrophage polarization during skin expansion. Our results showed that PEDF mRNA expression was increased in expanded human and mouse epidermis. PEDF protein levels were elevated in the subcutaneous exudates of a rat skin expansion model. Increased PEDF mRNA expression was accompanied by dermal thinning during a three-week expansion protocol. Subcutaneous injection of PEDF in vivo further resulted in dermal thinning and cell number increase of M1 macrophage in the expanded skin. PEDF also promoted macrophage polarization in vitro to the M1 subtype under hypoxic conditions. PEDF did not influence collagen gene expression in fibroblasts directly, but attenuated collagen synthesis in a macrophage-mediated manner. Additionally, blockage of PEDF receptors on macrophages with inhibitors rescued collagen synthesis in fibroblasts. Our research demonstrated PEDF elevation in expanded skin leads to dermal thinning through M1 macrophage-mediated collagen synthesis inhibition in fibroblasts. Our results could form a basis for the development of novel strategies to improve skin integrity in expanded skin by using PEDF.


Asunto(s)
Colágeno/biosíntesis , Proteínas del Ojo/metabolismo , Proteínas del Ojo/farmacología , Fibroblastos/metabolismo , Activación de Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Factores de Crecimiento Nervioso/farmacología , Serpinas/metabolismo , Serpinas/farmacología , Animales , Hipoxia de la Célula , Línea Celular , Colágeno/genética , Epidermis/metabolismo , Proteínas del Ojo/genética , Humanos , Hipoxia/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Modelos Animales , Factores de Crecimiento Nervioso/genética , Ratas Sprague-Dawley , Receptores de Neuropéptido/antagonistas & inhibidores , Serpinas/genética , Piel/irrigación sanguínea , Piel/efectos de los fármacos , Piel/metabolismo , Expansión de Tejido
10.
J Med Chem ; 64(11): 7555-7564, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-34008968

RESUMEN

RFamide-related peptide-3 (RFRP-3) and neuropeptide FF (NPFF) target two different receptor subtypes called neuropeptide FF1 (NPFF1R) and neuropeptide FF2 (NPFF2R) that modulate several functions. However, the study of their respective role is severely limited by the absence of selective blockers. We describe here the design of a highly selective NPFF1R antagonist called RF3286, which potently blocks RFRP-3-induced hyperalgesia in mice and luteinizing hormone release in hamsters. We then showed that the pharmacological blockade of NPFF1R in mice prevents the development of fentanyl-induced hyperalgesia while preserving its analgesic effect. Altogether, our data indicate that RF3286 represents a useful pharmacological tool to study the involvement of the NPFF1R/RFRP-3 system in different functions and different species. Thanks to this compound, we showed that this system is critically involved in the development of opioid-induced hyperalgesia, suggesting that NPFF1R antagonists might represent promising therapeutic tools to improve the use of opioids in the treatment of chronic pain.


Asunto(s)
Analgésicos Opioides/efectos adversos , Dipéptidos/química , Receptores de Neuropéptido/antagonistas & inhibidores , Animales , Cricetinae , Dipéptidos/metabolismo , Dipéptidos/farmacología , Dipéptidos/uso terapéutico , Femenino , Fentanilo/efectos adversos , Semivida , Humanos , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/etiología , Hormona Luteinizante/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neuropéptidos/química , Neuropéptidos/metabolismo , Neuropéptidos/farmacología , Neuropéptidos/uso terapéutico , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Receptores de Neuropéptido/metabolismo , Receptores Opioides/química , Receptores Opioides/metabolismo , Relación Estructura-Actividad
11.
J Med Chem ; 64(7): 4089-4108, 2021 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-33733768

RESUMEN

Neuropeptide S modulates important neurobiological functions including locomotion, anxiety, and drug abuse through interaction with its G protein-coupled receptor known as neuropeptide S receptor (NPSR). NPSR antagonists are potentially useful for the treatment of substance abuse disorders against which there is an urgent need for new effective therapeutic approaches. Potent NPSR antagonists in vitro have been discovered which, however, require further optimization of their in vivo pharmacological profile. This work describes a new series of NPSR antagonists of the oxazolo[3,4-a]pyrazine class. The guanidine derivative 16 exhibited nanomolar activity in vitro and 5-fold improved potency in vivo compared to SHA-68, a reference pharmacological tool in this field. Compound 16 can be considered a new tool for research studies on the translational potential of the NPSergic system. An in-depth molecular modeling investigation was also performed to gain new insights into the observed structure-activity relationships and provide an updated model of ligand/NPSR interactions.


Asunto(s)
Oxazoles/farmacología , Pirazinas/farmacología , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores de Neuropéptido/antagonistas & inhibidores , Animales , Células HEK293 , Humanos , Locomoción/efectos de los fármacos , Ratones Noqueados , Simulación del Acoplamiento Molecular , Estructura Molecular , Oxazoles/síntesis química , Oxazoles/metabolismo , Unión Proteica , Pirazinas/síntesis química , Pirazinas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropéptido/metabolismo , Relación Estructura-Actividad
12.
Neurosci Lett ; 751: 135746, 2021 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-33610674

RESUMEN

Symptoms that resemble allergic reactions, such as pruritus, flushing, and hypotension, are common side effects of therapeutic drugs. In a true allergic reaction, Immunoglobulin E (IgE) antibodies recognize the drug and trigger mediator release from mast cells through cross-linking of IgE receptors. However, many drugs can bypass this pathway and can activate mast cells directly through MRGPRX2, a G protein-coupled receptor that responds to a wide range of small molecules, peptides, and proteins that have little in common except for a net positive charge. This review will provide an overview of MRGPRX2, including its expression pattern, studies of its pharmacology, and its orthologs. It also will review evidence for MRGPRX2 activation by many drugs closely associated with these reactions.


Asunto(s)
Hipersensibilidad a las Drogas/metabolismo , Mastocitos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropéptido/metabolismo , Analgésicos/farmacología , Analgésicos/uso terapéutico , Animales , Antipruriginosos/farmacología , Antipruriginosos/uso terapéutico , Hipersensibilidad a las Drogas/tratamiento farmacológico , Humanos , Mastocitos/efectos de los fármacos , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/genética , Receptores de Neuropéptido/antagonistas & inhibidores , Receptores de Neuropéptido/genética
13.
Int J Med Sci ; 18(1): 18-28, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33390770

RESUMEN

Cardiac hypertrophy (CH) is a major risk factor for heart failure accompanied by maladaptive cardiac remodeling. The role and potential mechanism of neuropeptide Y (NPY) in CH are still unclear. We will explore the role and the mechanism of NPY inactivation (NPY-I) in CH caused by pressure overload. Abdominal aortic constriction (AAC) was used to induce CH model in rats. NPY or angiotensin II (Ang II) was used to trigger CH model in vitro in neonatal rat ventricular myocytes (NRVMs). We found that NPY was increased in the heart and plasma of hypertrophic rats. However, Ang II did not increase NPY expression in cardiomyocytes. NPY-I attenuated CH as decreasing CH-related markers (ANP, BNP and ß-MHC mRNA) level, reducing cell surface area, and restoring cardiac function. NPY inactivation increased miR-216b and decreased FoxO4 expression in CH heart. Moreover, NPY decreased miR-216b and increased FoxO4 expression in NRVMs which were reversed by NPY type 1 receptor (NPY1R) antagonist BIBO3304. MiR-216b mimic and FoxO4 siRNA (small interfering RNA) inhibited NPY/Ang II-induced myocardial hypertrophy in vitro. Meanwhile, BIBO3304 reversed the pro-hypertrophy effect of NPY in vitro. Collectively, NPY deficiency attenuated CH by NPY1R-miR-216b-FoxO4 axis. These findings suggested that NPY would be a potential therapeutic target for the prevention and treatment of cardiac hypertrophy.


Asunto(s)
Cardiomegalia/genética , Factores de Transcripción Forkhead/genética , MicroARNs/metabolismo , Miocardio/patología , Neuropéptido Y/metabolismo , Angiotensina II/metabolismo , Animales , Arginina/análogos & derivados , Arginina/farmacología , Cardiomegalia/patología , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Masculino , MicroARNs/agonistas , MicroARNs/antagonistas & inhibidores , Miocardio/citología , Miocitos Cardíacos/patología , Cultivo Primario de Células , Ratas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropéptido/antagonistas & inhibidores , Receptores de Neuropéptido/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
14.
J Cell Physiol ; 236(3): 1903-1912, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32710469

RESUMEN

Macrophage migration is thought to participate in obesity-related cardiovascular diseases. Matrix metalloproteinase-8 (MMP-8) possesses proteolytic activity on the extracellular matrix (ECM), which promotes macrophage migration to the site of vascular injury. Neuropeptide Y (NPY) is a bioactive peptide involved in MMP expression. However, it is uncertain whether NPY can regulate the expression of matrix metalloproteinase-8 (MMP-8) in macrophages. In this study, wild-type C57BL/6 and NPY-/- mice were fed a high-fat diet and subjected to subcutaneous carotid artery injury with ferric chloride, to observe the role of NPY and macrophages in neointima formation. In addition, Raw264.7 cells were treated with NPY and its antagonists to observe MMP-8 expression and macrophage migration. We found that NPY-/- mice exhibited significantly reduced neointima formation after carotid artery injury. The content of macrophages and MMP-8 in the neointima and media were also significantly reduced in NPY-/- mice compared with C57BL/6 mice. Moreover, the expression of MMP-8 in macrophages was also decreased in NPY-/- mice. NPY increased MMP-8 messenger RNA and protein expression in Raw264.7 cells in vitro, and this effect was abrogated by the Y1R antagonist. In addition, NPY increased the phosphorylation of ERK1/2, which was significantly attenuated by co-treatment with the Y1R antagonist. Moreover, NPY-induced MMP-8 expression could be decreased by the ERK1/2 inhibitor PD98059. Furthermore, NPY promoted macrophage migration across type I collagen in vitro. In conclusion, NPY promotes macrophage migration by upregulating MMP-8 expression, which we believe to be an underappreciated mechanism of the increased progression of neointima formation.


Asunto(s)
Movimiento Celular , Macrófagos/citología , Macrófagos/enzimología , Metaloproteinasa 8 de la Matriz/metabolismo , Neuropéptido Y/metabolismo , Animales , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neointima/metabolismo , Neointima/patología , Neuropéptido Y/deficiencia , Placa Aterosclerótica/patología , Células RAW 264.7 , Receptores de Neuropéptido/antagonistas & inhibidores , Receptores de Neuropéptido/metabolismo
15.
Int J Mol Sci ; 21(23)2020 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-33255594

RESUMEN

The opioid-induced analgesia is associated with a number of side effects such as addiction, tolerance and respiratory depression. The involvement of neuropeptide FF (NPFF) in modulation of pain perception, opioid-induced tolerance and dependence was well documented in contrast to respiratory depression. Therefore, the aim of the present study was to examine the potency of NPFF to block post-opioid respiratory depression, one of the main adverse effects of opioid therapy. Urethane-chloralose anaesthetized Wistar rats were injected either intravenously (iv) or intracerebroventricularly (icv) with various doses of NPFF prior to iv endomorphin-1 (EM-1) administration. Iv NPFF diminished the number of EM-1-induced apneas without affecting their length and without influence on the EM-1 induced blood pressure decline. Icv pretreatment with NPFF abolished the occurrence of post-EM-1 apneas and reduced also the maximal drop in blood pressure and heart rate. These effects were completely blocked by the NPFF receptor antagonist RF9, which was given as a mixture with NPFF before systemic EM-1 administration. In conclusion, our results showed that centrally administered neuropeptide FF is effective in preventing apnea evoked by stimulation of µ-opioid receptors and the effect was due to activation of central NPFF receptors. Our finding indicates a potential target for reversal of opioid-induced respiratory depression.


Asunto(s)
Apnea/tratamiento farmacológico , Enfermedades Cardiovasculares/tratamiento farmacológico , Oligopéptidos/farmacología , Receptores Opioides mu/genética , Analgesia/efectos adversos , Analgésicos Opioides/efectos adversos , Animales , Apnea/inducido químicamente , Apnea/genética , Apnea/patología , Enfermedades Cardiovasculares/inducido químicamente , Enfermedades Cardiovasculares/genética , Modelos Animales de Enfermedad , Humanos , Infusiones Intraventriculares , Oligopéptidos/efectos adversos , Oligopéptidos/genética , Percepción del Dolor/efectos de los fármacos , Ratas , Receptores de Neuropéptido/antagonistas & inhibidores , Receptores de Neuropéptido/genética , Receptores Opioides mu/antagonistas & inhibidores , Activación Transcripcional/efectos de los fármacos
16.
Bioorg Med Chem Lett ; 30(23): 127510, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32898693

RESUMEN

Here, we report the discovery of a new class of NPBWR1 antagonists identified from a fragment-based screen. Compound 1 (cAMP IC50 = 250 µM; LE = 0.29) emerged as an initial hit. Further optimization of 1 by SAR-by-catalogue and chemical modification produced 21a (cAMP IC50 = 30 nM; LE = 0.39) with a 6700-fold increase in potency from fragment 1. Somewhat surprisingly, Schild analysis of compound 21a suggested that in vitro inhibition of NPW-mediated effects on upon cAMP accumulation were saturable, and that compound 21a dose-dependently increased [125I]-hNPW23 dissociation rate constants from NPBWR1 in kinetic binding studies. Collectively, these data are inconsistent with a classic surmountable, orthosteric mechanism of inhibition. The benzimidazole inhibitors reported herein may therefore represent a mechanistically differentiated class of compounds with which to form a better appreciation of the pharmacology and physiological roles of this central neuropeptide system.


Asunto(s)
Bencimidazoles/farmacología , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores de Neuropéptido/antagonistas & inhibidores , Animales , Bencimidazoles/síntesis química , Células CHO , Cricetulus , Descubrimiento de Drogas , Humanos , Estructura Molecular , Relación Estructura-Actividad
17.
J Med Chem ; 63(21): 12387-12402, 2020 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-32673481

RESUMEN

The endogenous neuropeptide FF (NPFF) and its two cognate G protein-coupled receptors, Neuropeptide FF Receptors 1 and 2 (NPFFR1 and NPFFR2), represent a relatively new target system for many therapeutic applications including pain regulation, modulation of opioid side effects, drug reward, anxiety, cardiovascular conditions, and other peripheral effects. Since the cloning of NPFFR1 and NPFFR2 in 2000, significant progress has been made to understand their pharmacological roles and interactions with other receptor systems, notably the opioid receptors. A variety of NPFFR ligands with different mechanisms of action (agonists or antagonists) have been discovered although with limited subtype selectivities. Differential pharmacological effects have been observed for many of these NPFFR ligands, depending on assays/models employed and routes of administration. In this Perspective, we highlight the therapeutic potentials, current knowledge gaps, and latest updates of the development of peptidic and small molecule NPFFR ligands as tool compounds and therapeutic candidates.


Asunto(s)
Analgésicos Opioides/metabolismo , Ligandos , Oligopéptidos/metabolismo , Receptores de Neuropéptido/metabolismo , Analgésicos Opioides/química , Analgésicos Opioides/uso terapéutico , Animales , Ansiedad/tratamiento farmacológico , Ansiedad/patología , Humanos , Ratones , Oligopéptidos/química , Oligopéptidos/uso terapéutico , Dolor/tratamiento farmacológico , Dolor/patología , Peptidomiméticos , Ratas , Receptores de Neuropéptido/antagonistas & inhibidores , Receptores de Neuropéptido/química , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/metabolismo , Bibliotecas de Moléculas Pequeñas/uso terapéutico
18.
Front Immunol ; 11: 703, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32391014

RESUMEN

Mast cells are tissue-resident innate immune cells known for their prominent role in mediating allergic reactions. MAS-related G-protein coupled receptor-X2 (MRGPRX2) is a promiscuous G-protein coupled receptor (GPCR) expressed on mast cells that is activated by several ligands that share cationic and amphipathic properties. Interestingly, MRGPRX2 ligands include certain FDA-approved drugs, antimicrobial peptides, and neuropeptides. Consequently, this receptor has been implicated in causing mast cell-dependent pseudo-allergic reactions to these drugs and chronic inflammation associated with asthma, urticaria and rosacea in humans. In the current study we examined the role of osthole, a natural plant coumarin, in regulating mast cell responses when activated by the MRGPRX2 ligands, including compound 48/80, the neuropeptide substance P, and the cathelicidin LL-37. We demonstrate that osthole attenuates both the early (Ca2+ mobilization and degranulation) and delayed events (chemokine/cytokine production) of mast cell activation via MRGPRX2 in vitro. Osthole also inhibits MrgprB2- (mouse ortholog of human MRGPRX2) dependent inflammation in in vivo mouse models of pseudo-allergy. Molecular docking analysis suggests that osthole does not compete with the MRGPRX2 ligands for interaction with the receptor, but rather regulates MRGPRX2 activation via allosteric modifications. Furthermore, flow cytometry and confocal microscopy experiments reveal that osthole reduces both surface and intracellular expression levels of MRGPRX2 in mast cells. Collectively, our data demonstrate that osthole inhibits MRGPRX2/MrgprB2-induced mast cell responses and provides a rationale for the use of this natural compound as a safer alternative treatment for pseudo-allergic reactions in humans.


Asunto(s)
Cumarinas/administración & dosificación , Edema/tratamiento farmacológico , Mastocitos/inmunología , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Fitoterapia/métodos , Extractos Vegetales/administración & dosificación , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores de Neuropéptido/antagonistas & inhibidores , Animales , Señalización del Calcio/efectos de los fármacos , Degranulación de la Célula/efectos de los fármacos , Línea Celular Tumoral , Modelos Animales de Enfermedad , Edema/inmunología , Femenino , Humanos , Masculino , Mastocitos/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Ratas , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropéptido/química , Receptores de Neuropéptido/metabolismo , Donantes de Tejidos , Resultado del Tratamiento
19.
J Sep Sci ; 43(13): 2571-2578, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32281296

RESUMEN

Mas-related G protein-coupled receptor X2 was a mast cell-specific receptor mediating anaphylactoid reactions by activating mast cells degranulation, and it was also identified as a target for modulating mast cell-mediated anaphylactoid and inflammatory diseases. The anti-anaphylactoid drugs used clinically disturb the partial effect of partial mediators released by mast cells. The small molecule of Mas-related G protein-coupled receptor X2 specific antagonists may provide therapeutic action for the anaphylactoid and inflammatory diseases in the early stage. In this study, the Mas-related G protein-coupled receptor X2 high expression cell membrane chromatography was coupled online with liquid chromatography and mass spectrometry and successfully used to screen anti-anaphylactoid components from Magnolia biondii Pamp. Fargesin and pinoresinol dimethyl ether were identified as potential anti-anaphylactoid components. Bioactivity of these two components were investigated by ß hexosaminidase and histamine release assays on mast cells, and it was found that these two components could inhibit ß hexosaminidase and histamine release in a concentration-dependent manner. This Mas-related G protein-coupled receptor X2 high expression cell membrane chromatography coupled online with liquid chromatography and mass spectrometry system could be applied for screening potential anti-anaphylactoid components from natural medicinal herbs. This study also provided a powerful system for drug discovery in natural medicinal herbs.


Asunto(s)
Anafilaxia/tratamiento farmacológico , Membrana Celular/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Magnolia/química , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Extractos Vegetales/farmacología , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores de Neuropéptido/antagonistas & inhibidores , Anafilaxia/metabolismo , Membrana Celular/metabolismo , Células Cultivadas , Cromatografía Liquida , Medicamentos Herbarios Chinos/química , Células HEK293 , Humanos , Espectrometría de Masas , Mastocitos/efectos de los fármacos , Mastocitos/metabolismo , Medicina Tradicional China , Proteínas del Tejido Nervioso/metabolismo , Extractos Vegetales/química , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropéptido/metabolismo
20.
Eur J Pharmacol ; 878: 173104, 2020 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-32320700

RESUMEN

Anaphylaxis during general anaesthesia is a significant clinical challenge for anaesthesiologists. Approximately 50% of perioperative anaphylaxis cases lack the presence of specific IgE antibodies. Mas-related G-protein coupled receptor X2 (MRGPRX2) in humans and its mouse orthologue Mas-related G-protein coupled receptor B2 (Mrgprb2) are crucial receptors in non-IgE-dependent histamine release. Anaesthetics such as rocuronium and atracurium cause perioperative anaphylaxis by activating histamine release via the Mrgprb2 pathway. We hypothesized that antagonistic DNA aptamers that target MRGPRX2 can prevent perioperative anaphylaxis. Selection of a DNA aptamer that specifically binds MRGPRX2 was achieved by using our modified Systematic Evolution of Ligands by Exponential enrichment (SELEX) approach. Our SELEX process used MRGPRX2-proteoliposomes synthesised by a wheat germ cell-free system as templates. The activity of the selected aptamer to inhibit histamine release from MRGPRX2-activated mast cells and in an anaphylaxis rat model transplanted with this cell line was examined. Our selection process identified aptamer-X35 with the sequence 5'-ATGACCATGACCCTCCACACTGTAGGCACCACGGGTCCCTGGCAGTTAAAAGTACGTTTGTCAGACTGTGGCAGGGAAACA-3'. In silico 2D modelling of aptamer-X35 revealed a structure with a small loop and a long stem. Aptamer-X35 inhibited histamine release from mast cells by 70%. Subcutaneous injection of 30 nmol of aptamer-X35 inhibited the anaphylactic reaction in the rat anaphylaxis model. This study demonstrated that aptamer-X35 selected by the modified SELEX approach reduced histamine release by inhibiting the MRGPRX2 pathway. Overall, our findings establish aptamer-X35 as a potential therapeutic candidate against perioperative anaphylaxis.


Asunto(s)
Anafilaxia/tratamiento farmacológico , Anafilaxia/prevención & control , Aptámeros de Nucleótidos/farmacología , Liberación de Histamina/efectos de los fármacos , Mastocitos/metabolismo , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores de Neuropéptido/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Línea Celular , Simulación por Computador , Modelos Animales de Enfermedad , Diseño de Fármacos , Histamina/metabolismo , Humanos , Ligandos , Membrana Dobles de Lípidos/química , Masculino , Modelos Moleculares , Proteínas del Tejido Nervioso/genética , Unión Proteica , Conformación Proteica , Ratas , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuroquinina-1/metabolismo , Receptores de Neuropéptido/genética , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA