Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 478
Filtrar
1.
Nucleic Acids Res ; 49(22): 12970-12985, 2021 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-34878127

RESUMEN

Phosphorothioate (PS) modified antisense oligonucleotide (ASO) drugs that act on cellular RNAs must enter cells and be released from endocytic organelles to elicit antisense activity. It has been shown that PS-ASOs are mainly released by late endosomes. However, it is unclear how endosome movement in cells contributes to PS-ASO activity. Here, we show that PS-ASOs in early endosomes display Brownian type motion and migrate only short distances, whereas PS-ASOs in late endosomes (LEs) move linearly along microtubules with substantial distances. In cells with normal microtubules and LE movement, PS-ASO-loaded LEs tend to congregate perinuclearly. Disruption of perinuclear positioning of LEs by reduction of dynein 1 decreased PS-ASO activity, without affecting PS-ASO cellular uptake. Similarly, disruption of perinuclear positioning of PS-ASO-LE foci by reduction of ER tethering proteins RNF26, SQSTM1 and UBE2J1, or by overexpression of P50 all decreased PS-ASO activity. However, enhancing perinuclear positioning through reduction of USP15 or over-expression of RNF26 modestly increased PS-ASO activity, indicating that LE perinuclear positioning is required for ensuring efficient PS-ASO release. Together, these observations suggest that LE movement along microtubules and perinuclear positioning affect PS-ASO productive release.


Asunto(s)
Núcleo Celular/metabolismo , Endosomas/metabolismo , Oligonucleótidos Antisentido/metabolismo , Tionucleótidos/metabolismo , Animales , Transporte Biológico , Línea Celular Tumoral , Células Cultivadas , Dineínas/metabolismo , Retículo Endoplásmico/metabolismo , Células HeLa , Humanos , Ratones , Microscopía Confocal , Microtúbulos/metabolismo , Movimiento (Física) , Proteínas de Neoplasias/metabolismo , Oligonucleótidos Antisentido/genética , Tionucleótidos/genética
2.
Aging (Albany NY) ; 13(17): 21758-21777, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34520390

RESUMEN

Esophageal squamous cell carcinoma (ESCC) is a malignant tumor that commonly occurs worldwide. Usually, Asia, especially China, has a high incidence of esophageal cancer. ESCC often has a poor outcome because of a late diagnosis and lack of effective treatments. To build foundations for the early diagnosis and treatment of ESCC, we used the gene expression datasets GSE20347 and GSE17351 from the GEO database and a private dataset to uncover differentially expressed genes (DEGs) and key genes in ESCC. Notably, we found that replication factor C subunit 4 (RFC4) and guanine monophosphate synthase (GMPS) were upregulated but have been rarely studied in ESCC. In particular, to the best of our knowledge, our study is the first to explore GMPS and ESCC. Furthermore, we found that high levels of RFC4 and GMPS expression may result from an increase in DNA copy number alterations. Furthermore, RFC4 and GMPS were both upregulated in the early stage and early nodal metastases of esophageal carcinoma. The expression of RFC4 was strongly correlated with GMPS. In addition, we explored the relationship between RFC4 and GMPS expression and tumor-infiltrating immune cells (TILs) in esophageal carcinoma. The results showed that the levels of RFC4 and GMPS increased with a decrease in some tumor-infiltrating cells. Upregulated RFC4 and GMPS with high TILs indicate a worse prognosis. In summary, our study shows that RFC4 and GMPS have potential as biomarkers for the early diagnosis of ESCC and may played a crucial role in the process of tumor immunity in ESCC.


Asunto(s)
Biología Computacional/métodos , Variaciones en el Número de Copia de ADN , Neoplasias Esofágicas/genética , Carcinoma de Células Escamosas de Esófago/genética , Guanosina Monofosfato/genética , Proteína de Replicación C/genética , Tionucleótidos/genética , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , ADN de Neoplasias/genética , ADN de Neoplasias/metabolismo , Conjuntos de Datos como Asunto , Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas de Esófago/metabolismo , Femenino , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Guanosina Monofosfato/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Curva ROC , Proteína de Replicación C/metabolismo , Tionucleótidos/metabolismo , Regulación hacia Arriba
3.
Adv Sci (Weinh) ; 8(13): 2004929, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34258157

RESUMEN

The blood-brain barrier (BBB) is the most restrictive and complicated barrier that keeps most biomolecules and drugs from the brain. An efficient brain delivery strategy is urgently needed for the treatment of brain diseases. Based on the studies of brain-targeting extracellular vesicles (EVs), the potential of using small apoptotic bodies (sABs) from brain metastatic cancer cells for brain-targeting drug delivery is explored. It is found that anti-TNF-α antisense oligonucleotide (ASO) combined with cationic konjac glucomannan (cKGM) can be successfully loaded into sABs via a transfection/apoptosis induction process and that the sABs generated by B16F10 cells have an extraordinarily high brain delivery efficiency. Further studies suggest that ASO-loaded sABs (sCABs) are transcytosed by b. End3 (brain microvascular endothelial cells, BMECs) to penetrate the BBB, which is mediated by CD44v6, and eventually taken up by microglial cells in the brain. In a Parkinson's disease (PD) mouse model, sCABs dramatically ameliorate PD symptoms via the anti-inflammatory effect of ASO. This study suggests that sABs from brain metastatic cancer cells are excellent carriers for brain-targeted delivery, as they have not only an extraordinary delivery efficiency but also a much higher scale-up production potential than other EVs.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Neoplasias Encefálicas/tratamiento farmacológico , Vesículas Extracelulares/metabolismo , Mananos/farmacocinética , Oligonucleótidos Antisentido/farmacocinética , Animales , Neoplasias Encefálicas/metabolismo , Modelos Animales de Enfermedad , Masculino , Mananos/metabolismo , Ratones , Ratones Endogámicos C57BL , Oligonucleótidos Antisentido/metabolismo , Tionucleótidos/metabolismo , Tionucleótidos/farmacocinética
4.
Int J Mol Sci ; 22(2)2021 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-33435130

RESUMEN

Endothelial cells and astrocytes preferentially express metabotropic P2Y nucleotide receptors, which are involved in the maintenance of vascular and neural function. Among these, P2Y1 and P2Y2 receptors appear as main actors, since their stimulation induces intracellular calcium mobilization and activates signaling cascades linked to cytoskeletal reorganization. In the present work, we have analyzed, by means of atomic force microscopy (AFM) in force spectroscopy mode, the mechanical response of human umbilical vein endothelial cells (HUVEC) and astrocytes upon 2MeSADP and UTP stimulation. This approach allows for simultaneous measurement of variations in factors such as Young's modulus, maximum adhesion force and rupture event formation, which reflect the potential changes in both the stiffness and adhesiveness of the plasma membrane. The largest effect was observed in both endothelial cells and astrocytes after P2Y2 receptor stimulation with UTP. Such exposure to UTP doubled the Young's modulus and reduced both the adhesion force and the number of rupture events. In astrocytes, 2MeSADP stimulation also had a remarkable effect on AFM parameters. Additional studies performed with the selective P2Y1 and P2Y13 receptor antagonists revealed that the 2MeSADP-induced mechanical changes were mediated by the P2Y13 receptor, although they were negatively modulated by P2Y1 receptor stimulation. Hence, our results demonstrate that AFM can be a very useful tool to evaluate functional native nucleotide receptors in living cells.


Asunto(s)
Adenosina Difosfato/análogos & derivados , Astrocitos/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Receptores Purinérgicos P2Y1/metabolismo , Receptores Purinérgicos P2/metabolismo , Tionucleótidos/metabolismo , Uridina Trifosfato/metabolismo , Adenosina Difosfato/metabolismo , Adenosina Difosfato/farmacología , Astrocitos/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Microscopía de Fuerza Atómica , Transducción de Señal , Tionucleótidos/farmacología , Uridina Trifosfato/farmacología
5.
Sci Rep ; 10(1): 5045, 2020 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-32193508

RESUMEN

Osteosarcoma (OSA) is the most common type of cancer that originates in the bone and usually occurs in young children. OSA patients were treated with neoadjuvant chemotherapy and surgery, and the results were disappointing. Marine antimicrobial peptides (AMPs) have been the focus of antibiotic research because they are resistant to pathogen infection. Piscidin-1 is an AMP from the hybrid striped bass (Morone saxatilis × M. chrysops) and has approximately 22 amino acids. Research has shown that piscidin-1 can inhibit bacterial infections and has antinociception and anti-cancer properties; however, the regulatory effects of piscidin-1 on mitochondrial dysfunction in cancer cells are still unknown. We aimed to identify the effects of piscidin-1 on mitochondrial reactive oxygen species (mtROS) and apoptosis in OSA cells. Our analyses indicated that piscidin-1 has more cytotoxic effects against OSA cells than against lung and ovarian cancer cells; however, it has no effect on non-cancer cells. Piscidin-1 induces apoptosis in OSA cells, regulates mtROS, reduces mitochondrial antioxidant manganese superoxide dismutase and mitochondrial transmembrane potential, and decreases adenosine 5'-triphosphate production, thus leading to mitochondrial dysfunction and apoptosis. The mitochondrial antioxidant, mitoTempo, reduces the apoptosis induced by piscidin-1. Results suggest that piscidin-1 has potential for use in OSA treatment.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/farmacología , Antineoplásicos , Apoptosis/efectos de los fármacos , Neoplasias Óseas/patología , Proteínas de Peces/farmacología , Mitocondrias/metabolismo , Osteosarcoma/patología , Especies Reactivas de Oxígeno/metabolismo , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/metabolismo , Secuencia de Aminoácidos , Animales , Péptidos Catiónicos Antimicrobianos/química , Lubina , Proteínas de Peces/química , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/enzimología , Superóxido Dismutasa/metabolismo , Tionucleótidos/metabolismo , Células Tumorales Cultivadas
6.
Life Sci Alliance ; 3(1)2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31882444

RESUMEN

Epigenetic variation reflects the impact of a dynamic environment on chromatin. However, it remains elusive how environmental factors influence epigenetic events. Here, we show that G protein-coupled receptors (GPCRs) alter H3K4 methylation via oscillatory intracellular cAMP. Activation of Gs-coupled receptors caused a rapid decrease of H3K4me3 by elevating cAMP, whereas stimulation of Gi-coupled receptors increased H3K4me3 by diminishing cAMP. H3K4me3 gradually recovered towards baseline levels after the removal of GPCR ligands, indicating that H3K4me3 oscillates in tandem with GPCR activation. cAMP increased intracellular labile Fe(II), the cofactor for histone demethylases, through a non-canonical cAMP target-Rap guanine nucleotide exchange factor-2 (RapGEF2), which subsequently enhanced endosome acidification and Fe(II) release from the endosome via vacuolar H+-ATPase assembly. Removing Fe(III) from the media blocked intracellular Fe(II) elevation after stimulation of Gs-coupled receptors. Iron chelators and inhibition of KDM5 demethylases abolished cAMP-mediated H3K4me3 demethylation. Taken together, these results suggest a novel function of cAMP signaling in modulating histone demethylation through labile Fe(II).


Asunto(s)
AMP Cíclico/análogos & derivados , Desmetilación/efectos de los fármacos , Compuestos Ferrosos/metabolismo , Histonas/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Tionucleótidos/metabolismo , Animales , Células Cultivadas , AMP Cíclico/metabolismo , AMP Cíclico/farmacología , Silenciador del Gen , Histona Demetilasas con Dominio de Jumonji/genética , Histona Demetilasas con Dominio de Jumonji/metabolismo , Ligandos , Metilación/efectos de los fármacos , Oxidorreductasas N-Desmetilantes/genética , Oxidorreductasas N-Desmetilantes/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Procesamiento Proteico-Postraduccional/genética , Ratas , Receptores Acoplados a Proteínas G/metabolismo , Células de Schwann , Tionucleótidos/farmacología , Transfección
7.
Artículo en Inglés | MEDLINE | ID: mdl-31809961

RESUMEN

Individualized therapy involves genetic test of drug metabolism, which provides information about the initial dose and therapeutic drug monitoring for adjusting the subsequent dose. Consequently, toxic side effects are expected to be minimized and therapeutic effects to be maximized. In this study, an ultra-performance liquid chromatography tandem mass spectrometry method that was specific, accurate and sensitive was developed to simultaneously determine azathioprine two metabolites, 6-thioguanine nucleotides (6-TGN) and 6-methyl-mercaptopurine riboside (6-MMPr) in the whole blood lysate. We precipitated the sample by trifluoroacetic acid under the protection of dithiothreitol, with 6-MMPr and 6-TGN being hydrolyzed to produce 6-methymercaptopurine and 6-thioguanine. In the chromatographic separation, Waters ACQUITY BEH C18 (2.1 × 100 mm, 1.7 µm) chromatographic column was applied and gradient elution was conducted with 0.02 mol/L ammonium acetate buffer (which contains 0.3% formic acid) and acetonitrile at a flow rate of 0.4 ml/min. Tandem mass spectrometry in multiple reaction monitoring mode was applied for detection via electrospray ionization source in positive ionization mode. The analyzing process lasted for no more than 2 min. The calibration curve for each metabolite fitted a least squares model (weighed 1/X) from 1.25 to 5000 ng/ml (r2 > 0.99). The ion pairs were detected as 6-MMP m/z 167.07 â†’ 152.15, 6-TG m/z 168.06 â†’ 134.13, and internal standard m/z 171.07 â†’ 137.14. Under the guidance of FDA guidelines for bioanalytical method validation, we carried out validation and obtained satisfactory results. The method was successfully utilized for monitoring the concentrations of each metabolite from 65 affected patients who had received azathioprine maintenance therapy and achieved optimal results.


Asunto(s)
Azatioprina/sangre , Azatioprina/metabolismo , Cromatografía Líquida de Alta Presión/métodos , Monitoreo de Drogas/métodos , Espectrometría de Masas en Tándem/métodos , Adulto , Femenino , Nucleótidos de Guanina/sangre , Nucleótidos de Guanina/metabolismo , Humanos , Límite de Detección , Modelos Lineales , Masculino , Metiltioinosina/sangre , Metiltioinosina/metabolismo , Persona de Mediana Edad , Reproducibilidad de los Resultados , Tionucleótidos/sangre , Tionucleótidos/metabolismo
8.
Chembiochem ; 20(5): 727-733, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30452106

RESUMEN

Intracellular delivery and endosomal release of antisense oligonucleotides remain a significant challenge in the development of gene-targeted therapeutics. Previously, noncovalently cyclized TAT peptide (Cyc-TAT), in which the final ring-closing step is accomplished by hybridization of two short complementary γPNA segments, has been proven more efficient than its linear analogues at entering cells. As Cyc-TAT also readily accommodates a binding site, that is, an overhanging γPNA sequence, for codelivery of functional nucleic acid probes into cells, we were able to demonstrate that the overhang-Cyc-TAT penetrated into A549 cells when carrying an anti-telomerase γPNA that specifically reduced telomerase activity by over 97 %. Herein, we report that the cyclized TAT(FAM) can escape endosomes much more efficiently than the linear TAT(FAM) after LED illumination (490 nm). Based on this observation, the endosomal release of overhang-Cyc-TAT(FAM)/anti-telomerase γPNA complex can be greatly enhanced by photoactivation, thus shortening cell treatment time from 60 to 3 h, while keeping the same high efficiency in inhibiting telomerase activity inside A549 cells.


Asunto(s)
Endosomas/metabolismo , Productos del Gen tat/metabolismo , Oligonucleótidos Antisentido/metabolismo , Péptidos/metabolismo , Tionucleótidos/metabolismo , Células A549 , Ciclización , Citosol/metabolismo , Humanos
9.
Metab Brain Dis ; 33(5): 1501-1508, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29916183

RESUMEN

Chronic exposure to morphine can produce tolerance, dependence and addiction, but the underlying neurobiological basis is still incompletely understood. c-Jun, as an important component of the activator protein-1 transcription factor, is supposed to take part in regulating gene expression in AC/cAMP/PKA signaling. MicroRNA (miRNA) has emerged as a critical regulator of neuronal functions. Although a number of miRNAs have been reported to regulate the µ-opioid receptor expression, there has been no report about miRNAs to regulate chronic morphine-induced, naloxone-precipitated cAMP overshoot. Our results showed that chronic morphine pretreatment induced naloxone-precipitated cAMP overshoot in concentration- and time-dependent manners in HEK 293/µ cells. Chronic morphine pretreatment alone elevated both c-Jun protein and miR-139-5p expression levels, while dramatically artificial elevation of miR-139-5p inhibited c-Jun at the translational level. Furthermore, dramatically artificial upregulation of intracellular miR-139-5p limited chronic morphine-induced, naloxone-precipitated cAMP overshoot. These findings suggested that miR-139-5p was involved in regulating chronic morphine-induced, naloxone-precipitated cAMP overshoot in a negative feedback manner through its target c-Jun, which extends our understanding of neurobiological mechanisms underlying morphine dependence and addiction.


Asunto(s)
Adenilil Ciclasas/metabolismo , Analgésicos Opioides/farmacología , AMP Cíclico/análogos & derivados , MicroARNs/fisiología , Morfina/farmacología , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Proteínas Proto-Oncogénicas c-jun/metabolismo , Tionucleótidos/metabolismo , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Retroalimentación Fisiológica , Regulación de la Expresión Génica , Genes jun/genética , Células HEK293 , Humanos , MicroARNs/genética , Imitación Molecular , Dependencia de Morfina/genética , Dependencia de Morfina/metabolismo , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Factor de Transcripción AP-1/genética , Regulación hacia Arriba/efectos de los fármacos
10.
Braz J Med Biol Res ; 51(5): e6693, 2018 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-29561954

RESUMEN

Testosterone synthesis within Leydig cells is a calcium-dependent process. Intracellular calcium levels are regulated by different processes including ATP-activated P2X purinergic receptors, T-type Ca2+ channels modulated by the luteinizing hormone, and intracellular calcium storages recruited by a calcium-induced calcium release mechanism. On the other hand, nitric oxide (NO) is reported to have an inhibitory role in testosterone production. Based on these observations, we investigated the interaction between the purinergic and nitrergic systems in Leydig cells of adult mice. For this purpose, we recorded ATP-evoked currents in isolated Leydig cells using the whole cell patch clamp technique after treatment with L-NAME (300 µM and 1 mM), L-arginine (10, 100, 300, and 500 µM), ODQ (300 µM), and 8-Br-cGMP (100 µM). Our results show that NO produced by Leydig cells in basal conditions is insufficient to change the ATP-evoked currents and that extra NO provided by adding 300 µM L-arginine positively modulates the current through a mechanism involving the NO/cGMP signaling pathway. Thus, we report an interaction between the nitrergic and purinergic systems in Leydig cells and suggest that Ca2+ entry via the purinergic receptors can be regulated by NO.


Asunto(s)
Adenosina Trifosfato/fisiología , Células Intersticiales del Testículo/fisiología , Óxido Nítrico/fisiología , Receptores Purinérgicos/metabolismo , Potenciales de Acción , Animales , Arginina/administración & dosificación , Arginina/metabolismo , Células Cultivadas , GMP Cíclico/administración & dosificación , GMP Cíclico/análogos & derivados , GMP Cíclico/metabolismo , Masculino , Ratones , NG-Nitroarginina Metil Éster/administración & dosificación , NG-Nitroarginina Metil Éster/metabolismo , Óxido Nítrico/biosíntesis , Técnicas de Placa-Clamp , Tionucleótidos/administración & dosificación , Tionucleótidos/metabolismo
11.
Braz. j. med. biol. res ; 51(5): e6693, 2018. graf
Artículo en Inglés | LILACS | ID: biblio-889091

RESUMEN

Testosterone synthesis within Leydig cells is a calcium-dependent process. Intracellular calcium levels are regulated by different processes including ATP-activated P2X purinergic receptors, T-type Ca2+ channels modulated by the luteinizing hormone, and intracellular calcium storages recruited by a calcium-induced calcium release mechanism. On the other hand, nitric oxide (NO) is reported to have an inhibitory role in testosterone production. Based on these observations, we investigated the interaction between the purinergic and nitrergic systems in Leydig cells of adult mice. For this purpose, we recorded ATP-evoked currents in isolated Leydig cells using the whole cell patch clamp technique after treatment with L-NAME (300 μM and 1 mM), L-arginine (10, 100, 300, and 500 μM), ODQ (300 μM), and 8-Br-cGMP (100 μM). Our results show that NO produced by Leydig cells in basal conditions is insufficient to change the ATP-evoked currents and that extra NO provided by adding 300 μM L-arginine positively modulates the current through a mechanism involving the NO/cGMP signaling pathway. Thus, we report an interaction between the nitrergic and purinergic systems in Leydig cells and suggest that Ca2+ entry via the purinergic receptors can be regulated by NO.


Asunto(s)
Animales , Masculino , Ratones , Adenosina Trifosfato/fisiología , Receptores Purinérgicos/metabolismo , Células Intersticiales del Testículo/fisiología , Óxido Nítrico/fisiología , Arginina/administración & dosificación , Arginina/metabolismo , Tionucleótidos/administración & dosificación , Tionucleótidos/metabolismo , Potenciales de Acción , Células Cultivadas , GMP Cíclico/administración & dosificación , GMP Cíclico/análogos & derivados , GMP Cíclico/metabolismo , Técnicas de Placa-Clamp , NG-Nitroarginina Metil Éster/administración & dosificación , NG-Nitroarginina Metil Éster/metabolismo , Óxido Nítrico/biosíntesis
12.
Sci Rep ; 7(1): 13764, 2017 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-29062134

RESUMEN

The human P2Y1 receptor (P2Y1R) is a purinergic G-protein-coupled receptor (GPCR) that functions as a receptor for adenosine 5'-diphosphate (ADP). An antagonist of P2Y1R might potentially have antithrombotic effects, whereas agonists might serve as antidiabetic agents. On the basis of the antagonist-bound MRS2500-P2Y1R crystal structure, we constructed computational models of apo-P2Y1R and the agonist-receptor complex 2MeSADP-P2Y1R. We then performed conventional molecular dynamics (cMD) and accelerated molecular dynamics (aMD) simulations to study the conformational dynamics after binding with agonist/antagonist as well as the P2Y1R activation mechanism. We identified a new agonist-binding site of P2Y1R that is consistent with previous mutagenesis data. This new site is deeper than those of the agonist ADP in the recently simulated ADP-P2Y1R structure and the antagonist MRS2500 in the MRS2500-P2Y1R crystal structure. During P2Y1R activation, the cytoplasmic end of helix VI shifts outward 9.1 Å, the Ser1463.47-Tyr2375.58 hydrogen bond breaks, a Tyr2375.58-Val2626.37 hydrogen bond forms, and the conformation of the χ1 rotamer of Phe2696.44 changes from parallel to perpendicular to helix VI. The apo-P2Y1R system and the MRS2500-P2Y1R system remain inactive. The newly identified agonist binding site and activation mechanism revealed in this study may aid in the design of P2Y1R antagonists/agonists as antithrombotic/antidiabetic agents, respectively.


Asunto(s)
Adenosina Difosfato/análogos & derivados , Nucleótidos de Desoxiadenina/metabolismo , Receptores Purinérgicos P2Y1/química , Receptores Purinérgicos P2Y1/metabolismo , Tionucleótidos/metabolismo , Adenosina Difosfato/metabolismo , Sitios de Unión , Humanos , Simulación de Dinámica Molecular , Unión Proteica , Conformación Proteica
13.
Mol Pharmacol ; 92(5): 613-626, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28864555

RESUMEN

Traditionally, G protein-coupled receptor antagonists are classified as competitive or noncompetitive and surmountable or insurmountable based on functional antagonism. P2Y1 receptor (P2Y1R) structures showed two antagonists binding to two spatially distinct sites: nucleotide MRS2500 (orthosteric, contacting the helical bundle) and urea BPTU (allosteric, on the external receptor surface). However, the nature of their P2Y1R antagonism has not been characterized. Here we characterized BPTU antagonism at various signaling pathways activated by structurally diverse agonists. BPTU rightward shifted the concentration-response curves of both 2-methylthioadenosine 5'-diphosphate trisodium salt and MRS2365 (5'-diphosphates) in some signaling events, such as extracellular signal-regulated kinase 1/2 and label free, in a parallel manner without affecting the maximum agonist effect (Emax) but antagonized insurmountably (suppressed agonist Emax) in signaling events such as guanosine 5'-3-O-(thio)triphosphate binding and ß-arrestin2 recruitment. However, with dinucleotide Ap4A as an agonist, BPTU suppressed the Emax insurmountably in all signaling pathways. By comparison, MRS2500 behaved as surmountable antagonist rightward-shifting concentration-response curves of all three agonists in a parallel manner for all signaling pathways measured. Thus, we demonstrated a previously undocumented phenomenon that P2Y1R antagonism patterns could vary in different signaling pathways, which could be related to conformational selection, signaling amplification, and probe dependence. This phenomenon may apply generally to other receptors considering that antagonism by a specific ligand is often not compared at multiple signaling pathways. Thus, antagonism can be surmountable or insurmountable depending on the signaling pathways measured and the agonists used, which should be of broad relevance to drug discovery and disease treatment.


Asunto(s)
Antagonistas del Receptor Purinérgico P2Y/metabolismo , Antagonistas del Receptor Purinérgico P2Y/farmacología , Receptores Purinérgicos P2Y1/metabolismo , Transducción de Señal/fisiología , Adenosina Difosfato/análogos & derivados , Adenosina Difosfato/metabolismo , Adenosina Difosfato/farmacología , Regulación Alostérica/efectos de los fármacos , Regulación Alostérica/fisiología , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Humanos , Estructura Secundaria de Proteína , Receptores Purinérgicos P2Y1/química , Transducción de Señal/efectos de los fármacos , Tionucleótidos/metabolismo , Tionucleótidos/farmacología
14.
PLoS One ; 12(2): e0171335, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28192441

RESUMEN

During postnatal development rats demonstrate an age-dependent increase in NaCl chorda tympani (CT) responses and the number of functional apical amiloride-sensitive epithelial Na+ channels (ENaCs) in salt sensing fungiform (FF) taste receptor cells (TRCs). Currently, the intracellular signals that regulate the postnatal development of salt taste have not been identified. We investigated the effect of cAMP, a downstream signal for arginine vasopressin (AVP) action, on the postnatal development of NaCl responses in 19-23 day old rats. ENaC-dependent NaCl CT responses were monitored after lingual application of 8-chlorophenylthio-cAMP (8-CPT-cAMP) under open-circuit conditions and under ±60 mV lingual voltage clamp. Behavioral responses were tested using 2 bottle/24h NaCl preference tests. The effect of [deamino-Cys1, D-Arg8]-vasopressin (dDAVP, a specific V2R agonist) was investigated on ENaC subunit trafficking in rat FF TRCs and on cAMP generation in cultured adult human FF taste cells (HBO cells). Our results show that in 19-23 day old rats, the ENaC-dependent maximum NaCl CT response was a saturating sigmoidal function of 8-CPT-cAMP concentration. 8-CPT-cAMP increased the voltage-sensitivity of the NaCl CT response and the apical Na+ response conductance. Intravenous injections of dDAVP increased ENaC expression and γ-ENaC trafficking from cytosolic compartment to the apical compartment in rat FF TRCs. In HBO cells dDAVP increased intracellular cAMP and cAMP increased trafficking of γ- and δ-ENaC from cytosolic compartment to the apical compartment 10 min post-cAMP treatment. Control 19-23 day old rats were indifferent to NaCl, but showed clear preference for appetitive NaCl concentrations after 8-CPT-cAMP treatment. Relative to adult rats, 14 day old rats demonstrated significantly less V2R antibody binding in circumvallate TRCs. We conclude that an age-dependent increase in V2R expression produces an AVP-induced incremental increase in cAMP that modulates the postnatal increase in TRC ENaC and the neural and behavioral responses to NaCl.


Asunto(s)
Nervio de la Cuerda del Tímpano/efectos de los fármacos , AMP Cíclico/farmacología , Cloruro de Sodio/farmacología , Gusto/efectos de los fármacos , Adulto , Factores de Edad , Animales , Western Blotting , Células Cultivadas , Nervio de la Cuerda del Tímpano/fisiología , AMP Cíclico/análogos & derivados , AMP Cíclico/metabolismo , Desamino Arginina Vasopresina/farmacología , Canales Epiteliales de Sodio/genética , Canales Epiteliales de Sodio/metabolismo , Preferencias Alimentarias/efectos de los fármacos , Preferencias Alimentarias/fisiología , Expresión Génica/efectos de los fármacos , Humanos , Microscopía Confocal , Ratas Sprague-Dawley , Receptores de Vasopresinas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Gusto/fisiología , Papilas Gustativas/efectos de los fármacos , Papilas Gustativas/metabolismo , Papilas Gustativas/fisiología , Tionucleótidos/metabolismo , Tionucleótidos/farmacología
15.
Physiol Rep ; 5(1)2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28053225

RESUMEN

The cerebrospinal fluid (CSF) pH influences brain interstitial pH and, therefore, brain function. We hypothesized that the choroid plexus epithelium (CPE) expresses the vacuolar H+-ATPase (V-ATPase) as an acid extrusion mechanism in the luminal membrane to counteract detrimental elevations in CSF pH. The expression of mRNA corresponding to several V-ATPase subunits was demonstrated by RT-PCR analysis of CPE cells (CPECs) isolated by fluorescence-activated cell sorting. Immunofluorescence and electron microscopy localized the V-ATPase primarily in intracellular vesicles with only a minor fraction in the luminal microvillus area. The vesicles did not translocate to the luminal membrane in two in vivo models of hypocapnia-induced alkalosis. The Na+-independent intracellular pH (pHi) recovery from acidification was studied in freshly isolated clusters of CPECs. At extracellular pH (pHo) 7.4, the cells failed to display significant concanamycin A-sensitive pHi recovery (i.e., V-ATPase activity). The recovery rate in the absence of Na+ amounted to <10% of the pHi recovery rate observed in the presence of Na+ Recovery of pHi was faster at pHo 7.8 and was abolished at pHo 7.0. The concanamycin A-sensitive pHi recovery was stimulated by cAMP at pH 7.4 in vitro, but intraventricular infusion of the membrane-permeant cAMP analog 8-CPT-cAMP did not result in trafficking of the V-ATPase. In conclusion, we find evidence for the expression of a minor fraction of V-ATPase in the luminal membrane of CPECs. This fraction does not contribute to enhanced acid extrusion at high extracellular pH, but seems to be activated by cAMP in a trafficking-independent manner.


Asunto(s)
Membrana Celular/química , Plexo Coroideo/metabolismo , Concentración de Iones de Hidrógeno/efectos de los fármacos , Líquido Intracelular/química , ATPasas de Translocación de Protón Vacuolares/metabolismo , 8-Bromo Monofosfato de Adenosina Cíclica/administración & dosificación , 8-Bromo Monofosfato de Adenosina Cíclica/análogos & derivados , 8-Bromo Monofosfato de Adenosina Cíclica/metabolismo , Animales , Encéfalo/fisiología , Membrana Celular/metabolismo , Líquido Cefalorraquídeo/química , Líquido Cefalorraquídeo/enzimología , Líquido Cefalorraquídeo/fisiología , Plexo Coroideo/química , Plexo Coroideo/citología , Plexo Coroideo/ultraestructura , AMP Cíclico/análogos & derivados , AMP Cíclico/metabolismo , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/efectos adversos , Citometría de Flujo , Líquido Intracelular/enzimología , Líquido Intracelular/metabolismo , Macrólidos/administración & dosificación , Macrólidos/efectos adversos , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo , Sodio/metabolismo , Tionucleótidos/metabolismo
16.
Biol Pharm Bull ; 40(3): 365-374, 2017 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-27980245

RESUMEN

Guanosine 3',5'-cyclic monophosphate (cGMP)-dependent protein kinases (PKG) are kinases regulating diverse physiological functions including vascular smooth muscle relaxation, neuronal synaptic plasticity, and platelet activities. Certain PKG inhibitors, such as Rp-diastereomers of derivatives of guanosine 3',5'-cyclic monophosphorothioate (Rp-cGMPS), have been designed and used to study PKG-regulated cell signaling. 8-Nitroguanosine 3',5'-cyclic monophosphate (8-nitro-cGMP) is an endogenous cGMP derivative formed as a result of excess production of reactive oxygen species and nitric oxide. 8-Nitro-cGMP causes persistent activation of PKG1α through covalent attachment of cGMP moieties to cysteine residues of the enzyme (i.e., the process called protein S-guanylation). In this study, we synthesized a nitrated analogue of Rp-cGMPS, 8-nitroguanosine 3',5'-cyclic monophosphorothioate Rp-isomer (Rp-8-nitro-cGMPS), and investigated its effects on PKG1α activity. We synthesized Rp-8-nitro-cGMPS by reacting Rp-8-bromoguanosine 3',5'-cyclic monophosphorothioate (Rp-8-bromo-cGMPS) with sodium nitrite. Rp-8-Nitro-cGMPS reacted with the thiol compounds cysteine and glutathione to form Rp-8-thioalkoxy-cGMPS adducts to a similar extent as did 8-nitro-cGMP. As an important finding, a protein S-guanylation-like modification was clearly observed, by using Western blotting, in the reaction between recombinant PKG1α and Rp-8-nitro-cGMPS. Rp-8-Nitro-cGMPS inhibited PKG1α activity with an inhibitory constant of 22 µM in a competitive manner. An organ bath assay with mouse aorta demonstrated that Rp-8-nitro-cGMPS inhibited vascular relaxation induced by acetylcholine or 8-bromo-cGMP more than Rp-8-bromo-cGMPS did. These findings suggest that Rp-8-nitro-cGMPS inhibits PKG through induction of an S-guanylation-like modification by attaching the Rp-cGMPS moiety to the enzyme. Additional study is warranted to explore the potential application of Rp-8-nitro-cGMPS to biochemical and therapeutic research involving PKG1α activation.


Asunto(s)
Proteína Quinasa Dependiente de GMP Cíclico Tipo I/antagonistas & inhibidores , GMP Cíclico/análogos & derivados , Guanosina/análogos & derivados , Nitrocompuestos/farmacología , Tionucleótidos/farmacología , Vasodilatación/efectos de los fármacos , Acetilcolina , Animales , Aorta , GMP Cíclico/síntesis química , GMP Cíclico/metabolismo , GMP Cíclico/farmacología , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/metabolismo , Guanosina/metabolismo , Guanosina/farmacología , Isomerismo , Masculino , Ratones Endogámicos C57BL , Nitrocompuestos/metabolismo , Procesamiento Proteico-Postraduccional , Transducción de Señal , Tionucleótidos/síntesis química , Tionucleótidos/metabolismo
17.
Exp Neurol ; 283(Pt A): 365-74, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27401133

RESUMEN

Elevated levels of the second messenger molecule cyclic adenosine monophosphate (cAMP) are often associated with neuron sprouting and neurite extension (i.e., neuroplasticity). Phosphokinase A (PKA) is a prominent downstream target of cAMP that has been associated with neurite outgrowth. We hypothesized that rehabilitative motor training following spinal cord injuries promotes neuroplasticity via PKA activation. However, in two independent experiments, inhibition of cortical PKA using Rp-cAMPS throughout rehabilitative training robustly increased functional recovery and collateral sprouting of injured corticospinal tract axons, an indicator of neuroplasticity. Consistent with these in vivo findings, using cultured STHdh neurons, we found that Rp-cAMPS had no effect on the phosphorylation of CREB (cAMP response element-binding protein), a prominent downstream target of PKA, even with the concomitant application of the adenylate cyclase agonist forskolin to increase cAMP levels. Conversely, when cAMP levels were increased using the phosphodiesterase inhibitor IBMX, Rp-cAMPS potently inhibited CREB phosphorylation. Taken together, our results suggest that an alternate cAMP dependent pathway was involved in increasing CREB phosphorylation and neuroplasticity. This idea was supported by an in vitro neurite outgrowth assay, where inhibiting PKA did enhance neurite outgrowth. However, when PKA inhibition was combined with inhibition of EPAC2 (exchange protein directly activated by cAMP), another downstream target of cAMP in neurons, neurite outgrowth was significantly reduced. In conclusion, blocking PKA in cortical neurons of spinal cord injured rats increases neurite outgrowth of the lesioned corticospinal tract fibres and the efficacy of rehabilitative training, likely via EPAC.


Asunto(s)
Corteza Cerebral/enzimología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/rehabilitación , 1-Metil-3-Isobutilxantina/farmacología , Análisis de Varianza , Animales , Proteína de Unión a CREB/metabolismo , Línea Celular Transformada/metabolismo , Línea Celular Transformada/patología , Células Cultivadas , Corteza Cerebral/metabolismo , AMP Cíclico/análogos & derivados , AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Femenino , Ganglios Espinales/citología , Microglía/metabolismo , Microglía/patología , Neuritas/efectos de los fármacos , Neuritas/fisiología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Inhibidores de Fosfodiesterasa/farmacología , Tractos Piramidales/metabolismo , Ratas , Ratas Endogámicas Lew , Recuperación de la Función/fisiología , Tionucleótidos/metabolismo
18.
J Neuroimmunol ; 293: 82-85, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-27049566

RESUMEN

Myasthenia gravis (MG) is an autoimmune neuromuscular disorder characterised by fatigable voluntary skeletal muscle weakness. The underlying pathogenesis is complex involving adaptive autoimmune responses. Azathioprine remains a first line broad acting immunosuppressant for MG. Due to varied clinical responses to azathioprine we aimed to investigate the relationship between azathioprine metabolites and symptom control. Mild correlations between Quantitative Myasthenia Gravis Score (QMG) vs. 6-thioguanine nucleotides (R=-0.317 p=0.186) and QMG vs. lymphocyte count (R=0.402 p=0.08) were found. Azathioprine metabolite measurement should be considered in MG patients with; pancytopenia, deranged liver function or recurrent infections.


Asunto(s)
Azatioprina/metabolismo , Inmunosupresores/metabolismo , Miastenia Gravis/diagnóstico , Miastenia Gravis/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Recuento de Células , Estudios de Cohortes , Femenino , Nucleótidos de Guanina/metabolismo , Humanos , Linfocitos/patología , Masculino , Persona de Mediana Edad , Índice de Severidad de la Enfermedad , Estadística como Asunto , Tionucleótidos/metabolismo
19.
Mol Med Rep ; 12(4): 6370-6, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26260469

RESUMEN

The interstitial cells of Cajal (ICCs) are the pacemaker cells in the gastrointestinal (GI) tract. In the present study, the effects of Dangkwisoo­san (DS) on pacemaker potentials in cultured ICCs from the small intestine of the mouse were investigated. The whole­cell patch­clamp configuration was used to record pacemaker potentials from cultured ICCs and the increase in intracellular Ca2+ concentration ([Ca2+i) was analyzed in cultured ICCs using fura­2­acetoxymethyl ester. The generation of pacemaker potentials in the ICCs was observed. DS produced pacemaker depolarizations in a concentration dependent manner in current clamp mode. The 4­diphenylacetoxy­N­methyl­piperidine methiodide muscarinic M3 receptor antagonist inhibited DS­induced pacemaker depolarizations, whereas methoctramine, a muscarinic M2 receptor antagonist, did not. When guanosine 5'­[ß­thio] diphosphate (GDP­ß­S; 1 mM) was in the pipette solution, DS marginally induced pacemaker depolarizations, whereas low Na+ solution externally eliminated the generation of pacemaker potentials and inhibited the DS­induced pacemaker depolarizations. Additionally, the nonselective cation channel blocker, flufenamic acid, inhibited the DS­induced pacemaker depolarizations. Pretreatment with Ca2+­free solution and thapsigargin, a Ca2+­ATPase inhibitor in the endoplasmic reticulum, also eliminated the generation of pacemaker currents and suppressed the DS­induced pacemaker depolarizations. In addition, [Ca2+]i analysis revealed that DS increased [Ca2+]i. These results suggested that DS modulates pacemaker potentials through muscarinic M3 receptor activation in ICCs by G protein­dependent external and internal Ca2+ regulation and external Na+. Therefore, DS were observed to affect intestinal motility through ICCs.


Asunto(s)
Células Intersticiales de Cajal/efectos de los fármacos , Dolor/tratamiento farmacológico , Fitoterapia , Animales , ATPasas Transportadoras de Calcio/antagonistas & inhibidores , ATPasas Transportadoras de Calcio/metabolismo , Células Cultivadas , Diaminas/farmacología , Femenino , Motilidad Gastrointestinal/efectos de los fármacos , Guanosina Difosfato/análogos & derivados , Guanosina Difosfato/metabolismo , Intestino Delgado/citología , Intestino Delgado/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Piperidinas/farmacología , Plantas Medicinales/efectos adversos , Receptor Muscarínico M2/antagonistas & inhibidores , Receptor Muscarínico M2/metabolismo , Receptor Muscarínico M3/antagonistas & inhibidores , Receptor Muscarínico M3/metabolismo , Tapsigargina/farmacología , Tionucleótidos/metabolismo
20.
PLoS One ; 10(7): e0133244, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26226448

RESUMEN

6-Mercaptopurine (6-MP) is one of the key drugs in the treatment of many pediatric cancers, auto immune diseases and inflammatory bowel disease. 6-MP is a prodrug, converted to an active metabolite 6-thioguanine nucleotide (6-TGN) through enzymatic reaction involving thiopurine methyltransferase (TPMT). Pharmacogenomic variation observed in the TPMT enzyme produces a significant variation in drug response among the patient population. Despite 6-MP's widespread use and observed variation in treatment response, efforts at quantitative optimization of dose regimens for individual patients are limited. In addition, research efforts devoted on pharmacogenomics to predict clinical responses are proving far from ideal. In this work, we present a Bayesian population modeling approach to develop a pharmacological model for 6-MP metabolism in humans. In the face of scarcity of data in clinical settings, a global sensitivity analysis based model reduction approach is used to minimize the parameter space. For accurate estimation of sensitive parameters, robust optimal experimental design based on D-optimality criteria was exploited. With the patient-specific model, a model predictive control algorithm is used to optimize the dose scheduling with the objective of maintaining the 6-TGN concentration within its therapeutic window. More importantly, for the first time, we show how the incorporation of information from different levels of biological chain-of response (i.e. gene expression-enzyme phenotype-drug phenotype) plays a critical role in determining the uncertainty in predicting therapeutic target. The model and the control approach can be utilized in the clinical setting to individualize 6-MP dosing based on the patient's ability to metabolize the drug instead of the traditional standard-dose-for-all approach.


Asunto(s)
Teorema de Bayes , Mercaptopurina/administración & dosificación , Mercaptopurina/metabolismo , Modelos Biológicos , Profármacos/administración & dosificación , Profármacos/metabolismo , Antineoplásicos/administración & dosificación , Antineoplásicos/metabolismo , Estudios de Asociación Genética , Nucleótidos de Guanina/metabolismo , Humanos , Inmunosupresores/administración & dosificación , Inmunosupresores/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Medicina de Precisión , Tionucleótidos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA