Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
2.
Sci Transl Med ; 14(638): eabl6328, 2022 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-35353541

RESUMEN

Cyclic adenosine 3',5'-monophosphate (cAMP)-elevating agents, such as ß2-adrenergic receptor (ß2-AR) agonists and phosphodiesterase (PDE) inhibitors, remain a mainstay in the treatment of obstructive respiratory diseases, conditions characterized by airway constriction, inflammation, and mucus hypersecretion. However, their clinical use is limited by unwanted side effects because of unrestricted cAMP elevation in the airways and in distant organs. Here, we identified the A-kinase anchoring protein phosphoinositide 3-kinase γ (PI3Kγ) as a critical regulator of a discrete cAMP signaling microdomain activated by ß2-ARs in airway structural and inflammatory cells. Displacement of the PI3Kγ-anchored pool of protein kinase A (PKA) by an inhaled, cell-permeable, PI3Kγ mimetic peptide (PI3Kγ MP) inhibited a pool of subcortical PDE4B and PDE4D and safely increased cAMP in the lungs, leading to airway smooth muscle relaxation and reduced neutrophil infiltration in a murine model of asthma. In human bronchial epithelial cells, PI3Kγ MP induced unexpected cAMP and PKA elevations restricted to the vicinity of the cystic fibrosis transmembrane conductance regulator (CFTR), the ion channel controlling mucus hydration that is mutated in cystic fibrosis (CF). PI3Kγ MP promoted the phosphorylation of wild-type CFTR on serine-737, triggering channel gating, and rescued the function of F508del-CFTR, the most prevalent CF mutant, by enhancing the effects of existing CFTR modulators. These results unveil PI3Kγ as the regulator of a ß2-AR/cAMP microdomain central to smooth muscle contraction, immune cell activation, and epithelial fluid secretion in the airways, suggesting the use of a PI3Kγ MP for compartment-restricted, therapeutic cAMP elevation in chronic obstructive respiratory diseases.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística , Fosfatidilinositol 3-Quinasa , Animales , Fosfatidilinositol 3-Quinasa Clase Ib , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Humanos , Inflamación , Ratones , Péptidos/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo
3.
Pharmaceutics ; 15(1)2022 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-36678686

RESUMEN

G protein-coupled receptors (GPCRs) transfer extracellular signals across cell membranes by activating intracellular heterotrimeric G proteins. Several studies suggested G proteins as novel drug targets for the treatment of complex diseases, e.g., asthma and cancer. Recently, we developed specific radiotracers, [³H]PSB-15900-FR and [³H]PSB-16254-YM, for the Gαq family of G proteins by tritiation of the macrocyclic natural products FR900359 (FR) and YM-254890 (YM). In the present study, we utilized these potent radioligands to perform autoradiography studies in tissues of healthy mice, mouse models of disease, and human tissues. Specific binding was high, while non-specific binding was extraordinarily low, giving nearly identical results for both radioligands. High expression levels of Gαq proteins were detected in healthy mouse organs showing the following rank order of potency: kidney > liver > brain > pancreas > lung > spleen, while expression in the heart was low. Organ sub-structures, e.g., of mouse brain and lung, were clearly distinguishable. Whereas an acute asthma model in mice did not result in altered Gαq protein expressions as compared to control animals, a cutaneous melanoma model displayed significantly increased expression in comparison to healthy skin. These results suggest the future development of Gαq-protein-binding radio-tracers as novel diagnostics.

4.
ACS Pharmacol Transl Sci ; 4(2): 888-897, 2021 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-33860209

RESUMEN

Guanine nucleotide-binding proteins (G proteins) transduce extracellular signals received by G protein-coupled receptors (GPCRs) to intracellular signaling cascades. While GPCRs represent the largest class of drug targets, G protein inhibition has only recently been recognized as a novel strategy for treating complex diseases such as asthma, inflammation, and cancer. The structurally similar macrocyclic depsipeptides FR900359 (FR) and YM-254890 (YM) are potent selective inhibitors of the Gq subfamily of G proteins. FR and YM differ in two positions, FR being more lipophilic than YM. Both compounds are utilized as pharmacological tools to block Gq proteins in vitro and in vivo. However, no detailed characterization of FR and YM has been performed, which is a prerequisite for the compounds' translation into clinical application. Here, we performed a thorough study of both compounds' physicochemical, pharmacokinetic, and pharmacological properties. Chemical stability was high across a large range of pH values, with FR being somewhat more stable than YM. Oral bioavailability and brain penetration of both depsipeptides were low. FR showed lower plasma protein binding and was metabolized significantly faster than YM by human and mouse liver microsomes. FR accumulated in lung after chronic intratracheal or intraperitoneal application, while YM was more distributed to other organs. Most strikingly, the previously observed longer residence time of FR resulted in a significantly prolonged pharmacologic effect as compared to YM in a methacholine-induced bronchoconstriction mouse model. These results prove that changes within a molecule which seem marginal compared to its structural complexity can lead to crucial pharmacological differences.

5.
Front Chem ; 8: 833, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33173765

RESUMEN

The cyclic depsipeptide FR900359 (FR) isolated from the plant Ardisia crenata and produced by endosymbiotic bacteria acts as a selective Gq protein inhibitor. It is a powerful tool to study G protein-coupled receptor signaling, and has potential as a novel drug for the treatment of pulmonary diseases and cancer. For pharmacokinetic studies, sensitive quantitative measurements of drug levels are required. In the present study we established an LC-MS/MS method to detect nanomolar concentrations of FR and the structurally related natural product YM-254890 (YM) in biological samples. HPLC separation coupled to ESI-QTOF-MS and UV-VIS detection was applied. For identification and quantification, the extract ion chromatogram (EIC) of M+1 was evaluated. Limits of detection (LOD) of 0.53-0.55 nM and limits of quantification (LOQ) of 1.6-1.7 nM were achieved for both FR and YM. This protocol was subsequently applied to determine FR concentrations in mouse organs and tissues after peroral application of the drug. A three-step liquid-liquid extraction protocol was established, which resulted in adequate recovery rates of typically around 50%. The results indicated low peroral absorption of FR. Besides the gut, highest concentrations were determined in eye and kidney. The developed analytical method will be useful for preclinical studies to evaluate these potent Gq protein inhibitors, which may have potential as future drugs for complex diseases.

6.
Cell Metab ; 32(1): 56-70.e7, 2020 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-32589947

RESUMEN

The combination of aging populations with the obesity pandemic results in an alarming rise in non-communicable diseases. Here, we show that the enigmatic adenosine A2B receptor (A2B) is abundantly expressed in skeletal muscle (SKM) as well as brown adipose tissue (BAT) and might be targeted to counteract age-related muscle atrophy (sarcopenia) as well as obesity. Mice with SKM-specific deletion of A2B exhibited sarcopenia, diminished muscle strength, and reduced energy expenditure (EE), whereas pharmacological A2B activation counteracted these processes. Adipose tissue-specific ablation of A2B exacerbated age-related processes and reduced BAT EE, whereas A2B stimulation ameliorated obesity. In humans, A2B expression correlated with EE in SKM, BAT activity, and abundance of thermogenic adipocytes in white fat. Moreover, A2B agonist treatment increased EE from human adipocytes, myocytes, and muscle explants. Mechanistically, A2B forms heterodimers required for adenosine signaling. Overall, adenosine/A2B signaling links muscle and BAT and has both anti-aging and anti-obesity potential.


Asunto(s)
Envejecimiento/metabolismo , Obesidad/metabolismo , Receptor de Adenosina A2B/metabolismo , Adolescente , Adulto , Animales , Células Cultivadas , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Persona de Mediana Edad , Receptor de Adenosina A2B/deficiencia , Transducción de Señal , Adulto Joven
7.
Br J Pharmacol ; 177(8): 1898-1916, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31881095

RESUMEN

BACKGROUND AND PURPOSE: G proteins are intracellular switches that transduce and amplify extracellular signals from GPCRs. The Gq protein subtypes, which are coupled to PLC activation, can act as oncogenes, and their expression was reported to be up-regulated in cancer and inflammatory diseases. Gq inhibition may be an efficient therapeutic strategy constituting a new level of intervention. However, diagnostic tools and therapeutic drugs for Gq proteins are lacking. EXPERIMENTAL APPROACH: We have now developed Gq -specific, cell-permeable 3 H-labelled high-affinity probes based on the macrocyclic depsipeptides FR900359 (FR) and YM-254890 (YM). The tracers served to specifically label and quantify Gq proteins in their native conformation in cells and tissues with high accuracy. KEY RESULTS: FR and YM displayed low nanomolar affinity for Gαq , Gα11 and Gα14 expressed in CRISPR/Cas9 Gαq -knockout cells, but not for Gα15 . The two structurally very similar tracers showed strikingly different dissociation kinetics, which is predicted to result in divergent biological effects. Computational studies suggested a "dowel" effect of the pseudoirreversibly binding FR. A high-throughput binding assay led to the discovery of novel Gq inhibitors, which inhibited Gq signalling in recombinant cells and primary murine brown adipocytes, resulting in enhanced differentiation. CONCLUSIONS AND IMPLICATIONS: The Gq protein inhibitors YM and FR are pharmacologically different despite similar structures. The new versatile tools and powerful assays will contribute to the advancement of the rising field of G protein research.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gq-G11 , Transducción de Señal , Animales , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Cinética , Ratones
8.
J Control Release ; 305: 155-164, 2019 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-31121282

RESUMEN

Anti-angiogenic therapies are promising options for diseases with enhanced vessel formation such as tumors or retinopathies. In most cases, a site-specific local effect on vessel growth is required, while the current focus on systemic distribution of angiogenesis inhibitors may cause severe unwanted side-effects. Therefore, in the current study we have developed an approach for the local inhibition of vascularization, using complexes of lentivirus and magnetic nanoparticles in combination with magnetic fields. Using this strategy in the murine embryonic stem cell (ESC) system, we were able to site-specifically downregulate the protein tyrosine phosphatase SHP2 by RNAi technology in areas with active vessel formation. This resulted in a reduction of vessel development, as shown by reduced vascular tube length, branching points and vascular loops. The anti-angiogenic effect could also be recapitulated in the dorsal skinfold chamber of mice in vivo. Here, site-specific downregulation of SHP2 reduced re-vascularization after wound induction. Thus, we have developed a magnet-assisted, RNAi-based strategy for the efficient local inhibition of angiogenesis in ESCs in vitro and also in vivo.


Asunto(s)
Regulación hacia Abajo , Vectores Genéticos/genética , Lentivirus/genética , Células Madre Embrionarias de Ratones/metabolismo , Neovascularización Fisiológica , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Animales , Línea Celular , Vectores Genéticos/administración & dosificación , Imanes/química , Ratones , Neovascularización Patológica/genética , Neovascularización Patológica/terapia , Interferencia de ARN , Transducción Genética/métodos
9.
Sci Rep ; 8(1): 17582, 2018 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-30514882

RESUMEN

For the monitoring of vascular growth as well as adaptive or therapeutic (re)vascularization endothelial-specific reporter mouse models are valuable tools. However, currently available mouse models have limitations, because not all endothelial cells express the reporter in all developmental stages. We have generated PECAM/eGFP embryonic stem (ES) cell and mouse lines where the reporter gene labels PECAM+ endothelial cells and vessels with high specificity. Native eGFP expression and PECAM staining were highly co-localized in vessels of various organs at embryonic stages E9.5, E15.5 and in adult mice. Expression was found in large and small arteries, capillaries and in veins but not in lymphatic vessels. Also in the bone marrow arteries and sinusoidal vessel were labeled, moreover, we could detect eGFP in some CD45+ hematopoietic cells. We also demonstrate that this labeling is very useful to monitor sprouting in an aortic ring assay as well as vascular remodeling in a murine injury model of myocardial infarction. Thus, PECAM/eGFP transgenic ES cells and mice greatly facilitate the monitoring and quantification of endothelial cells ex vivo and in vivo during development and injury.


Asunto(s)
Proteínas Fluorescentes Verdes/metabolismo , Modelos Animales , Células Madre Embrionarias de Ratones/citología , Neovascularización Patológica , Neovascularización Fisiológica , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Animales , Médula Ósea/metabolismo , Línea Celular , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Genes Reporteros , Ratones , Ratones Transgénicos , Remodelación Vascular
10.
Life Sci ; 215: 96-105, 2018 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-30403990

RESUMEN

AIMS: Pulmonary hypertension (PH) leads to right ventricular (RV) adaptation and remodeling and has deleterious long-term effects on RV function. The endocannabinoid receptor CB2 has been associated with protective effects in adaptation and remodeling of the left ventricle after ischemia. Therefore, we investigated the role of CB2 receptor in RV adaptation after occlusion of the left pulmonary artery (LPA) in a murine model. MAIN METHODS: C57/Bl6 (WT)- and CB2 receptor-deficient (Cnr2-/-)-mice underwent paramedian sternotomy and LPA was occluded using a metal clip. Right heart hemodynamic study (Millar®) preceded organ harvesting for immunohistochemistry and mRNA analysis 7 and 21 days (d) post-occlusion. KEY FINDINGS: LPA occlusion led to higher RV systolic pressure in Cnr2-/--hearts, while hemodynamics were comparable with WT-hearts after 21d. Cnr2-/--hearts showed higher macrophage infiltration and lower interleukin-10 expression after 7 d, but otherwise a comparable inflammatory mediator expression profile. Cardiomyocyte-hypertrophy was stronger in Cnr2-/--mice, presenting with higher tenascin-C expression than WT-hearts. Planimetry revealed higher collagen area in Cnr2-/--hearts and small areas of cardiomyocyte-loss. Surrounding cardiomyocytes were cleaved caspase-3- and TUNEL positive in Cnr2-/--hearts. This was associated by maladaptation of myosin heavy-chain isoforms and lower reactive oxygen scavenger enzymes induction in Cnr2-/--hearts. We found comparable morphological changes in both lungs between the two genotypes. SIGNIFICANCE: LPA occlusion led to increased systolic pressure and adaptation of RV in CB2-deficient mice. CB2 receptor seems to modulate RV adaptation through expression of contractile elements, reactive oxygen scavenger enzymes, and inflammatory response in order to prevent cardiomyocyte apoptosis.


Asunto(s)
Hipertensión Pulmonar/fisiopatología , Hipertrofia Ventricular Derecha/fisiopatología , Miocitos Cardíacos/patología , Arteria Pulmonar/fisiopatología , Receptor Cannabinoide CB2/genética , Animales , Apoptosis/fisiología , Modelos Animales de Enfermedad , Hipertrofia Ventricular Derecha/genética , Inflamación/patología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Especies Reactivas de Oxígeno/metabolismo , Función Ventricular Derecha/fisiología
11.
Cell Stem Cell ; 22(2): 262-276.e7, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29451855

RESUMEN

Despite much work studying ex vivo multipotent stromal cells (MSCs), the identity and characteristics of MSCs in vivo are not well defined. Here, we generated a CD73-EGFP reporter mouse to address these questions and found EGFP+ MSCs in various organs. In vivo, EGFP+ mesenchymal cells were observed in fetal and adult bones at proliferative ossification sites, while in solid organs EGFP+ cells exhibited a perivascular distribution pattern. EGFP+ cells from the bone compartment could be clonally expanded ex vivo from single cells and displayed trilineage differentiation potential. Moreover, in the central bone marrow CD73-EGFP+ specifically labeled sinusoidal endothelial cells, thought to be a critical component of the hematopoietic stem cell niche. Purification and molecular characterization of this CD73-EGFP+ population revealed an endothelial subtype that also displays a mesenchymal signature, highlighting endothelial cell heterogeneity in the marrow. Thus, the CD73-EGFP mouse is a powerful tool for studying MSCs and sinusoidal endothelium.


Asunto(s)
5'-Nucleotidasa/metabolismo , Células de la Médula Ósea/metabolismo , Células Endoteliales/metabolismo , Células Madre Multipotentes/metabolismo , Coloración y Etiquetado , Nicho de Células Madre , Animales , Médula Ósea/metabolismo , Células de la Médula Ósea/citología , Condrogénesis , Células Endoteliales/citología , Femenino , Genes Reporteros , Proteínas Fluorescentes Verdes/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Madre Multipotentes/citología , Especificidad de Órganos , Células del Estroma/citología , Células del Estroma/metabolismo
12.
Angiogenesis ; 21(2): 349-361, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29417260

RESUMEN

Endothelial cell proliferation is a key process during vascular growth but its kinetics could only be assessed in vitro or ex vivo so far. To enable the monitoring and quantification of cell cycle kinetics in vivo, we have generated transgenic mice expressing an eGFP-anillin construct under control of the endothelial-specific Flt-1 promoter. This construct labels the nuclei of endothelial cells in late G1, S and G2 phase and changes its localization during the different stages of M phase, thereby enabling the monitoring of EC proliferation and cytokinesis. In Flt-1/eGFP-anillin mice, we found eGFP+ signals specifically in Ki67+/PECAM+ endothelial cells during vascular development. Quantification using this cell cycle reporter in embryos revealed a decline in endothelial cell proliferation between E9.5 to E12.5. By time-lapse microscopy, we determined the length of different cell cycle phases in embryonic endothelial cells in vivo and found a M phase duration of about 80 min with 2/3 covering karyokinesis and 1/3 cytokinesis. Thus, we have generated a versatile transgenic system for the accurate assessment of endothelial cell cycle dynamics in vitro and in vivo.


Asunto(s)
Ciclo Celular , Proteínas Contráctiles/metabolismo , Embrión de Mamíferos/metabolismo , Células Endoteliales/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Proteínas Contráctiles/genética , Embrión de Mamíferos/citología , Células Endoteliales/citología , Proteínas Fluorescentes Verdes/genética , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética
13.
Biomaterials ; 155: 176-190, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29179133

RESUMEN

Cell replacement in the heart is considered a promising strategy for the treatment of post-infarct heart failure. Direct intramyocardial injection of cells proved to be the most effective application route, however, engraftment rates are very low (<5%) strongly hampering its efficacy. Herein we combine magnetic nanoparticle (MNP) loading of EGFP labeled embryonic cardiomyocytes (eCM) and embryonic stem cell-derived cardiomyocytes (ES-CM) with application of custom designed magnets to enhance their short and long-term engraftment. To optimize cellular MNP uptake and magnetic force within the infarct area, first numerical simulations and experiments were performed in vitro. All tested cell types could be loaded efficiently with SOMag5-MNP (200 pg/cell) without toxic side effects. Application of a 1.3 T magnet at 5 mm distance from the heart for 10 min enhanced engraftment of both eCM and ES-CM by approximately 7 fold at 2 weeks and 3.4 fold (eCM) at 8 weeks after treatment respectively and also strongly improved left ventricular function at all time points. As underlying mechanisms we found that application of the magnetic field prevented the initial dramatic loss of cells via the injection channel. In addition, grafted eCM displayed higher proliferation and lower apoptosis rates. Electron microscopy revealed better differentiation of engrafted eCM, formation of cell to cell contacts and more physiological matrix formation in magnet-treated grafts. These results were corroborated by gene expression data. Thus, combination of MNP-loaded cells and magnet-application strongly increases long-term engraftment of cells addressing a major shortcoming of cardiomyoplasty.


Asunto(s)
Infarto del Miocardio/terapia , Miocitos Cardíacos/citología , Animales , Nanopartículas de Magnetita/efectos adversos , Trasplante de Células Madre
14.
Cancer Immunol Res ; 5(6): 455-467, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28468914

RESUMEN

A hypoxic tumor microenvironment is linked to poor prognosis. It promotes tumor cell dedifferentiation and metastasis and desensitizes tumor cells to type-I IFN, chemotherapy, and irradiation. The cytoplasmic immunoreceptor retinoic acid-inducible gene-I (RIG-I) is ubiquitously expressed in tumor cells and upon activation by 5'-triphosphate RNA (3pRNA) drives the induction of type I IFN and immunogenic cell death. Here, we analyzed the impact of hypoxia on the expression of RIG-I in various human and murine tumor and nonmalignant cell types and further investigated its function in hypoxic murine melanoma. 3pRNA-inducible RIG-I-expression was reduced in hypoxic melanoma cells compared with normoxic controls, a phenomenon that depended on the hypoxia-associated transcription factor HIF1α. Still, RIG-I functionality was conserved in hypoxic melanoma cells, whereas responsiveness to recombinant type-I IFN was abolished, due to hypoxia-induced loss of type I IFN receptor expression. Likewise, RIG-I activation in hypoxic melanoma cells, but not exposure to recombinant IFNα, provoked melanocyte antigen-specific CD8+ T-cell and NK-cell attack. Scavenging of hypoxia-induced reactive oxygen species by vitamin C restored the inducible expression of RIG-I under hypoxia in vitro, boosted in vitro anti-melanoma NK- and CD8+ T-cell attack, and augmented 3pRNA antitumor efficacy in vivo These results demonstrate that RIG-I remains operational under hypoxia and that RIG-I function is largely insensitive to lower cell surface expression of the IFNα receptor. RIG-I function could be fortified under hypoxia by the combined use of 3pRNA with antioxidants. Cancer Immunol Res; 5(6); 455-67. ©2017 AACR.


Asunto(s)
Hipoxia/metabolismo , Tolerancia Inmunológica , Melanoma/metabolismo , Receptores de Ácido Retinoico/metabolismo , Animales , Antioxidantes/farmacología , Ácido Ascórbico/farmacología , Línea Celular , Línea Celular Tumoral , Femenino , Técnicas de Inactivación de Genes , Humanos , Ratones Endogámicos C57BL , ARN/farmacología , Especies Reactivas de Oxígeno/metabolismo , Receptores de Ácido Retinoico/genética , Bazo/citología
15.
J Pharmacol Exp Ther ; 360(2): 289-299, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28082514

RESUMEN

Drug discovery strives for selective ligands to achieve targeted modulation of tissue function. Here we introduce engineered context-sensitive agonism as a postreceptor mechanism for tissue-selective drug action through a G protein-coupled receptor. Acetylcholine M2-receptor activation is known to mediate, among other actions, potentially dangerous slowing of the heart rate. This unwanted side effect is one of the main reasons that limit clinical application of muscarinic agonists. Herein we show that dualsteric (orthosteric/allosteric) agonists induce less cardiac depression ex vivo and in vivo than conventional full agonists. Exploration of the underlying mechanism in living cells employing cellular dynamic mass redistribution identified context-sensitive agonism of these dualsteric agonists. They translate elevation of intracellular cAMP into a switch from full to partial agonism. Designed context-sensitive agonism opens an avenue toward postreceptor pharmacologic selectivity, which even works in target tissues operated by the same subtype of pharmacologic receptor.


Asunto(s)
Descubrimiento de Drogas , Agonistas Muscarínicos/farmacología , Receptor Muscarínico M2/agonistas , Receptor Muscarínico M2/metabolismo , Regulación Alostérica/efectos de los fármacos , Animales , Células CHO , Cricetinae , Cricetulus , AMP Cíclico/metabolismo , Femenino , Corazón/efectos de los fármacos , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Masculino , Ratones , Agonistas Muscarínicos/efectos adversos , Transducción de Señal/efectos de los fármacos
16.
Biochim Biophys Acta Mol Cell Res ; 1864(1): 62-75, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27756573

RESUMEN

The cochaperone BAG3 is a central protein homeostasis factor in mechanically strained mammalian cells. It mediates the degradation of unfolded and damaged forms of the actin-crosslinker filamin through chaperone-assisted selective autophagy (CASA). In addition, BAG3 stimulates filamin transcription in order to compensate autophagic disposal and to maintain the actin cytoskeleton under strain. Here we demonstrate that BAG3 coordinates protein synthesis and autophagy through spatial regulation of the mammalian target of rapamycin complex 1 (mTORC1). The cochaperone utilizes its WW domain to contact a proline-rich motif in the tuberous sclerosis protein TSC1 that functions as an mTORC1 inhibitor in association with TSC2. Interaction with BAG3 results in a recruitment of TSC complexes to actin stress fibers, where the complexes act on a subpopulation of mTOR-positive vesicles associated with the cytoskeleton. Local inhibition of mTORC1 is essential to initiate autophagy at sites of filamin unfolding and damage. At the same time, BAG3-mediated sequestration of TSC1/TSC2 relieves mTORC1 inhibition in the remaining cytoplasm, which stimulates protein translation. In human muscle, an exercise-induced association of TSC1 with the cytoskeleton coincides with mTORC1 activation in the cytoplasm. The spatial regulation of mTORC1 exerted by BAG3 apparently provides the basis for a simultaneous induction of autophagy and protein synthesis to maintain the proteome under mechanical strain.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Reguladoras de la Apoptosis/genética , Autofagia/genética , Complejos Multiproteicos/genética , Músculo Esquelético/metabolismo , Miocitos del Músculo Liso/metabolismo , Estrés Mecánico , Serina-Treonina Quinasas TOR/genética , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/ultraestructura , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Fenómenos Biomecánicos , Línea Celular , Clonación Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Filaminas/genética , Filaminas/metabolismo , Expresión Génica , Regulación de la Expresión Génica , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Complejos Multiproteicos/metabolismo , Músculo Esquelético/citología , Miocitos del Músculo Liso/ultraestructura , Unión Proteica , Biosíntesis de Proteínas , Ratas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
17.
J Control Release ; 241: 164-173, 2016 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-27667178

RESUMEN

Gene therapy is a promising approach for chronic disorders that require continuous treatment such as cardiovascular disease. Overexpression of vasoprotective genes has generated encouraging results in animal models, but not in clinical trials. One major problem in humans is the delivery of sufficient amounts of genetic vectors to the endothelium which is impeded by blood flow, whereas prolonged stop-flow conditions impose the risk of ischemia. In the current study we have therefore developed a strategy for the efficient circumferential lentiviral gene transfer in the native endothelium under constant flow conditions. For that purpose we perfused vessels that were exposed to specially designed magnetic fields with complexes of lentivirus and magnetic nanoparticles thereby enabling overexpression of therapeutic genes such as endothelial nitric oxide synthase (eNOS) and vascular endothelial growth factor (VEGF). This treatment enhanced NO and VEGF production in the transduced endothelium and resulted in a reduction of vascular tone and increased angiogenesis. Thus, the combination of MNPs with magnetic fields is an innovative strategy for site-specific and efficient vascular gene therapy.


Asunto(s)
Endotelio Vascular/fisiología , Técnicas de Transferencia de Gen , Vectores Genéticos , Lentivirus/genética , Nanopartículas de Magnetita/química , Animales , Aorta/metabolismo , Aorta/fisiología , Endotelio Vascular/enzimología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Tono Muscular/genética , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiología , Neovascularización Fisiológica/genética , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética
18.
Nat Commun ; 6: 10156, 2015 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-26658454

RESUMEN

Despite the discovery of heterotrimeric αßγ G proteins ∼25 years ago, their selective perturbation by cell-permeable inhibitors remains a fundamental challenge. Here we report that the plant-derived depsipeptide FR900359 (FR) is ideally suited to this task. Using a multifaceted approach we systematically characterize FR as a selective inhibitor of Gq/11/14 over all other mammalian Gα isoforms and elaborate its molecular mechanism of action. We also use FR to investigate whether inhibition of Gq proteins is an effective post-receptor strategy to target oncogenic signalling, using melanoma as a model system. FR suppresses many of the hallmark features that are central to the malignancy of melanoma cells, thereby providing new opportunities for therapeutic intervention. Just as pertussis toxin is used extensively to probe and inhibit the signalling of Gi/o proteins, we anticipate that FR will at least be its equivalent for investigating the biological relevance of Gq.


Asunto(s)
Depsipéptidos/farmacología , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Animales , Ardisia/química , Línea Celular Tumoral , Depsipéptidos/química , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Humanos , Melanoma/metabolismo , Ratones , Modelos Moleculares , Estructura Molecular , Conformación Proteica , Isoformas de Proteínas , Transducción de Señal , Cola (estructura animal)/irrigación sanguínea , Vasoconstricción/efectos de los fármacos
19.
Rev. bras. saúde matern. infant ; 14(3): 241-249, Jul-Sep/2014. tab
Artículo en Portugués | LILACS, BVSAM | ID: lil-725705

RESUMEN

Identificar os fatores que determinam o aleitamento materno nas diferentes regiões do país. Métodos: os dados fazem parte da Pesquisa de Orçamento Familiar de 2002-2003 realizada no Brasil. A amostra foi de 2958 crianças de zero a um ano, representativas da população nacional. Um modelo de regressão linear (GLM) com família binomial e ligação logarítmica foi criado para cada região, com um intervalo de confiança de 90 por cento. Resultados: o aumento do número de moradores no domicilio é o principal fator negativo para a amamentação em todas as regiões do Brasil. Foram considerados como fatores desfavoráveis, no conjunto da amostra: mães com idade avançada, ter quatro ou mais moradores no domicílio, maior renda, maior escolaridade materna e uso de creche. Os fatores favoráveis foram: ter dois ou mais filhos menores de cinco anos em casa e mães de cor preta ou parda. Conclusões: as regiões que apresentaram um número maior de fatores desfavoráveis ao aleitamento materno foram Sudeste e Sul, sendo que o maior número de moradores no domicílio foi a pior situação para a efetivação da amamentação nas regiões do Brasil...


To identify factors associated with maternal breastfeeding in different regions of the country. Methods: the data form part of the Brazilian 2002-2003 Family Budget Study. The sample comprised 2958 children aged between 0 and 1 year, representative of the national population. And linear regression model with a binomial family and logarithmic connection was created for each region, with a confidence interval of 90 percent. Results: an increase in the number of people living in a household is the principal negative factor for breastfeeding in all regions of Brazil. In this sample, negative factors were found to be: mothers of advanced age, having four or more people living in the household, higher income, higher level of education of the mother and the use of a crèche. Positive factors were having two or more children at home under the age of five years and being black or of mixed race. Conclusions: the regions that had the highest number of negative factors associated with maternal breastfeeding were the Southeast and the South, with a large number of household members being the worst situation for encouraging breastfeeding in all regions of Brazil...


Asunto(s)
Humanos , Femenino , Recién Nacido , Lactante , Lactancia Materna/estadística & datos numéricos , Brasil , Factores Epidemiológicos , Topografía Médica , Demografía , Estudios Transversales
20.
Atherosclerosis ; 236(2): 400-10, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25150938

RESUMEN

OBJECTIVE: Obstructive sleep apnoea (OSA) has been implicated as a risk factor for atherosclerosis. The aim of our study was to examine the effects of chronic intermittent hypoxia in apoE-/- mice serving as model of OSA on endothelial dysfunction and oxidative stress and to evaluate the reversibility of hypoxia-induced changes under anti-inflammatory infliximab and anti-oxidative l-glutathione. METHODS: ApoE-/- mice were divided into 4 groups (n = 9 each): 1. intermittent hypoxia 8 h/day for 6 weeks, 2. intermittent hypoxia + injections of infliximab, 3. intermittent hypoxia + injections of l-glutathione, 4. normoxia = control. RESULTS: Endothelial function was impaired under hypoxia compared to control. Application of infliximab and l-glutathione improved it to a level of control. The percentage of endothelial microparticles increased under hypoxia compared to other groups. Levels of NADPH oxidase 2-derived reactive oxygen species were approximately 9 times higher in the hypoxia group. The number of sca-1/flk-1+ endothelial progenitor cells was higher in bone marrow and lower in blood under hypoxia vs. other groups. Stromal cell derived factor-1alpha- and matrix metalloproteinase-9-dependent release of these cells from bone marrow was attenuated under hypoxia. The number of DilacLDL+/lectin + early outgrowth progenitor cells and that of colony forming units from these cells were higher under hypoxia. Atherosclerotic plaques in the aorta were more frequent under hypoxia and control in comparison with both drug groups. CONCLUSION: Intermittent hypoxia contributes to endothelial dysfunction by the local increase in reactive oxygen species and reduction of the peripheral repair capacity. Infliximab and l-glutathione prevent hypoxia-induced vascular and extravascular changes.


Asunto(s)
Antiinflamatorios/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Antioxidantes/uso terapéutico , Endotelio Vascular/fisiopatología , Glutatión/uso terapéutico , Hipoxia/fisiopatología , Animales , Enfermedades de la Aorta/etiología , Apolipoproteínas E/deficiencia , Médula Ósea/fisiopatología , Micropartículas Derivadas de Células , Células Cultivadas , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Células Progenitoras Endoteliales/fisiología , Femenino , Hipoxia/sangre , Hipoxia/tratamiento farmacológico , Infliximab , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estrés Oxidativo , Placa Aterosclerótica/etiología , Especies Reactivas de Oxígeno/sangre , Apnea Obstructiva del Sueño , Bazo/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA