Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Infect Dis ; 221(10): 1598-1606, 2020 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-31282542

RESUMEN

BACKGROUND: Adenosine is a potent immunoregulatory nucleoside produced during inflammatory states to limit tissue damage. We hypothesized that dipyridamole, which inhibits cellular adenosine uptake, could raise the extracellular adenosine concentration and dampen chronic inflammation associated with human immunodeficiency virus (HIV) type 1. METHODS: Virally suppressed participants receiving antiretroviral therapy were randomized 1:1 for 12 weeks of dipyridamole (100 mg 4 times a day) versus placebo capsules. All participants took open-label dipyridamole during weeks 12-24. Study end points included changes in markers of systemic inflammation (soluble CD163 and CD14, and interleukin 6) and levels of T-cell immune activation (HLA-DR+CD38+). RESULTS: Of 40 participants who were randomized, 17 dipyridamole and 18 placebo recipients had baseline and week 12 data available for analyses. There were no significant changes in soluble markers, apart from a trend toward decreased levels of soluble CD163 levels (P = .09). There was a modest decrease in CD8+ T-cell activation (-17.53% change for dipyridamole vs +13.31% for placebo; P = .03), but the significance was lost in the pooled analyses (P = .058). Dipyridamole also reduced CD4+ T-cell activation (-11.11% change; P = .006) in the pooled analyses. In post hoc analysis, detectable plasma dipyridamole levels were associated with higher levels of inosine, an adenosine surrogate, and of cyclic adenosine monophosphate. CONCLUSION: Dipyridamole increased extracellular adenosine levels and decreased T-cell activation significantly among persons with HIV-1 infection receiving virally suppressive therapy.


Asunto(s)
Dipiridamol/uso terapéutico , Infecciones por VIH/complicaciones , Inflamación/tratamiento farmacológico , Inflamación/etiología , Inhibidores de Fosfodiesterasa/uso terapéutico , Adolescente , Adulto , Biomarcadores/sangre , Enfermedad Crónica , Método Doble Ciego , Infecciones por VIH/tratamiento farmacológico , VIH-1 , Humanos , Persona de Mediana Edad , Proyectos Piloto , Adulto Joven
2.
Angiogenesis ; 23(4): 599-610, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32419057

RESUMEN

RATIONALE: One hallmark of tumor-derived exosomes (TEX) is the promotion of cancer progression by stimulating angiogenesis. This study was performed to evaluate the role of adenosine receptors in TEX-induced angiogenesis. METHODS: TEX produced by UMSCC47 head and neck cancer cell line were isolated by mini size exclusion chromatography (mini-SEC). Enzymatic activity of ectonucleotidases CD39/CD73 carried by TEX was measured by HPLC. Adenosine content of TEX was measured by UPLC-MS/MS. Primary human macrophages were co-incubated with TEX or exosomes derived from the plasma of head and neck cancer patients and their marker expression profile was analyzed by flow cytometry. The macrophage secretome was analyzed by angiogenesis arrays. The in vitro angiogenic potential of TEX was evaluated in endothelial growth studies. Results were validated in vivo using basement membrane extract plug assays in A1R-/-, A2AR-/- and A2BR-/- rats. Vascularization was analyzed by hemoglobin quantification and immunohistology with vessel and macrophage markers. RESULTS: TEX carried enzymatically active CD39/CD73 and adenosine. TEX promoted A2BR-mediated polarization of macrophages toward an M2-like phenotype (p < 0.05) and enhanced their secretion of angiogenic factors. Growth of endothelial cells was stimulated directly by TEX and indirectly via macrophage-reprogramming dependent on A2BR signaling (p < 0.01). In vivo, TEX stimulated the formation of defined vascular structures and macrophage infiltration. This response was absent in A2BR-/- rats (p < 0.05). CONCLUSION: This report provides the first evidence for adenosine production by TEX to promote angiogenesis via A2BR. A2BR antagonism emerges as a potential strategy to block TEX-induced angiogenesis.


Asunto(s)
Exosomas/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Receptor de Adenosina A2B/metabolismo , Transducción de Señal , Animales , Línea Celular Tumoral , Proliferación Celular , Reprogramación Celular , Exosomas/ultraestructura , Femenino , Neoplasias de Cabeza y Cuello/patología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Macrófagos/metabolismo , Masculino , Modelos Biológicos , Fenotipo , Ratas
3.
J Pharmacol Exp Ther ; 373(1): 135-148, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32015161

RESUMEN

By reducing their metabolism, dipeptidyl peptidase 4 inhibition (DPP4I) enhances the effects of numerous peptides including neuropeptide Y1-36 (NPY1-36), peptide YY1-36 (PYY1-36), and SDF-1α Studies show that separately NPY1-36, PYY1-36 and SDF-1α stimulate proliferation of, and collagen production by, cardiac fibroblasts (CFs), preglomerular vascular smooth muscle cells (PGVSMCs), and glomerular mesangial cells (GMCs), particularly in cells isolated from genetically hypertensive rats. Whether certain combinations of these factors, in the absence or presence of DPP4I, are more profibrotic than others is unknown. Here we contrasted 24 different combinations of conditions (DPP4I, hypertensive genotype and physiologic levels [3 nM] of NPY1-36, PYY1-36, or SDF-1α) on proliferation of, and [3H]-proline incorporation by, CFs, PGVSMCs, and GMCs. In all three cell types, the various treatment conditions differentially increased proliferation and [3H]-proline incorporation, with a hypertensive genotype + DPP4I + NPY1-36 + SDF-1α being the most efficacious combination. Although the effects of this four-way combination were similar in male versus female CFs, physiologic (1 nM) concentrations of 2-methoxyestradiol (2ME; nonestrogenic metabolite of 17ß-estradiol), abolished the effects of this combination in both male and female CFs. In conclusion, this study demonstrates that CFs, PGVSMCs, and GMCs are differentially activated by various combinations of NPY1-36, PYY1-36, SDF-1α, a hypertensive genetic background and DPP4I. We hypothesize that as these progrowth conditions accumulate, a tipping point would be reached that manifests in the long term as organ fibrosis and that 2ME would obviate any profibrotic effects of DPP4I, even under the most profibrotic conditions (i.e., hypertensive genotype with high NPY1-36 + SDF-1α levels and low 2ME levels). SIGNIFICANCE STATEMENT: This work elucidates combinations of factors that could contribute to long-term profibrotic effects of dipeptidyl peptidase 4 inhibitors and suggests a novel drug combination that could prevent any potential profibrotic effects of dipeptidyl peptidase 4 inhibitors while augmenting the protective effects of this class of antidiabetic agents.


Asunto(s)
2-Metoxiestradiol/farmacología , Quimiocina CXCL12/sangre , Colágeno/biosíntesis , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Hipertensión/sangre , Neuropéptido Y/sangre , Fragmentos de Péptidos/sangre , 2-Metoxiestradiol/uso terapéutico , Animales , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Células Cultivadas , Inhibidores de la Dipeptidil-Peptidasa IV/uso terapéutico , Femenino , Hipertensión/genética , Hipertensión/patología , Masculino , Péptido YY/sangre , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY
4.
Purinergic Signal ; 16(2): 187-211, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32367441

RESUMEN

The goal of this study was to determine the validity of using N6-etheno-bridged adenine nucleotides to evaluate ecto-nucleotidase activity. We observed that the metabolism of N6-etheno-ATP versus ATP was quantitatively similar when incubated with recombinant CD39, ENTPD2, ENTPD3, or ENPP-1, and the quantitative metabolism of N6-etheno-AMP versus AMP was similar when incubated with recombinant CD73. This suggests that ecto-nucleotidases process N6-etheno-bridged adenine nucleotides similarly to endogenous adenine nucleotides. Four cell types rapidly (t1/2, 0.21 to 0.66 h) metabolized N6-etheno-ATP. Applied N6-etheno-ATP was recovered in the medium as N6-etheno-ADP, N6-etheno-AMP, N6-etheno-adenosine, and surprisingly N6-etheno-adenine; intracellular N6-etheno compounds were undetectable. This suggests minimal cellular uptake, intracellular metabolism, or deamination of these compounds. N6-etheno-ATP, N6-etheno-ADP, N6-etheno-AMP, N6-etheno-adenosine, and N6-etheno-adenine had little affinity for recombinant A1, A2A, or A2B receptors, for a subset of P2X receptors (3H-α,ß-methylene-ATP binding to rat bladder membranes), or for a subset of P2Y receptors (35S-ATP-αS binding to rat brain membranes), suggesting minimal pharmacological activity. N6-etheno-adenosine was partially converted to N6-etheno-adenine in four different cell types; this was blocked by purine nucleoside phosphorylase (PNPase) inhibition. Intravenous N6-etheno-ATP was quickly metabolized, with N6-etheno-adenine being the main product in naïve rats, but not in rats pretreated with a PNPase inhibitor. PNPase inhibition reduced the urinary excretion of endogenous adenine and attenuated the conversion of exogenous adenosine to adenine in the renal cortex. The N6-etheno-bridge method is a valid technique to assess extracellular metabolism of adenine nucleotides by ecto-nucleotidases. Also, rats express an enzyme with PNPase-like activity that metabolizes N6-etheno-adenosine to N6-etheno-adenine.


Asunto(s)
Nucleótidos de Adenina/metabolismo , Adenosina Trifosfatasas/metabolismo , Adenosina/metabolismo , Purina-Nucleósido Fosforilasa/metabolismo , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/metabolismo , Animales , Masculino , Nucleotidasas/metabolismo , Ratas
5.
Purinergic Signal ; 16(2): 231-240, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32440820

RESUMEN

Exosomes, small-sized extracellular vesicles, carry components of the purinergic pathway. The production by cells of exosomes carrying this pathway remains poorly understood. Here, we asked whether type 1, 2A, or 2B adenosine receptors (A1Rs, A2ARs, and A2BRs, respectively) expressed by producer cells are involved in regulating exosome production. Preglomerular vascular smooth muscle cells (PGVSMCs) were isolated from wildtype, A1R-/-, A2AR-/-, and A2BR-/- rats, and exosome production was quantified under normal or metabolic stress conditions. Exosome production was also measured in various cancer cells treated with pharmacologic agonists/antagonists of A1Rs, A2ARs, and A2BRs in the presence or absence of metabolic stress or cisplatin. Functional activity of exosomes was determined in Jurkat cell apoptosis assays. In PGVSMCs, A1R and A2AR constrained exosome production under normal conditions (p = 0.0297; p = 0.0409, respectively), and A1R, A2AR, and A2BR constrained exosome production under metabolic stress conditions. Exosome production from cancer cells was reduced (p = 0.0028) by the selective A2AR agonist CGS 21680. These exosomes induced higher levels of Jurkat apoptosis than exosomes from untreated cells or cells treated with A1R and A2BR agonists (p = 0.0474). The selective A2AR antagonist SCH 442416 stimulated exosome production under metabolic stress or cisplatin treatment, whereas the selective A2BR antagonist MRS 1754 reduced exosome production. Our findings indicate that A2ARs suppress exosome release in all cell types examined, whereas effects of A1Rs and A2BRs are dependent on cell type and conditions. Pharmacologic targeting of cancer with A2AR antagonists may inadvertently increase exosome production from tumor cells.


Asunto(s)
Agonistas del Receptor de Adenosina A2/farmacología , Exosomas/efectos de los fármacos , Receptor de Adenosina A1/metabolismo , Receptor de Adenosina A2A/metabolismo , Adenosina/análogos & derivados , Adenosina/farmacología , Animales , Exosomas/metabolismo , Masculino , Fenetilaminas/farmacología , Ratas , Receptor de Adenosina A1/efectos de los fármacos , Receptor de Adenosina A2A/efectos de los fármacos , Células Tumorales Cultivadas/metabolismo
6.
Am J Physiol Endocrinol Metab ; 310(5): E313-22, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26732685

RESUMEN

17ß-Estradiol (estradiol) inhibits microglia proliferation. 2-Methoxyestradiol (2-ME) is an endogenous metabolite of estradiol with little affinity for estrogen receptors (ERs). We hypothesize that 2-ME inhibits microglial proliferation and activation and contributes to estradiol's inhibitory effects on microglia. We compared the effects of estradiol, 2-hydroxyestradiol [2-OE; estradiol metabolite produced by cytochrome P450 (CYP450)], and 2-ME [formed by catechol-O-methyltransferase (COMT) acting upon 2-OE] on microglial (BV2 cells) DNA synthesis, cell proliferation, activation, and phagocytosis. 2-ME and 2-OE were approximately three- and 10-fold, respectively, more potent than estradiol in inhibiting microglia DNA synthesis. The antimitogenic effects of estradiol were reduced by pharmacological inhibitors of CYP450 and COMT. Inhibition of COMT blocked the conversion of 2-OE to 2-ME and the antimitogenic effects of 2-OE but not 2-ME. Microglia expressed ERß and GPR30 but not ERα. 2,3-Bis(4-hydroxyphenyl)-propionitrile (ERß agonist), but not 4,4',4''-(4-propyl-[1H]-pyrazole-1,3,5-triyl)trisphenol (ERα agonist) or G1 (GPR30 agonist), inhibited microglial proliferation. The antiproliferative effects of estradiol, but not 2-OE or 2-ME, were partially reversed by ICI-182,780 (ERα/ß antagonist) but not by 1,3-bis(4-hydroxyphenyl)-4-methyl-5-[4-(2-piperidinylethoxy)phenol]-1H-pyrazole (ERα antagonist) or G15 (GPR30 antagonist). Lipopolysaccharide increased microglia iNOS and COX-2 expression and phagocytosing activity of microglia; these effects were inhibited by 2-ME. We conclude that in microglia, 2-ME inhibits proliferation, proinflammatory responses, and phagocytosis. 2-ME partially mediates the effects of estradiol via ER-independent mechanisms involving sequential metabolism of estradiol to 2-OE and 2-ME. 2-ME could be of potential therapeutic use in postischemic stroke injuries. Interindividual differences in estradiol metabolism might affect the individual's ability to recover from stroke.


Asunto(s)
Proliferación Celular/efectos de los fármacos , ADN/biosíntesis , Estradiol/análogos & derivados , Estradiol/farmacología , Estrógenos/farmacología , Microglía/efectos de los fármacos , Fagocitosis/efectos de los fármacos , 2-Metoxiestradiol , Animales , Catecol O-Metiltransferasa , Inhibidores de Catecol O-Metiltransferasa/farmacología , Inhibidores Enzimáticos del Citocromo P-450/farmacología , ADN/efectos de los fármacos , Receptor alfa de Estrógeno/antagonistas & inhibidores , Receptor alfa de Estrógeno/efectos de los fármacos , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/antagonistas & inhibidores , Receptor beta de Estrógeno/efectos de los fármacos , Receptor beta de Estrógeno/metabolismo , Ratones , Microglía/metabolismo , Moduladores de Tubulina/farmacología
7.
J Pharmacol Exp Ther ; 359(3): 420-435, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27679494

RESUMEN

In vivo, guanine moieties in DNA, RNA, guanine nucleotides, or guanosine or guanine per se can undergo nitration (for example, by peroxynitrite) or hydroxylation (for example, by superoxide anion) on position 8 of the purine ring. Subsequent catabolism of these modified biomolecules leads to the production of a diverse group of 8-nitro, 8-amino, and 8-hydroxy guanosine and guanine compounds. Indeed, studies suggest the in vivo existence of 8-nitroguanosine, 8-nitroguanine, 8-aminoguanosine, 8-aminoguanine, 8-hydroxyguanosine, 8-hydroxy-2'-deoxyguanosine, and 8-hydroxyguanine. Since a multitude of these compounds exist in vivo, and since the renal effects of 8-substituted guanosine and guanine compounds are entirely unknown, we examined the effects of guanosine, guanine, 8-nitroguanosine, 8-nitroguanine, 8-hydroxyguanosine, 8-hydroxyguanine, 8-hydroxy-2'-deoxyguanosine, 8-aminoguanosine, and 8-aminoguanine (33.5 µmol/kg/min; intravenous infusion for 115 minutes) on excretion of sodium, potassium, and glucose in rats. Guanosine, 8-nitroguanosine, and 8-hydroxy-2'-deoxyguanosine had minimal natriuretic activity. Guanine, 8-nitroguanine, 8-hydroxyguanosine, and 8-hydroxyguanine had moderate natriuretic activity (increased sodium excretion by 9.4-, 7.8-, 7.1-, and 8.6-fold, respectively). In comparison with all other compounds, 8-aminoguanosine and 8-aminoguanine were highly efficacious and increased sodium excretion by 26.6- and 17.2-fold, respectively, exceeding that of a matched dose of amiloride (13.6-fold increase). 8-Aminoguanosine and 8-aminoguanine also increased glucose excretion by 12.1- and 12.2-fold, respectively, and decreased potassium excretion by 69.1 and 71.0%, respectively. Long-term radiotelemetry studies demonstrated that oral 8-aminoguanosine and 8-aminoguanine (5 mg/kg/day) suppressed deoxycorticosterone/salt-induced hypertension. These experiments demonstrate that some naturally occurring 8-substitued guanosine and guanine compounds, particularly 8-aminoguanosine and 8-aminoguanine, are potent and efficacious potassium-sparing diuretics/natriuretics that may represent a novel class of antihypertensive diuretics.


Asunto(s)
Antihipertensivos/farmacología , Diuréticos/farmacología , Glucosuria/tratamiento farmacológico , Guanina/análogos & derivados , Guanosina/análogos & derivados , Natriuresis/efectos de los fármacos , Animales , Antihipertensivos/uso terapéutico , Diuréticos/uso terapéutico , Guanina/farmacología , Guanina/uso terapéutico , Guanosina/farmacología , Guanosina/uso terapéutico , Masculino , Ratas , Ratas Sprague-Dawley
8.
J Virol ; 89(18): 9616-30, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26178986

RESUMEN

UNLABELLED: The role of the adenosine (ADO) pathway in human immunodeficiency virus type 1/simian immunodeficiency virus (HIV-1/SIV) infection remains unclear. We compared SIVsab-induced changes of markers related to ADO production (CD39 and CD73) and breakdown (CD26 and adenosine deaminase) on T cells from blood, lymph nodes, and intestine collected from pigtailed macaques (PTMs) and African green monkeys (AGMs) that experience different SIVsab infection outcomes. We also measured ADO and inosine (INO) levels in tissues by mass spectrometry. Finally, we assessed the suppressive effect of ADO on proinflammatory cytokine production after T cell receptor stimulation. The baseline level of both CD39 and CD73 coexpression on regulatory T cells and ADO levels were higher in AGMs than in PTMs. Conversely, high INO levels associated with dramatic increases in CD26 expression and adenosine deaminase activity were observed in PTMs during chronic SIV infection. Immune activation and inflammation markers in the gut and periphery inversely correlated with ADO and directly correlated with INO. Ex vivo administration of ADO significantly suppressed proinflammatory cytokine production by T cells in both species. In conclusion, the opposite dynamics of ADO pathway-related markers and contrasting ADO/INO levels in species with divergent proinflammatory responses to SIV infection support a key role of ADO in controlling immune activation/inflammation in nonprogressive SIV infections. Changes in ADO levels predominately occurred in the gut, suggesting that the ADO pathway may be involved in sparing natural hosts of SIVs from developing SIV-related gut dysfunction. Focusing studies of the ADO pathway on mucosal sites of viral replication is warranted. IMPORTANCE: The mechanisms responsible for the severe gut dysfunction characteristic of progressive HIV and SIV infection in humans and macaques are not completely elucidated. We report that ADO may play a key role in controlling immune activation/inflammation in nonprogressive SIV infections by limiting SIV-related gut inflammation. Conversely, in progressive SIV infection, significant degradation of ADO occurs, possibly due to an early increase of ADO deaminase complexing protein 2 (CD26) and adenosine deaminase. Our study supports therapeutic interventions to offset alterations of this pathway during progressive HIV/SIV infections. These potential approaches to control chronic immune activation and inflammation during pathogenic SIV infection may prevent HIV disease progression.


Asunto(s)
5'-Nucleotidasa/inmunología , Adenosina/inmunología , Antígenos CD/inmunología , Apirasa/inmunología , Dipeptidil Peptidasa 4/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Linfocitos T/inmunología , Animales , Chlorocebus aethiops , Enfermedad Crónica , Citocinas/inmunología , Humanos , Macaca nemestrina , Masculino , Receptores de Antígenos de Linfocitos T/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/patología , Linfocitos T/patología
9.
Am J Physiol Heart Circ Physiol ; 309(9): H1528-42, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26371160

RESUMEN

Cardiac sympathetic nerves release neuropeptide Y (NPY)1-36, and peptide YY (PYY)1-36 is a circulating peptide; therefore, these PP-fold peptides could affect cardiac fibroblasts (CFs). We examined the effects of NPY1-36 and PYY1-36 on the proliferation of and collagen production ([(3)H]proline incorporation) by CFs isolated from Wistar-Kyoto (WKY) normotensive rats and spontaneously hypertensive rats (SHRs). Experiments were performed with and without sitagliptin, an inhibitor of dipeptidyl peptidase 4 [DPP4; an ectoenzyme that metabolizes NPY1-36 and PYY1-36 (Y1 receptor agonists) to NPY3-36 and PYY3-36 (inactive at Y1 receptors), respectively]. NPY1-36 and PYY1-36, but not NPY3-36 or PYY3-36, stimulated proliferation of CFs, and these effects were more potent than ANG II, enhanced by sitagliptin, blocked by BIBP3226 (Y1 receptor antagonist), and greater in SHR CFs. SHR CF membranes expressed more receptor for activated C kinase (RACK)1 [which scaffolds the Gi/phospholipase C (PLC)/PKC pathway] compared with WKY CF membranes. RACK1 knockdown (short hairpin RNA) and inhibition of Gi (pertussis toxin), PLC (U73122), and PKC (GF109203X) blocked the proliferative effects of NPY1-36. NPY1-36 and PYY1-36 stimulated collagen production more potently than did ANG II, and this was enhanced by sitagliptin and greater in SHR CFs. In conclusion, 1) NPY1-36 and PYY1-36, via the Y1 receptor/Gi/PLC/PKC pathway, activate CFs, and this pathway is enhanced in SHR CFs due to increased localization of RACK1 in membranes; and 2) DPP4 inhibition enhances the effects of NPY1-36 and PYY1-36 on CFs, likely by inhibiting the metabolism of NPY1-36 and PYY1-36. The implications are that endogenous NPY1-36 and PYY1-36 could adversely affect cardiac structure/function by activating CFs, and this may be exacerbated in genetic hypertension and by DPP4 inhibitors.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Colágeno/efectos de los fármacos , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Fibroblastos/efectos de los fármacos , Hipertensión/genética , Neuropéptido Y/farmacología , Péptido YY/farmacología , Fosfato de Sitagliptina/farmacología , Angiotensina II/farmacología , Animales , Colágeno/biosíntesis , Estrenos/farmacología , Fibroblastos/metabolismo , Proteínas de Unión al GTP/metabolismo , Indoles/farmacología , Maleimidas/farmacología , Miocardio/citología , Fragmentos de Péptidos/farmacología , Toxina del Pertussis/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Pirrolidinonas/farmacología , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Receptores de Cinasa C Activada , Transducción de Señal , Fosfolipasas de Tipo C/antagonistas & inhibidores
10.
Am J Physiol Renal Physiol ; 307(1): F14-24, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24808540

RESUMEN

Energy depletion increases the renal production of 2',3'-cAMP (a positional isomer of 3',5'-cAMP that opens mitochondrial permeability transition pores) and 2',3'-cAMP is converted to 2'-AMP and 3'-AMP, which in turn are metabolized to adenosine. Because the enzymes involved in this "2',3'-cAMP-adenosine pathway" are unknown, we examined whether 2',3'-cyclic nucleotide 3'-phosphodiesterase (CNPase) participates in the renal metabolism of 2',3'-cAMP. Western blotting and real-time PCR demonstrated expression of CNPase in rat glomerular mesangial, preglomerular vascular smooth muscle and endothelial, proximal tubular, thick ascending limb and collecting duct cells. Real-time PCR established the expression of CNPase in human glomerular mesangial, proximal tubular and vascular smooth muscle cells; and the level of expression of CNPase was greater than that for phosphodiesterase 4 (major enzyme for the metabolism of 3',5'-cAMP). Overexpression of CNPase in rat preglomerular vascular smooth muscle cells increased the metabolism of exogenous 2',3'-cAMP to 2'-AMP. Infusions of 2',3'-cAMP into isolated CNPase wild-type (+/+) kidneys increased renal venous 2'-AMP, and this response was diminished by 63% in CNPase knockout (-/-) kidneys, whereas the conversion of 3',5'-cAMP to 5'-AMP was similar in CNPase +/+ vs. -/- kidneys. In CNPase +/+ kidneys, energy depletion (metabolic poisons) increased kidney tissue levels of adenosine and its metabolites (inosine, hypoxanthine, xanthine, and uric acid) without accumulation of 2',3'-cAMP. In contrast, in CNPase -/- kidneys, energy depletion increased kidney tissue levels of 2',3'-cAMP and abolished the increase in adenosine and its metabolites. In conclusion, kidneys express CNPase, and renal CNPase mediates in part the renal 2',3'-cAMP-adenosine pathway.


Asunto(s)
2',3'-Nucleótido Cíclico 3'-Fosfodiesterasa/metabolismo , Adenosina Monofosfato/metabolismo , AMP Cíclico/metabolismo , Riñón/metabolismo , Músculo Liso Vascular/metabolismo , Transducción de Señal , Adenosina/metabolismo , Animales , Células Cultivadas , Humanos , Ratones , Ratas , Transducción de Señal/fisiología
11.
Dig Dis Sci ; 59(9): 2118-25, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24711075

RESUMEN

BACKGROUND: Intraperitoneal adenosine reduces abdominal adhesions. However, because of the ultra-short half-life and low solubility of adenosine, optimal efficacy requires multiple dosing. AIM: Here, we compared the ability of potential adenosine prodrugs to inhibit post-surgical abdominal adhesions after a single intraperitoneal dose. METHODS: Abdominal adhesions were induced in mice using an electric toothbrush to damage the cecum. Also, 20 µL of 95 % ethanol was applied to the cecum to cause chemically induced injury. After injury, mice received intraperitoneally either saline (n = 18) or near-solubility limit of adenosine (23 mmol/L; n = 12); 5'-adenosine monophosphate (75 mmol/L; n = 11); 3'-adenosine monophosphate (75 mmol/L; n = 12); 2'-adenosine monophosphate (75 mmol/L; n = 12); 3',5'-cyclic adenosine monophosphate (75 mmol/L; n = 19); or 2',3'-cyclic adenosine monophosphate (75 mmol/L; n = 20). After 2 weeks, adhesion formation was scored by an observer blinded to the treatments. In a second study, intraperitoneal adenosine levels were measured using tandem mass spectrometry for 3 h after instillation of 2',3'-cyclic adenosine monophosphate (75 mmol/L) into the abdomen. RESULTS: The order of efficacy for attenuating adhesion formation was: 2',3'-cyclic adenosine monophosphate > 3',5'-cyclic adenosine monophosphate ≈ adenosine > 5'-adenosine monophosphate ≈ 3'-adenosine monophosphate ≈ 2'-adenosine monophosphate. The groups were compared using a one-factor analysis of variance, and the overall p value for differences between groups was p < 0.000001. Intraperitoneal administration of 2',3'-cAMP yielded pharmacologically relevant levels of adenosine in the abdominal cavity for >3 h. CONCLUSION: Administration of 2',3'-cyclic adenosine monophosphate into the surgical field is a unique, convenient and effective method of preventing post-surgical adhesions by acting as an adenosine prodrug.


Asunto(s)
Nucleótidos de Adenina/uso terapéutico , Complicaciones Posoperatorias/prevención & control , Adherencias Tisulares/prevención & control , Nucleótidos de Adenina/metabolismo , Nucleótidos de Adenina/farmacocinética , Adenosina/farmacocinética , Adenosina/uso terapéutico , Adenosina Monofosfato/farmacocinética , Adenosina Monofosfato/uso terapéutico , Administración Intravaginal , Animales , AMP Cíclico/farmacocinética , AMP Cíclico/uso terapéutico , Ratones Endogámicos C57BL , Factores de Tiempo
12.
Sci Rep ; 14(1): 22652, 2024 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-39349636

RESUMEN

The metabolic syndrome is characterized by obesity, insulin resistance, dyslipidemia and hypertension and predisposes to cardiorenal injury. Here, we tested our hypothesis that 8-aminoguanine, an endogenous purine, exerts beneficial effects in Zucker Diabetic-Sprague Dawley (ZDSD) rats, a preclinical model of the metabolic syndrome. ZDSD rats were instrumented for blood pressure radiotelemetry and randomized to vehicle or 8-aminoguanine (10 mg/kg/day, po). The protocol was divided into four phases: Phase 1: 17 days of tap water/normal diet; Phase 2: 30 days of 1% saline/normal diet; Phase 3: 28 days of 1% saline/diabetogenic diet; Phase 4: acute/terminal measurements. 8-Aminoguanine: (1) decreased mean arterial blood pressure (P = 0.0004; 119.5 ± 1.0 (vehicle) versus 116.3 ± 1.0 (treated) mmHg) throughout all three phases of the radiotelemetry study; (2) rebalanced the purine metabolome away from hypoxanthine (pro-inflammatory) and towards inosine (anti-inflammatory); (3) reduced by 71% circulating IL-1ß, a cytokine that contributes to hypertension-induced adverse cardiovascular events and type 2 diabetes; (4) attenuated renovascular responses to angiotensin II; (5) improved cardiac and renal histopathology; (6) attenuated diet-induced polydipsia/polyuria; and (7) reduced HbA1c. In the metabolic syndrome, 8-aminoguanine lowers blood pressure, improves diabetes and reduces organ damage, likely by rebalancing the purine metabolome leading to reductions in injurious cytokines such as IL-1ß.


Asunto(s)
Síndrome Metabólico , Ratas Zucker , Animales , Síndrome Metabólico/metabolismo , Síndrome Metabólico/tratamiento farmacológico , Ratas , Masculino , Presión Sanguínea/efectos de los fármacos , Ratas Sprague-Dawley , Guanina/análogos & derivados , Guanina/metabolismo , Guanina/farmacología , Modelos Animales de Enfermedad
13.
J Leukoc Biol ; 2024 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-39217408

RESUMEN

Twelve weeks of dipyridamole increased extracellular adenosine levels and decreased T cell activation in people with HIV. In this analysis, we investigated the effect of dipyridamole on HIV-specific T cell responses. We compared changes in Gag- and Env-specific T cell responses using intracellular cytokine staining, following 12 weeks of dipyridamole treatment vs placebo. We evaluated whether frequencies of polyfunctional HIV-specific T cells were associated with purines in the adenosine pathway and with measures of HIV persistence and chronic inflammation. There was a significant decrease in CD4+ polyfunctional T cell responses to Gag (-62.6% vs -23.0%; p<0.001) and Env (-56.1% vs -6.0%; p<0.001) in the dipyridamole arm. In the dipyridamole group, lower frequencies of polyfunctional Env-specific CD4+ T cells were associated with higher plasma levels of adenosine (r= -0.85; p<0.01) and inosine (r= -0.70; p=0.04). Higher adenosine levels induced by dipyridamole treatment is associated with decreased HIV-specific CD4+ T cell polyfunctional responses in people with HIV on antiretroviral therapy.

14.
Am J Physiol Cell Physiol ; 304(5): C406-21, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23242185

RESUMEN

The aim of this investigation was to test the hypothesis that extracellular guanosine regulates extracellular adenosine levels. Rat preglomerular vascular smooth muscle cells were incubated with adenosine, guanosine, or both. Guanosine (30 µmol/l) per se had little effect on extracellular adenosine levels. Extracellular adenosine levels 1 h after addition of adenosine (3 µmol/l) were 0.125 ± 0.020 µmol/l, indicating rapid disposition of extracellular adenosine. Extracellular adenosine levels 1 h after addition of adenosine (3 µmol/l) plus guanosine (30 µmol/l) were 1.173 ± 0.061 µmol/l, indicating slow disposition of extracellular adenosine. Cell injury increased extracellular levels of endogenous adenosine and guanosine, and the effects of cell injury on endogenous extracellular adenosine were modulated by altering the levels of endogenous extracellular guanosine with exogenous purine nucleoside phosphorylase (converts guanosine to guanine) or 8-aminoguanosine (inhibits purine nucleoside phosphorylase). Extracellular guanosine also slowed the disposition of extracellular adenosine in rat preglomerular vascular endothelial cells, mesangial cells, cardiac fibroblasts, and kidney epithelial cells and in human aortic and coronary artery vascular smooth muscle cells and coronary artery endothelial cells. The effects of guanosine on adenosine levels were not mimicked or attenuated by 5-iodotubericidin (adenosine kinase inhibitor), erythro-9-(2-hydroxy-3-nonyl)-adenine (adenosine deaminase inhibitor), 5-aminoimidazole-4-carboxamide (guanine deaminase inhibitor), aristeromycin (S-adenosylhomocysteine hydrolase inhibitor), low sodium (inhibits concentrative nucleoside transporters), S-(4-nitrobenzyl)-6-thioinosine [inhibits equilibrative nucleoside transporter (ENT) type 1], zidovudine (inhibits ENT type 2), or acadesine (known modulator of adenosine levels). Guanosine also increases extracellular inosine, uridine, thymidine, and cytidine, yet decreases extracellular uric acid. In conclusion, extracellular guanosine regulates extracellular adenosine levels.


Asunto(s)
Adenosina/metabolismo , Guanosina/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Adenosina/farmacología , Animales , Aorta/metabolismo , Células Cultivadas , Vasos Coronarios/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Fibroblastos/metabolismo , Guanosina/análogos & derivados , Guanosina/farmacología , Humanos , Riñón/irrigación sanguínea , Riñón/citología , Riñón/metabolismo , Masculino , Células Mesangiales/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Miocardio/citología , Miocitos del Músculo Liso/efectos de los fármacos , Purina-Nucleósido Fosforilasa/metabolismo , Ratas , Ratas Endogámicas WKY
15.
Glia ; 61(10): 1595-606, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23922219

RESUMEN

Extracellular adenosine 3',5'-cyclic monophosphate (3',5'-cAMP) is an endogenous source of localized adenosine production in many organs. Recent studies suggest that extracellular 2',3'-cAMP (positional isomer of 3',5'-cAMP) is also a source of adenosine, particularly in the brain in vivo post-injury. Moreover, in vitro studies show that both microglia and astrocytes can convert extracellular 2',3'-cAMP to adenosine. Here, we examined the ability of primary mouse oligodendrocytes and neurons to metabolize extracellular 2',3'-cAMP and their respective adenosine monophosphates (2'-AMP and 3'-AMP). Cells were also isolated from mice deficient in 2',3'-cyclic nucleotide-3'-phosphodiesterase (CNPase). Oligodendrocytes metabolized 2',3'-cAMP to 2'-AMP with 10-fold greater efficiency than did neurons (and also more than previously examined microglia and astrocytes); whereas, the production of 3'-AMP was minimal in both oligodendrocytes and neurons. The production of 2'-AMP from 2',3'-cAMP was reduced by 65% in CNPase -/- versus CNPase +/+ oligodendrocytes. Oligodendrocytes also converted 2'-AMP to adenosine, and this was also attenuated in CNPase -/- oligodendrocytes. Inhibition of classic 3',5'-cAMP-3'-phosphodiesterases with 3-isobutyl-1-methylxanthine did not block metabolism of 2',3'-cAMP to 2'-AMP and inhibition of classic ecto-5'-nucleotidase (CD73) with α,ß-methylene-adenosine-5'-diphosphate did not attenuate the conversion of 2'-AMP to adenosine. These studies demonstrate that oligodendrocytes express the extracellular 2',3'-cAMP-adenosine pathway (2',3'-cAMP → 2'-AMP → adenosine). This pathway is more robustly expressed in oligodendrocytes than in all other CNS cell types because CNPase is the predominant enzyme that metabolizes 2',3'-cAMP to 2-AMP in CNS cells. By reducing levels of 2',3'-cAMP (a mitochondrial toxin) and increasing levels of adenosine (a neuroprotectant), oligodendrocytes may protect axons from injury.


Asunto(s)
2',3'-Nucleótido Cíclico Fosfodiesterasas/metabolismo , Adenosina/metabolismo , Líquido Extracelular/metabolismo , Oligodendroglía/metabolismo , Transducción de Señal/fisiología , 1-Metil-3-Isobutilxantina/farmacología , 2',3'-Nucleótido Cíclico Fosfodiesterasas/deficiencia , Nucleótidos de Adenina/metabolismo , Adenosina Monofosfato/metabolismo , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/farmacología , Animales , Células Cultivadas , Embrión de Mamíferos , Ratones , Ratones Transgénicos , Neuronas/efectos de los fármacos , Neuronas/fisiología , Oligodendroglía/efectos de los fármacos , Inhibidores de Fosfodiesterasa/farmacología , Transducción de Señal/genética
16.
Am J Physiol Renal Physiol ; 304(1): F49-55, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23077101

RESUMEN

In a previous study, we demonstrated that human proximal tubular epithelial cells obtained from a commercial source metabolized extracellular 2',3'-cAMP to 2'-AMP and 3'-AMP and extracellular 2'-AMP and 3'-AMP to adenosine (the extracellular 2',3'-cAMP-adenosine pathway; extracellular 2',3'-cAMP → 2'-AMP + 3'-AMP → adenosine). The purpose of this study was to investigate the metabolism of extracellular 2',3'-cAMP in proximal tubular vs. thick ascending limb vs. collecting duct epithelial cells freshly isolated from their corresponding nephron segments obtained from rat kidneys. In epithelial cells from all three nephron segments, 1) extracellular 2',3'-cAMP was metabolized to 2'-AMP and 3'-AMP, with 2'-AMP > 3'-AMP, 2) the metabolism of extracellular 2',3'-cAMP to 2'-AMP and 3'-AMP was not inhibited by either 3-isobutyl-1-methylxanthine (phosphodiesterase inhibitor) or 1,3-dipropyl-8-p-sulfophenylxanthine (ecto-phosphodiesterase inhibitor), 3) extracellular 2',3'-cAMP increased extracellular adenosine levels, 4) 3'-AMP and 2'-AMP were metabolized to adenosine with an efficiency similar to that of 5'-AMP, and 5) the metabolism of 5'-AMP, 3'-AMP, and 2'-AMP was not inhibited by α,ß-methylene-adenosine-5'-diphosphate (CD73 inhibitor). These results support the conclusion that renal epithelial cells all along the nephron can metabolize extracellular 2',3'-cAMP to 2'-AMP and 3'-AMP and can efficiently metabolize extracellular 2'-AMP and 3'-AMP to adenosine and that the metabolic enzymes involved are not the classical phosphodiesterases nor ecto-5'-nucleotidase (CD73). Because 2',3'-cAMP is released by injury and because previous studies demonstrate that the extracellular 2',3'-cAMP-adenosine pathway stimulates epithelial cell proliferation via adenosine A(2B) receptors, the present results suggest that the extracellular 2',3'-cAMP-adenosine pathway may help restore epithelial cells along the nephron following kidney injury.


Asunto(s)
Nucleótidos de Adenina/metabolismo , Adenosina/biosíntesis , Túbulos Renales Colectores/metabolismo , Túbulos Renales Proximales/metabolismo , Asa de la Nefrona/metabolismo , 1-Metil-3-Isobutilxantina/farmacología , Adenosina Difosfato/análogos & derivados , Adenosina Difosfato/farmacología , Adenosina Monofosfato/metabolismo , Animales , Células Epiteliales/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Xantinas/farmacología
17.
Am J Physiol Renal Physiol ; 304(6): F770-80, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23303411

RESUMEN

Previous studies show that neuropeptide Y(1-36) (NPY(1-36)) and peptide YY(1-36) (PYY(1-36)), by engaging Y1 receptors, stimulate proliferation of spontaneous hypertensive rat (SHR) preglomerular vascular smooth muscle cells (PGVSMCs). In contrast, these peptides have little effect on proliferation of Wistar-Kyoto (WKY) PGVSMCs. Why SHR and WKY PGVSMCs differ in this regard is unknown. Because receptor for activated C kinase 1 (RACK1) can modulate cell proliferation, we tested the hypothesis that differences in RACK1 levels/localization may explain the differential response of SHR vs. WKY PGVSMCs to NPY(1-36) and PYY(1-36). Western blotting for RACK1 in subcellular fractions of cultured SHR and WKY PGVSMCs demonstrated increased levels of RACK1 in the membrane and cytoskeletal subcellular fractions of SHR vs. WKY PGVSMCs. NPY(1-36) and PYY(1-36) stimulated proliferation of SHR PGVSMCs, and siRNA knockdown of RACK1 abrogated this effect. Neither NPY(1-36) nor PYY(1-36) stimulated the proliferation of WKY PGVSMCs. However, in WKY PGVSMCs treated with a RACK1 plasmid, both NPY(1-36) and PYY(1-36) stimulated proliferation. In SHR PGVSMCs, inhibitors of the G(i)/phospholipase C/PKC pathway (a pathway known to be organized by RACK1) attenuated the ability of NPY(1-36) to stimulate the proliferation of SHR PGVSMCs. Our results suggest that RACK1 modulates the ability of PGVSMCs to respond to the proliferative actions of NPY(1-36) and PYY(1-36)and differences in RACK1 levels/localization account for, in part, differential proliferative responses to NPY(1-36) and PYY(1-36) in SHR vs. WKY PGVSMCs. Because dipeptidyl peptidase IV inhibitors increase NPY(1-36) and PYY(1-36) levels, our findings have implications for the use of such drugs in diabetic patients.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Riñón/irrigación sanguínea , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/fisiología , Neuropéptido Y/fisiología , Péptido YY/fisiología , Animales , Proliferación Celular , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Masculino , Músculo Liso Vascular/fisiología , Plásmidos , Proteína Quinasa C/metabolismo , ARN Interferente Pequeño , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Receptores de Cinasa C Activada , Transducción de Señal , Timidina/metabolismo , Tritio/análisis , Fosfolipasas de Tipo C/metabolismo
18.
J Pharmacol Exp Ther ; 347(2): 516-28, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24023368

RESUMEN

Pleckstrin homology domain and leucine-rich repeat protein phosphatase 1 (PHLPP1) inhibits protein kinase B (AKT) survival signaling in neurons. Small molecule pan-PHLPP inhibitors (selective for PHLPP1 and PHLPP2) may offer a translatable method to induce AKT neuroprotection. We tested several recently discovered PHLPP inhibitors (NSC117079 and NSC45586; benzoic acid, 5-[2-[4-[2-(2,4-diamino-5-methylphenyl)diazenyl]phenyl]diazenyl]-2-hydroxy-,sodium salt.) in rat cortical neurons and astrocytes and compared the biochemical response of these agents with short hairpin RNA (shRNA)-mediated PHLPP1 knockdown (KD). In neurons, both PHLPP1 KD and experimental PHLPP inhibitors activated AKT and ameliorated staurosporine (STS)-induced cell death. Unexpectedly, in astrocytes, both inhibitors blocked AKT activation, and NSC117079 reduced viability. Only PHLPP2 KD mimicked PHLPP inhibitors on astrocyte biochemistry. This suggests that these inhibitors could have possible detrimental effects on astrocytes by blocking novel PHLPP2-mediated prosurvival signaling mechanisms. Finally, because PHLPP1 levels are reportedly high in the hippocampus (a region prone to ischemic death), we characterized hippocampal changes in PHLPP and several AKT targeting prodeath phosphatases after cardiac arrest (CA)-induced brain injury. PHLPP1 levels increased in rat brains subjected to CA. None of the other AKT inhibitory phosphatases increased after global ischemia (i.e., PHLPP2, PTEN, PP2A, and PP1). Selective PHLPP1 inhibition (such as by shRNA KD) activates AKT survival signaling in neurons and astrocytes. Nonspecific PHLPP inhibition (by NSC117079 and NSC45586) only activates AKT in neurons. Taken together, these results suggest that selective PHLPP1 inhibitors should be developed and may yield optimal strategies to protect injured hippocampal neurons and astrocytes-namely from global brain ischemia.


Asunto(s)
Antraquinonas/farmacología , Astrocitos/efectos de los fármacos , Compuestos Azo/farmacología , Fármacos Neuroprotectores/farmacología , Proteínas Nucleares/antagonistas & inhibidores , Fenilendiaminas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sulfonamidas/farmacología , Animales , Antraquinonas/química , Astrocitos/metabolismo , Astrocitos/patología , Compuestos Azo/química , Isquemia Encefálica/etiología , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Isquemia Encefálica/prevención & control , Técnicas de Cultivo de Célula , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Células HEK293 , Paro Cardíaco/complicaciones , Paro Cardíaco/metabolismo , Paro Cardíaco/patología , Humanos , Estructura Molecular , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Fármacos Neuroprotectores/química , Proteínas Nucleares/genética , Fenilendiaminas/química , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Sulfonamidas/química
19.
Hypertension ; 80(5): 981-994, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36802842

RESUMEN

BACKGROUND: The endogenous purine 8-aminoguanine induces diuresis/natriuresis/glucosuria by inhibiting PNPase (purine nucleoside phosphorylase); however, mechanistic details are unknown. METHODS: Here, we further explored in rats 8-aminoguanine's effects on renal excretory function by combining studies using intravenous 8-aminoguanine, intrarenal artery infusions of PNPase substrates (inosine and guanosine), renal microdialysis, mass spectrometry, selective adenosine receptor ligands, adenosine receptor knockout rats, laser doppler blood flow analysis, cultured renal microvascular smooth muscle cells, HEK293 cells expressing A2B receptors and homogeneous time resolved fluorescence assay for adenylyl cyclase activity. RESULTS: Intravenous 8-aminoguanine caused diuresis/natriuresis/glucosuria and increased renal microdialysate levels of inosine and guanosine. Intrarenal inosine, but not guanosine, exerted diuretic/natriuretic/glucosuric effects. In 8-aminoguanine-pretreated rats, intrarenal inosine did not induce additional diuresis/natriuresis/glucosuria. 8-Aminoguanine did not induce diuresis/natriuresis/glucosuria in A2B-receptor knockout rats, yet did so in A1- and A2A-receptor knockout rats. Inosine's effects on renal excretory function were abolished in A2B knockout rats. Intrarenal BAY 60-6583 (A2B agonist) induced diuresis/natriuresis/glucosuria and increased medullary blood flow. 8-Aminoguanine increased medullary blood flow, a response blocked by pharmacological inhibition of A2B, but not A2A, receptors. In HEK293 cells expressing A2B receptors, inosine activated adenylyl cyclase, and this was abolished by MRS 1754 (A2B antagonist). In renal microvascular smooth muscle cells, 8-aminoguanine and forodesine (PNPase inhibitor) increased inosine and 3',5'-cAMP; however, in cells from A2B knockout rats, 8-aminoguanine and forodesine did not augment 3',5'-cAMP yet increased inosine. CONCLUSIONS: 8-Aminoguanine induces diuresis/natriuresis/glucosuria by increasing renal interstitial levels of inosine which, via A2B receptor activation, increases renal excretory function, perhaps in part by increasing medullary blood flow.


Asunto(s)
Adenilil Ciclasas , Diuresis , Ratas , Humanos , Animales , Adenilil Ciclasas/farmacología , Células HEK293 , Diuréticos/farmacología , Natriuresis , Receptores Purinérgicos P1 , Inosina/farmacología
20.
Am J Physiol Renal Physiol ; 303(7): F954-62, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22791337

RESUMEN

Kidneys release into the extracellular compartment 3',5'-cAMP and its positional isomer 2',3'-cAMP. The purpose of the present study was to investigate the metabolism of extracellular 2',3'-cAMP and 3',5'-cAMP in preglomular vascular endothelial and proximal tubular epithelial cells and to determine whether these cAMPs and their downstream metabolites affect cellular proliferation. In preglomerular vascular endothelial and proximal tubular epithelial cells, 1) extracellular 2',3'-cAMP increased extracellular levels of 3'-AMP and 2'-AMP, whereas extracellular 3',5'-cAMP increased extracellular levels of 5'-AMP; 2) extracellular 5'-AMP, 3'-AMP, and 2'-AMP increased extracellular adenosine; 3) α,ß-methylene-adenosine-5'-diphosphate (CD73 inhibitor) prevented the 5'-AMP-induced increase in extracellular adenosine in preglomerular vascular endothelial cells, but did not affect the 5'-AMP-induced increase in extracellular adenosine in proximal tubular cells or the 3'-AMP-induced or 2'-AMP-induced increase in extracellular adenosine in either cell type; 4) extracellular 2',3'-cAMP, 3'-AMP, 2'-AMP, 3',5'-cAMP, 5'-AMP, and adenosine stimulated proliferation of both preglomerular vascular endothelial and proximal tubular cells; and 5) MRS-1754 (selective A(2B) receptor antagonist) abolished the progrowth effects of extracellular 2',3'-cAMP, 3'-AMP, 2'-AMP, 3',5'-cAMP, 5'-AMP, and adenosine in both cell types. Extracellular 2',3'-cAMP and 3',5'-cAMP stimulate proliferation of preglomerular vascular endothelial cells and proximal tubular cells. The mechanism by which the cAMPs increase cell proliferation entails 1) metabolism to their respective AMPs, 2) metabolism of their respective AMPs to adenosine (which for 5'-AMP in preglomerular vascular endothelial cells is mediated by CD73), and 3) activation of A(2B) receptors. Both extracellular 2',3'-cAMP and 3',5'-cAMP may help restore architecture of the preglomerular microcirculation and tubular system following kidney injury.


Asunto(s)
Proliferación Celular/efectos de los fármacos , AMP Cíclico/metabolismo , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Células Epiteliales/metabolismo , Riñón/metabolismo , Adenosina/metabolismo , Animales , Línea Celular , AMP Cíclico/farmacología , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Humanos , Riñón/citología , Riñón/efectos de los fármacos , Masculino , Ratas , Ratas Endogámicas WKY
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA