Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int J Nephrol Renovasc Dis ; 13: 19-26, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32110087

RESUMO

AIM OF STUDY: To investigate podocyte density in aging diabetic Ins2± and Ins2±, A1AR-/- mouse models in C57Bl/6 background. METHODS: Ins2± mice and especially Ins2±, adenosine A1 receptor knockout mice (Ins2±, A1AR-/-) are mouse models with a phenotype of diabetic nephropathy. Aged mice (at ~40 weeks) were assessed for glomerular filtration barrier function by measuring albuminuria, glomerular filtration, glomerular damage by electron microscopy, and podocyte numbers by Wilms Tumor protein (WT-1) staining. RESULTS: Compared to healthy wild-type mice, both diabetic mouse models developed diabetic nephropathy, including hyperfiltration (p<0.01) and albuminuria (p<0.05). Typical diabetic structural glomerular and podocyte damage was visualized by electron microscopy. Podocyte count per glomerular area (podocyte density) was significantly decreased in both diabetic mouse models (p<0.01). In contrast, no significant correlation was detected between albuminuria and absolute podocyte count per glomerulus. CONCLUSION: The amount of albuminuria as marker of diabetic nephropathy does not correlate with the podocytes density; however, a relative podocyte deficiency became evident with an increase in glomerular area in the diabetic animals, suggesting a relative podocytopenia.

2.
Pflugers Arch ; 469(2): 339-347, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27975140

RESUMO

Administration of the nucleoside adenosine has been shown to induce hypothermia in a number of species, an effect mediated predominantly by the adenosine 1 receptor (A1AR) subtype. The present experiments were performed to explore the possibility that the rise of intracellular adenosine levels expected to accompany adenosine administration may contribute to the hypothermic effect of adenosine independent of A1AR activation. Since phosphorylation of adenosine by adenosine kinase (ADK) is causal in the maintenance of low intracellular adenosine, we have examined the effect of ADK inhibition on core body temperature (CBT). Our data show that inhibition of ADK by A-134974 causes a long-lasting deep hypothermia in wild-type mice. Since there was an about 4-fold increase of adenosine plasma levels, experiments were repeated in A1AR-/- mice. ADK inhibition caused deep hypothermia despite the absence of A1AR, although the effect was significantly reduced compared to WT. Furthermore, the dose-dependent hypothermia caused by adenosine administration in WT mice was found to be reduced, but not abolished in A1AR-/- mice. To assess the possible role of A2AR and A3AR activation in our experimental setting, we compared the effects of the agonists CPA (A1AR), CGS21680 (A2AR), and IB-MECA (A3AR) on CBT. Hypothermia induced by CPA was much greater than that caused by CGS21680 or IB-MECA indicating that A1AR activation is the major receptor-dependent pathway for adenosine-induced hypothermia under our experimental conditions. Induction of deep hypothermia by inhibition of ADK, maintenance of this effect in A1AR-/- mice, and maintenance of adenosine-induced hypothermia in A1AR-deficient mice suggest that a receptor-independent action of adenosine requiring intact function of adenosine kinase contributes importantly to the hypothermia induced by adenosine.


Assuntos
Adenosina Quinase/antagonistas & inibidores , Adenosina/metabolismo , Hipotermia/metabolismo , Receptor A1 de Adenosina/metabolismo , Adenosina/análogos & derivados , Adenosina/farmacologia , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nucleosídeos/farmacologia , Fenetilaminas/farmacologia
3.
Annu Rev Physiol ; 77: 301-22, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25668021

RESUMO

Tubuloglomerular feedback (TGF) describes the negative relationship between (a) NaCl concentration at the macula densa and (b) glomerular filtration rate or glomerular capillary pressure. TGF-induced vasoconstriction of the afferent arteriole results from the enhanced effect of several vasoconstrictors with an effect size sequence of adenosine = 20-HETE > angiotensin II > thromboxane = superoxide > renal nerves > ATP. TGF-mediated vasoconstriction is limited by the simultaneous release of several vasodilators with an effect size sequence of nitric oxide > carbon monoxide = kinins > adenosine. The sum of the constrictor effects exceeds that of the dilator effects by the magnitude of the TGF response. The validity of the additive model used in this analysis can be tested by determining the effect of combined inhibition of some or all agents contributing to TGF. Multiple independent contributors to TGF are consistent with the variability of TGF and of the factors contributing to TGF resetting.


Assuntos
Retroalimentação Fisiológica/fisiologia , Glomérulos Renais/fisiologia , Túbulos Renais/fisiologia , Transdução de Sinais/fisiologia , Vasoconstrição/fisiologia , Animais , Arteríolas/fisiologia , Humanos , Rim/irrigação sanguínea , Modelos Animais , Vasoconstritores , Vasodilatadores
4.
Am J Physiol Renal Physiol ; 307(8): F939-48, 2014 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-25143457

RESUMO

Acute kidney injury (AKI) dramatically increases sepsis mortality, but AKI diagnosis is delayed when based on serum creatinine (SCr) changes, due in part, to decreased creatinine production. During experimental sepsis, we compared serum cystatin C (sCysC), SCr, and blood urea nitrogen (BUN) to inulin glomerular filtration rate (iGFR) before or 3-18 h after cecal ligation and puncture (CLP)-induced sepsis in CD-1 mice. sCysC had a faster increase and reached peak levels more rapidly than SCr in both sepsis and bilateral nephrectomy (BiNx) models. sCysC was a better surrogate of iGFR than SCr during sepsis. Combining sCysC with SCr values into a composite biomarker improved correlation with iGFR better than any biomarker alone or any other combination. We determined the renal contribution to sCysC handling with BiNx. sCysC and SCr were lower post-BiNx/CLP than post-BiNx alone, despite increased inflammatory and nonrenal organ damage biomarkers. Sepsis decreased CysC production in nephrectomized mice without changing body weight or CysC space. Sepsis decreased sCysC production and increased nonrenal clearance, similar to effects of sepsis on SCr. sCysC, SCr, and BUN were measured 6 h postsepsis to link AKI with mortality. Mice with above-median sCysC, BUN, or SCr values 6 h postsepsis died earlier than mice with below-median values, corresponding to a substantial AKI association with sepsis mortality in this model. sCysC performs similarly to SCr in classifying mice at risk for early mortality. We conclude that sCysC detects AKI early and better reflects iGFR in CLP-induced sepsis. This study shows that renal biomarkers need to be evaluated in specific contexts.


Assuntos
Injúria Renal Aguda/mortalidade , Biomarcadores/sangue , Creatinina/sangue , Cistatina C/sangue , Sepse/mortalidade , Injúria Renal Aguda/sangue , Injúria Renal Aguda/fisiopatologia , Animais , Nitrogênio da Ureia Sanguínea , Ceco/lesões , Taxa de Filtração Glomerular , Inulina , Ligadura , Masculino , Camundongos , Nefrectomia , Punções , Sepse/complicações
5.
Am J Physiol Renal Physiol ; 304(4): F382-9, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23269643

RESUMO

A(1) adenosine receptors (A1AR) are required for the modulation of afferent arteriolar tone by changes in luminal NaCl concentration implying that extracellular adenosine concentrations need to change in synchrony with NaCl. The present experiments were performed in mice with a null mutation in the gene for the major equilibrative nucleoside transporter ENT1 to test whether interference with adenosine disposition by cellular uptake of adenosine may modify TGF characteristics. Responses of stop flow pressure (P(SF)) to maximum flow stimulation were measured in mice with either C57Bl/6 or SWR/J genetic backgrounds. Maximum flow stimulation reduced P(SF) in ENT1(-/-) compared with wild-type (WT) mice by 1.6 ± 0.4 mmHg (n = 28) and 5.8 ± 1.1 mmHg (n = 17; P < 0.001) in C57Bl/6 and by 1.4 ± 0.4 mmHg (n = 15) and 9 ± 1.5 mmHg (n = 9; P < 0.001) in SWR/J. Plasma concentrations of adenosine and inosine were markedly higher in ENT1(-/-) than WT mice (ado: 1,179 ± 78 and 225 ± 48 pmol/ml; ino: 179 ± 24 and 47.5 ± 9 pmol/ml). Renal mRNA expressions of the four adenosine receptors, ENT2, and adenosine deaminase were not significantly different between WT and ENT1(-/-) mice. No significant differences of glomerular filtration rate or mean arterial blood pressure were found while plasma renin concentration, and heart rates were significantly lower in ENT1(-/-) animals. In conclusion, TGF responsiveness is significantly attenuated in the absence of ENT1, pointing to a role of nucleoside transport in the NaCl-synchronous changes of extracellular adenosine levels in the juxtaglomerular apparatus interstitium.


Assuntos
Transportador Equilibrativo 1 de Nucleosídeo/fisiologia , Deleção de Genes , Túbulos Renais/fisiologia , Adenosina/sangue , Adenosina Desaminase/biossíntese , Animais , Pressão Arterial/genética , Transportador Equilibrativo 1 de Nucleosídeo/genética , Transportador Equilibrativo 2 de Nucleosídeo/biossíntese , Feminino , Taxa de Filtração Glomerular/genética , Frequência Cardíaca/genética , Inosina/sangue , Glomérulos Renais/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores Purinérgicos P1/biossíntese , Renina/sangue
6.
Pflugers Arch ; 465(1): 39-51, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22665048

RESUMO

The intratubular composition of fluid at the tubulovascular contact site of the juxtaglomerular apparatus serves as regulatory input for secretion and synthesis of renin. Experimental evidence, mostly from in vitro perfused preparations, indicates an inverse relation between luminal NaCl concentration and renin secretion. The cellular transduction mechanism is initiated by concentration-dependent NaCl uptake through the Na-K-2Cl cotransporter (NKCC2) with activation of NKCC2 causing inhibition and deactivation of NKCC2 causing stimulation of renin release. Changes in NKCC2 activity are coupled to alterations in the generation of paracrine factors that interact with granular cells. Among these factors, generation of PGE2 in a COX-2-dependent fashion appears to play a dominant role in the stimulatory arm of tubular control of renin release. [NaCl] is a determinant of local PG release over an appropriate concentration range, and blockade of COX-2 activity interferes with the NaCl dependency of renin secretion. The complex array of local paracrine controls also includes nNOS-mediated synthesis of nitric oxide, with NO playing the role of a modifier of the intracellular signaling pathway. A role of adenosine may be particularly important when [NaCl] is increased, and at least some of the available evidence is consistent with an important suppressive effect of adenosine at higher salt concentrations.


Assuntos
Túbulos Renais/metabolismo , Renina/biossíntese , Animais , Ciclo-Oxigenase 2/metabolismo , Humanos , Sistema Justaglomerular/metabolismo , Túbulos Renais/anatomia & histologia , Túbulos Renais/fisiologia , Prostaglandinas/metabolismo , Renina/metabolismo , Sistema Renina-Angiotensina , Simportadores de Cloreto de Sódio-Potássio/metabolismo
7.
Ultrasonics ; 53(3): 727-38, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23218908

RESUMO

Ultrasound and Duplex ultrasonography in particular are routinely used to diagnose cardiovascular disease (CVD), which is the leading cause of morbidity and mortality worldwide. However, these techniques may not be able to characterize vascular tissue compositional changes due to CVD. This work describes an ultrasound-based hybrid imaging technique that can be used for vascular tissue characterization and the diagnosis of atherosclerosis. Ultrasound radiofrequency (RF) data were acquired and processed in time, frequency, and wavelet domains to extract six parameters including time integrated backscatter (T(IB)), time variance (T(var)), time entropy (T(E)), frequency integrated backscatter (F(IB)), wavelet root mean square value (W(rms)), and wavelet integrated backscatter (W(IB)). Each parameter was used to reconstruct an image co-registered to morphological B-scan. The combined set of hybrid images were used to characterize vascular tissue in vitro and in vivo using three mouse models including control (C57BL/6), and atherosclerotic apolipoprotein E-knockout (APOE-KO) and APOE/A(1) adenosine receptor double knockout (DKO) mice. The technique was tested using high-frequency ultrasound including single-element (center frequency=55 MHz) and commercial array (center frequency=40 MHz) systems providing superior spatial resolutions of 24 µm and 40 µm, respectively. Atherosclerotic vascular lesions in the APOE-KO mouse exhibited the highest values (contrast) of -10.11±1.92 dB, -12.13±2.13 dB, -7.54±1.45 dB, -5.10±1.06 dB, -5.25±0.94 dB, and -10.23±2.12 dB in T(IB), T(var), T(E), F(IB), W(rms), W(IB) hybrid images (n=10, p<0.05), respectively. Control segments of normal vascular tissue showed the lowest values of -20.20±2.71 dB, -22.54±4.54 dB, -14.94±2.05 dB, -9.64±1.34 dB, -10.20±1.27 dB, and -19.36±3.24 dB in same hybrid images (n=6, p<0.05). Results from both histology and optical images showed good agreement with ultrasound findings within a maximum error of 3.6% in lesion estimation. This study demonstrated the feasibility of a high-resolution hybrid imaging technique to diagnose atherosclerosis and characterize plaque components in mouse. In the future, it can be easily implemented on commercial ultrasound systems and eventually translated into clinics as a screening tool for atherosclerosis and the assessment of vulnerable plaques.


Assuntos
Doenças da Aorta/diagnóstico por imagem , Aterosclerose/diagnóstico por imagem , Ultrassonografia Doppler/instrumentação , Animais , Doenças da Aorta/patologia , Aterosclerose/patologia , Modelos Animais de Doenças , Processamento de Imagem Assistida por Computador , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Processamento de Sinais Assistido por Computador , Transdutores
8.
Transplantation ; 94(9): 894-902, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23073466

RESUMO

BACKGROUND: Liver ischemia-reperfusion injury (IRI) is a known risk factor for the postoperative outcome of patients undergoing liver surgery/transplantation. Attempts to protect from organ damage require multidisciplinary strategies and are of emerging interest in view of patients with higher age and American Society of Anesthesiology status. Ischemic preconditioning has been successfully applied to prevent from IRI during liver resection/transplantation. Because even short periods of ischemia during preconditioning inevitably lead to hypoxia and formation of anti-inflammatory/cytoprotective acting adenosine, we reasoned that short nonischemic hypoxia also protects against hepatic IRI. METHODS: Mice underwent hypoxic preconditioning (HPC) by breathing 10% oxygen for 10 min followed by 10 min of 21% oxygen before left liver lobe ischemia (45 min) and reperfusion (4 hr). The interactions of hypoxia→adenosine→adenosine receptors were tested by pharmacologic antagonism at adenosine receptor (AR) sites in wild-type mice and in mice with genetic deletions at the A1, A2A, A2B, and A3 ARs. Hepatocellular damage, inflammation, and metabolic effects were quantified by enzyme activities, cytokines, liver myeloperoxidase, blood adenosine, and tissue AMP, respectively. RESULTS: Hepatoprotection by HPC was significant in wild-type and A1, A2A, and A3 AR knockout mice as quantified by lower alanine aminotransferase serum activities, cytokine levels, histologic damage scores, tissue myeloperoxidase concentrations, and preserved AMP concentrations. Protection by HPC was blunted in mice pretreated with the A2B AR antagonist MRS1754 or in A2B AR knockout mice. CONCLUSIONS: Because liver protective effects of HPC are negated when the A2B receptor is nonfunctional, the hypoxia→adenosine→A2B receptor pathway plays a critical role in the prevention of warm IRI in vivo. Hypoxic activation of this pathway warrants use of selective A2B AR agonists or even intermittent hypoxia (e.g., in deceased organ donors) to protect from liver IRI.


Assuntos
Hipóxia/fisiopatologia , Precondicionamento Isquêmico , Fígado/irrigação sanguínea , Receptor A2B de Adenosina/fisiologia , Traumatismo por Reperfusão/prevenção & controle , Isquemia Quente , Acetamidas/farmacologia , Adenosina/fisiologia , Animais , Hepatócitos/patologia , Hepatócitos/fisiologia , Fígado/patologia , Fígado/fisiopatologia , Transplante de Fígado/patologia , Transplante de Fígado/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Purinas/farmacologia , Receptor A2B de Adenosina/deficiência , Receptor A2B de Adenosina/efeitos dos fármacos , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/fisiopatologia , Transdução de Sinais/fisiologia
9.
Am J Physiol Renal Physiol ; 303(8): F1166-75, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22896040

RESUMO

Adenosine 1 receptors (A1AR) have been shown in previous experiments to play a major role in the tubuloglomerular feedback (TGF) constrictor response of afferent arterioles (AA) to increased loop of Henle flow. Overexpression studies have pointed to a critical role of vascular A1AR, but it has remained unclear whether selective deletion of A1AR from smooth muscle cells is sufficient to abolish TGF responsiveness. To address this question, we have determined TGF response magnitude in mice in which vascular A1AR deletion was achieved using the loxP recombination approach with cre recombinase being controlled by a smooth muscle actin promoter (SmCre/A1ARff). Effective vascular deletion of A1AR was affirmed by absence of vasoconstrictor responses to adenosine or cyclohexyl adenosine (CHA) in microperfused AA. Elevation of loop of Henle flow from 0 to 30 nl/min caused a 22.1 ± 3.1% reduction of stop flow pressure in control mice and of 7.2 ± 1.5% in SmCre/A1ARff mice (P < 0.001). Maintenance of residual TGF activity despite absence of A1AR-mediated responses in AA suggests participation of extravascular A1AR in TGF. Support for this notion comes from the observation that deletion of A1ARff by nestin-driven cre causes an identical TGF response reduction (7.3 ± 2.4% in NestinCre/A1ARff vs. 20.3 ± 2.7% in controls), whereas AA responsiveness was reduced but not abolished. A1AR on AA smooth muscle cells are primarily responsible for TGF activation, but A1AR on extravascular cells, perhaps mesangial cells, appear to contribute to the TGF response.


Assuntos
Arteríolas/fisiologia , Pressão Sanguínea/fisiologia , Taxa de Filtração Glomerular/fisiologia , Rim/fisiologia , Receptor A1 de Adenosina/genética , Adenosina/farmacologia , Animais , Arteríolas/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Taxa de Filtração Glomerular/efeitos dos fármacos , Frequência Cardíaca/efeitos dos fármacos , Frequência Cardíaca/fisiologia , Rim/irrigação sanguínea , Rim/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Receptor A1 de Adenosina/metabolismo , Vasoconstrição/efeitos dos fármacos , Vasoconstrição/fisiologia
10.
Proc Natl Acad Sci U S A ; 109(16): 6265-70, 2012 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-22421436

RESUMO

Adenosine is a potent anticonvulsant acting on excitatory synapses through A1 receptors. Cellular release of ATP, and its subsequent extracellular enzymatic degradation to adenosine, could provide a powerful mechanism for astrocytes to control the activity of neural networks during high-intensity activity. Despite adenosine's importance, the cellular source of adenosine remains unclear. We report here that multiple enzymes degrade extracellular ATP in brain tissue, whereas only Nt5e degrades AMP to adenosine. However, endogenous A1 receptor activation during cortical seizures in vivo or heterosynaptic depression in situ is independent of Nt5e activity, and activation of astrocytic ATP release via Ca(2+) photolysis does not trigger synaptic depression. In contrast, selective activation of postsynaptic CA1 neurons leads to release of adenosine and synaptic depression. This study shows that adenosine-mediated synaptic depression is not a consequence of astrocytic ATP release, but is instead an autonomic feedback mechanism that suppresses excitatory transmission during prolonged activity.


Assuntos
Adenosina/metabolismo , Potenciais Pós-Sinápticos Excitadores/fisiologia , Retroalimentação Fisiológica/fisiologia , Neurônios/metabolismo , 5'-Nucleotidase/antagonistas & inibidores , 5'-Nucleotidase/genética , 5'-Nucleotidase/metabolismo , Difosfato de Adenosina/análogos & derivados , Difosfato de Adenosina/farmacologia , Monofosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Astrócitos/metabolismo , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Região CA1 Hipocampal/citologia , Região CA1 Hipocampal/metabolismo , Região CA1 Hipocampal/fisiologia , Proteínas Ligadas por GPI/antagonistas & inibidores , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Depressão Sináptica de Longo Prazo/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Técnicas de Patch-Clamp , Receptor A1 de Adenosina/metabolismo , Convulsões/metabolismo , Convulsões/fisiopatologia
11.
Kidney Int ; 81(6): 529-38, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22258323

RESUMO

The juxtaglomerular (JG) cell product renin is rate limiting in the generation of the bioactive octapeptide angiotensin II. Rates of synthesis and secretion of the aspartyl protease renin by JG cells are controlled by multiple afferent and efferent pathways originating in the CNS, cardiovascular system, and kidneys, and making critical contributions to the maintenance of extracellular fluid volume and arterial blood pressure. Since both excesses and deficits of angiotensin II have deleterious effects, it is not surprising that control of renin is secured by a complex system of feedforward and feedback relationships. Mice with genetic alterations have contributed to a better understanding of the networks controlling renin synthesis and secretion. Essential input for the setting of basal renin generation rates is provided by ß-adrenergic receptors acting through cyclic adenosine monophosphate, the primary intracellular activation mechanism for renin mRNA generation. Other major control mechanisms include COX-2 and nNOS affecting renin through PGE2, PGI2, and nitric oxide. Angiotensin II provides strong negative feedback inhibition of renin synthesis, largely an indirect effect mediated by baroreceptor and macula densa inputs. Adenosine appears to be a dominant factor in the inhibitory arms of the baroreceptor and macula densa mechanisms. Targeted gene mutations have also shed light on a number of novel aspects related to renin processing and the regulation of renin synthesis and secretion.


Assuntos
Sistema Justaglomerular/enzimologia , Sistema Justaglomerular/metabolismo , Mutação , Sistema Renina-Angiotensina/genética , Renina/biossíntese , Renina/metabolismo , Animais , Animais Geneticamente Modificados , Conexinas/genética , Conexinas/metabolismo , Retroalimentação Fisiológica , Regulação da Expressão Gênica , Genótipo , Camundongos , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase/metabolismo , Fenótipo , Prostaglandina-Endoperóxido Sintases/genética , Prostaglandina-Endoperóxido Sintases/metabolismo , Receptores Adrenérgicos/genética , Receptores Adrenérgicos/metabolismo , Receptores de Prostaglandina/genética , Receptores de Prostaglandina/metabolismo , Receptores Purinérgicos P1/genética , Receptores Purinérgicos P1/metabolismo , Transdução de Sinais/genética
12.
Clin Exp Nephrol ; 16(1): 17-24, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22124804

RESUMO

Control of the renin system by physiological mechanisms such as the baroreceptor or the macula densa (MD) is characterized by asymmetry in that the capacity for renin secretion and expression to increase is much larger than the magnitude of the inhibitory response. The large stimulatory reserve of the renin-angiotensin system may be one of the causes for the remarkable salt-conserving power of the mammalian kidney. Physiological stimulation of renin secretion and expression relies on the activation of regulatory pathways that converge on the cyclic adenosine monophosphate/protein kinase A (cAMP/PKA) pathway. Mice with selective Gs-alpha (Gsα) deficiency in juxtaglomerular granular cells show a marked reduction of basal renin secretion, and an almost complete unresponsiveness of renin release to furosemide, hydralazine, or isoproterenol. Cyclooxygenase-2 generating prostaglandin E(2) (PGE(2)) and prostacyclin (PGI(2)) in MD and thick ascending limb cells is one of the main effector systems utilizing Gsα-coupled receptors to stimulate the renin-angiotensin system. In addition, ß-adrenergic receptors are critical for the expression of high basal levels of renin and for its release response to lowering blood pressure or MD sodium chloride concentration. Nitric oxide generated by nitric oxide synthases in the MD and in endothelial cells enhances cAMP-dependent signaling by stabilizing cAMP through cyclic guanosine monophosphate-dependent inhibition of phosphodiesterase 3. The stimulation of renin secretion by drugs that inhibit angiotensin II formation or action results from the convergent activation of cAMP probably through indirect augmentation of the activity of PGE(2) and PGI(2) receptors, ß-adrenergic receptors, and nitric oxide.


Assuntos
AMP Cíclico/fisiologia , Subunidades alfa Gs de Proteínas de Ligação ao GTP/fisiologia , Renina/metabolismo , Transdução de Sinais/fisiologia , Animais , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Ciclo-Oxigenase 2/fisiologia , Diuréticos/farmacologia , Furosemida/farmacologia , Subunidades alfa Gs de Proteínas de Ligação ao GTP/deficiência , Sistema Justaglomerular/citologia , Sistema Justaglomerular/metabolismo , Túbulos Renais Distais/metabolismo , Camundongos , Óxido Nítrico Sintase/metabolismo , Prostaglandinas/fisiologia , Receptores Adrenérgicos beta/metabolismo , Sistema Renina-Angiotensina/fisiologia , Transdução de Sinais/efeitos dos fármacos
13.
Kidney Int ; 80(11): 1198-211, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21832986

RESUMO

We have shown that folate-induced kidney dysfunction and interstitial fibrosis predisposes mice to sepsis mortality. Agents that increase survival in normal septic mice were ineffective in a two-stage kidney disease model. Here we used the 5/6 nephrectomy mouse model of progressive chronic kidney disease (CKD) to study how CKD affects acute kidney injury (AKI) induced by sepsis. We induced sepsis using cecal ligation and puncture and found that the presence of CKD intensified the severity of kidney and liver injury, cytokine release, and splenic apoptosis. Accumulation of High Mobility Group Box Protein-1 (HMGB1; a late proinflammatory cytokine released from apoptotic cells), vascular endothelial growth factor (VEGF), tumor necrosis factor (TNF)-α, interleukin (IL)-6, or IL-10 was increased in CKD or sepsis alone and to a greater extent in CKD-sepsis. Only part of the increase was explained by decreased renal clearance. Surprisingly, we found splenic apoptosis in CKD, even in the absence of sepsis. Although VEGF neutralization with soluble fms-like tyrosine kinase 1 (sFLT-1) (a soluble VEGF receptor) effectively treated sepsis, it was ineffective against CKD-sepsis. A single dose of HMGB1-neutralizing antiserum administered 6 h after sepsis alone was ineffective; however, CKD-sepsis was attenuated by anti-HMGB1. Splenectomy transiently decreased circulating HMGB1 levels, reversing the effectiveness of anti-HMGB1 treatment on CKD-sepsis. Thus, progressive CKD increases the severity of sepsis, in part, by reducing the renal clearance of several cytokines. CKD-induced splenic apoptosis and HMGB1 release could be important common mediators for both CKD and sepsis.


Assuntos
Proteína HMGB1/metabolismo , Nefropatias/etiologia , Insuficiência Renal Crônica/patologia , Sepse/complicações , Animais , Anticorpos/administração & dosagem , Anticorpos/uso terapêutico , Apoptose , Progressão da Doença , Proteína HMGB1/imunologia , Nefropatias/complicações , Nefropatias/terapia , Camundongos , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/metabolismo , Baço/patologia , Esplenectomia , Resultado do Tratamento
14.
Diabetes ; 60(10): 2578-87, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21831968

RESUMO

OBJECTIVE: The role of adenosine (ADO) in the regulation of glucose homeostasis is not clear. In the current study, we used A1-ADO receptor (A1AR)-deficient mice to investigate the role of ADO/A1AR signaling for glucose homeostasis. RESEARCH DESIGN AND METHODS: After weaning, A1AR(-/-) and wild-type mice received either a standard diet (12 kcal% fat) or high-fat diet (HFD; 45 kcal% fat). Body weight, fasting plasma glucose, plasma insulin, and intraperitoneal glucose tolerance tests were performed in 8-week-old mice and again after 12-20 weeks of subsequent observation. Body composition was quantified by magnetic resonance imaging and epididymal fat-pad weights. Glucose metabolism was investigated by hyperinsulinemic-euglycemic clamp studies. To describe pathophysiological mechanisms, adipokines and Akt phosphorylation were measured. RESULTS: A1AR(-/-) mice were significantly heavier than wild-type mice because of an increased fat mass. Fasting plasma glucose and insulin were significantly higher in A1AR(-/-) mice after weaning and remained higher in adulthood. An intraperitoneal glucose challenge disclosed a significantly slower glucose clearance in A1AR(-/-) mice. An HFD enhanced this phenotype in A1AR(-/-) mice and unmasked a dysfunctional insulin secretory mechanism. Insulin sensitivity was significantly impaired in A1AR(-/-) mice on the standard diet shortly after weaning. Clamp studies detected a significant decrease of net glucose uptake in A1AR(-/-) mice and a reduced glucose uptake in muscle and white adipose tissue. Effects were not triggered by leptin deficiency but involved a decreased Akt phosphorylation. CONCLUSIONS: ADO/A1AR signaling contributes importantly to insulin-controlled glucose homeostasis and insulin sensitivity in C57BL/6 mice and is involved in the metabolic regulation of adipose tissue.


Assuntos
Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo , Receptor A1 de Adenosina/genética , Receptor A1 de Adenosina/metabolismo , Transdução de Sinais/fisiologia , Adiposidade , Animais , Glicemia/metabolismo , Peso Corporal , Gorduras na Dieta/administração & dosagem , Gorduras na Dieta/efeitos adversos , Ingestão de Alimentos , Feminino , Insulina/sangue , Insulina/metabolismo , Masculino , Camundongos , Camundongos Knockout , Atividade Motora , Transdução de Sinais/genética
15.
Handb Exp Pharmacol ; (200): 391-412, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-20859805

RESUMO

This chapter describes the effects of the natural methylxanthines caffeine and theophylline on kidney function. Theophylline in particular was used traditionally to increase urine out put until more potent diuretics became available in the middle of the last century. The mildly diuretic actions of both methylxanthines are mainly the result of inhibition of tubular fluid reabsorption along the renal proximal tubule. Based upon the use of specific adenosine receptor antagonists and the observation of a complete loss of diuresis in mice with targeted deletion of the A1AR gene, transport inhibition by methylxanthines is mediated mainly by antagonism of adenosine A1 receptors (A1AR) in the proximal tubule. Methylxanthines are weak renal vasodilators, and they act as competitive antagonists against adenosine-induced preglomerular vasoconstriction. Caffeine and theophylline stimulate the secretion of renin by inhibition of adenosine receptors and removal of the general inhibitory brake function of endogenous adenosine. Since enhanced intrarenal adenosine levels lead to reduced glomerular filtration rate in several pathological conditions theophylline has been tested for its therapeutic potential in the renal impairment following administration of nephrotoxic substances such as radiocontrast media, cisplatin, calcineurin inhibitors or following ischemia-reperfusion injury. In experimental animals functional improvements have been observed in all of these conditions, but available clinical data in humans are insufficient to affirm a definite therapeutic efficacy of methylxanthines in the prevention of nephrotoxic or postischemic renal injury.


Assuntos
Cafeína/farmacologia , Rim/efeitos dos fármacos , Teofilina/farmacologia , Animais , Meios de Contraste/toxicidade , Diurese/efeitos dos fármacos , Hemodinâmica/efeitos dos fármacos , Humanos , Rim/fisiologia , Natriurese/efeitos dos fármacos , Renina/metabolismo
16.
Am J Physiol Renal Physiol ; 300(2): F339-44, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21147842

RESUMO

Tubuloglomerular feedback (TGF), the change of afferent arteriolar resistance initiated by changes of luminal NaCl concentration, is thought to be related to NaCl-dependent release of ATP by macula densa cells. In the present study, we have explored the possibility that the released ATP may directly interact with vasoconstrictor P2 purinergic receptors in the vicinity of the glomerular vascular pole. In two different strains of wild-type mice (SWR/J and FVB), TGF responses were determined in vivo by measuring the stop flow pressure (P(SF)) change caused by a saturating increase in loop of Henle flow rate before and during the administration of the P2 receptor inhibitors PPADS (12 mg/kg + 35 mg·kg(-1)·h(-1) iv) or suramin (50 mg/kg + 150 mg·kg(-1)·h(-1)). Both agents significantly reduced the blood pressure response to the P2X agonist α,ß-methylene ATP. In SWR/J and FVB mice, elevating flow to 30 nl/min reduced P(SF) by 16.4 ± 2.2 and 17.1 ± 1.8%. During infusion of PPADS, P(SF) fell by 18.8 ± 2 (P = 0.4) and 16.5 ± 1.5% (P = 0.82) in the two strains of mice. During suramin infusion, P(SF) decreased by 14.7 ± 2.4 (P = 0.62) and 15 ± 1.3% (P = 0.4) in SWR/J and FVB mice, respectively. Including PPADS (10(-4) M) in the loop perfusate did not significantly alter the P(SF) response (18.9 ± 1.8%; P = 0.54). Arterial blood pressure was not systematically affected by the P2 inhibitors. As measured by free-flow micropuncture, PPADS significantly reduced proximal tubular fluid reabsorption in both fractional and absolute terms. These results indicate that the direct activation of P2 purinergic receptors by ATP is not a major cause of TGF-induced vasoconstriction in vivo.


Assuntos
Retroalimentação Fisiológica , Glomérulos Renais/fisiologia , Túbulos Renais/fisiologia , Receptores Purinérgicos P2/fisiologia , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/farmacologia , Animais , Arteríolas/efeitos dos fármacos , Arteríolas/fisiologia , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Taxa de Filtração Glomerular/efeitos dos fármacos , Taxa de Filtração Glomerular/fisiologia , Glomérulos Renais/efeitos dos fármacos , Túbulos Renais/efeitos dos fármacos , Masculino , Camundongos , Agonistas do Receptor Purinérgico P2/farmacologia , Antagonistas do Receptor Purinérgico P2/farmacologia , Fosfato de Piridoxal/análogos & derivados , Fosfato de Piridoxal/farmacologia , Suramina/farmacologia , Vasoconstrição/efeitos dos fármacos , Vasoconstrição/fisiologia
17.
Nat Neurosci ; 13(7): 883-8, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20512135

RESUMO

Acupuncture is an invasive procedure commonly used to relieve pain. Acupuncture is practiced worldwide, despite difficulties in reconciling its principles with evidence-based medicine. We found that adenosine, a neuromodulator with anti-nociceptive properties, was released during acupuncture in mice and that its anti-nociceptive actions required adenosine A1 receptor expression. Direct injection of an adenosine A1 receptor agonist replicated the analgesic effect of acupuncture. Inhibition of enzymes involved in adenosine degradation potentiated the acupuncture-elicited increase in adenosine, as well as its anti-nociceptive effect. These observations indicate that adenosine mediates the effects of acupuncture and that interfering with adenosine metabolism may prolong the clinical benefit of acupuncture.


Assuntos
Analgesia por Acupuntura , Adenosina/metabolismo , Vias Aferentes/metabolismo , Dor/metabolismo , Doenças do Sistema Nervoso Periférico/metabolismo , Receptores Purinérgicos P1/metabolismo , Análise de Variância , Animais , Modelos Animais de Doenças , Camundongos , Nucleotidases/metabolismo , Dor/complicações , Manejo da Dor , Doenças do Sistema Nervoso Periférico/complicações , Doenças do Sistema Nervoso Periférico/terapia , Ciática/complicações , Ciática/metabolismo , Ciática/terapia , Estatísticas não Paramétricas
18.
J Am Soc Nephrol ; 21(6): 986-92, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20395378

RESUMO

Angiotensin II converting enzyme inhibitors (ACEI) or angiotensin II receptor blockers (ARB) presumably stimulate renin secretion by interrupting angiotensin II feedback inhibition. The increase in cytosolic calcium caused by activation of Gq-coupled AT1 receptors may mediate the renin-inhibitory effect of angiotensin II at the cellular level, implying that ACEI and ARB may work by reducing intracellular calcium. Here, we investigated whether angiotensin II blockade acts predominantly through Gs-mediated stimulation of adenylyl cyclase (AC) by testing the effect of ACEI and ARB in mice with juxtaglomerular cell-specific deficiency of the AC-stimulatory Gsalpha. The ACEI captopril and quinaprilate and the ARB candesartan significantly increased plasma renin concentration (PRC) to 20 to 40 times basal PRC in wild-type mice but did not significantly alter PRC in Gsalpha-deficient mice. Captopril also completely abrogated renin stimulation in wild-type mice after co-administration of propranolol, indomethacin, and L-NAME. Treatment with enalapril and a low-NaCl diet for 7 days led to a 35-fold increase in PRC among wild-type mice but no significant change in PRC among Gsalpha-deficient mice. Three different pharmacologic inhibitors of AC reduced the stimulatory effect of captopril by 70% to 80%. In conclusion, blockade of angiotensin II stimulates renin synthesis and release indirectly through the action of ligands that activate the cAMP/PKA pathway in a Gsalpha-dependent fashion, including catecholamines, prostaglandins, and nitric oxide.


Assuntos
Adenilil Ciclases/metabolismo , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Angiotensina II/antagonistas & inibidores , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Sistema Justaglomerular/metabolismo , Renina/metabolismo , Angiotensina II/metabolismo , Animais , Benzimidazóis/farmacologia , Compostos de Bifenilo , Captopril/farmacologia , Catecolaminas/metabolismo , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Enalapril/farmacologia , Feminino , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Sistema Justaglomerular/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Modelos Animais , Óxido Nítrico/metabolismo , Prostaglandinas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tetra-Hidroisoquinolinas/farmacologia , Tetrazóis/farmacologia
19.
J Clin Invest ; 120(5): 1429-40, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20407207

RESUMO

Sphingosine kinase 1 (SphK1) and SphK2 are ubiquitous enzymes that generate sphingosine-1-phosphate (S1P), a ligand for a family of G protein-coupled receptors (S1PR1-S1PR5) with important functions in the vascular and immune systems. Here we explore the role of these kinases and receptors in recovery from anaphylaxis in mice. We found that Sphk2-/- mice had a rapid recovery from anaphylaxis. In contrast, Sphk1-/- mice showed poor recovery from anaphylaxis and delayed histamine clearance. Injection of S1P into Sphk1-/- mice increased histamine clearance and promoted recovery from anaphylaxis. Adoptive cell transfer experiments demonstrated that SphK1 activity was required in both the hematopoietic and nonhematopoietic compartments for recovery from anaphylaxis. Mice lacking the S1P receptor S1PR2 also showed a delay in plasma histamine clearance and a poor recovery from anaphylaxis. However, S1P did not promote the recovery of S1pr2-/- mice from anaphylaxis, whereas S1pr2+/- mice showed partial recovery. Unlike Sphk2-/- mice, Sphk1-/- and S1pr2-/- mice had severe hypotension during anaphylaxis. Thus, SphK1-produced S1P regulates blood pressure, histamine clearance, and recovery from anaphylaxis in a manner that involves S1PR2. This suggests that specific S1PR2 agonists may serve to counteract the vasodilation associated with anaphylactic shock.


Assuntos
Anafilaxia/enzimologia , Regulação Enzimológica da Expressão Gênica , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Animais , Transplante de Medula Óssea , Taxa de Filtração Glomerular , Células-Tronco Hematopoéticas/metabolismo , Histamina/metabolismo , Rim/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Biológicos , Permeabilidade
20.
J Neurotrauma ; 27(5): 901-10, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20121416

RESUMO

We reported that adenosine A(1) receptor (A(1)AR) knockout (KO) mice develop lethal status epilepticus after experimental traumatic brain injury (TBI), which is not seen in wild-type (WT) mice. Studies in epilepsy, multiple sclerosis, and neuro-oncology suggest enhanced neuro-inflammation and/or neuronal death in A(1)AR KO. We hypothesized that A(1)AR deficiency exacerbates the microglial response and neuronal damage after TBI. A(1)AR KO and WT littermates were subjected to mild controlled cortical impact (3 m/sec; 0.5 mm depth) to left parietal cortex, an injury level below the acute seizure threshold in the KO. At 24 h or 7 days, mice were sacrificed and serial sections prepared. Iba-1 immunostaining was used to quantify microglia at 7 days. To assess neuronal injury, sections were stained with Fluoro-Jade C (FJC) at 24 h to evaluate neuronal death in the hippocampus and cresyl violet staining at 7 days to analyze cortical lesion volumes. We also studied the effects of adenosine receptor agonists and antagonists on (3)H-thymidine uptake (proliferation index) by BV-2 cells (immortalized mouse microglial). There was no neuronal death in CA1 or CA3 quantified by FJC. A(1)AR KO mice exhibited enhanced microglial response; specifically, Iba-1 + microglia were increased 20-50% more in A(1)AR KO versus WT in ipsilateral cortex, CA3, and thalamus, and contralateral cortex, CA1, and thalamus (p < 0.05). However, contusion and cortical volumes did not differ between KO and WT. Pharmacological studies in cultured BV-2 cells indicated that A(1)AR activation inhibits microglial proliferation. A(1)AR activation is an endogenous inhibitor of the microglial response to TBI, likely via inhibition of proliferation, and this may represent a therapeutic avenue to modulate microglia after TBI.


Assuntos
Lesões Encefálicas/metabolismo , Gliose/metabolismo , Inibidores do Crescimento/fisiologia , Microglia/metabolismo , Receptor A1 de Adenosina/metabolismo , Adenosina/metabolismo , Animais , Lesões Encefálicas/complicações , Lesões Encefálicas/patologia , Linhagem Celular Transformada , Córtex Cerebral/lesões , Córtex Cerebral/patologia , Modelos Animais de Doenças , Feminino , Gliose/tratamento farmacológico , Gliose/patologia , Inibidores do Crescimento/deficiência , Inibidores do Crescimento/genética , Hipocampo/metabolismo , Hipocampo/patologia , Masculino , Camundongos , Camundongos Knockout , Microglia/efeitos dos fármacos , Microglia/patologia , Degeneração Neural/etiologia , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Receptor A1 de Adenosina/deficiência , Receptor A1 de Adenosina/genética , Tálamo/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA