Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Am J Physiol Renal Physiol ; 309(5): F414-28, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26136559

RESUMEN

Renal hypoxia contributes to chronic kidney disease (CKD) progression, as validated in experimental and human CKD. In the early stages, increased oxygen consumption causes oxygen demand/supply mismatch, leading to hypoxia. Hence, early targeting of the determinants and regulators of oxygen consumption in CKD may alter the disease course before permanent damage ensues. Here, we focus on hypoxia inducible factor-1α (HIF-1α) and AMP-activated protein kinase (AMPK) and on the mechanisms by which they may facilitate cellular hypoxia adaptation. We found that HIF-1α activation in the subtotal nephrectomy (STN) model of CKD limits protein synthesis, inhibits apoptosis, and activates autophagy, presumably for improved cell survival. AMPK activation was diminished in the STN kidney and was remarkably restored by HIF-1α activation, demonstrating a novel role for HIF-1α in the regulation of AMPK activity. We also investigated the independent and combined effects of HIF-1α and AMPK on cell survival and death pathways by utilizing pharmacological and knockdown approaches in cell culture models. We found that the effect of HIF-1α activation on autophagy is independent of AMPK, but on apoptosis it is partially AMPK dependent. The effects of HIF-1α and AMPK activation on inhibiting protein synthesis via the mTOR pathway appear to be additive. These various effects were also observed under hypoxic conditions. In conclusion, HIF-1α and AMPK appear to be linked at a molecular level and may act as components of a concerted cellular response to hypoxic stress in the pathophysiology of CKD.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Adaptación Fisiológica/fisiología , Hipoxia de la Célula/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Insuficiencia Renal Crónica/metabolismo , Animales , Apoptosis/fisiología , Autofagia/fisiología , Modelos Animales de Enfermedad , Masculino , Nefrectomía , Ratas , Ratas Wistar , Circulación Renal/fisiología , Serina-Treonina Quinasas TOR/metabolismo
2.
Nephron Exp Nephrol ; 2014 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-25503637

RESUMEN

Background: Kidney ischemia-reperfusion is a form of acute kidney injury resulting in a cascade of cellular events prompting rapid cellular damage and suppression of kidney function. A cellular response to ischemic stress is the activation of AMP-activated protein kinase (AMPK), where AMPK induces a number of homeostatic and renoprotective mechanisms, including autophagy. However, whether autophagy is beneficial or detrimental in ischemia-reperfusion remains controversial. We investigated the effects of agonist induction of AMPK activity on autophagy and cell stress proteins in the model of kidney ischemia-reperfusion. Methods: AMPK agonists, AICAR (0.1 g/kg) and metformin (0.3 g/kg), were administered 24 h prior to ischemia, with kidneys harvested at 24 h of reperfusion. Results: We observed a paradoxical decrease in AMPK activity accompanied by increases in mammalian target of rapamycin (mTOR) C1 activity and p62/SQSTM1 expression. These results led us to propose that AMPK and autophagy are insufficient to properly counter the cellular insults in ischemia-reperfusion. Agonist induction of AMPK activity with AICAR or metformin increased macroautophagy protein LC3 and normalized p62/SQSTM1 expression and mTOR activity. Ischemia-reperfusion increases in Beclin-1 and PINK1 expressions, consistent with increased mitophagy, were also mitigated with AMPK agonists. Stress-responsive and apoptotic marker expressions increase in ischemia-reperfusion and are significantly attenuated with agonist administration, as are early indicators of fibrosis. Conclusions: Our data suggest that levels of renoprotective AMPK activity and canonical autophagy are insufficient to maintain cellular homeostasis, contributing to the progression of ischemia-reperfusion injury. We further demonstrate that induction of AMPK activity can provide beneficial cellular effects in containing injury in ischemia-reperfusion. © 2014 S. Karger AG, Basel.

3.
Am J Physiol Renal Physiol ; 306(2): F194-204, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24226524

RESUMEN

Our previous work has shown that gene knockout of the sodium-glucose cotransporter SGLT2 modestly lowered blood glucose in streptozotocin-diabetic mice (BG; from 470 to 300 mg/dl) and prevented glomerular hyperfiltration but did not attenuate albuminuria or renal growth and inflammation. Here we determined effects of the SGLT2 inhibitor empagliflozin (300 mg/kg of diet for 15 wk; corresponding to 60-80 mg·kg(-1)·day(-1)) in type 1 diabetic Akita mice that, opposite to streptozotocin-diabetes, upregulate renal SGLT2 expression. Akita diabetes, empagliflozin, and Akita + empagliflozin similarly increased renal membrane SGLT2 expression (by 38-56%) and reduced the expression of SGLT1 (by 33-37%) vs. vehicle-treated wild-type controls (WT). The diabetes-induced changes in SGLT2/SGLT1 protein expression are expected to enhance the BG-lowering potential of SGLT2 inhibition, and empagliflozin strongly lowered BG in Akita (means of 187-237 vs. 517-535 mg/dl in vehicle group; 100-140 mg/dl in WT). Empagliflozin modestly reduced GFR in WT (250 vs. 306 µl/min) and completely prevented the diabetes-induced increase in glomerular filtration rate (GFR) (255 vs. 397 µl/min). Empagliflozin attenuated increases in kidney weight and urinary albumin/creatinine ratio in Akita in proportion to hyperglycemia. Empagliflozin did not increase urinary glucose/creatinine ratios in Akita, indicating the reduction in filtered glucose balanced the inhibition of glucose reabsorption. Empagliflozin attenuated/prevented the increase in systolic blood pressure, glomerular size, and molecular markers of kidney growth, inflammation, and gluconeogenesis in Akita. We propose that SGLT2 inhibition can lower GFR independent of reducing BG (consistent with the tubular hypothesis of diabetic glomerular hyperfiltration), while attenuation of albuminuria, kidney growth, and inflammation in the early diabetic kidney may mostly be secondary to lower BG.


Asunto(s)
Albuminuria/tratamiento farmacológico , Compuestos de Bencidrilo/farmacología , Diabetes Mellitus/genética , Nefropatías Diabéticas/prevención & control , Glucósidos/farmacología , Hiperglucemia/metabolismo , Glomérulos Renales/metabolismo , Riñón/patología , Inhibidores del Cotransportador de Sodio-Glucosa 2 , Adipocitos/efectos de los fármacos , Adipocitos/ultraestructura , Animales , Biomarcadores/metabolismo , Glucemia/metabolismo , Presión Sanguínea/fisiología , Western Blotting , Nefropatías Diabéticas/metabolismo , Ingestión de Líquidos/fisiología , Ingestión de Alimentos/fisiología , Tasa de Filtración Glomerular/efectos de los fármacos , Frecuencia Cardíaca/fisiología , Hiperglucemia/complicaciones , Inflamación/metabolismo , Riñón/efectos de los fármacos , Riñón/crecimiento & desarrollo , Glomérulos Renales/crecimiento & desarrollo , Ratones , Ratones Endogámicos , Reacción en Cadena en Tiempo Real de la Polimerasa , Transportador 1 de Sodio-Glucosa/biosíntesis , Transportador 1 de Sodio-Glucosa/genética , Transportador 2 de Sodio-Glucosa
4.
Kidney Int ; 85(3): 611-23, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24304883

RESUMEN

AMP-activated protein kinase (AMPK) is an important energy sensor that may be critical in regulating renal lipid accumulation. To evaluate the role of AMPK in mediating renal lipid accumulation, C57BL/6J mice were randomized to a standard diet, a high-fat diet, or a high-fat diet plus AICAR (an AMPK activator) for 14 weeks. Renal functional and structural studies along with electron microscopy were performed. Mice given the high-fat diet had proximal tubule injury with the presence of enlarged clear vacuoles, and multilaminar inclusions concurrent with an increase of tissue lipid and overloading of the lysosomal system. The margins of the clear vacuoles were positive for the endolysosomal marker, LAMP1, suggesting lysosome accumulation. Characterization of vesicles by special stains (Oil Red O, Nile Red, Luxol Fast Blue) and by electron microscopy showed they contained onion skin-like accumulations consistent with phospholipids. Moreover, cholesteryl esters and phosphatidylcholine-containing phospholipids were significantly increased in the kidneys of mice on a high-fat diet. AMPK activation with chronic AICAR treatment prevented the clinical and structural effects of high-fat diet. Thus, high-fat diet contributes to a dysfunction of the lysosomal system and altered lipid metabolism characterized by cholesterol and phospholipid accumulation in the kidney. AMPK activation normalizes the changes in renal lipid content despite chronic exposure to lipid challenge.


Asunto(s)
Proteínas Quinasas Activadas por AMP/fisiología , Riñón/metabolismo , Metabolismo de los Lípidos , Albuminuria/prevención & control , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Colesterol/metabolismo , Dieta Alta en Grasa , Resistencia a la Insulina , Riñón/patología , Ratones Endogámicos C57BL , Mitocondrias/fisiología , Obesidad/prevención & control , Ribonucleótidos/farmacología
5.
Nephrol Dial Transplant ; 28 Suppl 4: iv29-36, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23901047

RESUMEN

Obesity is a long-term source of cellular stress that predisposes to chronic kidney disease (CKD). Autophagy is a homeostatic mechanism for cellular quality control through the disposal and recycling of cellular components. During times of cellular stress, autophagy affords mechanisms to manage stress by selectively ridding the cell of the accumulation of potentially toxic proteins, lipids and organelles. The adaptive processes employed may vary between cell types and selectively adjust to the insult by inducing components of the basic autophagy machinery utilized by the cells while not under duress. In this review, we will discuss the autophagic responses of organs to cellular stressors, such as high-fat diet, obesity and diabetes, and how these mechanisms may prevent or promote the progression of disease. The identification of early cellular mechanisms in the advent of obesity- and diabetes-related renal complications could afford avenues for future therapeutic interventions.


Asunto(s)
Autofagia/fisiología , Obesidad/metabolismo , Insuficiencia Renal Crónica/metabolismo , Animales , Humanos , Obesidad/complicaciones , Insuficiencia Renal Crónica/etiología
6.
Am J Physiol Renal Physiol ; 305(5): F727-33, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23825068

RESUMEN

The rat kidney ablation and infarction (A/I) model of subtotal or 5/6th nephrectomy is the most commonly studied model of nondiabetic chronic kidney disease (CKD). The A/I kidney at 1 wk exhibits reductions in kidney function, as determined by glomerular filtration rate, and diminished metabolic efficiency as determined by oxygen consumption per sodium transport (QO2/TNa). As renoprotective AMPK activity is affected by metabolic changes and cellular stress, we evaluated AMPK activity in this model system. We show that these early pathophysiological changes are accompanied by a paradoxical decrease in AMPK activity. Over time, these kidney parameters progressively worsen with extensive kidney structural, functional, metabolic, and fibrotic changes observed at 4 wk after A/I. We show that induction of AMPK activity with either metformin or 5-aminoimidazole-4-carboxamide ribonucleotide increases AMPK activity in this model and also corrects kidney metabolic inefficiency, improves kidney function, and ameliorates kidney fibrosis and structural alterations. We conclude that AMPK activity is reduced in the subtotal nephrectomy model of nondiabetic CKD, that altered regulation of AMPK is coincident with the progression of disease parameters, and that restoration of AMPK activity can suppress the progressive loss of function characteristic of this model. We propose that induction of AMPK activity may prove an effective therapeutic target for the treatment of nondiabetic CKD.


Asunto(s)
Adenilato Quinasa/biosíntesis , Insuficiencia Renal Crónica/fisiopatología , Animales , Modelos Animales de Enfermedad , Inducción Enzimática , Masculino , Metformina/farmacología , Nefrectomía , Ratas , Ratas Wistar
7.
Drug Discov Today ; 18(17-18): 880-93, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23769988

RESUMEN

Agmatine (decarboxylated arginine) has been known as a natural product for over 100 years, but its biosynthesis in humans was left unexplored owing to long-standing controversy. Only recently has the demonstration of agmatine biosynthesis in mammals revived research, indicating its exceptional modulatory action at multiple molecular targets, including neurotransmitter systems, nitric oxide (NO) synthesis and polyamine metabolism, thus providing bases for broad therapeutic applications. This timely review, a concerted effort by 16 independent research groups, draws attention to the substantial preclinical and initial clinical evidence, and highlights challenges and opportunities, for the use of agmatine in treating a spectrum of complex diseases with unmet therapeutic needs, including diabetes mellitus, neurotrauma and neurodegenerative diseases, opioid addiction, mood disorders, cognitive disorders and cancer.


Asunto(s)
Agmatina/uso terapéutico , Descubrimiento de Drogas , Agmatina/química , Agmatina/historia , Agmatina/metabolismo , Agmatina/farmacología , Animales , Descubrimiento de Drogas/historia , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Transducción de Señal/efectos de los fármacos , Distribución Tisular
8.
Am J Physiol Renal Physiol ; 304(2): F156-67, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23152292

RESUMEN

The Na-glucose cotransporter SGLT2 mediates high-capacity glucose uptake in the early proximal tubule and SGLT2 inhibitors are developed as new antidiabetic drugs. We used gene-targeted Sglt2 knockout (Sglt2(-/-)) mice to elucidate the contribution of SGLT2 to blood glucose control, glomerular hyperfiltration, kidney growth, and markers of renal growth and injury at 5 wk and 4.5 mo after induction of low-dose streptozotocin (STZ) diabetes. The absence of SGLT2 did not affect renal mRNA expression of glucose transporters SGLT1, NaGLT1, GLUT1, or GLUT2 in response to STZ. Application of STZ increased blood glucose levels to a lesser extent in Sglt2(-/-) vs. wild-type (WT) mice (∼300 vs. 470 mg/dl) but increased glucosuria and food and fluid intake to similar levels in both genotypes. Lack of SGLT2 prevented STZ-induced glomerular hyperfiltration but not the increase in kidney weight. Knockout of SGLT2 attenuated the STZ-induced renal accumulation of p62/sequestosome, an indicator of impaired autophagy, but did not attenuate the rise in renal expression of markers of kidney growth (p27 and proliferating cell nuclear antigen), oxidative stress (NADPH oxidases 2 and 4 and heme oxygenase-1), inflammation (interleukin-6 and monocyte chemoattractant protein-1), fibrosis (fibronectin and Sirius red-sensitive tubulointerstitial collagen accumulation), or injury (renal/urinary neutrophil gelatinase-associated lipocalin). SGLT2 deficiency did not induce ascending urinary tract infection in nondiabetic or diabetic mice. The results indicate that SGLT2 is a determinant of hyperglycemia and glomerular hyperfiltration in STZ-induced diabetes mellitus but is not critical for the induction of renal growth and markers of renal injury, inflammation, and fibrosis.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Nefropatías Diabéticas/metabolismo , Hiperglucemia/metabolismo , Riñón/crecimiento & desarrollo , Transportador 2 de Sodio-Glucosa/metabolismo , Animales , Glucemia , Diabetes Mellitus Experimental/sangre , Nefropatías Diabéticas/genética , Tasa de Filtración Glomerular , Hiperglucemia/sangre , Riñón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Tamaño de los Órganos , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transportador 2 de Sodio-Glucosa/genética
9.
Amino Acids ; 42(2-3): 775-81, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21805293

RESUMEN

Agmatine is a novel neuromodulator that plays a protective role in the CNS in several models of cellular damage. However, the mechanisms involved in these protective effects in neurodegenerative diseases are poorly understood. Fourier transform infrared (FTIR) spectroscopy analysis detects biomolecular changes in disordered cells and tissues. In this report, we utilize FTIR spectroscopy to characterize the changes in rotenone-induced damage in neuronal-like differentiated SH-SY5Y neuroblastoma cells in the presence or absence of agmatine. The analysis of the FTIR spectra demonstrates significant alterations in rotenone-treated cells, whereas the FTIR spectra obtained after pre-incubation with agmatine (250 nM) significantly reduces these redox alterations and more closely resembles those of the control cells. In particular, rotenone-damaged cells demonstrate spectral alterations related to amide I, which correspond to an increase in ß-sheet components, and decreases in the amide II absorption intensity, suggesting a loss of N-H bending and C-N stretching. These alterations were also evident by Fourier self-deconvolution analysis. Thus, rotenone-induced increases in the levels of stretching vibration band related to the protein carboxyl group would account for a significant amount of misfolded proteins in the cell. Agmatine effectively reduces these effects of rotenone on protein structure. In conclusion, antioxidant and scavenging properties of agmatine reduce rotenone-produced cellular damage at the level of protein structure. These, together with other previous observations, demonstrate the therapeutic potential of agmatine in the treatment of Parkinson's disease.


Asunto(s)
Agmatina/farmacología , Modelos Biológicos , Neuroblastoma/patología , Enfermedad de Parkinson/patología , Rotenona/toxicidad , Línea Celular Tumoral , Humanos , Espectroscopía Infrarroja por Transformada de Fourier
10.
J Neurochem ; 116(1): 67-75, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21044082

RESUMEN

Agmatine, an endogenous arginine metabolite, has been proposed as a novel neuromodulator that plays protective roles in the CNS in several models of cellular damage. However, the mechanisms involved in these protective effects in neurodegenerative diseases are poorly understood. The present study was undertaken to investigate the effects of agmatine on cell injury induced by rotenone, commonly used in establishing in vivo and in vitro models of Parkinson's disease, in human-derived dopaminergic neuroblastoma cell line (SH-SY5Y). We report that agmatine dose-dependently suppressed rotenone-induced cellular injury through a reduction of oxidative stress. Similar effects were obtained by spermine, suggesting a scavenging effect for these compounds. However, unlike spermine, agmatine also prevented rotenone-induced nuclear factor-κB nuclear translocation and mitochondrial membrane potential dissipation. Furthermore, rotenone-induced increase in apoptotic markers, such as caspase 3 activity, Bax expression and cytochrome c release, was significantly attenuated with agmatine treatment. These findings demonstrate mitochondrial preservation with agmatine in a rotenone model of apoptotic cell death, and that the neuroprotective action of agmatine appears because of suppressing apoptotic signalling mechanisms. Thus, agmatine may have therapeutic potential in the treatment of Parkinson's disease by protecting dopaminergic neurons.


Asunto(s)
Agmatina/farmacología , Potencial de la Membrana Mitocondrial/fisiología , FN-kappa B/metabolismo , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Rotenona/toxicidad , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Línea Celular Tumoral , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Neuronas/efectos de los fármacos
11.
Am J Physiol Renal Physiol ; 299(6): F1365-73, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20881034

RESUMEN

The 5/6(th) nephrectomy or ablation/infarction (A/I) preparation has been used as a classic model of chronic kidney disease (CKD). We observed increased kidney oxygen consumption (Q(O2)) and altered renal hemodynamics in the A/I kidney that were normalized after combined angiotensin II (ANG II) blockade. Studies suggest hypoxia inducible factor as a protective influence in A/I. We induced hypoxia-inducible factor (HIF) and HIF target proteins by two different methods, cobalt chloride (CoCl(2)) and dimethyloxalyglycine (DMOG), for the first week after creation of A/I and compared the metabolic and renal hemodynamic outcomes to combined ANG II blockade. We also examined the HIF target proteins expressed by using Western blots and real-time PCR. Treatment with DMOG, CoCl(2), and ANG II blockade normalized kidney oxygen consumption factored by Na reabsorption and increased both renal blood flow and glomerular filtration rate. At 1 wk, CoCl(2) and DMOG increased kidney expression of HIF by Western blot. In the untreated A/I kidney, VEGF, heme oxygenase-1, and GLUT1 were all modestly increased. Both ANG II blockade and CoCl(2) therapy increased VEGF and GLUT1 but the cobalt markedly so. ANG II blockade decreased heme oxygenase-1 expression while CoCl(2) increased it. By real-time PCR, erythropoietin and GLUT1 were only increased by CoCl(2) therapy. Cell proliferation was modestly increased by ANG II blockade but markedly after cobalt therapy. Metabolic and hemodynamic abnormalities were corrected equally by ANG II blockade and HIF therapies. However, the molecular patterns differed significantly between ANG II blockade and cobalt therapy. HIF induction may prove to be protective in this model of CKD.


Asunto(s)
Angiotensina II/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Fallo Renal Crónico/metabolismo , Riñón/efectos de los fármacos , Aminoácidos Dicarboxílicos/farmacología , Animales , Anhidrasa Carbónica IX , Anhidrasas Carbónicas/metabolismo , Cobalto/farmacología , Inducción Enzimática , Transportador de Glucosa de Tipo 1/biosíntesis , Masculino , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ratas , Ratas Wistar
12.
Am J Physiol Cell Physiol ; 299(2): C374-80, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20505038

RESUMEN

Diabetic nephropathy is the commonest cause of end-stage renal disease. Inordinate kidney growth and glomerular hyperfiltration at the very early stages of diabetes are putative antecedents to this disease. The kidney is the only organ that grows larger with the onset of diabetes mellitus, yet there remains confusion about the mechanism and significance of this growth. Here we show that kidney proximal tubule cells in culture transition to senescence in response to oxidative stress. We further determine the temporal expression of G(1) phase cell cycle components in rat kidney cortex at days 4 and 10 of streptozotocin diabetes to evaluate changes in this growth response. In diabetic rats we observe increases in kidney weight-to-body weight ratios correlating with increases in expression of the growth-related proteins in the kidney at day 4 after induction of diabetes. However, at day 10 we find a decrease in this profile in diabetic animals coincident with increased cyclin-dependent kinase inhibitor expressions. We observe no change in caspase-3 expression in the diabetic kidneys at these early time points; however, diabetic animals demonstrate reduced kidney connexin 43 and increased plasminogen activator inhibitor-1 expressions and increased senescence-associated beta-galactosidase activity in cortical tubules. In summary, diabetic kidneys exhibit an early temporal induction of growth phase components followed by their suppression concurrent with the induction of cyclin-dependent kinase inhibitors and markers of senescence. These data delineate a phenotypic change in cortical tubules early in the pathogenesis of diabetes that may contribute to further downstream complications of the disease.


Asunto(s)
Senescencia Celular/fisiología , Diabetes Mellitus Experimental/patología , Túbulos Renales Proximales/patología , Fenotipo , Animales , Transporte Biológico/fisiología , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/fisiopatología , Túbulos Renales Proximales/enzimología , Túbulos Renales Proximales/metabolismo , Masculino , Zarigüeyas , Estrés Oxidativo/fisiología , Ratas , Ratas Sprague-Dawley , Cloruro de Sodio Dietético/metabolismo , Factores de Tiempo , beta-Galactosidasa/metabolismo
13.
Nephron Physiol ; 111(3): p30-8, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19276628

RESUMEN

BACKGROUND: In early type 1 diabetes mellitus, changes in proximal reabsorption influence glomerular filtration rate (GFR) through tubuloglomerular feedback (TGF). Due to TGF, a primary increase in proximal reabsorption causes early diabetic hyperfiltration, while a heightened sensitivity of the proximal tubule to dietary salt leads to the so-called salt paradox, where a change in dietary salt causes a reciprocal change in GFR ('tubulocentric principle'). Here, experiments were performed in adenosine A(1) receptor knockout mice (A(1)R-/-), which lack an immediate TGF response, to determine whether A(1)Rs are essential for early diabetic hyperfiltration and the salt paradox. METHODS: GFR was measured by inulin disappearance in conscious A(1)R-/- and wild-type (WT) mice after 4 weeks of streptozotocin diabetes on a control NaCl diet (1%), and measurements were repeated after 6 days of equilibration on a low-NaCl (0.1%) or a high-NaCl (4%) diet. RESULTS: A(1)R-/- and WT were similar with respect to blood glucose, dietary intakes and body weight changes on a given diet. Diabetic hyperfiltration occurred in WT, but was blunted in A(1)R-/-. A reciprocal relationship between GFR and dietary salt was found in WT diabetics, but not A(1)R-/- diabetics or nondiabetics of either strain. CONCLUSION: A(1)Rs determine glomerular hyperfiltration and the salt paradox in early diabetes, which is consistent with the tubulocentric principle.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Nefropatías Diabéticas/metabolismo , Tasa de Filtración Glomerular , Túbulos Renales Proximales/metabolismo , Receptor de Adenosina A1/metabolismo , Cloruro de Sodio Dietético/metabolismo , Aldosterona/sangre , Animales , Glucemia/metabolismo , Peso Corporal , Diabetes Mellitus Experimental/fisiopatología , Nefropatías Diabéticas/fisiopatología , Dieta Hiposódica , Ingestión de Líquidos , Ingestión de Alimentos , Retroalimentación Fisiológica , Túbulos Renales Proximales/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Adenosina A1/deficiencia , Receptor de Adenosina A1/genética , Factores de Tiempo
14.
Am J Physiol Cell Physiol ; 296(6): C1411-9, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19321739

RESUMEN

Agmatine, an endogenous metabolite of arginine, selectively suppresses growth in cells with high proliferative kinetics, such as transformed cells, through depletion of intracellular polyamine levels. In the present study, we depleted intracellular polyamine content with agmatine to determine if attrition by cell death contributes to the growth-suppressive effects. We did not observe an increase in necrosis, DNA fragmentation, or chromatin condensation in Ha-Ras-transformed NIH-3T3 cells administered agmatine. In response to Ca(2+)-induced oxidative stress in kidney mitochondrial preparations, agmatine demonstrated attributes of a free radical scavenger by protecting against the oxidation of sulfhydryl groups and decreasing hydrogen peroxide content. The functional outcome was a protective effect against Ca(2+)-induced mitochondrial swelling and mitochondrial membrane potential collapse. We also observed decreased expression of proapoptotic Bcl-2 family members and of execution caspase-3, implying antiapoptotic potential. Indeed, we found that apoptosis induced by camptothecin or 5-fluorourocil was attenuated in cells administered agmatine. Agmatine may offer an alternative to the ornithine decarboxylase inhibitor difluoromethyl ornithine for depletion of intracellular polyamine content while avoiding the complications of increasing polyamine import and reducing the intracellular free radical scavenger capacity of polyamines. Depletion of intracellular polyamine content with agmatine suppressed cell growth, yet its antioxidant capacity afforded protection from mitochondrial insult and resistance to cellular apoptosis. These results could explain the beneficial outcomes observed with agmatine in models of injury and disease.


Asunto(s)
Agmatina/metabolismo , Antioxidantes/metabolismo , Apoptosis , Arginina/metabolismo , Riñón/metabolismo , Mitocondrias/metabolismo , Animales , Apoptosis/efectos de los fármacos , Calcio/metabolismo , Camptotecina/toxicidad , Caspasa 3/metabolismo , Proliferación Celular , Citoprotección , Fragmentación del ADN , Fluorouracilo/toxicidad , Riñón/efectos de los fármacos , Riñón/patología , Potencial de la Membrana Mitocondrial , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Dilatación Mitocondrial , Células 3T3 NIH , Poliaminas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas
15.
Am J Physiol Renal Physiol ; 294(3): F638-44, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18199602

RESUMEN

Activation of adenosine A(1) receptors (A(1)R) can inhibit arginine vasopressin (AVP)-induced cAMP formation in isolated cortical and medullary collecting ducts. To assess the in vivo consequences of the absence of A(1)R, we performed experiments in mice lacking A(1)R (A(1)R(-/-)). We assessed the effects of the vasopressin V(2) receptor (V(2)R) agonist 1-desamino-8-d-arginine vasopressin (dDAVP) on cAMP formation in isolated inner medullary collecting ducts (IMCD) and on water excretion in conscious water-loaded mice. dDAVP-induced cAMP formation in isolated IMCD was significantly greater ( approximately 2-fold) in A(1)R(-/-) compared with wild-type mice (WT) and, in contrast to WT, was not inhibited by the A(1)R agonist N6-cyclohexyladenosine. A(1)R(-/-) and WT had similar basal urinary excretion of vasopressin, expression of aquaporin-2 protein in renal cortex and medulla, and acute increases in urinary flow rate and electrolyte-free water clearance in response to the V(2)R antagonist SR121463 or acute water loading; the latter increased inner medullary A(1)R expression in WT. Dose dependence of dDAVP-induced antidiuresis after acute water loading was not different between the genotypes. However, A(1)R(-/-) had greater inner medullary expression of cyclooxygenase-1 under basal conditions and of the P2Y(2) and EP(3) receptor in response to water loading compared with WT mice. Thus vasopressin-induced cAMP formation is enhanced in isolated IMCD of mice lacking A(1)R, but the adenosine-A(1)R/V(2)R interaction demonstrated in vitro is likely compensated in vivo by multiple mechanisms, a number of which can be "uncovered" by water loading.


Asunto(s)
Arginina Vasopresina/metabolismo , Túbulos Renales Colectores/metabolismo , Receptor de Adenosina A1/metabolismo , Agua/metabolismo , Animales , AMP Cíclico/metabolismo , Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2/metabolismo , Desamino Arginina Vasopresina/farmacología , Endotelina-1/metabolismo , Técnicas In Vitro , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo III/metabolismo , Reacción en Cadena de la Polimerasa , Receptor de Adenosina A1/genética , Receptores de Prostaglandina E/metabolismo , Subtipo EP3 de Receptores de Prostaglandina E , Receptores Purinérgicos P2/metabolismo , Receptores Purinérgicos P2Y2 , Receptores de Vasopresinas/agonistas
16.
Am J Physiol Renal Physiol ; 294(4): F795-800, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18199604

RESUMEN

Agmatine, decarboxylated arginine, is produced in the kidney and can increase nephron and kidney filtration rate via renal vasodilatation and increases in plasma flow. This increase in filtration rate after agmatine is prevented by administration of nitric oxide synthase (NOS) inhibitors. In endothelial cells, agmatine-stimulated nitrite production is accompanied by induction of cytosolic calcium. NOS activity requires calcium for activation; however, the source of this calcium remains unknown. Ryanodine receptor (RyR) calcium-activated calcium release channels are present in the kidney cortex, and we evaluated if RyR contributes to the agmatine response. Agmatine microperfused into Bowman's space reversibly increases nephron filtration rate (SNGFR) by approximately 30%. cADP-ribose (cADPR) regulates RyR channel activity. Concurrent infusion of agmatine with the cADPR blocker 8-bromo-cADPR (2 microM) prevents the increase in filtration rate. Furthermore, direct activation of the RyR channel with ryanodine at agonist concentrations (5 microM) increases SNGFR, and, like agmatine, this increase is prevented by administration of N(G)-monomethyl-l-arginine, a nonselective NOS blocker. We demonstrate that agmatine does not elicit ADPR cyclase activity in vascular smooth muscle membranes and does not directly affect RyR calcium channel responses using sea urchin egg homogenates. These results imply interplay between endothelial cell cADPR/RyR/Ca(2+)/NO and the cADPR/RyR/Ca(2+) pathways in vascular smooth muscle cells in arterioles in the regulation of kidney filtration rate. In conclusion, we show that agmatine-induced effects require activation of cADPR and RyR calcium release channels for NO generation, vasodilation, and increased filtration rate.


Asunto(s)
Agmatina/farmacología , Tasa de Filtración Glomerular/efectos de los fármacos , Óxido Nítrico/biosíntesis , Canal Liberador de Calcio Receptor de Rianodina/fisiología , Animales , Glomérulos Renales/efectos de los fármacos , Glomérulos Renales/fisiología , Masculino , Nefronas/efectos de los fármacos , Nefronas/fisiología , Óxido Nítrico/fisiología , Óxido Nítrico Sintasa/metabolismo , Ratas , Ratas Wistar , Rianodina/farmacología
17.
Am J Physiol Cell Physiol ; 293(2): C705-11, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17475661

RESUMEN

Polyamines are small cationic molecules required for cellular proliferation. Agmatine is a biogenic amine unique in its capacity to arrest proliferation in cell lines by depleting intracellular polyamine levels. We previously demonstrated that agmatine enters mammalian cells via the polyamine transport system. As polyamine transport is positively correlated with the rate of cellular proliferation, the current study examines the antiproliferative effects of agmatine on cells with varying proliferative kinetics. Herein, we evaluate agmatine transport, intracellular accumulation, and its effects on antizyme expression and cellular proliferation in nontransformed cell lines and their transformed variants. H-ras- and Src-transformed murine NIH/3T3 cells (Ras/3T3 and Src/3T3, respectively) that were exposed to exogenous agmatine exhibit increased uptake and intracellular accumulation relative to the parental NIH/3T3 cell line. Similar increases were obtained for human primary foreskin fibroblasts relative to a human fibrosarcoma cell line, HT1080. Agmatine increases expression of antizyme, a protein that inhibits polyamine biosynthesis and transport. Ras/3T3 and Src/3T3 cells demonstrated augmented increases in antizyme protein expression relative to NIH/3T3 in response to agmatine. All transformed cell lines were significantly more sensitive to the antiproliferative effects of agmatine than nontransformed lines. These effects were attenuated in the presence of exogenous polyamines or inhibitors of polyamine transport. In conclusion, the antiproliferative effects of agmatine preferentially target transformed cell lines due to the increased agmatine uptake exhibited by cells with short cycling times.


Asunto(s)
Agmatina/metabolismo , Proliferación Celular , Fibroblastos/metabolismo , Biosíntesis de Proteínas , Proteínas/metabolismo , Agmatina/farmacología , Animales , Unión Competitiva , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/metabolismo , Línea Celular Transformada , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Genes ras , Genes src , Humanos , Cinética , Ratones , Células 3T3 NIH , Ornitina Descarboxilasa/metabolismo , Biosíntesis de Proteínas/efectos de los fármacos , Putrescina/metabolismo
18.
Tohoku J Exp Med ; 210(2): 145-51, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17023768

RESUMEN

Polyamines play an essential role in the growth and differentiation of mammalian cells. The depletion of intracellular polyamines results in the suppression of growth. Proliferation of glomerular mesangial cells (MC) is the most common pathologic change in many forms of glomerulonephritis. Agmatine is a metabolite of arginine via arginine decarboxylase (ADC), highly expressed in the kidney, and unique in its capacity to suppress intracellular polyamine levels required for proliferation. As agmatine enters mammalian cells via the polyamine transport system, its antiproliferative effects may preferentially target cells with increased proliferative kinetics. In the present study, we evaluated the antiproliferative effects of agmatine on human MC in vitro. MC proliferation was stimulated with 20% fetal bovine serum (FBS) or platelet-derived growth factor (PDGF-BB, 20 ng/ml). Cell proliferation was measured using the (4.3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide) (MTT) proliferation assay. Intracellular polyamine levels were assayed by high performance liquid chromatography, and cell death was assessed by cellular DNA fragmentation enzyme-linked immunosorbent assay. The MTT proliferation assay showed that agmatine significantly suppressed proliferation of human MC treated with 20% FBS or 5% FBS + PDGF as compared to human MC treated with 5% FBS. Polyamine levels were markedly lower in cells treated with agmatine, and proliferation was rescued by administration of putrescine. The fragmented DNA was hardly detected in agmatine-treated human MC. In summary, human MC stimulated to increase their proliferative kinetics are significantly more sensitive to the antiproliferative effects of agmatine than normally cultured cells. Suppressed proliferation of the agmatine-treated human MC is not due to increased cell death. These results suggest that agmatine is a promising drug candidate for the treatment of human mesangial proliferative glomerulonephritis.


Asunto(s)
Agmatina/farmacología , Proliferación Celular/efectos de los fármacos , Células Mesangiales/efectos de los fármacos , Poliaminas/metabolismo , Línea Celular , Glomerulonefritis/tratamiento farmacológico , Humanos
19.
Am J Physiol Renal Physiol ; 291(5): F1078-82, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16705150

RESUMEN

The tubuloglomerular feedback (TGF) system serves to establish an appropriate balance between tubular reabsorption and glomerular filtration rate (GFR). High salt at the macula densa activates TGF to decrease GFR. Effector molecules for the TGF signal include ATP and adenosine. Over time, the GFR will adapt by increasing even if a high salt concentration persists. A potential modulator of this TGF adaptation is nitric oxide synthase-1-derived nitric oxide (NO). In isolated glomerular preparations, we developed a system for evaluating the effects of changing dietary salt on ecto-5'-nucleotidase (ecto-5'-NT) activity, the final enzyme in the conversion of ATP to adenosine. We found observable ecto-5'-NT activity in isolated glomeruli and that this activity can be regulated by dietary salt, with high salt increasing activity. Conversely, NO decreases ecto-5'-NT activity in glomerular preparations. Moreover, NO inhibition of ecto-5'-NT activity is suppressed in the presence of dithiothreitol, suggesting nitrosylation as a reversible, oxidative stress-sensitive mechanism. The salt-induced activation of ecto-5'-NT correlates with high salt resetting of TGF. NO inhibition of enzymatic activity could be part of the adaptive phase.


Asunto(s)
5'-Nucleotidasa/metabolismo , Glomérulos Renales/enzimología , Túbulos Renales/metabolismo , Óxido Nítrico/metabolismo , Cloruro de Sodio/metabolismo , Adaptación Fisiológica/fisiología , Adenosina/metabolismo , Animales , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Retroalimentación Fisiológica/fisiología , Tasa de Filtración Glomerular/fisiología , Masculino , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo I , Óxidos de Nitrógeno/farmacología , Ratas , Ratas Wistar , Cloruro de Sodio/farmacología , Espermina/análogos & derivados , Espermina/farmacología
20.
Nephron Physiol ; 102(2): p17-26, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16230862

RESUMEN

BACKGROUND/AIMS: Inducible nitric oxide (NO) synthase (iNOS) generated NO increases in the early phase of Thy-1 glomerulonephritis concurrently with mesangiolysis and reduction in glomerular filtration rate (GFR). Activation of ornithine decarboxylase (ODC), the rate-limiting enzyme of polyamine biosynthesis, is upregulated to allow mesangial cell proliferation which constitutes the repair phase in this model. Antiproliferative high-output NO generation inhibits proproliferative ODC activity, thereby temporally separating the early 'bactericidal' phase from the later 'growth' repair phase. METHODS: Renal function, ODC protein expression, arginine, ornithine, and polyamines by high-performance liquid chromatography, and histological changes were assessed in rats after induction of Thy-1 nephritis with and without NOS inhibition. RESULTS: Thy-1 significantly reduced the GFR relative to untreated controls. Treatment with a nonspecific NOS inhibitor, but not a selective iNOS inhibitor, further decreased the GFR at day 1. This implys a protective role for constitutive NOS in the early phase of this inflammatory model. Selective iNOS inhibition abrogated increased plasma NO(2)/NO(3) levels in Thy-1 glomerulonephritis, but did not significantly reduce mesangiolysis. However, inhibition of iNOS did result in significantly more nuclei/glomerulus during the proliferative phase, increasing the hypercellularity component of this disease model. This correlates with increased levels of polyamines, ornithine, and arginine beyond those observed with Thy-1 administration alone. CONCLUSIONS: These studies provide evidence that NO generation from different NOS isoforms can be protective in the temporal course of Thy-1 glomerulonephritis. The finding that iNOS attenuates hypercellularity in the repair phase of this inflammatory model adds cautionary insight in the therapeutic use of selective iNOS inhibition in vivo.


Asunto(s)
Glomerulonefritis/metabolismo , Riñón/metabolismo , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico/metabolismo , Antígenos Thy-1/metabolismo , Animales , Masculino , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...