Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 555
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 117(1): 464-471, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31852821

RESUMEN

Metabolites are increasingly appreciated for their roles as signaling molecules. To dissect the roles of metabolites, it is essential to understand their signaling pathways and their enzymatic regulations. From an RNA interference (RNAi) screen for regulators of intestinal stem cell (ISC) activity in the Drosophila midgut, we identified adenosine receptor (AdoR) as a top candidate gene required for ISC proliferation. We demonstrate that Ras/MAPK and Protein Kinase A (PKA) signaling act downstream of AdoR and that Ras/MAPK mediates the major effect of AdoR on ISC proliferation. Extracellular adenosine, the ligand for AdoR, is a small metabolite that can be released by various cell types and degraded in the extracellular space by secreted adenosine deaminase. Interestingly, down-regulation of adenosine deaminase-related growth factor A (Adgf-A) from enterocytes is necessary for extracellular adenosine to activate AdoR and induce ISC overproliferation. As Adgf-A expression and its enzymatic activity decrease following tissue damage, our study provides important insights into how the enzymatic regulation of extracellular adenosine levels under tissue-damage conditions facilitates ISC proliferation.


Asunto(s)
Adenosina Desaminasa/metabolismo , Proteínas de Drosophila/metabolismo , Enterocitos/fisiología , Células Madre Multipotentes/fisiología , Receptores Purinérgicos P1/metabolismo , Adenosina/metabolismo , Animales , Animales Modificados Genéticamente , Diferenciación Celular , Proliferación Celular , Regulación hacia Abajo , Drosophila , Proteínas de Drosophila/genética , Técnicas de Sustitución del Gen , Técnicas de Silenciamiento del Gen , Sistema de Señalización de MAP Quinasas/genética , Interferencia de ARN , Receptores Purinérgicos P1/genética
2.
Int J Mol Sci ; 24(24)2023 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-38139439

RESUMEN

Viral infections trigger inflammation by controlling ATP release. CD39 ectoenzymes hydrolyze ATP/ADP to AMP, which is converted by CD73 into anti-inflammatory adenosine (ADO). ADO is an anti-inflammatory and immunosuppressant molecule which can enhance viral persistence and severity. The CD39-CD73-adenosine axis contributes to the immunosuppressive T-reg microenvironment and may affect COVID-19 disease progression. Here, we investigated the link between CD39 expression, mostly on T-regs, and levels of CD73, adenosine, and adenosine receptors with COVID-19 severity and progression. Our study included 73 hospitalized COVID-19 patients, of which 33 were moderately affected and 40 suffered from severe infection. A flow cytometric analysis was used to analyze the frequency of T-regulatory cells (T-regs), CD39+ T-regs, and CD39+CD4+ T-cells. Plasma concentrations of adenosine, IL-10, and TGF-ß were quantified via an ELISA. An RT-qPCR was used to analyze the gene expression of CD73 and adenosine receptors (A1, A2A, A2B, and A3). T-reg cells were higher in COVID-19 patients compared to healthy controls (7.4 ± 0.79 vs. 2.4 ± 0.28; p < 0.0001). Patients also had a higher frequency of the CD39+ T-reg subset. In addition, patients who suffered from a severe form of the disease had higher CD39+ T-regs compared with moderately infected patients. CD39+CD4+ T cells were increased in patients compared to the control group. An analysis of serum adenosine levels showed a marked decrease in their levels in patients, particularly those suffering from severe illness. However, this was paralleled with a marked decline in the expression levels of CD73. IL-10 and TGF-ß levels were higher in COVID-19; in addition, their values were also higher in the severe group. In conclusion, there are distinct immunological alterations in CD39+ lymphocyte subsets and a dysregulation in the adenosine signaling pathway in COVID-19 patients which may contribute to immune dysfunction and disease progression. Understanding these immunological alterations in the different immune cell subsets and adenosine signaling provides valuable insights into the pathogenesis of the disease and may contribute to the development of novel therapeutic approaches targeting specific immune mechanisms.


Asunto(s)
Adenosina , COVID-19 , Linfocitos T Reguladores , Humanos , 5'-Nucleotidasa/genética , 5'-Nucleotidasa/metabolismo , Adenosina/metabolismo , Adenosina Trifosfato/metabolismo , Antiinflamatorios , Antígenos CD/genética , Antígenos CD/metabolismo , Progresión de la Enfermedad , Interleucina-10 , Receptores Purinérgicos P1/genética , Factor de Crecimiento Transformador beta/genética , Linfocitos T Reguladores/metabolismo
3.
PLoS Biol ; 17(4): e3000213, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30951527

RESUMEN

Adenosine modulation is considered both a paracrine signal coordinating different cells in a tissue and a stress signal. Both functions are ensured by 4 types of adenosine receptors (ARs), which have been studied individually. Mice with knockout of all ARs (quad-AR-KO) now allow enquiring the overall function of the adenosine modulation system. The observed "normal" physiology of quad-AR-KO mice indicates that ARs do not regulate homeostasis and are likely recruited to selectively control allostasis.


Asunto(s)
Alostasis/fisiología , Homeostasis/fisiología , Receptores Purinérgicos P1/metabolismo , Adenosina/fisiología , Alostasis/genética , Animales , Homeostasis/genética , Ratones , Ratones Noqueados , Receptores Purinérgicos P1/genética , Receptores Purinérgicos P1/fisiología , Transducción de Señal/fisiología
4.
Am J Physiol Cell Physiol ; 320(5): C892-C901, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33689481

RESUMEN

Adenosine receptors (ADORs) are G protein-coupled purinoceptors that have several functions including regulation of chloride secretion via cystic fibrosis transmembrane conductance regulator (CFTR) in human airway and kidney. We cloned an ADOR from Squalus acanthias (shark) that likely regulates CFTR in the rectal gland. Phylogenic and expression analyses indicate that elasmobranch ADORs are nonolfactory and appear to represent extant predecessors of mammalian ADORs. We therefore designate the shark ADOR as the A0 receptor. We coexpressed A0 with CFTR in Xenopus laevis oocytes and characterized the coupling of A0 to the chloride channel. Two-electrode voltage clamping was performed, and current-voltage (I-V) responses were recorded to monitor CFTR status. Only in A0- and CFTR-coinjected oocytes did adenosine analogs produce a significant concentration-dependent activation of CFTR consistent with its electrophysiological signature. A pharmacological profile for A0 was obtained for ADOR agonists and antagonists that differed markedly from all mammalian ADOR subtypes [agonists: R-phenyl-isopropyl adenosine (R-PIA) > S-phenyl-isopropyl adenosine (S-PIA) > CGS21680 > N6-cyclopentyladenosine (CPA) > 2-chloroadenosine (2ClAdo) > CV1808 = N6-[2-(3,5-dimethoxyphenyl)-2-(2-methylphenyl)ethyl]adenosine (DPMA) > N-ethyl-carboxyl adenosine (NECA); and antagonists: 8-cyclopentyl-1,3-dipropylxanthine (DPCPX) > PD115199 > 1,3-dimethyl-8-phenylxanthine (8PT) > CGS15943]. Structures of human ADORs permitted a high-confidence homology model of the shark A0 core that revealed unique structural features of ancestral receptors. We conclude that 1) A0 is a novel and unique adenosine receptor ancestor by functional and structural criteria; 2) A0 likely activates CFTR in vivo, and this receptor activates CFTR in oocytes, indicating an evolutionary coupling between ADORs and chloride secretion; and 3) A0 appears to be a nonolfactory evolutionary ancestor of all four mammalian ADOR subtypes.


Asunto(s)
Cloruros/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Proteínas de Peces/metabolismo , Receptores Purinérgicos P1/metabolismo , Glándula de Sal/metabolismo , Squalus acanthias/metabolismo , Animales , Clonación Molecular , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Evolución Molecular , Femenino , Proteínas de Peces/genética , Humanos , Masculino , Potenciales de la Membrana , Filogenia , Conformación Proteica , Agonistas del Receptor Purinérgico P1/farmacología , Antagonistas de Receptores Purinérgicos P1/farmacología , Receptores Purinérgicos P1/efectos de los fármacos , Receptores Purinérgicos P1/genética , Squalus acanthias/genética , Relación Estructura-Actividad , Xenopus laevis
5.
Nat Immunol ; 10(2): 195-202, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19122655

RESUMEN

The neuronal guidance molecule netrin-1 is linked to the coordination of inflammatory responses. Given that mucosal surfaces are particularly prone to hypoxia-elicited inflammation, we sought to determine the function of netrin-1 in hypoxia-induced inflammation. We detected hypoxia-inducible factor 1alpha (HIF-1alpha)-dependent induction of expression of the gene encoding netrin-1 (Ntn1) in hypoxic epithelia. Neutrophil transepithelial migration studies showed that by engaging A2B adenosine receptor (A2BAR) on neutrophils, netrin-1 attenuated neutrophil transmigration. Exogenous netrin-1 suppressed hypoxia-elicited inflammation in wild-type but not in A2BAR-deficient mice, and inflammatory hypoxia was enhanced in Ntn1(+/-) mice relative to that in Ntn1(+/+) mice. Our studies demonstrate that HIF-1alpha-dependent induction of netrin-1 attenuates hypoxia-elicited inflammation at mucosal surfaces.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia/metabolismo , Inflamación/inmunología , Factores de Crecimiento Nervioso/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Células CACO-2 , Quimiotaxis de Leucocito/inmunología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Expresión Génica , Humanos , Hipoxia/complicaciones , Hipoxia/inmunología , Subunidad alfa del Factor 1 Inducible por Hipoxia/inmunología , Immunoblotting , Inmunohistoquímica , Inmunoprecipitación , Inflamación/genética , Inflamación/metabolismo , Ratones , Membrana Mucosa/inmunología , Membrana Mucosa/metabolismo , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/inmunología , Netrina-1 , Infiltración Neutrófila/inmunología , Receptores Purinérgicos P1/genética , Receptores Purinérgicos P1/inmunología , Receptores Purinérgicos P1/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/inmunología
6.
Methods ; 180: 19-26, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32061675

RESUMEN

Membrane proteins (MPs) are important drug discovery targets for a wide range of diseases. Conventional detergents such as n-Dodecyl ß-D-maltoside have been used largely and efficiently to solubilize MPs with varying degrees of success concerning MPs functionality and stability. Fluorinated surfactants (FSs) have shown a great potential for the stabilization of various MPs. However, so far only a limited number of reports have demonstrated the ability of FSs to solubilize MPs from biological membranes. We report herein the use of a fluorinated lactobionamide-based detergent named FLAC6 for functional and structural stabilization of membrane proteins. We first demonstrated that FLAC6 efficiently solubilized three membrane proteins i.e. the native adenosine receptor A2AR, a G protein-coupled receptor, and two native transporters AcrB and BmrA. The resulting affinity purified MPs were highly pure, homogenous and aggregates free. Furthermore, the functionality of each MP was well maintained. Finally, striking overstabilization features were observed. Indeed, the Tm of native A2AR, AcrB and BmrA could be improved by 7, ~9 and ~ 23 °C, respectively when FLAC6 was used instead of the reference detergent. This work illustrates that FLAC6 is an efficient tool to maintain structural and functional integrities of different MPs belonging to different classes, providing a new avenue for functional stabilization of highly druggable and challenging membrane proteins involved in unmet medical needs.


Asunto(s)
Detergentes/química , Disacáridos/química , Proteínas de la Membrana/química , Animales , Cromatografía en Gel , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Expresión Génica , Halogenación , Proteínas de la Membrana/genética , Proteínas de la Membrana/aislamiento & purificación , Proteínas de la Membrana/metabolismo , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/química , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores Purinérgicos P1/química , Receptores Purinérgicos P1/genética , Receptores Purinérgicos P1/metabolismo , Células Sf9 , Solubilidad , Tensoactivos/química
7.
Am J Physiol Cell Physiol ; 318(5): C832-C835, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32159362

RESUMEN

Danger sensing is one of the most fundamental evolutionary features enabling multicellular organisms to perceive potential threats, escape from risky situations, fight actual intruders, and repair damage. Several endogenous molecules are used to "signal damage," currently referred to as "alarmins" or "damage-associated molecular patterns" (DAMPs), most being already present within all cells (preformed DAMPs), and thus ready to be released, and others neosynthesized following injury. Over recent years it has become overwhelmingly clear that adenosine 5'-triphosphate (ATP) is a ubiquitous and extremely efficient DAMP (thus promoting inflammation), and its main metabolite, adenosine, is a strong immunosuppressant (thus dampening inflammation). Extracellular ATP ligates and activates the P2 purinergic receptors (P2Rs) and is then degraded by soluble and plasma membrane ecto-nucleotidases to generate adenosine acting at P1 purinergic receptors (P1Rs). Extracellular ATP, P2Rs, ecto-nucleotidases, adenosine, and P1Rs are basic elements of the purinergic signaling network and fundamental pillars of inflammation.


Asunto(s)
Alarminas/genética , Inflamación/metabolismo , Receptores Purinérgicos P1/genética , Receptores Purinérgicos P2/genética , Adenosina/metabolismo , Adenosina Trifosfatasas/genética , Adenosina Trifosfato/metabolismo , Alarminas/metabolismo , Animales , Membrana Celular/metabolismo , Humanos , Inmunosupresores/metabolismo , Inflamación/fisiopatología , Receptores Purinérgicos P2/metabolismo , Transducción de Señal/genética
8.
Int J Mol Sci ; 21(20)2020 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-33050467

RESUMEN

Adenosine is an extracellular signaling molecule that is particularly relevant in times of cellular stress, inflammation and metabolic disturbances when the levels of the purine increase. Adenosine acts on two G-protein-coupled stimulatory and on two G-protein-coupled inhibitory receptors, which have varying expression profiles in different tissues and conditions, and have different affinities for the endogenous ligand. Studies point to significant roles of adenosine and its receptors in metabolic disease and bone health, implicating the receptors as potential therapeutic targets. This review will highlight our current understanding of the dichotomous effects of adenosine and its receptors on adipogenesis versus osteogenesis within the bone marrow to maintain bone health, as well as its relationship to obesity. Therapeutic implications will also be reviewed.


Asunto(s)
Adipogénesis/genética , Tejido Adiposo/metabolismo , Médula Ósea/metabolismo , Osteogénesis/genética , Receptores Purinérgicos P1/genética , Receptores Purinérgicos P1/metabolismo , Animales , Células de la Médula Ósea/metabolismo , Diferenciación Celular/genética , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Transducción de Señal
9.
J Cell Physiol ; 234(3): 2329-2336, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30191994

RESUMEN

The hypoxic niche of tumor leads to a tremendous increase in the extracellular adenosine concentration through alteration of adenosine metabolism in the tumor microenvironment (TME). This consequently affects cancer progression, local immune responses, and apoptosis of tumor cells. Regulatory effect of adenosine on apoptosis in TME depends on the cancer cell type, pharmacological characteristics of adenosine receptor subtypes, and the adenosine concentration in the tumor niche. Exploiting specific pharmacological adenosine receptor agonist and antagonist inducing apoptosis in cancer cells can be considered as a proper procedure to control cancer progression. This review summarizes the regulatory role of adenosine in cancer cell apoptosis for a better understanding, and hence better management of the disease.


Asunto(s)
Neoplasias/tratamiento farmacológico , Agonistas del Receptor Purinérgico P1/uso terapéutico , Antagonistas de Receptores Purinérgicos P1/uso terapéutico , Receptores Purinérgicos P1/genética , Adenosina Trifosfato/genética , Apoptosis/efectos de los fármacos , Neoplasias/genética , Neoplasias/patología , Transducción de Señal/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos
10.
J Cell Physiol ; 234(5): 5863-5879, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-29271489

RESUMEN

Maintenance of the endothelial cell (EC) barrier is critical to vascular homeostasis and a loss of barrier integrity results in increased vascular permeability. While the mechanisms that govern increased EC permeability have been under intense investigation over the past several decades, the processes regulating the preservation/restoration of the EC barrier remain poorly understood. Herein we show that the extracellular purines, adenosine (Ado) and adenosine 5'-[γ-thio]-triphosphate (ATPγS) can strengthen the barrier function of human lung microvascular EC (HLMVEC). This ability involves protein kinase A (PKA) activation and decreases in myosin light chain 20 (MLC20) phosphorylation secondary to the involvement of MLC phosphatase (MLCP). In contrast to Ado, ATPγS-induced PKA activation is accompanied by a modest, but significant decrease in cyclic adenosine monophosphate (cAMP) levels supporting the existence of an unconventional cAMP-independent pathway of PKA activation. Furthermore, ATPγS-induced EC barrier strengthening does not involve the Rap guanine nucleotide exchange factor 3 (EPAC1) which is directly activated by cAMP but is instead dependent upon PKA-anchor protein 2 (AKAP2) expression. We also found that AKAP2 can directly interact with the myosin phosphatase-targeting protein MYPT1 and that depletion of AKAP2 abolished ATPγS-induced increases in transendothelial electrical resistance. Ado-induced strengthening of the HLMVEC barrier required the coordinated activation of PKA and EPAC1 in a cAMP-dependent manner. In summary, ATPγS-induced enhancement of the EC barrier is EPAC1-independent and is instead mediated by activation of PKA which is then guided by AKAP2, in a cAMP-independent mechanism, to activate MLCP which dephosphorylates MLC20 resulting in reduced EC contraction and preservation.


Asunto(s)
Adenosina Trifosfato/análogos & derivados , Permeabilidad Capilar/efectos de los fármacos , Microvasos/efectos de los fármacos , Agonistas del Receptor Purinérgico P1/farmacología , Receptores Purinérgicos P1/efectos de los fármacos , Proteínas de Anclaje a la Quinasa A/genética , Proteínas de Anclaje a la Quinasa A/metabolismo , Adenosina Trifosfato/farmacología , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Impedancia Eléctrica , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Células HEK293 , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Microvasos/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Fosfatasa de Miosina de Cadena Ligera/genética , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Fosforilación , Receptores Purinérgicos P1/genética , Receptores Purinérgicos P1/metabolismo , Transducción de Señal
11.
Tumour Biol ; 42(4): 1010428319837138, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30957676

RESUMEN

The mechanisms leading to immune escape of melanoma have been largely investigated in relation to its tumour immunogenicity and features of inflamed microenvironment that promote the immune suppression during the disease progression. These findings have recently led to advantages in terms of immunotherapy-based approaches as rationale for overcoming the immune escape. However, besides immune checkpoints, other mechanisms including the adenosine produced by ectonucleotidases CD39 and CD73 contribute to the melanoma progression due to the immunosuppression induced by the tumour milieu. On the other hand, CD73 has recently emerged as both promising therapeutic target and unfavourable prognostic biomarker. Here, we review the major mechanisms of immune escape activated by the CD39/CD73/adenosine pathway in melanoma and focus potential therapeutic strategies based on the control of CD39/CD73 downstream adenosine receptor signalling. These evidences provide the basis for translational strategies of immune combination, while CD73 would serve as potential prognostic biomarker in metastatic melanoma.


Asunto(s)
5'-Nucleotidasa/inmunología , Adenosina/inmunología , Melanoma/inmunología , Microambiente Tumoral/inmunología , 5'-Nucleotidasa/genética , Adenosina/genética , Apirasa/genética , Apirasa/inmunología , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/inmunología , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/inmunología , Humanos , Tolerancia Inmunológica/genética , Terapia de Inmunosupresión , Inmunoterapia/métodos , Melanoma/patología , Receptores Purinérgicos P1/genética , Receptores Purinérgicos P1/inmunología , Transducción de Señal/inmunología
12.
BMC Biol ; 16(1): 24, 2018 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-29486745

RESUMEN

BACKGROUND: G-protein-coupled receptor (GPCR) heteromeric complexes have distinct properties from homomeric GPCRs, giving rise to new receptor functionalities. Adenosine receptors (A1R or A2AR) can form A1R-A2AR heteromers (A1-A2AHet), and their activation leads to canonical G-protein-dependent (adenylate cyclase mediated) and -independent (ß-arrestin mediated) signaling. Adenosine has different affinities for A1R and A2AR, allowing the heteromeric receptor to detect its concentration by integrating the downstream Gi- and Gs-dependent signals. cAMP accumulation and ß-arrestin recruitment assays have shown that, within the complex, activation of A2AR impedes signaling via A1R. RESULTS: We examined the mechanism by which A1-A2AHet integrates Gi- and Gs-dependent signals. A1R blockade by A2AR in the A1-A2AHet is not observed in the absence of A2AR activation by agonists, in the absence of the C-terminal domain of A2AR, or in the presence of synthetic peptides that disrupt the heteromer interface of A1-A2AHet, indicating that signaling mediated by A1R and A2AR is controlled by both Gi and Gs proteins. CONCLUSIONS: We identified a new mechanism of signal transduction that implies a cross-communication between Gi and Gs proteins guided by the C-terminal tail of the A2AR. This mechanism provides the molecular basis for the operation of the A1-A2AHet as an adenosine concentration-sensing device that modulates the signals originating at both A1R and A2AR.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores Purinérgicos P1/metabolismo , Transducción de Señal/fisiología , Secuencia de Aminoácidos , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/química , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Subunidades alfa de la Proteína de Unión al GTP Gs/química , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Células HEK293 , Humanos , Estructura Terciaria de Proteína , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , Receptores Purinérgicos P1/química , Receptores Purinérgicos P1/genética
13.
Int J Mol Sci ; 20(12)2019 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-31216722

RESUMEN

Hearing impairment is the most common sensory deficit, affecting more than 400 million people worldwide. Sensorineural hearing losses currently lack any specific or efficient pharmacotherapy largely due to the insufficient knowledge of the pathomechanism. Purinergic signaling plays a substantial role in cochlear (patho)physiology. P2 (ionotropic P2X and the metabotropic P2Y) as well as adenosine receptors expressed on cochlear sensory and non-sensory cells are involved mostly in protective mechanisms of the cochlea. They are implicated in the sensitivity adjustment of the receptor cells by a K+ shunt and can attenuate the cochlear amplification by modifying cochlear micromechanics. Cochlear blood flow is also regulated by purines. Here, we propose to comprehend this field with the purine-immune interactions in the cochlea. The role of harmful immune mechanisms in sensorineural hearing losses has been emerging in the horizon of cochlear pathologies. In addition to decreasing hearing sensitivity and increasing cochlear blood supply, influencing the immune system can be the additional avenue for pharmacological targeting of purinergic signaling in the cochlea. Elucidating this complexity of purinergic effects on cochlear functions is necessary and it can result in development of new therapeutic approaches in hearing disabilities, especially in the noise-induced ones.


Asunto(s)
Cóclea/inmunología , Cóclea/metabolismo , Enfermedades Cocleares/etiología , Enfermedades Cocleares/metabolismo , Transducción de Señal , Animales , Calcio/metabolismo , Cóclea/fisiología , Cóclea/ultraestructura , Enfermedades Cocleares/tratamiento farmacológico , Enfermedades Cocleares/fisiopatología , Expresión Génica , Pérdida Auditiva Sensorineural/etiología , Pérdida Auditiva Sensorineural/metabolismo , Pérdida Auditiva Sensorineural/fisiopatología , Humanos , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Purinérgicos/metabolismo , Receptores Purinérgicos/genética , Receptores Purinérgicos/metabolismo , Receptores Purinérgicos P1/genética , Receptores Purinérgicos P1/metabolismo
14.
Int J Mol Sci ; 20(23)2019 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-31775376

RESUMEN

Ischemic postconditioning (IPoC) reduces reperfusion arrhythmias but the antiarrhythmic mechanisms remain unknown. The aim of this study was to analyze IPoC electrophysiological effects and the role played by adenosine A1, A2A and A3 receptors, protein kinase C, ATP-dependent potassium (KATP) channels, and connexin 43. IPoC reduced reperfusion arrhythmias (mainly sustained ventricular fibrillation) in isolated rat hearts, an effect associated with a transient delay in epicardial electrical activation, and with action potential shortening. Electrical impedance measurements and Lucifer-Yellow diffusion assays agreed with such activation delay. However, this delay persisted during IPoC in isolated mouse hearts in which connexin 43 was replaced by connexin 32 and in mice with conditional deletion of connexin 43. Adenosine A1, A2A and A3 receptor blockade antagonized the antiarrhythmic effect of IPoC and the associated action potential shortening, whereas exogenous adenosine reduced reperfusion arrhythmias and shortened action potential duration. Protein kinase C inhibition by chelerythrine abolished the protective effect of IPoC but did not modify the effects on action potential duration. On the other hand, glibenclamide, a KATP inhibitor, antagonized the action potential shortening but did not interfere with the antiarrhythmic effect. The antiarrhythmic mechanisms of IPoC involve adenosine receptor activation and are associated with action potential shortening. However, this action potential shortening is not essential for protection, as it persisted during protein kinase C inhibition, a maneuver that abolished IPoC protection. Furthermore, glibenclamide induced the opposite effects. In addition, IPoC delays electrical activation and electrical impedance recovery during reperfusion, but these effects are independent of connexin 43.


Asunto(s)
Arritmias Cardíacas/prevención & control , Conexina 43/fisiología , Poscondicionamiento Isquémico/métodos , Canales KATP/metabolismo , Isquemia Miocárdica/complicaciones , Proteína Quinasa C/metabolismo , Receptores Purinérgicos P1/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Arritmias Cardíacas/etiología , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/patología , Canales KATP/genética , Ratones , Ratones Transgénicos , Proteína Quinasa C/genética , Ratas , Ratas Sprague-Dawley , Receptores Purinérgicos P1/genética
15.
Int J Mol Sci ; 20(6)2019 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-30909368

RESUMEN

BACKGROUND: Vascular endothelial injury during ischemia generates apoptotic cell death and precedes apoptosis of underlying tissues. We aimed at studying the role of extracellular adenosine triphosphate (ATP) on endothelial cells protection against hypoxia injury. METHODS: In a hypoxic model on endothelial cells, we quantified the extracellular concentration of ATP and adenosine. The expression of mRNA (ectonucleotidases, adenosine, and P2 receptors) was measured. Apoptosis was evaluated by the expression of cleaved caspase 3. The involvement of P2 and adenosine receptors and signaling pathways was investigated using selective inhibitors. RESULTS: Hypoxic stress induced a significant increase in extracellular ATP and adenosine. After a 2-h hypoxic injury, an increase of cleaved caspase 3 was observed. ATP anti-apoptotic effect was prevented by suramin, pyridoxalphosphate-6-azophenyl-2',4'-disulfonic acid (PPADS), and CGS15943, as well as by selective A2A, A2B, and A3 receptor antagonists. P2 receptor-mediated anti-apoptotic effect of ATP involved phosphoinositide 3-kinase (PI3K), extracellular signal-regulated kinases (ERK1/2), mitoKATP, and nitric oxide synthase (NOS) pathways whereas adenosine receptor-mediated anti-apoptotic effect involved ERK1/2, protein kinase A (PKA), and NOS. CONCLUSIONS: These results suggest a complementary role of P2 and adenosine receptors in ATP-induced protective effects against hypoxia injury of endothelial. This could be considered therapeutic targets to limit the development of ischemic injury of organs such as heart, brain, and kidney.


Asunto(s)
Adenosina Trifosfato/metabolismo , Apoptosis , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Hipoxia/metabolismo , Receptores Purinérgicos P1/metabolismo , Receptores Purinérgicos P2/metabolismo , Adenosina/metabolismo , Apoptosis/genética , Biomarcadores , Espacio Extracelular/metabolismo , Expresión Génica , Humanos , Hipoxia/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Óxido Nítrico Sintasa/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , ARN Mensajero/genética , Receptores Purinérgicos P1/genética , Receptores Purinérgicos P2/genética , Transducción de Señal , Estrés Fisiológico/genética
16.
Biochim Biophys Acta Mol Cell Res ; 1864(12): 2438-2448, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28965824

RESUMEN

Theobromine, a methylxanthine derived from cacao beans, reportedly has various health-promoting properties but molecular mechanism by which effects of theobromine on adipocyte differentiation and adipogenesis remains unclear. In this study, we aimed to clarify the molecular mechanisms of the anti-adipogenic effect of theobromine in vitro and in vivo. ICR mice (4week-old) were administered with theobromine (0.1g/kg) for 7days. Theobromine administration attenuated gains in body and epididymal adipose tissue weights in mice and suppressed expression of adipogenic-associated genes in mouse adipose tissue. In 3T3-L1 preadipocytes, theobromine caused degradation of C/EBPß protein by the ubiquitin-proteasome pathway. Pull down assay showed that theobromine selectively interacts with adenosine receptor A1 (AR1), and AR1 knockdown inhibited theobromine-induced C/EBPß degradation. Theobromine increased sumoylation of C/EBPß at Lys133. Expression of the small ubiquitin-like modifier (SUMO)-specific protease 2 (SENP2) gene, coding for a desumoylation enzyme, was suppressed by theobromine. In vivo knockdown studies showed that AR1 knockdown in mice attenuated the anti-adipogenic effects of theobromine in younger mice. Theobromine suppresses adipocyte differentiation and induced C/EBPß degradation by increasing its sumoylation. Furthermore, the inhibition of AR1 signaling is important for theobromine-induced C/EBPß degradation.


Asunto(s)
Adipogénesis/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Cisteína Endopeptidasas/genética , Receptores Purinérgicos P1/genética , Células 3T3-L1 , Adipocitos/metabolismo , Adipogénesis/efectos de los fármacos , Animales , Diferenciación Celular/genética , Cisteína Endopeptidasas/metabolismo , Ratones , Proteolisis/efectos de los fármacos , Transducción de Señal , Sumoilación/genética , Teobromina/administración & dosificación
17.
J Biol Chem ; 292(4): 1211-1217, 2017 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-27974461

RESUMEN

T follicular helper (TFH) cells have been shown to be critically required for the germinal center (GC) reaction where B cells undergo class switch recombination and clonal selection to generate high affinity neutralizing antibodies. However, detailed knowledge of the physiological cues within the GC microenvironment that regulate T cell help is limited. The cAMP-elevating, Gs protein-coupled A2a adenosine receptor (A2aR) is an evolutionarily conserved receptor that limits and redirects cellular immunity. However, the role of A2aR in humoral immunity and B cell differentiation is unknown. We hypothesized that the hypoxic microenvironment within the GC facilitates an extracellular adenosine-rich milieu, which serves to limit TFH frequency and function, and also promotes immunosuppressive T follicular regulatory cells (TFR). In support of this hypothesis, we found that following immunization, mice lacking A2aR (A2aRKO) exhibited a significant expansion of T follicular cells, as well as increases in TFH to TFR ratio, GC T cell frequency, GC B cell frequency, and class switching of GC B cells to IgG1. Transfer of CD4 T cells from A2aRKO or wild type donors into T cell-deficient hosts revealed that these increases were largely T cell-intrinsic. Finally, injection of A2aR agonist, CGS21680, following immunization suppressed T follicular differentiation, GC B cell frequency, and class switching of GC B cells to IgG1. Taken together, these observations point to a previously unappreciated role of GS protein-coupled A2aR in regulating humoral immunity, which may be pharmacologically targeted during vaccination or pathological states in which GC-derived autoantibodies contribute to the pathology.


Asunto(s)
Autoanticuerpos/inmunología , Centro Germinal/inmunología , Inmunidad Humoral , Cambio de Clase de Inmunoglobulina/inmunología , Inmunoglobulina G/inmunología , Receptores Purinérgicos P1/inmunología , Linfocitos T Reguladores/inmunología , Adenosina/análogos & derivados , Adenosina/farmacología , Animales , Autoanticuerpos/genética , Linfocitos B/inmunología , Cambio de Clase de Inmunoglobulina/efectos de los fármacos , Inmunoglobulina G/genética , Ratones , Ratones Noqueados , Fenetilaminas/farmacología , Receptores Purinérgicos P1/genética
18.
Am J Physiol Regul Integr Comp Physiol ; 315(3): R500-R508, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29791204

RESUMEN

Fetal anemia causes rapid and profound changes in cardiac structure and function, stimulating proliferation of the cardiac myocytes, expansion of the coronary vascular tree, and impairing early contraction and relaxation. Although hypoxia-inducible factor-1α is sure to play a role, adenosine, a metabolic byproduct that increases coronary flow and growth, is implicated as a major stimulus for these adaptations. We hypothesized that genes involved in myocardial adenosine signaling would be upregulated in chronically anemic fetuses and that calcium-handling genes would be downregulated. After sterile surgical instrumentation under anesthesia, gestationally timed fetal sheep were made anemic by isovolumetric hemorrhage for 1 wk (16% vs. 35% hematocrit). At 87% of gestation, necropsy was performed to collect heart tissue for PCR and immunohistochemical analysis. Anemia increased mRNA expression levels of adenosine receptors ADORA 1, ADORA2A, and ADORA2B in the left and right ventricles (adenosine receptor ADORA3 was unchanged). In both ventricles, anemia also increased expression of ectonucleoside triphosphate diphosphohydrolase 1 and ecto-5'-nucleotidase. The genes for both equilibrative nucleoside transporters 1 and 2 were expressed more abundantly in the anemic right ventricle but were not different in the left ventricle. Neither adenosine deaminase nor adenosine kinase cardiac levels were significantly changed by chronic fetal anemia. Chronic fetal anemia did not significantly change cardiac mRNA expression levels of the voltage-dependent L-type calcium channel, ryanodine receptor 1, sodium-calcium exchanger, sarcoplasmic/endoplasmic reticulum calcium transporting ATPase 2, phospholamban, or cardiac calsequestrin. These data support local metabolic integration of vascular and myocyte function through adenosine signaling in the anemic fetal heart.


Asunto(s)
Adenosina/metabolismo , Anemia/metabolismo , Señalización del Calcio , Vasos Coronarios/metabolismo , Enfermedades Fetales/metabolismo , Miocitos Cardíacos/metabolismo , Neovascularización Fisiológica , 5'-Nucleotidasa/genética , 5'-Nucleotidasa/metabolismo , Anemia/sangre , Anemia/embriología , Anemia/genética , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Apirasa/genética , Apirasa/metabolismo , Señalización del Calcio/genética , Enfermedad Crónica , Vasos Coronarios/embriología , Modelos Animales de Enfermedad , Proteínas de Transporte de Nucleósido Equilibrativas/genética , Proteínas de Transporte de Nucleósido Equilibrativas/metabolismo , Femenino , Enfermedades Fetales/sangre , Enfermedades Fetales/genética , Regulación del Desarrollo de la Expresión Génica , Neovascularización Fisiológica/genética , Embarazo , Receptores Purinérgicos P1/genética , Receptores Purinérgicos P1/metabolismo , Oveja Doméstica
19.
J Cell Biochem ; 118(8): 2371-2379, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28106278

RESUMEN

Diabetic macular edema (DME) is the major cause of vision loss in patients affected by diabetic retinopathy. Hyperglycemia and hypoxia represent the key elements in the progression of these pathologies, leading to breakdown of the blood-retinal barrier (BRB). Caffeine, a psychoactive substance largely consumed in the world, is a nonselective antagonist of adenosine receptors (AR) and it possesses a protective effect in various diseases, including eye pathologies. Here, we have investigated the effect of this substance on BRB integrity following exposure to hyperglycemic/hypoxic insult. Retinal pigmented epithelial cells, ARPE-19, have been grown on semi-permeable supports mimicking an experimental model, in vitro, of outer BRB. Caffeine treatment has reduced cell monolayer permeability after exposure to high glucose and desferoxamine as shown by TEER and FITC-dextran permeability assays. This effect is also mediated through the restoration of membrane's tight junction expression, ZO-1. Moreover, we have demonstrated that caffeine is able to prevent outer BRB damage by inhibiting apoptotic cell death induced by hyperglycemic/hypoxic insult since it downregulates the proapoptotic Bax and upregulates the anti-apoptotic Bcl-2 genes. Although further studies are needed to better comprise the beneficial effect of caffeine, we can speculate that it might be used as an innovative drug for DME treatment. J. Cell. Biochem. 118: 2371-2379, 2017. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Barrera Hematorretinal/efectos de los fármacos , Barrera Hematorretinal/metabolismo , Cafeína/farmacología , Edema Macular/metabolismo , Western Blotting , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Línea Celular , Humanos , Hiperglucemia/metabolismo , Antagonistas de Receptores Purinérgicos P1/farmacología , ARN Mensajero/genética , Receptores Purinérgicos P1/genética , Receptores Purinérgicos P1/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo , Hipoxia Tumoral/fisiología
20.
Clin Exp Rheumatol ; 35(6): 943-947, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28598776

RESUMEN

OBJECTIVES: We sought further understanding of the association between methotrexate (MTX) therapy and accelerated development of subcutaneous rheumatoid nodules. The objective was to establish expression of genes involved in the transport, metabolism, and mechanism of action of MTX within nodule tissue. We also examined for differences in gene expression between nodules from patients actively receiving MTX compared to those not receiving MTX. METHODS: Subcutaneous nodule tissues (n=23) were obtained from 21 patients with RA, undergoing elective surgery. Expression of genes important to the transport (SLC19A1, ABCB1, ABCC1, ABCG2), metabolism (FPGS, GGH), and mechanism of action (TYMS, MTR, MTRR) of MTX, including for the adenosine receptors ADORA1, ADORA2A, ADORA2B, ADORA3 and ADORA3variant were quantitated by real-time PCR in each nodule sample. RESULTS: Transcripts for all genes were found in all nodules. Expression of MTR was significantly reduced in nodules from patients receiving MTX therapy. Patterns of gene expression differed, with those metabolising MTX more prominent in nodules from patients receiving MTX when compared to nodules from those not receiving MTX, where genes involved in MTX transport were more prominent. CONCLUSIONS: Genes involved in MTX handling are expressed in rheumatoid nodules, providing further evidence that metabolism of MTX within nodules could exert a local effect. Furthermore the profile of gene expression in nodules differed from that previously observed in rheumatoid synovial membrane. The significant reduction of MTR expression in nodules has implications for MTR- and MTRR-mediated re-methylation reactions. Our data suggest that in contrast to synovium, downstream methylation reactions involving methionine and the biosynthesis of S-adenosylmethionine (SAM) could be reduced in nodule tissue. This could help explain differing responses to MTX in rheumatoid nodules and synovium and warrants further investigation.


Asunto(s)
Artritis Reumatoide/tratamiento farmacológico , Metotrexato/efectos adversos , Nódulo Reumatoide/inducido químicamente , 5-Metiltetrahidrofolato-Homocisteína S-Metiltransferasa/genética , Anciano , Femenino , Humanos , Masculino , Metotrexato/metabolismo , Metotrexato/farmacología , Metilación , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Receptores Purinérgicos P1/genética , Nódulo Reumatoide/genética , S-Adenosilmetionina/metabolismo , Transcriptoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA