Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Gene ; 570(1): 1-7, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26079571

RESUMEN

Bone development and homeostasis require the interplay between several cell types, including mesenchymal osteoblasts and osteocytes, as well as hematopoietic osteoclasts. Recent evidence suggests that cell proliferation, differentiation and apoptosis of both mesenchymal and hematopoietic stem cells, which are fundamental for tissue regeneration and treatment of degenerative diseases, are controlled by P2 receptors (i.e., P2X and P2Y receptors). Both types of P2 receptors are versatile transducers of diverse signals activated by extracellular nucleotides like ATP that are released in response to tissue injury, infection or shear stress. The P2X family of receptors has been shown to mediate multiple signaling events including the influx of calcium, activation of mitogen activated protein kinases (MAPKs) and induction of AP-1 family members known to regulate bone development. Support for the significance of P2X7 in regulating bone development and homeostasis has been provided by several studies focusing on animal models and single nucleotide polymorphisms. P2 receptors are functionally expressed in both bone forming osteoblasts and bone resorbing osteoclasts, while recent findings also suggest that these receptors translate mechanical stimuli in osteocytes. Their ability to respond to external nucleotide analogs renders these cell surface proteins excellent targets for skeletal regenerative therapies. This overview summarizes mechanisms by which nucleotide receptors control skeletal cells and contribute to bone tissue development remodeling and repair.


Asunto(s)
Desarrollo Óseo , Células Madre Hematopoyéticas/fisiología , Células Madre Mesenquimatosas/fisiología , Receptores Purinérgicos P2X/fisiología , Receptores Purinérgicos P2Y/fisiología , Animales , Humanos , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteogénesis , Transducción de Señal
2.
PLoS One ; 8(5): e63789, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23691096

RESUMEN

BACKGROUND: The P2X7 receptor binds extracellular ATP to mediate numerous inflammatory responses and is considered a potential biomarker and therapeutic target for diverse inflammatory and neurological diseases. P2X7 contains many single nucleotide polymorphisms, including several mutations located within its intracellular C-terminal trafficking domain. Mutations within the trafficking domain result in attenuated receptor activity and cell surface presentation, but the mechanisms by which amino acid changes within this region promote altered P2X7 function have not been elucidated. METHODS AND RESULTS: We analyzed the amino acid sequence of P2X7 for any potential trafficking signals and found that P2X7 contains putative Arg-X-Arg ER retention sequences. Alanine substitutions near or within these sequences were constructed, and we determined that single mutation of R574 and R578 but not R576 or K579 attenuates P2X7-stimulated activation of ERK1/2 and induction of the transcription factors FosB and ΔFosB. We found that mutation of R578 within the trafficking domain to the naturally occurring Gln substitution disrupts P2X7 localization at the plasma membrane and results in R578Q displaying a higher apparent molecular weight in comparison to wild-type receptor. We used the glycosidase endoglycosidase H to determine that this difference in mass is due in part to the R578Q mutant possessing a larger mass of oligosaccharides, indicative of improper N-linked glycosylation addition and/or trimming. Chemical cross-linking experiments were also performed and suggest that the R578Q variant also does not form trimers as well as wild-type receptor, a function required for its full activity. CONCLUSIONS: These data demonstrate the distal C-terminus of P2X7 is important for oligomerization and post-translational modification of the receptor, providing a mechanism by which mutations in the trafficking domain disrupt P2X7 activity and localization at the plasma membrane.


Asunto(s)
Nitrógeno/metabolismo , Multimerización de Proteína , Procesamiento Proteico-Postraduccional , Receptores Purinérgicos P2X7/química , Receptores Purinérgicos P2X7/metabolismo , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/farmacología , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Glicosilación , Células HEK293 , Humanos , Mutación , Polimorfismo de Nucleótido Simple , Porosidad , Multimerización de Proteína/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Transporte de Proteínas/efectos de los fármacos , Receptores Purinérgicos P2X7/genética , Transducción de Señal/efectos de los fármacos
3.
Am J Respir Cell Mol Biol ; 47(3): 363-71, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22493010

RESUMEN

Airway epithelial cell defenses to viral infections are often compromised in disease or injury. Danger molecules, including ATP, are released during infection and contribute to nucleotide receptor-dependent inflammatory responses, largely through P2X(7). Although respiratory epithelium has been shown to express a variety of nucleotide receptors, the functional contribution of P2X(7) to the epithelial cell inflammatory response is unclear. We used human donor bronchial epithelial cells (BECs) and primary brushed epithelium to explore responses upon nucleotide and Toll-like receptor stimulation. P2X(7) messenger RNA and protein were observed in unprimed BECs, whereas inflammatory cytokine stimulation increased both messenger RNA and protein. Functional pore activity characteristic of P2X(7) was observed in BECs, and IL-1ß was rapidly released by BECs after Toll-like receptor 3 agonist, polyinosine-polycytidylic acid, priming followed by ATP administration, although no change was observed in IL-18 release. BECs produced more IL-1ß after stimulation with polyinosine-polycytidylic acid than LPS, showing a different preferential response than monocytes. In addition, blockade of nucleotide receptors with oxidized ATP significantly increased human rhinovirus (HRV) recovered 24 hours after infection in BECs, whereas 2'-3'-O-(4-benzoylbenzoyl) ATP treatment of brushed epithelial cells and respiratory cell lines nonsignificantly decreased HRV recovery. IL-1ß release was detected after HRV infection in both BECs and brushed cells, but BzATP did not significantly increase IL-1ß release further. BEC processing of pro-IL-1ß to the mature, cleaved, 17-kD form was confirmed by Western blotting. These results support the expression of functional P2X(7) in human lung epithelium, although its role in epithelial pathogen defense is likely independent of IL-1 family cytokine processing.


Asunto(s)
Bronquios/metabolismo , Interleucina-1beta/metabolismo , Receptores Purinérgicos P2X7/fisiología , Rhinovirus/fisiología , Bronquios/citología , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Células Epiteliales/metabolismo , Humanos , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética , Receptores Purinérgicos P2X7/genética , Receptores Toll-Like/agonistas , Receptores Toll-Like/metabolismo
4.
Immunol Res ; 50(1): 22-38, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21298493

RESUMEN

The nucleotide receptor P2X(7) is an attractive therapeutic target and potential biomarker for multiple inflammatory and neurologic disorders, and it is expressed in several immune, osteogenic, and neurologic cell types. Aside from its role in the nervous system, it is activated by ATP released at sites of tissue damage, inflammation, and infection. Ligand binding to P2X(7) stimulates many cell responses, including calcium fluxes, MAPK activation, inflammatory mediator release, and apoptosis. Much work has centered on P2X(7) action in cell death and mediator processing (e.g., pro-interleukin-1 cleavage by the inflammasome), but the contribution of P2X(7) to transcriptional regulation is less well defined. This review will focus on the growing evidence for the importance of nucleotide-mediated gene expression, highlight several animal models, human genetic, and clinical studies that support P2X(7) as a therapeutic target, and discuss the latest developments in anti-P2X(7) clinical trials.


Asunto(s)
Regulación de la Expresión Génica , Osteogénesis/genética , Osteogénesis/inmunología , Receptores Purinérgicos P2X7/inmunología , Adenosina Trifosfato/metabolismo , Animales , Biomarcadores/metabolismo , Humanos , Factores Inmunológicos/inmunología , Inflamación/inmunología , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptores Purinérgicos P2X7/genética , Transducción de Señal/genética , Transducción de Señal/inmunología
5.
J Biol Chem ; 285(44): 34288-98, 2010 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-20813842

RESUMEN

Activation of the ionotropic P2RX7 nucleotide receptor by extracellular ATP has been implicated in modulating inflammatory disease progression. Continuous exposure of P2RX7 to ligand can result in apoptosis in many cell types, including monocytic cells, whereas transient activation of P2RX7 is linked to inflammatory mediator production and the promotion of cell growth. Given the rapid hydrolysis of ATP in the circulation and interstitial space, transient activation of P2RX7 appears critically important for its action, yet its effects on gene expression are unclear. The present study demonstrates that short-term stimulation of human and mouse monocytic cells as well as mouse osteoblasts with P2RX7 agonists substantially induces the expression of several activating protein-1 (AP-1) members, particularly FosB. The potent activation of FosB after P2RX7 stimulation is especially noteworthy considering that little is known concerning the role of FosB in immunological regulation. Interestingly, the magnitude of FosB activation induced by P2RX7 stimulation appears greater than that observed with other known inducers of FosB expression. In addition, we have identified a previously unrecognized role for FosB in osteoblasts with respect to nucleotide-induced expression of cyclooxygenase-2 (COX-2), which is the rate-limiting enzyme in prostaglandin biosynthesis from arachidonic acid and is critical for osteoblastic differentiation and immune behavior. The present studies are the first to link P2RX7 action to FosB/AP-1 regulation in multiple cell types, including a role in nucleotide-induced COX-2 expression, and support a role for FosB in the control of immune and osteogenic function by P2RX7.


Asunto(s)
Monocitos/metabolismo , Osteoblastos/metabolismo , Receptores Purinérgicos P2/química , Factor de Transcripción AP-1/metabolismo , Animales , Línea Celular , Supervivencia Celular , Ciclooxigenasa 2/metabolismo , Humanos , Sistema Inmunológico , Macrófagos/metabolismo , Ratones , Modelos Biológicos , Receptores Purinérgicos P2X7 , Transducción de Señal
6.
J Immunol ; 185(5): 3028-34, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20668222

RESUMEN

Extracellular ATP has been proposed to act as a danger signal to alert the immune system of cell damage. Release of high local concentrations of ATP activates the nucleotide receptor, purinergic receptor X7 (P2RX7), on monocytic cells, which promotes the processing/release of proinflammatory mediators. Although the proinflammatory actions of P2RX7 are well recognized, little is known regarding the potential function of P2RX7 in repair responses. Because the resolution of inflammation is characterized by monocytic cell-dependent production of proangiogenic factors, we evaluated the contribution of P2RX7 to this process. We observed that both short-term and long-term P2RX7 activation promotes the robust release of vascular endothelial growth factor from primary human monocytes. This vascular endothelial growth factor release is calcium dependent and associated with reactive oxygen species production. This previously unrecognized action of P2RX7 suggests that it may not only participate in inflammation and cell death, but that it is also likely to be important in the control of angiogenesis and wound repair.


Asunto(s)
Adenosina Trifosfato/análogos & derivados , Monocitos/inmunología , Monocitos/metabolismo , Receptores Purinérgicos P2/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Cicatrización de Heridas/inmunología , Adenosina Trifosfato/fisiología , Muerte Celular/inmunología , Líquido Extracelular/inmunología , Líquido Extracelular/metabolismo , Humanos , Mediadores de Inflamación/agonistas , Mediadores de Inflamación/metabolismo , Mediadores de Inflamación/fisiología , Ligandos , Lipopolisacáridos/farmacología , Monocitos/citología , Agonistas del Receptor Purinérgico P2 , Receptores Purinérgicos P2/metabolismo , Receptores Purinérgicos P2X7 , Factor A de Crecimiento Endotelial Vascular/biosíntesis
7.
Biochemistry ; 49(22): 4611-9, 2010 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-20450227

RESUMEN

The nucleotide receptor P2X(7) is an immunomodulatory cation channel and a potential therapeutic target. P2X(7) is expressed in immune cells such as monocytes and macrophages and is activated by extracellular ATP following tissue injury or infection. Ligand binding to P2X(7) can stimulate ERK1/2, the transcription factor CREB, enzymes linked to the production of reactive oxygen species and interleukin-1 isoforms, and the formation of a nonspecific pore. However, little is known about the biochemistry of P2X(7), including whether the receptor is N-linked glycosylated and if this modification affects receptor function. Here we provide evidence that P2X(7) is sensitive to the glycosidases EndoH and PNGase F and that the human receptor appears glycosylated at N187, N202, N213, N241, and N284. Mutation of N187 results in weakened P2X(7) agonist-induced phosphorylation of ERK1/2, CREB, and p90 ribosomal S6 kinase, as well as a decreased level of pore formation. In further support of a role for glycosylation in receptor function, treatment of RAW 264.7 macrophages with the N-linked glycosylation synthesis inhibitor tunicamycin attenuates P2X(7) agonist-induced, but not phorbol ester-induced, ERK1/2 phosphorylation. Interestingly, residue N187 belongs to an N-linked glycosylation consensus sequence found in six of the seven P2X family members, suggesting this site is fundamentally important to P2X receptor function. To address the mechanism whereby N187 mutation attenuates receptor activity, we developed a live cell proteinase K digestion assay that demonstrated altered cell surface expression of P2X(7) N187A. This is the first report to map human P2X(7) glycosylation sites and reveal residue N187 is critical for receptor trafficking and function.


Asunto(s)
Asparagina/genética , Mutación Puntual , Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2/metabolismo , Secuencia de Aminoácidos , Animales , Asparagina/metabolismo , Células COS , Línea Celular , Chlorocebus aethiops , Secuencia Conservada , Regulación hacia Abajo/genética , Espacio Extracelular/genética , Glicosilación , Humanos , Ratones , Datos de Secuencia Molecular , Polimorfismo de Nucleótido Simple , Procesamiento Proteico-Postraduccional/genética , Transporte de Proteínas/genética , Agonistas del Receptor Purinérgico P2 , Receptores Purinérgicos P2/fisiología , Receptores Purinérgicos P2X7 , Transducción de Señal/genética
8.
Purinergic Signal ; 5(2): 175-87, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19263245

RESUMEN

Extracellular nucleotides can act as important intercellular signals in diverse biological processes, including the enhanced production of factors that are key to immune response regulation. One receptor that binds extracellular adenosine triphosphate released at sites of infection and injury is P2X(7), which is an ionotrophic receptor that can also lead to the formation of a non-specific pore, activate multiple mitogen-activated protein kinases (MAPKs), and stimulate the production of immune mediators including interleukin family members and reactive oxygen species (ROS). In the present report, we have investigated the signaling mechanisms by which P2X(7) promotes monocytic cell mediator production and induces transcription factor expression/phosphorylation, as well as how receptor-associated pore activity is regulated by intracellular trafficking. We report that P2X(7) stimulates ROS production in macrophages through the MAPKs ERK1/2 and the nicotinamide adenine dinucleotide phosphate oxidase complex, activates several transcription factors including cyclic-AMP response element-binding protein and components of the activating protein-1 complex, and contains specific sequences within its intracellular C-terminus that appear critical for its activity. Altogether, these data further implicate P2X(7) activation and signaling as a fundamental modulator of macrophage immune responses.

9.
J Biol Chem ; 280(29): 26653-8, 2005 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-15883153

RESUMEN

WNKs are large serine/threonine protein kinases structurally distinct from all other members of the protein kinase superfamily. Of the four human WNK family members, WNK1 and WNK4 have been linked to a hereditary form of hypertension, pseudohypoaldosteronism type II. We characterized the biochemical properties and regulation of WNK1 that may contribute to its physiological activities and abnormal function in disease. We showed that WNK1 is activated by hypertonic stress in kidney epithelial cells and in breast and colon cancer cell lines. In addition, hypotonic stress also led to a modest increase in WNK1 activity. Gel filtration suggested that WNK1 exists as a tetramer, and yeast two-hybrid data showed that the N terminus of WNK1 (residues 1-222) interacts with residues 481-660, which includes the WNK1 autoinhibitory domain and a C-terminal coiled-coil domain. Although cell biological studies have suggested a functional interaction between WNK1 and WNK4, we found no evidence of stable interactions between these kinases. However, WNK1 phosphorylated both WNK4 and WNK2. In addition, the WNK1 autoinhibitory domain inhibited the catalytic activity of these WNKs. These findings suggest potential mechanisms for interconnected regulation of WNK family members.


Asunto(s)
Proteínas Serina-Treonina Quinasas/fisiología , Animales , Línea Celular , Línea Celular Tumoral , Células Epiteliales/patología , Humanos , Hipertensión/etiología , Soluciones Hipotónicas/farmacología , Péptidos y Proteínas de Señalización Intracelular , Antígenos de Histocompatibilidad Menor , Fosforilación , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Terciaria de Proteína , Estrés Fisiológico/metabolismo , Proteína Quinasa Deficiente en Lisina WNK 1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA