Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Kidney Int ; 105(1): 132-149, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38069998

RESUMEN

Glucagon like peptide-1 (GLP-1) is a hormone produced and released by cells of the gastrointestinal tract following meal ingestion. GLP-1 receptor agonists (GLP-1RA) exhibit kidney-protective actions through poorly understood mechanisms. Here we interrogated whether the receptor for advanced glycation end products (RAGE) plays a role in mediating the actions of GLP-1 on inflammation and diabetic kidney disease. Mice with deletion of the GLP-1 receptor displayed an abnormal kidney phenotype that was accelerated by diabetes and improved with co-deletion of RAGE in vivo. Activation of the GLP-1 receptor pathway with liraglutide, an anti-diabetic treatment, downregulated kidney RAGE, reduced the expansion of bone marrow myeloid progenitors, promoted M2-like macrophage polarization and lessened markers of kidney damage in diabetic mice. Single cell transcriptomics revealed that liraglutide induced distinct transcriptional changes in kidney endothelial, proximal tubular, podocyte and macrophage cells, which were dominated by pathways involved in nutrient transport and utilization, redox sensing and the resolution of inflammation. The kidney-protective action of liraglutide was corroborated in a non-diabetic model of chronic kidney disease, the subtotal nephrectomised rat. Thus, our findings identify a novel glucose-independent kidney-protective action of GLP-1-based therapies in diabetic kidney disease and provide a valuable resource for exploring the cell-specific kidney transcriptional response ensuing from pharmacological GLP-1R agonism.


Asunto(s)
Diabetes Mellitus Experimental , Nefropatías Diabéticas , Ratas , Ratones , Animales , Receptor para Productos Finales de Glicación Avanzada/genética , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Nefropatías Diabéticas/etiología , Nefropatías Diabéticas/genética , Liraglutida/farmacología , Liraglutida/uso terapéutico , Receptor del Péptido 1 Similar al Glucagón/genética , Diabetes Mellitus Experimental/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Péptido 1 Similar al Glucagón/farmacología , Péptido 1 Similar al Glucagón/uso terapéutico , Inflamación
2.
Clin Sci (Lond) ; 130(9): 711-20, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26831938

RESUMEN

Oxidative phosphorylation (OXPHOS) drives ATP production by mitochondria, which are dynamic organelles, constantly fusing and dividing to maintain kidney homoeostasis. In diabetic kidney disease (DKD), mitochondria appear dysfunctional, but the temporal development of diabetes-induced adaptations in mitochondrial structure and bioenergetics have not been previously documented. In the present study, we map the changes in mitochondrial dynamics and function in rat kidney mitochondria at 4, 8, 16 and 32 weeks of diabetes. Our data reveal that changes in mitochondrial bioenergetics and dynamics precede the development of albuminuria and renal histological changes. Specifically, in early diabetes (4 weeks), a decrease in ATP content and mitochondrial fragmentation within proximal tubule epithelial cells (PTECs) of diabetic kidneys were clearly apparent, but no changes in urinary albumin excretion or glomerular morphology were evident at this time. By 8 weeks of diabetes, there was increased capacity for mitochondrial permeability transition (mPT) by pore opening, which persisted over time and correlated with mitochondrial hydrogen peroxide (H2O2) generation and glomerular damage. Late in diabetes, by week 16, tubular damage was evident with increased urinary kidney injury molecule-1 (KIM-1) excretion, where an increase in the Complex I-linked oxygen consumption rate (OCR), in the context of a decrease in kidney ATP, indicated mitochondrial uncoupling. Taken together, these data show that changes in mitochondrial bioenergetics and dynamics may precede the development of the renal lesion in diabetes, and this supports the hypothesis that mitochondrial dysfunction is a primary cause of DKD.


Asunto(s)
Adaptación Fisiológica , Diabetes Mellitus Experimental/patología , Riñón/patología , Mitocondrias/metabolismo , Albuminuria , Animales , ADN Mitocondrial/genética , Diabetes Mellitus Experimental/genética , Metabolismo Energético , Riñón/metabolismo , Túbulos Renales/patología , Masculino , Dinámicas Mitocondriales , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Estrés Oxidativo , Fenotipo , Ratas Sprague-Dawley , Factores de Tiempo , Regulación hacia Arriba
3.
Kidney Int ; 80(2): 190-8, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21412218

RESUMEN

Obesity is highly prevalent in Western populations and is considered a risk factor for the development of renal impairment. Interventions that reduce the tissue burden of advanced glycation end-products (AGEs) have shown promise in stemming the progression of chronic disease. Here we tested if treatments that lower tissue AGE burden in patients and mice would improve obesity-related renal dysfunction. Overweight and obese individuals (body mass index (BMI) 26-39 kg/m(2)) were recruited to a randomized, crossover clinical trial involving 2 weeks each on a low- and a high-AGE-containing diet. Renal function and an inflammatory profile (monocyte chemoattractant protein-1 (MCP-1) and macrophage migration inhibitory factor (MIF)) were improved following the low-AGE diet. Mechanisms of advanced glycation-related renal damage were investigated in a mouse model of obesity using the AGE-lowering pharmaceutical, alagebrium, and mice in which the receptor for AGE (RAGE) was deleted. Obesity, resulting from a diet high in both fat and AGE, caused renal impairment; however, treatment of the RAGE knockout mice with alagebrium improved urinary albumin excretion, creatinine clearance, the inflammatory profile, and renal oxidative stress. Alagebrium treatment, however, resulted in decreased weight gain and improved glycemic control compared with wild-type mice on a high-fat Western diet. Thus, targeted reduction of the advanced glycation pathway improved renal function in obesity.


Asunto(s)
Productos Finales de Glicación Avanzada/efectos adversos , Enfermedades Renales/dietoterapia , Riñón/fisiopatología , Obesidad/dietoterapia , Obesidad/fisiopatología , Receptores Inmunológicos/efectos de los fármacos , Adolescente , Adulto , Animales , Estudios Cruzados , Dieta , Productos Finales de Glicación Avanzada/administración & dosificación , Humanos , Inflamación/prevención & control , Riñón/efectos de los fármacos , Enfermedades Renales/inducido químicamente , Enfermedades Renales/tratamiento farmacológico , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Obesidad/tratamiento farmacológico , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/deficiencia , Tiazoles/farmacología , Tiazoles/uso terapéutico , Adulto Joven
4.
Am J Nephrol ; 34(4): 347-55, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21876347

RESUMEN

BACKGROUND/AIMS: The formation of advanced glycation end products (AGEs) is accelerated in patients with diabetic nephropathy. The aim of this study was to ascertain if the urinary excretion of proteins modified by advanced glycation can be used as biomarkers for albuminuria in individuals with type 1 or type 2 diabetes. METHODS: Community-based patients with type 1 (n = 68) or type 2 diabetes (n = 216) attending a diabetes clinic of a tertiary referral hospital were classified as having normoalbuminuria (Normo, albumin excretion rate (AER) <20 µg/min), microalbuminuria (Micro, AER 20-200 µg/min) or macroalbuminuria (Macro, AER ≥200 µg/min). Serum and urine AGE-modified proteins were measured. RESULTS: In patients with both type 1 diabetes and type 2 diabetes, there was a clear association between the degree of albuminuria and urinary AGE-modified proteins (p < 0.0001). Exclusive to patients with type 1 diabetes, urinary excretion of the AGE carboxymethyllysine correlated with AER, whereas patients with type 2 diabetes and macroalbuminuria had an increase in urinary methylglyoxal, an AGE intermediate. These changes were independent of isotopic glomerular filtration rate levels. Serum concentrations of AGEs or soluble receptor for AGEs were not consistently associated with albuminuria in either type 1 or type 2 diabetes. CONCLUSIONS: Urinary excretion of proteins modified by AGEs may be useful biomarkers of albuminuria in individuals with type 1 and type 2 diabetes, warranting prospective investigation in larger diabetic cohorts.


Asunto(s)
Albuminuria/orina , Biomarcadores/metabolismo , Nefropatías Diabéticas/orina , Productos Finales de Glicación Avanzada/orina , Adulto , Albuminuria/metabolismo , Índice de Masa Corporal , Estudios de Cohortes , Diabetes Mellitus Tipo 1/orina , Diabetes Mellitus Tipo 2/orina , Ensayo de Inmunoadsorción Enzimática/métodos , Femenino , Tasa de Filtración Glomerular , Humanos , Masculino , Persona de Mediana Edad , Estrés Oxidativo
5.
Endocrinol Diabetes Metab ; 4(3): e00278, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34277994

RESUMEN

AIMS: The accumulation of advanced glycation end products is implicated in the development and progression of diabetic kidney disease. No study has examined whether stimulating advanced glycation clearance via receptor manipulation is reno-protective in diabetes. Podocytes, which are early contributors to diabetic kidney disease and could be a target for reno-protection. MATERIALS AND METHODS: To examine the effects of increased podocyte oligosaccharyltransferase-48 on kidney function, glomerular sclerosis, tubulointerstitial fibrosis and proteome (PXD011434), we generated a mouse with increased oligosaccharyltransferase-48kDa subunit abundance in podocytes driven by the podocin promoter. RESULTS: Despite increased urinary clearance of advanced glycation end products, we observed a decline in renal function, significant glomerular damage including glomerulosclerosis, collagen IV deposition, glomerular basement membrane thickening and foot process effacement and tubulointerstitial fibrosis. Analysis of isolated glomeruli identified enrichment in proteins associated with collagen deposition, endoplasmic reticulum stress and oxidative stress. Ultra-resolution microscopy of podocytes revealed denudation of foot processes where there was co-localization of oligosaccharyltransferase-48kDa subunit and advanced glycation end-products. CONCLUSIONS: These studies indicate that increased podocyte expression of oligosaccharyltransferase-48 kDa subunit results in glomerular endoplasmic reticulum stress and a decline in kidney function.


Asunto(s)
Nefropatías Diabéticas , Podocitos , Animales , Nefropatías Diabéticas/etiología , Nefropatías Diabéticas/metabolismo , Membrana Basal Glomerular/metabolismo , Productos Finales de Glicación Avanzada/metabolismo , Ratones , Podocitos/metabolismo , Receptor para Productos Finales de Glicación Avanzada/metabolismo
6.
Sci Adv ; 7(14)2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33789895

RESUMEN

Intake of processed foods has increased markedly over the past decades, coinciding with increased microvascular diseases such as chronic kidney disease (CKD) and diabetes. Here, we show in rodent models that long-term consumption of a processed diet drives intestinal barrier permeability and an increased risk of CKD. Inhibition of the advanced glycation pathway, which generates Maillard reaction products within foods upon thermal processing, reversed kidney injury. Consequently, a processed diet leads to innate immune complement activation and local kidney inflammation and injury via the potent proinflammatory effector molecule complement 5a (C5a). In a mouse model of diabetes, a high resistant starch fiber diet maintained gut barrier integrity and decreased severity of kidney injury via suppression of complement. These results demonstrate mechanisms by which processed foods cause inflammation that leads to chronic disease.


Asunto(s)
Inflamación , Insuficiencia Renal Crónica , Animales , Dieta , Femenino , Alimentos , Humanos , Inflamación/etiología , Masculino , Ratones , Permeabilidad
7.
Circulation ; 119(15): 2103-11, 2009 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-19349317

RESUMEN

BACKGROUND: Low plasma high-density lipoprotein (HDL) is associated with elevated cardiovascular risk and aspects of the metabolic syndrome. We hypothesized that HDL modulates glucose metabolism via elevation of plasma insulin and through activation of the key metabolic regulatory enzyme, AMP-activated protein kinase, in skeletal muscle. METHODS AND RESULTS: Thirteen patients with type 2 diabetes mellitus received both intravenous reconstituted HDL (rHDL: 80 mg/kg over 4 hours) and placebo on separate days in a double-blind, placebo-controlled crossover study. A greater fall in plasma glucose from baseline occurred during rHDL than during placebo (at 4 hours rHDL=-2.6+/-0.4; placebo=-2.1+/-0.3 mmol/L; P=0.018). rHDL increased plasma insulin (at 4 hours rHDL=3.4+/-10.0; placebo= -19.2+/-7.4 pmol/L; P=0.034) and also the homeostasis model assessment beta-cell function index (at 4 hours rHDL=18.9+/-5.9; placebo=8.6+/-4.4%; P=0.025). Acetyl-CoA carboxylase beta phosphorylation in skeletal muscle biopsies was increased by 1.7+/-0.3-fold after rHDL, indicating activation of the AMP-activated protein kinase pathway. Both HDL and apolipoprotein AI increased glucose uptake (by 177+/-12% and 144+/-18%, respectively; P<0.05 for both) in primary human skeletal muscle cell cultures established from patients with type 2 diabetes mellitus (n=5). The mechanism is demonstrated to include stimulation of the ATP-binding cassette transporter A1 with subsequent activation of the calcium/calmodulin-dependent protein kinase kinase and the AMP-activated protein kinase pathway. CONCLUSIONS: rHDL reduced plasma glucose in patients with type 2 diabetes mellitus by increasing plasma insulin and activating AMP-activated protein kinase in skeletal muscle. These findings suggest a role for HDL-raising therapies beyond atherosclerosis to address type 2 diabetes mellitus.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Glucosa/metabolismo , Lipoproteínas HDL/uso terapéutico , Proteínas Quinasas Activadas por AMP/metabolismo , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/fisiología , Animales , Apolipoproteína A-I/farmacología , Apolipoproteína A-I/uso terapéutico , Señalización del Calcio/efectos de los fármacos , Proteínas Quinasas Dependientes de Calcio-Calmodulina/fisiología , Células Cultivadas/efectos de los fármacos , Células Cultivadas/metabolismo , Estudios Cruzados , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/metabolismo , Método Doble Ciego , Ácidos Grasos/metabolismo , Femenino , Humanos , Infusiones Intravenosas , Insulina/sangre , Insulina/metabolismo , Insulina/farmacología , Secreción de Insulina , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Lipoproteínas HDL/administración & dosificación , Lipoproteínas HDL/farmacología , Lipoproteínas LDL/farmacología , Masculino , Ratones , Persona de Mediana Edad , Células Musculares/efectos de los fármacos , Células Musculares/metabolismo , Músculo Esquelético/metabolismo , Fenformina/farmacología , Transducción de Señal/efectos de los fármacos
8.
Kidney Int ; 78(3): 287-95, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20463655

RESUMEN

The accumulation of advanced glycation end products is thought to be a key factor in the initiation and progression of diabetic nephropathy. Here we determined whether the size of the ligands for the receptor for advanced glycation end products (RAGEs) that were present in the serum of patients with type 2 diabetes modulates their pathogenic potential. Serum was collected from control subjects and patients with type 2 diabetes with varying degrees of renal disease (normo-, micro-, or macroalbuminuria). The titers of the RAGE ligands N-carboxymethyllysine (CML), S100A, S100B, and high-mobility group box 1 (HMGB1) were measured by enzyme-linked immunosorbent assay in serum as well as in pooled size-fractionated serum. We also measured cellular binding of serum fractions to mesangial cells transfected with RAGE and examined the downstream signaling pathways. Circulating CML was increased in patients with type 2 diabetes, whereas HMGB1 was decreased. S100A8, S100BA9, and soluble RAGE were unchanged. The high-molecular-weight (over 50 kDa) serum fraction contained the greatest proportion of RAGE ligands, with all immunoreactivity and cellular binding observed only with serum fractions over 30 kDa. High-molecular-weight serum from macroalbuminuric patients showed greater RAGE binding capacity, modulation of cell-surface RAGE expression, increased phospho-protein kinase C-alpha, and p65 nuclear factor kappaB DNA-binding activity, which were competitively inhibited by soluble RAGE or CML neutralizing antibodies. These data show that ligands that activate RAGE present in the circulation of patients with type 2 diabetes and nephropathy are predominantly of high molecular weight.


Asunto(s)
Nefropatías Diabéticas/metabolismo , Receptores Inmunológicos/metabolismo , Anciano , Anticuerpos Neutralizantes/metabolismo , Estudios de Casos y Controles , Diabetes Mellitus Tipo 2/sangre , Nefropatías Diabéticas/sangre , Ensayo de Inmunoadsorción Enzimática , Femenino , Proteína HMGB1/análisis , Proteína HMGB1/metabolismo , Humanos , Ligandos , Lisina/análogos & derivados , Lisina/análisis , Lisina/metabolismo , Masculino , Persona de Mediana Edad , Peso Molecular , Proteína Quinasa C-alfa/metabolismo , Receptor para Productos Finales de Glicación Avanzada , Transcripción Genética
9.
J Am Soc Nephrol ; 20(4): 742-52, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19158353

RESUMEN

Damaged mitochondria generate an excess of superoxide, which may mediate tissue injury in diabetes. We hypothesized that in diabetic nephropathy, advanced glycation end-products (AGEs) lead to increases in cytosolic reactive oxygen species (ROS), which facilitate the production of mitochondrial superoxide. In normoglycemic conditions, exposure of primary renal cells to AGEs, transient overexpression of the receptor for AGEs (RAGE) with an adenoviral vector, and infusion of AGEs to healthy rodents each induced renal cytosolic oxidative stress, which led to mitochondrial permeability transition and deficiency of mitochondrial complex I. Because of a lack of glucose-derived NADH, which is the substrate for complex I, these changes did not lead to excess production of mitochondrial superoxide; however, when we performed these experiments in hyperglycemic conditions in vitro or in diabetic rats, we observed significant generation of mitochondrial superoxide at the level of complex I, fueled by a sustained supply of NADH. Pharmacologic inhibition of AGE-RAGE-induced mitochondrial permeability transition in vitro abrogated production of mitochondrial superoxide; we observed a similar effect in vivo after inhibiting cytosolic ROS production with apocynin or lowering AGEs with alagebrium. Furthermore, RAGE deficiency prevented diabetes-induced increases in renal mitochondrial superoxide and renal cortical apoptosis in mice. Taken together, these studies suggest that AGE-RAGE-induced cytosolic ROS production facilitates mitochondrial superoxide production in hyperglycemic environments, providing further evidence of a role for the advanced glycation pathway in the development and progression of diabetic nephropathy.


Asunto(s)
Diabetes Mellitus Experimental/fisiopatología , Productos Finales de Glicación Avanzada/fisiología , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores Inmunológicos/fisiología , Superóxidos/metabolismo , Animales , Transporte de Electrón , Femenino , Riñón/fisiopatología , Corteza Renal/fisiopatología , Fosforilación Oxidativa , Ratas , Ratas Sprague-Dawley , Receptor para Productos Finales de Glicación Avanzada
10.
Sci Rep ; 9(1): 13664, 2019 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-31541173

RESUMEN

The accumulation of advanced glycation end products (AGEs) have been implicated in the development and progression of diabetic kidney disease (DKD). There has been interest in investigating the potential of AGE clearance receptors, such as oligosaccharyltransferase-48 kDa subunit (OST48) to prevent the detrimental effects of excess AGE accumulation seen in the diabetic kidney. Here the objective of the study was to increase the expression of OST48 to examine if this slowed the development of DKD by facilitating the clearance of AGEs. Groups of 8-week-old heterozygous knock-in male mice (n = 9-12/group) over-expressing the gene encoding for OST48, dolichyl-diphosphooligosaccharide-protein glycosyltransferase (DDOST+/-) and litter mate controls were randomised to either (i) no diabetes or (ii) diabetes induced via multiple low-dose streptozotocin and followed for 24 weeks. By the study end, global over expression of OST48 increased glomerular OST48. This facilitated greater renal excretion of AGEs but did not affect circulating or renal AGE concentrations. Diabetes resulted in kidney damage including lower glomerular filtration rate, albuminuria, glomerulosclerosis and tubulointerstitial fibrosis. In diabetic mice, tubulointerstitial fibrosis was further exacerbated by global increases in OST48. There was significantly insulin effectiveness, increased acute insulin secretion, fasting insulin concentrations and AUCinsulin observed during glucose tolerance testing in diabetic mice with global elevations in OST48 when compared to diabetic wild-type littermates. Overall, this study suggested that despite facilitating urinary-renal AGE clearance, there were no benefits observed on kidney functional and structural parameters in diabetes afforded by globally increasing OST48 expression. However, the improvements in insulin secretion seen in diabetic mice with global over-expression of OST48 and their dissociation from effects on kidney function warrant future investigation.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Nefropatías Diabéticas/genética , Productos Finales de Glicación Avanzada/sangre , Hexosiltransferasas/genética , Insulina/metabolismo , Animales , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/fisiopatología , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Hexosiltransferasas/metabolismo , Pruebas de Función Hepática , Masculino , Ratones , Estreptozocina
11.
Front Physiol ; 10: 309, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31040788

RESUMEN

Patients with diabetic hypertensive nephropathy have accelerated disease progression. Diabetes and hypertension have both been associated with changes in renal catecholamines and reactive oxygen species. With a specific focus on renal catecholamines and oxidative stress we examined a combined model of hypertension and diabetes using normotensive BPN/3J and hypertensive BPH/2J Schlager mice. Induction of diabetes (5 × 55 mg/kg streptozotocin i.p.) did not change the hypertensive status of BPH/2J mice (telemetric 24 h avg. MAP, non-diabetic 131 ± 2 vs. diabetic 129 ± 1 mmHg, n.s at 9 weeks of study). Diabetes-associated albuminuria was higher in BPH/2J vs. diabetic BPN/3J (1205 + 196/-169 versus 496 + 67/-59 µg/24 h, p = 0.008). HPLC measurement of renal cortical norepinephrine and dopamine showed significantly greater levels in hypertensive mice whilst diabetes was associated with significantly lower catecholamine levels. Diabetic BPH/2J also had greater renal catecholamine levels than diabetic BPN/3J (diabetic: norepinephrine BPN/3J 40 ± 4, BPH/2J 91 ± 5, p = 0.010; dopamine: BPN/3J 2 ± 1; BPH/2J 3 ± 1 ng/mg total protein, p < 0.001 after 10 weeks of study). Diabetic BPH/2J showed greater cortical tubular immunostaining for monoamine oxidase A and cortical mitochondrial hydrogen peroxide formation was greater in both diabetic and non-diabetic BPH/2J. While cytosolic catalase activity was greater in non-diabetic BPH/2J it was significantly lower in diabetic BPH/2J (cytosolic: BPH/2J 127 ± 12 vs. 63 ± 6 nmol/min/ml, p < 0.001). We conclude that greater levels of renal norepinephrine and dopamine associated with hypertension, together with diabetes-associated compromised anti-oxidant systems, contribute to increased renal oxidative stress in diabetes and hypertension. Elevations in renal cortical catecholamines and reactive oxygen species have important therapeutic implications for hypertensive diabetic patients.

12.
Diabetes ; 65(4): 1085-98, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26822084

RESUMEN

Apoptosis-inducing factor (AIF) is a mitochondrial flavoprotein with dual roles in redox signaling and programmed cell death. Deficiency in AIF is known to result in defective oxidative phosphorylation (OXPHOS), via loss of complex I activity and assembly in other tissues. Because the kidney relies on OXPHOS for metabolic homeostasis, we hypothesized that a decrease in AIF would result in chronic kidney disease (CKD). Here, we report that partial knockdown of Aif in mice recapitulates many features of CKD, in association with a compensatory increase in the mitochondrial ATP pool via a shift toward mitochondrial fusion, excess mitochondrial reactive oxygen species production, and Nox4 upregulation. However, despite a 50% lower AIF protein content in the kidney cortex, there was no loss of complex I activity or assembly. When diabetes was superimposed onto Aif knockdown, there were extensive changes in mitochondrial function and networking, which augmented the renal lesion. Studies in patients with diabetic nephropathy showed a decrease in AIF within the renal tubular compartment and lower AIFM1 renal cortical gene expression, which correlated with declining glomerular filtration rate. Lentiviral overexpression of Aif1m rescued glucose-induced disruption of mitochondrial respiration in human primary proximal tubule cells. These studies demonstrate that AIF deficiency is a risk factor for the development of diabetic kidney disease.


Asunto(s)
Factor Inductor de la Apoptosis/genética , Diabetes Mellitus Experimental/complicaciones , Nefropatías Diabéticas/genética , Mitocondrias/metabolismo , Insuficiencia Renal Crónica/genética , Animales , Respiración de la Célula/genética , Células Cultivadas , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Nefropatías Diabéticas/metabolismo , Predisposición Genética a la Enfermedad , Homeostasis/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Fosforilación Oxidativa , Insuficiencia Renal Crónica/metabolismo , Factores de Riesgo
13.
Nat Rev Endocrinol ; 9(2): 113-23, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23296171

RESUMEN

Glycaemic control, reduction of blood pressure using agents that block the renin-angiotensin system and control of dyslipidaemia are the major strategies used in the clinical management of patients with diabetes mellitus. Each of these approaches interrupts a number of pathological pathways, which directly contributes to the vascular complications of diabetes mellitus, including renal disease, blindness, neuropathy and cardiovascular disease. However, research published over the past few years has indicated that many of the pathological pathways important in the development of the vascular complications of diabetes mellitus are equally relevant to the initiation of diabetes mellitus itself. These pathways include insulin signalling, generation of cellular energy, post-translational modifications and redox imbalances. This Review will examine how the development of diabetes mellitus has come full circle from initiation to complications and suggests that the development of diabetes mellitus and the progression to chronic complications both require the same mechanistic triggers.


Asunto(s)
Enfermedades Cardiovasculares/etiología , Complicaciones de la Diabetes/etiología , Diabetes Mellitus/etiología , Diabetes Mellitus/fisiopatología , Animales , Glucemia/metabolismo , Enfermedades Cardiovasculares/tratamiento farmacológico , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/microbiología , Tracto Gastrointestinal/microbiología , Humanos , Insulina/metabolismo
14.
Antioxid Redox Signal ; 19(4): 331-43, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23320803

RESUMEN

AIMS: Defects in the activity of enzyme complexes of the mitochondrial respiratory chain are thought to be responsible for several disorders, including renal impairment. Gene mutations that result in complex I deficiency are the most common oxidative phosphorylation disorders in humans. To determine whether an abnormality in mitochondrial complex I per se is associated with development of renal disease, mice with a knockdown of the complex I gene, Ndufs6 were studied. RESULTS: Ndufs6 mice had a partial renal cortical complex I deficiency; Ndufs6gt/gt, 32% activity and Ndufs6gt/+, 83% activity compared with wild-type mice. Both Ndufs6gt/+ and Ndufs6gt/gt mice exhibited hallmarks of renal disease, including albuminuria, urinary excretion of kidney injury molecule-1 (Kim-1), renal fibrosis, and changes in glomerular volume, with decreased capacity to generate mitochondrial ATP and superoxide from substrates oxidized via complex I. However, more advanced renal defects in Ndufs6gt/gt mice were observed in the context of a disruption in the inner mitochondrial electrochemical potential, 3-nitrotyrosine-modified mitochondrial proteins, increased urinary excretion of 15-isoprostane F2t, and up-regulation of antioxidant defence. Juvenile Ndufs6gt/gt mice also exhibited signs of early renal impairment with increased urinary Kim-1 excretion and elevated circulating cystatin C. INNOVATION: We have identified renal impairment in a mouse model of partial complex I deficiency, suggesting that even modest deficits in mitochondrial respiratory chain function may act as risk factors for chronic kidney disease. CONCLUSION: These studies identify for the first time that complex I deficiency as the result of interruption of Ndufs6 is an independent cause of renal impairment.


Asunto(s)
Complejo I de Transporte de Electrón/deficiencia , Complejo I de Transporte de Electrón/metabolismo , Enfermedades Renales/metabolismo , Enfermedades Mitocondriales/metabolismo , NADH Deshidrogenasa/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Antioxidantes/metabolismo , Complejo I de Transporte de Electrón/genética , Enfermedades Renales/genética , Ratones , Ratones Noqueados , Enfermedades Mitocondriales/genética , NADH Deshidrogenasa/genética , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/metabolismo
15.
Free Radic Biol Med ; 52(3): 716-723, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22172526

RESUMEN

Cardiovascular benefits of ubiquinone have been previously demonstrated, and we administered it as a novel therapy in an experimental model of type 2 diabetic nephropathy. db/db and dbH mice were followed for 10 weeks, after randomization to receive either vehicle or ubiquinone (CoQ10; 10mg/kg/day) orally. db/db mice had elevated urinary albumin excretion rates and albumin:creatinine ratio, not seen in db/db CoQ10-treated mice. Renal cortices from db/db mice had lower total and oxidized CoQ10 content, compared with dbH mice. Mitochondria from db/db mice also contained less oxidized CoQ10(ubiquinone) compared with dbH mice. Diabetes-induced increases in total renal collagen but not glomerulosclerosis were significantly decreased with CoQ10 therapy. Mitochondrial superoxide and ATP production via complex II in the renal cortex were increased in db/db mice, with ATP normalized by CoQ10. However, excess renal mitochondrial hydrogen peroxide production and increased mitochondrial membrane potential seen in db/db mice were attenuated with CoQ10. Renal superoxide dismutase activity was also lower in db/db mice compared with dbH mice. Our results suggest that a deficiency in mitochondrial oxidized CoQ10 (ubiquinone) may be a likely precipitating factor for diabetic nephropathy. Therefore CoQ10 supplementation may be renoprotective in type 2 diabetes, via preservation of mitochondrial function.


Asunto(s)
Diabetes Mellitus Tipo 2/complicaciones , Nefropatías Diabéticas/tratamiento farmacológico , Riñón/fisiopatología , Mitocondrias/efectos de los fármacos , Ubiquinona/análogos & derivados , Adenosina Trifosfato/biosíntesis , Albuminuria/tratamiento farmacológico , Animales , Citrato (si)-Sintasa/metabolismo , Creatinina/orina , Cistatina C/orina , Diabetes Mellitus Tipo 2/orina , Nefropatías Diabéticas/etiología , Nefropatías Diabéticas/orina , Femenino , Receptor Celular 1 del Virus de la Hepatitis A , Riñón/efectos de los fármacos , Riñón/enzimología , Riñón/patología , Pruebas de Función Renal , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Proteínas de la Membrana/orina , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Mitocondrias/enzimología , Mitocondrias/metabolismo , Oxidación-Reducción , Distribución Aleatoria , Especies Reactivas de Oxígeno/metabolismo , Ubiquinona/farmacología , Ubiquinona/uso terapéutico
16.
Exp Diabetes Res ; 2010: 974681, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21318189

RESUMEN

BACKGROUND: Advanced glycation end-products (AGEs) and their receptors are prominent contributors to diabetic kidney disease. METHODS: Flow cytometry was used to measure the predictive capacity for kidney impairment of the AGE receptors RAGE, AGE-R1, and AGE-R3 on peripheral blood mononuclear cells (PBMCs) in experimental models of type 2 diabetes (T2DM) fed varied AGE containing diets and in obese type 2 diabetic and control human subjects. RESULTS: Diets high in AGE content fed to diabetic mice decreased cell surface RAGE on PBMCs and in type 2 diabetic patients with renal impairment (RI). All diabetic mice had elevated Albumin excretion rates (AERs), and high AGE fed dbdb mice had declining Glomerular filtration rate (GFR). Cell surface AGE-R1 expression was also decreased by high AGE diets and with diabetes in dbdb mice and in humans with RI. PBMC expression of AGE R3 was decreased in diabetic dbdb mice or with a low AGE diet. CONCLUSIONS: The most predictive PBMC profile for renal disease associated with T2DM was an increase in the cell surface expression of AGE-R1, in the context of a decrease in membranous RAGE expression in humans, which warrants further investigation as a biomarker for progressive DN in larger patient cohorts.


Asunto(s)
Diabetes Mellitus Tipo 2/complicaciones , Nefropatías Diabéticas/sangre , Productos Finales de Glicación Avanzada/sangre , Animales , Diabetes Mellitus Tipo 2/sangre , Citometría de Flujo , Masculino , Ratones , Ratones Endogámicos C57BL , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/sangre
17.
Metabolism ; 59(11): 1556-61, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20199785

RESUMEN

Decreased gene expression of heat shock protein 72 (HSP72) in skeletal muscle is associated with insulin resistance in humans. We aimed to determine whether HSP72 protein expression in insulin-sensitive tissues is related to criterion standard measures of adiposity and insulin resistance in a young healthy human population free of hyperglycemia. Healthy participants (N = 17; age, 30 ± 3 years) underwent measurement of body composition (dual-energy x-ray absorptiometry), a maximum aerobic capacity test (VO(2max)), an oral glucose tolerance test, and a hyperinsulinemic-euglycemic clamp (M) to access insulin sensitivity. Skeletal muscle and subcutaneous adipose tissue biopsies were obtained by percutaneous needle biopsy. HSP72 protein expression in skeletal muscle was inversely related to percentage body fat (r = -0.54, P < .05) and remained significant after adjustment for age and sex (P < .05). Insulin sensitivity was also related to HSP72 protein expression in skeletal muscle (r = 0.52, P < .05); however, this relationship disappeared after adjustment for percentage body fat (P = .2). In adipose tissue, HSP72 protein expression was not related to adiposity or insulin sensitivity. Physical activity and aerobic fitness did not show any association with HSP72 protein expression in either tissue studied. A lower expression of HSP72 protein in human skeletal muscle was associated with increased adiposity and decreased insulin sensitivity in healthy individuals. These findings are consistent with rodent data suggesting that HSP72 stimulates fat oxidation with consequent reduction in fat storage and adiposity.


Asunto(s)
Adiposidad/genética , Proteínas del Choque Térmico HSP72/biosíntesis , Resistencia a la Insulina/genética , Músculo Esquelético/metabolismo , Absorciometría de Fotón , Tejido Adiposo , Adulto , Composición Corporal , Prueba de Esfuerzo , Prueba de Tolerancia a la Glucosa , Humanos , Músculo Esquelético/química
18.
Diabetes ; 57(2): 460-9, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17959934

RESUMEN

OBJECTIVE: Excessive production of reactive oxygen species (ROS) via NADPH oxidase has been implicated in the pathogenesis of diabetic nephropathy. Since NADPH oxidase activation is closely linked to other putative pathways, its interaction with changes in protein kinase C (PKC) and increased advanced glycation was examined. RESEARCH DESIGN AND METHODS: Streptozotocin-induced diabetic or nondiabetic Sprague Dawley rats were followed for 32 weeks, with groups randomized to no treatment or the NADPH oxidase assembly inhibitor apocynin (15 mg . kg(-1) . day(-1); weeks 16-32). Complementary in vitro studies were performed in which primary rat mesangial cells, in the presence and absence of advanced glycation end products (AGEs)-BSA, were treated with either apocynin or the PKC-alpha inhibitor Ro-32-0432. RESULTS; Apocynin attenuated diabetes-associated increases in albuminuria and glomerulosclerosis. Circulating, renal cytosolic, and skin collagen-associated AGE levels in diabetic rats were not reduced by apocynin. Diabetes-induced translocation of PKC, specifically PKC-alpha to renal membranes, was associated with increased NADPH-dependent superoxide production and elevated renal, serum, and urinary vascular endothelial growth factor (VEGF) concentrations. In both diabetic rodents and in AGE-treated mesangial cells, blockade of NADPH oxidase or PKC-alpha attenuated cytosolic superoxide and PKC activation and increased VEGF. Finally, renal extracellular matrix accumulation of fibronectin and collagen IV was decreased by apocynin. CONCLUSIONS: In the context of these and previous findings by our group, we conclude that activation of NADPH oxidase via phosphorylation of PKC-alpha is downstream of the AGE-receptor for AGE interaction in diabetic renal disease and may provide a novel therapeutic target for diabetic nephropathy.


Asunto(s)
Acetofenonas/uso terapéutico , Diabetes Mellitus Experimental/enzimología , Nefropatías Diabéticas/enzimología , Productos Finales de Glicación Avanzada/antagonistas & inhibidores , NADPH Oxidasas/antagonistas & inhibidores , Proteína Quinasa C-alfa/metabolismo , Animales , Diabetes Mellitus Experimental/patología , Nefropatías Diabéticas/patología , Inhibidores Enzimáticos/uso terapéutico , Ensayo de Inmunoadsorción Enzimática , Mesangio Glomerular/efectos de los fármacos , Mesangio Glomerular/enzimología , Mesangio Glomerular/patología , Lisina/análogos & derivados , Lisina/análisis , Masculino , Ratas , Ratas Sprague-Dawley , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/efectos de los fármacos , Receptores Inmunológicos/fisiología , Superóxidos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA