Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Nat Commun ; 10(1): 4427, 2019 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-31562314

RESUMEN

Insulin and IGF-1 actions in vascular smooth muscle cells (VSMC) are associated with accelerated arterial intima hyperplasia and restenosis after angioplasty, especially in diabetes. To distinguish their relative roles, we delete insulin receptor (SMIRKO) or IGF-1 receptor (SMIGF1RKO) in VSMC and in mice. Here we report that intima hyperplasia is attenuated in SMIRKO mice, but not in SMIGF1RKO mice. In VSMC, deleting IGF1R increases homodimers of IR, enhances insulin binding, stimulates p-Akt and proliferation, but deleting IR decreases responses to insulin and IGF-1. Studies using chimeras of IR(extracellular domain)/IGF1R(intracellular-domain) or IGF1R(extracellular domain)/IR(intracellular-domain) demonstrate homodimer IRα enhances insulin binding and signaling which is inhibited by IGF1Rα. RNA-seq identifies hyaluronan synthase2 as a target of homo-IR, with its expression increases by IR activation in SMIGF1RKO mice and decreases in SMIRKO mice. Enhanced intima hyperplasia in diabetes is mainly due to insulin signaling via homo-IR, associated with increased Has2 expression.


Asunto(s)
Diabetes Mellitus/metabolismo , Hiperplasia/metabolismo , Resistencia a la Insulina/fisiología , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/metabolismo , Animales , Modelos Animales de Enfermedad , Arteria Femoral/lesiones , Arteria Femoral/metabolismo , Arteria Femoral/patología , Homocigoto , Hialuronano Sintasas/metabolismo , Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Liso Vascular/metabolismo , Receptor IGF Tipo 1/química , Receptor de Insulina/química , Transducción de Señal
2.
Diabetes Care ; 42(7): 1263-1273, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31076418

RESUMEN

OBJECTIVE: Elevated glycolytic enzymes in renal glomeruli correlated with preservation of renal function in the Medalist Study, individuals with ≥50 years of type 1 diabetes. Specifically, pyruvate kinase M2 (PKM2) activation protected insulin-deficient diabetic mice from hyperglycemia-induced glomerular pathology. This study aims to extend these findings in a separate cohort of individuals with type 1 and type 2 diabetes and discover new circulatory biomarkers for renal protection through proteomics and metabolomics of Medalists' plasma. We hypothesize that increased glycolytic flux and improved mitochondrial biogenesis will halt the progression of diabetic nephropathy. RESEARCH DESIGN AND METHODS: Immunoblots analyzed selected glycolytic and mitochondrial enzymes in postmortem glomeruli of non-Medalists with type 1 diabetes (n = 15), type 2 diabetes (n = 19), and no diabetes (n = 5). Plasma proteomic (SOMAscan) (n = 180) and metabolomic screens (n = 214) of Medalists with and without stage 3b chronic kidney disease (CKD) were conducted and significant markers validated by ELISA. RESULTS: Glycolytic (PKM1, PKM2, and ENO1) and mitochondrial (MTCO2) enzymes were significantly elevated in glomeruli of CKD- versus CKD+ individuals with type 2 diabetes. Medalists' plasma PKM2 correlated with estimated glomerular filtration rate (r 2 = 0.077; P = 0.0002). Several glucose and mitochondrial enzymes in circulation were upregulated with corresponding downregulation of toxic metabolites in CKD-protected Medalists. Amyloid precursor protein was also significantly upregulated, tumor necrosis factor receptors downregulated, and both confirmed by ELISA. CONCLUSIONS: Elevation of enzymes involved in the metabolism of intracellular free glucose and its metabolites in renal glomeruli is connected to preserving kidney function in both type 1 and type 2 diabetes. The renal profile of elevated glycolytic enzymes and reduced toxic glucose metabolites is reflected in the circulation, supporting their use as biomarkers for endogenous renal protective factors in people with diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 2/complicaciones , Nefropatías Diabéticas/metabolismo , Enzimas/metabolismo , Glucosa/metabolismo , Piruvato Quinasa/metabolismo , Anciano , Anciano de 80 o más Años , Autopsia , Biomarcadores/sangre , Estudios de Casos y Controles , Estudios de Cohortes , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Nefropatías Diabéticas/sangre , Nefropatías Diabéticas/patología , Progresión de la Enfermedad , Enzimas/análisis , Femenino , Tasa de Filtración Glomerular , Humanos , Riñón/metabolismo , Riñón/patología , Riñón/fisiopatología , Glomérulos Renales/metabolismo , Glomérulos Renales/patología , Glomérulos Renales/fisiopatología , Masculino , Redes y Vías Metabólicas/fisiología , Metabolómica/métodos , Persona de Mediana Edad , Mitocondrias/metabolismo , Proteómica/métodos , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/patología , Insuficiencia Renal Crónica/fisiopatología
3.
J Mol Med (Berl) ; 96(5): 373-381, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29574544

RESUMEN

Diabetic nephropathy (DN) affects approximately 30-40% of patients with type 1 (T1DM) and type 2 diabetes (T2DM). It is a major cause of end-stage renal disease (ESRD) for the developed world. Hyperglycemia and genetics are major causal factors for the initiation and progression of DN. Multiple abnormalities in glucose and mitochondrial metabolism induced by diabetes likely contribute to the severity of DN. Recent clinical studies in people with extreme duration of T1DM (> 50 years, Joslin Medalist Study) have supported the importance of endogenous protective factors to neutralize the toxic effects of hyperglycemia on renal and other vascular tissues. Using renal glomeruli from these patients (namely Medalists) with and without DN, we have shown the importance of increased glycolytic flux in decreasing the accumulation of glucose toxic metabolites, improving mitochondrial function, survival of glomerular podocytes, and reducing glomerular pathology. Activation of a key glycolytic enzyme, pyruvate kinase M2 (PKM2), resulted in the normalization of renal hemodynamics and mitochondrial and glomerular dysfunction, leading to the mitigation of glomerular pathologies in several mouse models of DN.


Asunto(s)
Nefropatías Diabéticas/metabolismo , Glucosa/metabolismo , Riñón/metabolismo , Animales , Enfermedad Crónica , Nefropatías Diabéticas/fisiopatología , Humanos , Riñón/fisiopatología
4.
Circ Res ; 121(10): 1153-1167, 2017 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-28855204

RESUMEN

RATIONALE: Activation of monocytes/macrophages by hyperlipidemia associated with diabetes mellitus and obesity contributes to the development of atherosclerosis. PKCδ (protein kinase C δ) expression and activity in monocytes were increased by hyperlipidemia and diabetes mellitus with unknown consequences to atherosclerosis. OBJECTIVE: To investigate the effect of PKCδ activation in macrophages on the severity of atherosclerosis. METHODS AND RESULTS: PKCδ expression and activity were increased in Zucker diabetic rats. Mice with selective deletion of PKCδ in macrophages were generated by breeding PKCδ flox/flox mice with LyzM-Cre and ApoE-/- mice (MPKCδKO/ApoE-/- mice) and studied in atherogenic (AD) and high-fat diet (HFD). Mice fed AD and HFD exhibited hyperlipidemia, but only HFD-fed mice had insulin resistance and mild diabetes mellitus. Surprisingly, MPKCδKO/ApoE-/- mice exhibited accelerated aortic atherosclerotic lesions by 2-fold versus ApoE-/- mice on AD or HFD. Splenomegaly was observed in MPKCδKO/ApoE-/- mice on AD and HFD but not on regular chow. Both the AD or HFD increased macrophage number in aortic plaques and spleen by 1.7- and 2-fold, respectively, in MPKCδKO/ApoE-/- versus ApoE-/- mice because of decreased apoptosis (62%) and increased proliferation (1.9-fold), and not because of uptake, with parallel increased expressions of inflammatory cytokines. Mechanisms for the increased macrophages in MPKCδKO/ApoE-/- were associated with elevated phosphorylation levels of prosurvival cell-signaling proteins, Akt and FoxO3a, with reduction of proapoptotic protein Bim associated with PKCδ induced inhibition of P85/PI3K. CONCLUSIONS: Accelerated development of atherosclerosis induced by insulin resistance and hyperlipidemia may be partially limited by PKCδ isoform activation in the monocytes, which decreased its number and inflammatory responses in the arterial wall.


Asunto(s)
Apoptosis/fisiología , Aterosclerosis/metabolismo , Dieta Alta en Grasa/efectos adversos , Hiperlipidemias/metabolismo , Macrófagos/metabolismo , Proteína Quinasa C-delta/metabolismo , Animales , Aterosclerosis/etiología , Aterosclerosis/patología , Activación Enzimática/fisiología , Hiperlipidemias/etiología , Hiperlipidemias/patología , Resistencia a la Insulina/fisiología , Isoenzimas/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratas , Ratas Zucker
5.
Nat Med ; 23(6): 753-762, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28436957

RESUMEN

Diabetic nephropathy (DN) is a major cause of end-stage renal disease, and therapeutic options for preventing its progression are limited. To identify novel therapeutic strategies, we studied protective factors for DN using proteomics on glomeruli from individuals with extreme duration of diabetes (l50 years) without DN and those with histologic signs of DN. Enzymes in the glycolytic, sorbitol, methylglyoxal and mitochondrial pathways were elevated in individuals without DN. In particular, pyruvate kinase M2 (PKM2) expression and activity were upregulated. Mechanistically, we showed that hyperglycemia and diabetes decreased PKM2 tetramer formation and activity by sulfenylation in mouse glomeruli and cultured podocytes. Pkm-knockdown immortalized mouse podocytes had higher levels of toxic glucose metabolites, mitochondrial dysfunction and apoptosis. Podocyte-specific Pkm2-knockout (KO) mice with diabetes developed worse albuminuria and glomerular pathology. Conversely, we found that pharmacological activation of PKM2 by a small-molecule PKM2 activator, TEPP-46, reversed hyperglycemia-induced elevation in toxic glucose metabolites and mitochondrial dysfunction, partially by increasing glycolytic flux and PGC-1α mRNA in cultured podocytes. In intervention studies using DBA2/J and Nos3 (eNos) KO mouse models of diabetes, TEPP-46 treatment reversed metabolic abnormalities, mitochondrial dysfunction and kidney pathology. Thus, PKM2 activation may protect against DN by increasing glucose metabolic flux, inhibiting the production of toxic glucose metabolites and inducing mitochondrial biogenesis to restore mitochondrial function.


Asunto(s)
Diabetes Mellitus/metabolismo , Nefropatías Diabéticas/metabolismo , Glucosa/metabolismo , Potencial de la Membrana Mitocondrial , Mitocondrias/metabolismo , Podocitos/metabolismo , Piruvato Quinasa/genética , Anciano , Anciano de 80 o más Años , Animales , Western Blotting , Línea Celular , Diabetes Mellitus Experimental , Femenino , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Glucólisis , Humanos , Riñón/metabolismo , Glomérulos Renales/metabolismo , Masculino , Metabolómica , Ratones , Ratones Noqueados , Persona de Mediana Edad , Óxido Nítrico Sintasa de Tipo III/genética , Biogénesis de Organelos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Proteómica , Piruvato Quinasa/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
6.
Diabetologia ; 60(3): 585-596, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27933336

RESUMEN

AIMS/HYPOTHESIS: Accelerated migration and proliferation of vascular smooth muscle cells (VSMCs) enhances arterial restenosis after angioplasty in insulin resistance and diabetes. Elevation of Src homology 2-containing protein tyrosine phosphatase 1 (SHP-1) induces apoptosis in the microvasculature. However, the role of SHP-1 in intimal hyperplasia and restenosis has not been clarified in insulin resistance and diabetes. METHODS: We used a femoral artery wire injury mouse model, rodent models with insulin resistance and diabetes, and patients with type 2 diabetes. Further, we modulated SHP-1 expression using a transgenic mouse that overexpresses SHP-1 in VSMCs (Shp-1-Tg). SHP-1 agonists were also employed to study the molecular mechanisms underlying the regulation of SHP-1 by oxidised lipids. RESULTS: Mice fed a high-fat diet (HFD) exhibited increased femoral artery intimal hyperplasia and decreased arterial SHP-1 expression compared with mice fed a regular diet. Arterial SHP-1 expression was also decreased in Zucker fatty rats, Zucker diabetic fatty rats and in patients with type 2 diabetes. In primary cultured VSMCs, oxidised LDL suppressed SHP-1 expression by activating Mek-1 (also known as Map2k1) and increased DNA methylation of the Shp-1 promoter. VSMCs from Shp-1-Tg mice exhibited impaired platelet-derived growth factor (PDGF)-stimulated tyrosine phosphorylation with a concomitant decrease in PDGF-stimulated VSMC proliferation and migration. Similarly, HFD-fed Shp-1-Tg mice and mice treated with the SHP-1 inducer, Icariside II, were protected from the development of intimal hyperplasia following wire injury. CONCLUSIONS/INTERPRETATION: Suppression of SHP-1 by oxidised lipids may contribute to the excessive VSMC proliferation, inflammatory cytokine production and intimal hyperplasia observed in arteries from diabetes and insulin resistance. Augmenting SHP-1 levels is a potential therapeutic strategy to maintain stent patency in patients with insulin resistance and diabetes.


Asunto(s)
Hiperplasia/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Túnica Íntima/patología , Animales , Western Blotting , Ciclo Celular/genética , Ciclo Celular/fisiología , Movimiento Celular/genética , Movimiento Celular/fisiología , Proliferación Celular/genética , Proliferación Celular/fisiología , Humanos , Resistencia a la Insulina/genética , Resistencia a la Insulina/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Ratas , Ratas Zucker , Reacción en Cadena en Tiempo Real de la Polimerasa , Túnica Íntima/metabolismo
7.
J Clin Invest ; 126(3): 837-53, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26808499

RESUMEN

Abnormal fibroblast function underlies poor wound healing in patients with diabetes; however, the mechanisms that impair wound healing are poorly defined. Here, we evaluated fibroblasts from individuals who had type 1 diabetes (T1D) for 50 years or more (Medalists, n = 26) and from age-matched controls (n = 7). Compared with those from controls, Medalist fibroblasts demonstrated a reduced migration response to insulin, lower VEGF expression, and less phosphorylated AKT (p-AKT), but not p-ERK, activation. Medalist fibroblasts were also functionally less effective at wound closure in nude mice. Activation of the δ isoform of protein kinase C (PKCδ) was increased in postmortem fibroblasts from Medalists, fibroblasts from living T1D subjects, biopsies of active wounds of living T1D subjects, and granulation tissues from mice with streptozotocin-induced diabetes. Diabetes-induced PKCD mRNA expression was related to a 2-fold increase in the mRNA half-life. Pharmacologic inhibition and siRNA-mediated knockdown of PKCδ or expression of a dominant-negative isoform restored insulin signaling of p-AKT and VEGF expression in vitro and improved wound healing in vivo. Additionally, increasing PKCδ expression in control fibroblasts produced the same abnormalities as those seen in Medalist fibroblasts. Our results indicate that persistent PKCδ elevation in fibroblasts from diabetic patients inhibits insulin signaling and function to impair wound healing and suggest PKCδ inhibition as a potential therapy to improve wound healing in diabetic patients.


Asunto(s)
Diabetes Mellitus Tipo 1/enzimología , Pie Diabético/enzimología , Fibroblastos/fisiología , Proteína Quinasa C-delta/fisiología , Anciano , Anciano de 80 o más Años , Animales , Hipoxia de la Célula , Movimiento Celular , Proliferación Celular , Células Cultivadas , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 1/patología , Pie Diabético/patología , Femenino , Técnicas de Silenciamiento del Gen , Semivida , Humanos , Insulina/fisiología , Masculino , Ratones Desnudos , Persona de Mediana Edad , Proteína Quinasa C-delta/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Cicatrización de Heridas
8.
Circ Res ; 113(4): 418-27, 2013 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-23759514

RESUMEN

RATIONALE: Loss of insulin action in the endothelium can cause endothelial dysfunction and atherosclerosis. Hyperglycemia and elevated fatty acids induced by diabetes mellitus can activate protein kinase C-ß isoforms and selectively inhibit insulin signaling via phosphatidylinositol 3-kinase/Akt pathway to inhibit the activation of endothelial nitric oxide synthase and metabolic actions. OBJECTIVE: To demonstrate that overexpressing protein kinase C-ß2 isoform in endothelial cells can cause selective insulin resistance and exacerbate atherosclerosis in the aorta. METHODS AND RESULTS: Protein kinase C-ß2 isoform was overexpressed in endothelial cells using a promoter of vascular endothelial cell cadherin. These mice were cross-bred with apoE-/- mice [Tg (Prkcb)apoE-/-]. On a Western diet, Tg(Prkcb)apoE-/- and apoE-/- mice did not differ in systemic insulin sensitivity, glucose tolerance, plasma lipid, or blood pressure. Insulin action in endothelial cells and femoral artery from Tg(Prkcb)apoE-/- mice was impaired by ≈40% with respect to Akt/endothelial nitric oxide synthase activation, and leukocyte-endothelial cell binding increased in cultured lung endothelial cells from Tg(Prkcb)apoE-/- mice compared with that from apoE-/- mice. Basal and angiotensin-stimulated big endothelin-1 levels were elevated in Tg(Prkcb)apoE-/- mice compared with apoE-/- mice. The severity of atherosclerosis in the aorta from Tg(Prkcb)apoE-/- mice increased by ≈70% as measured by en face fat staining and plaque content of the number of smooth muscle cells, macrophages, and extracellular matrix. CONCLUSIONS: Specific protein kinase C-ß2 activation in the endothelial cells caused dysfunction and accelerated atherosclerosis because of loss of insulin-stimulated Akt/endothelial nitric oxide synthase activation and angiotensin-induced increases in endothelin-1 expression.


Asunto(s)
Aterosclerosis/fisiopatología , Endotelina-1/fisiología , Endotelio Vascular/fisiopatología , Resistencia a la Insulina/fisiología , Proteína Quinasa C beta/fisiología , Regulación hacia Arriba/fisiología , Animales , Aorta/patología , Aorta/fisiopatología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Apolipoproteínas E/fisiología , Aterosclerosis/patología , Modelos Animales de Enfermedad , Endotelina-1/genética , Endotelio Vascular/patología , Femenino , Isoenzimas/genética , Isoenzimas/fisiología , Masculino , Ratones , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo III/fisiología , Proteína Quinasa C beta/genética , Proteínas Proto-Oncogénicas c-akt/fisiología , Molécula 1 de Adhesión Celular Vascular/fisiología
9.
Invest Ophthalmol Vis Sci ; 53(13): 8424-32, 2012 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-23197686

RESUMEN

PURPOSE: To correlate changes between VEGF expression with systemic and retinal oxidative stress and inflammation in rodent models of obesity induced insulin resistance and diabetes. METHODS: Retinal VEGF mRNA and protein levels were assessed by RT-PCR and VEGF ELISA, respectively. Urinary 8-hydroxydeoxyguanosine (8-OHdG), blood levels of C-reactive protein (CRP), malondialdehyde (MDA), and CD11b/c positive cell ratio were used as systemic inflammatory markers. Retinal expression of Nox2, Nox4, and p47phox mRNA levels were measured as oxidative stress markers. TNF-α, inter-cellular adhesion molecule-1 (ICAM-1), IL1ß, and activation of nuclear factor κB (NF-κB) were used as retinal inflammatory markers. RESULTS: Retinal VEGF mRNA and protein expression increased in Zucker diabetic fatty (ZDF(fa/fa)) rats and streptozotosin (STZ) induced diabetic Sprague-Dawley rats, after two months of disease, but not in Zucker fatty (ZF) rats. Systemic markers of oxidative stress and inflammation were elevated in insulin resistant and diabetic rats. Some oxidative stress and inflammatory markers (TNF-α, IL-6, ICAM-1, and IL1-ß) were upregulated in the retina of ZDF(fa/fa) and STZ diabetic rats after 4 months of disease. In contrast, activation of NF-κB in the retina was observed in high fat fed nondiabetic and diabetic cis-NF-κB(EGFP) mice, ZF, ZDF(fa/fa), and STZ-induced diabetic rats. CONCLUSIONS: Only persistent hyperglycemia and diabetes increased retinal VEGF expression. Some markers of inflammation and oxidative stress were elevated in the retina and systemic circulation of obese and insulin resistant rodents with and without diabetes. Induction of VEGF and its associated retinal pathologies by diabetes requires chronic hyperglycemia and factors in addition to inflammation and oxidative stress.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Retinopatía Diabética/metabolismo , Resistencia a la Insulina/fisiología , Estrés Oxidativo/fisiología , Retina/metabolismo , Estrés Fisiológico/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , 8-Hidroxi-2'-Desoxicoguanosina , Animales , Biomarcadores/metabolismo , Proteína C-Reactiva/metabolismo , Antígeno CD11b/metabolismo , Antígeno CD11c/metabolismo , Desoxiguanosina/análogos & derivados , Desoxiguanosina/orina , Ensayo de Inmunoadsorción Enzimática , Inflamación/metabolismo , Masculino , Malondialdehído/sangre , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Obesidad/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Ratas Zucker , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor A de Crecimiento Endotelial Vascular/genética
10.
Semin Nephrol ; 32(5): 471-8, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23062988

RESUMEN

Diabetes results in vascular changes and dysfunction, and vascular complications are the leading cause of morbidity and mortality in diabetic patients. There has been a continual increase in the number of diabetic nephropathy patients and epidemic increases in the number of patients progressing to end-stage renal diseases. To identify targets for therapeutic intervention, most studies have focused on understanding how abnormal levels of glucose metabolites cause diabetic nephropathy, which is of paramount importance in devising strategies to combat the development and progression of diabetic nephropathy. However, less studied than the systemic toxic mechanisms, hyperglycemia and dyslipidemia might inhibit the endogenous vascular protective factors such as insulin, vascular endothelial growth factor, and platelet-derived growth factor. In this review, we highlight the importance of enhancing endogenous protective factors to prevent or delay diabetic nephropathy.


Asunto(s)
Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/prevención & control , Dislipidemias/metabolismo , Hiperglucemia/metabolismo , Antiinflamatorios/uso terapéutico , Antioxidantes/uso terapéutico , Nefropatías Diabéticas/etiología , Dislipidemias/complicaciones , Productos Finales de Glicación Avanzada/antagonistas & inhibidores , Productos Finales de Glicación Avanzada/metabolismo , Humanos , Hiperglucemia/complicaciones , Insulina/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
11.
FASEB J ; 26(7): 2963-74, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22499584

RESUMEN

This study characterizes the effect of glucose-induced activation of protein kinase Cδ (PKCδ) and Src homology-2 domain-containing phosphatase-1 (SHP-1) expression on vascular endothelial growth factor (VEGF) actions in glomerular podocytes in cultures and in glomeruli of diabetic rodents. Elevation of glucose levels induced PKCδ and p38 mitogen-activated protein kinase (p38 MAPK) to increase SHP-1 expression, increased podocyte apoptosis, and inhibited VEGF activation in podocytes and glomerular endothelial cells. The adverse effects of high glucose levels can be negated by molecular inhibitors of PKCδ, p38MAPK, and SHP-1 and only partially reduced by antioxidants and nuclear factor-κB (NF-κB) inhibitor. Increased PKCδ activation and SHP-1 expression correlated with loss of VEGF signaling and podocyte numbers in the glomeruli of diabetic rats and mice. In contrast, diabetic PKCδ-knockout (Prkcd(-/-)) mice did not exhibit activation of p38 MAPK and SHP-1 or inhibition of VEGF signaling in renal glomeruli. Functionally, diabetic Prkcd(-/-) mice had decreased expressions of TGFß, VEGF, and extracellular matrix and less albuminuria than diabetic Prkcd(+/+) mice. Hyperglycemia and diabetes can cause glomerular podocyte apoptosis and endothelial dysfunction partly due to increased PKCδ/p38 MAPK activation and the expression of SHP-1 to cause VEGF resistance, independent of NF-κB activation.


Asunto(s)
Nefropatías Diabéticas/etiología , Nefropatías Diabéticas/metabolismo , Proteína Quinasa C-delta/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Apoptosis , Secuencia de Bases , Células Cultivadas , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/patología , Células Endoteliales/metabolismo , Activación Enzimática , Femenino , Glucosa/metabolismo , Glomérulos Renales/metabolismo , Masculino , Ratones , Ratones Noqueados , FN-kappa B/metabolismo , Podocitos/metabolismo , Podocitos/patología , Proteína Quinasa C-delta/antagonistas & inhibidores , Proteína Quinasa C-delta/deficiencia , Proteína Quinasa C-delta/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/antagonistas & inhibidores , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , ARN Interferente Pequeño/genética , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
12.
PLoS One ; 6(7): e22777, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21829510

RESUMEN

BACKGROUND: Diabetic cardiomyopathy (DCM) is an increasingly recognized cause of chronic heart failure amongst diabetic patients. Both increased reactive oxygen species (ROS) generation and impaired ROS scavenging have been implicated in the pathogenesis of hyperglycemia-induced left ventricular dysfunction, cardiac fibrosis, apoptosis and hypertrophy. We hypothesized that 3',4'-dihydroxyflavonol (DiOHF), a small highly lipid soluble synthetic flavonol, may prevent DCM by scavenging ROS, thus preventing ROS-induced cardiac damage. METHODOLOGY/PRINCIPAL FINDINGS: Six week old homozygous Ren-2 rats were randomized to receive either streptozotocin or citrate buffer, then further randomized to receive either DiOHF (1 mg/kg/day) by oral gavage or vehicle for six weeks. Cardiac function was assessed via echocardiography and left ventricular cardiac catheterization before the animals were sacrificed and hearts removed for histological and molecular analyses. Diabetic Ren-2 rats showed evidence of diastolic dysfunction with prolonged deceleration time, reduced E/A ratio, and increased slope of end-diastolic pressure volume relationship (EDPVR) in association with marked interstitial fibrosis and oxidative stress (all P<0.05 vs control Ren-2). Treatment with DiOHF prevented the development of diastolic dysfunction and was associated with reduced oxidative stress and interstitial fibrosis (all P<0.05 vs untreated diabetic Ren-2 rats). In contrast, few changes were seen in non-diabetic treated animals compared to untreated counterparts. CONCLUSIONS: Inhibition of ROS production and action by DiOHF improved diastolic function and reduced myocyte hypertrophy as well as collagen deposition. These findings suggest the potential clinical utility of antioxidative compounds such as flavonols in the prevention of diabetes-associated cardiac dysfunction.


Asunto(s)
Antioxidantes/uso terapéutico , Diabetes Mellitus Experimental/tratamiento farmacológico , Cardiomiopatías Diabéticas/tratamiento farmacológico , Flavonoles/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo , Disfunción Ventricular Izquierda/tratamiento farmacológico , Animales , Autorradiografía , Cateterismo Cardíaco , Ecocardiografía , Homocigoto , Hibridación in Situ , Masculino , NADPH Oxidasas/metabolismo , Estrés Oxidativo , Ratas , Ratas Transgénicas
13.
Am J Physiol Renal Physiol ; 301(1): F84-93, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21478477

RESUMEN

Hyperglycemia and hypoxia have independent and convergent roles in the development of renal disease. Transforming growth factor-ß(1) (TGF-ß(1)) is a key cytokine promoting the production of extracellular matrix proteins. The cationic-independent mannose 6-phosphate receptor (CI-M6PR) is a membrane protein that binds M6P-containing proteins. A key role is to activate latent TGF-ß(1). PXS25, a novel CI-MPR inhibitor, has antifibrotic properties in skin fibroblasts, but its role in renal fibrosis is unclear. The aim was to study the role of PXS25 in matrix protein production under high glucose ± hypoxic conditions in human proximal tubule (HK-2) cells. HK-2 cells were exposed to high glucose (30 mM) ± 100 µM PXS25 in both normoxic (20% O(2)) and hypoxic (1% O(2)) conditions for 72 h. Cellular fibronectin, collagen IV, and matrix metalloproteinase-2 (MMP-2) and MMP-9 were assessed. Total and active TGF-ß(1) were measured by ELISA. High glucose and hypoxia independently induced TGF-ß(1) production. Active TGF-ß(1), but not total TGF-ß(1) was reduced with concurrent PXS25 in the presence of high glucose, but not in hyperglycemia+hypoxia conditions. Hyperglycemia induced fibronectin and collagen IV production (P < 0.05), as did hypoxia, but only hyperglycemia-induced increases in matrix proteins were suppressed by concurrent PXS25 exposure. High glucose induced MMP-2 and -9 in normoxic and hypoxic conditions, which was not modified in the presence of PXS25. High glucose and hypoxia can independently induce endogenous active TGF-ß(1) production in human proximal tubular cells. PXS25 inhibits conversion of high glucose-induced release of active TGF-ß(1), only in the absence of hypoxia.


Asunto(s)
Túbulos Renales Proximales/patología , Manosafosfatos/farmacología , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Factor de Crecimiento Transformador beta1/metabolismo , Western Blotting , Línea Celular , Proliferación Celular , Supervivencia Celular , Colágeno Tipo IV/antagonistas & inhibidores , Colágeno Tipo IV/biosíntesis , Nefropatías Diabéticas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Fibronectinas/biosíntesis , Fibronectinas/fisiología , Fibrosis , Glucosa/farmacología , Humanos , Hiperglucemia/metabolismo , Hipoxia/metabolismo , Túbulos Renales Proximales/enzimología , Túbulos Renales Proximales/metabolismo , Metaloproteinasa 9 de la Matriz/biosíntesis , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptor IGF Tipo 2 , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína Smad2/metabolismo
14.
Int J Biochem Cell Biol ; 43(3): 383-92, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21109018

RESUMEN

Macrophage inflammatory protein-3 alpha (MIP-3α) is known to be upregulated early in the development of diabetic nephropathy (DN). However, the transcriptional regulation of MIP-3α is unknown. We previously demonstrated that the transcription factors KLF6 and PPAR-γ play key roles in regulating renal fibrotic and inflammatory responses to factors inherent in diabetes mellitus. Hence we determined the role of these transcription factors in regulating MIP-3α expression. HK-2 cells and STZ-induced diabetic rats were used. siRNAs, over-expressing constructs and CHIP promoter binding assays were used to determine the role of KLF6 and PPAR-γ in MIP-3α transcriptional regulation. KLF6 overexpression increased MIP-3α which was inhibited by concurrent exposure to PPAR-γ agonists. PPAR-γ agonists attenuated high glucose-induced MIP-3α secretion. Furthermore, MIP-3α secretion was up-regulated in PPAR-γ silenced cells, suggesting both KLF6 and PPAR-γ antagonistically regulate high glucose-induced MIP-3α secretion. The CHIP promoter binding assay confirmed that PPAR-γ binds to the MIP-3α promoter and negatively regulates MIP-3α expression. PPAR-γ agonists increased the binding activity of the PPAR-γ-MIP-3α promoter. In contrast, promoter binding activity decreased in KLF6 over-expressing cells. PPAR-γ decreased in KLF6 over-expressing cells and increased in KLF6 silenced cells, while PPAR-γ siRNA had no effect on KLF6 expression, suggesting that KLF6 acted upstream of PPAR-γ in the regulation of MIP-3α. In diabetic rats, renal MIP-3α and the macrophage marker ED-1 expression increased, which was inhibited by exposure to PPAR-γ agonists. The recognition of MIP-3α as a significant pathogenic mediator in diabetic nephropathy reaffirms the increasingly recognized role of inflammation in the progression of DN. Targeting pro-inflammatory chemokine MIP-3α and its signaling pathways will provide novel strategy to treat diabetic kidney disease.


Asunto(s)
Quimiocina CCL20/genética , Diabetes Mellitus Experimental/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , PPAR gamma/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Quimiocina CCL20/metabolismo , Diabetes Mellitus Experimental/patología , Regulación de la Expresión Génica/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Glucosa/farmacología , Humanos , Corteza Renal/efectos de los fármacos , Corteza Renal/metabolismo , Corteza Renal/patología , Factor 6 Similar a Kruppel , Masculino , PPAR gamma/agonistas , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Tiazolidinedionas/farmacología
15.
Br J Pharmacol ; 162(3): 722-32, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20958293

RESUMEN

BACKGROUND AND PURPOSE: Aortic valve stenosis (AVS) is associated with significant cardiovascular morbidity and mortality. To date, no therapeutic modality has been shown to be effective in retarding AVS progression. We evaluated the effect of angiotensin-converting enzyme inhibition with ramipril on disease progression in a recently developed rabbit model of AVS. EXPERIMENTAL APPROACH: The effects of 8 weeks of treatment with either vitamin D2 at 25,000 IU for 4 days a week alone or in combination with ramipril (0.5 mg·kg⁻¹) on aortic valve structure and function were examined in New Zealand white rabbits. Echocardiographic aortic valve backscatter (AV(BS)) and aortic valve:outflow tract flow velocity ratio were utilized to quantify changes in valve structure and function. KEY RESULTS: Treatment with ramipril significantly reduced AV(BS) and improved aortic valve :outflow tract flow velocity ratio. The intravalvular content of the pro-oxidant thioredoxin-interacting protein was decreased significantly with ramipril treatment. Endothelial function, as measured by asymmetric dimethylarginine concentrations and vascular responses to ACh, was improved significantly with ramipril treatment. CONCLUSIONS AND IMPLICATIONS: Ramipril retards the development of AVS, reduces valvular thioredoxin-interacting protein accumulation and limits endothelial dysfunction in this animal model. These findings provide important insights into the mechanisms of AVS development and an impetus for future human studies of AVS retardation using an angiotensin-converting enzyme inhibitor.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Estenosis de la Válvula Aórtica/tratamiento farmacológico , Estenosis de la Válvula Aórtica/prevención & control , Ramipril/farmacología , Animales , Válvula Aórtica/efectos de los fármacos , Válvula Aórtica/fisiología , Estenosis de la Válvula Aórtica/diagnóstico por imagen , Estenosis de la Válvula Aórtica/fisiopatología , Arginina/análogos & derivados , Arginina/sangre , Proteínas Portadoras/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Ecocardiografía , Inhibidores Enzimáticos/sangre , Ergocalciferoles/uso terapéutico , Humanos , Masculino , Conejos , Vitaminas/uso terapéutico
16.
Nephrol Dial Transplant ; 26(1): 100-10, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20573806

RESUMEN

BACKGROUND: Diabetic nephropathy is the leading cause of kidney failure in the developed world. Tranilast has been reported to not only act as an anti-inflammatory and anti-fibrotic compound, but it also exerts anti-oxidative stress effects in diabetic nephropathy. Thioredoxin-interacting protein (Txnip) is the endogenous inhibitor of the anti-oxidant thioredoxin and is highly up-regulated in diabetic nephropathy, leading to oxidative stress and fibrosis. In this study, we aimed to investigate whether tranilast exerts its anti-oxidant properties through the inhibition of Txnip. METHODS: Heterozygous Ren-2 rats were rendered diabetic with streptozotocin. Another group of rats were injected with citrate buffer alone and treated as non-diabetic controls. After 6 weeks of diabetes, diabetic rats were divided into two groups: one group gavaged with tranilast at 200 mg/kg/day and another group with vehicle. RESULTS: Diabetic rats had a significant increase in albuminuria, tubulointerstitial fibrosis, peritubular collagen IV accumulation, reactive oxygen species (ROS) and macrophage infiltration (all P < 0.05). These changes were associated with an increase in Txnip mRNA and protein expression in the tubules and glomeruli of diabetic kidney. Treatment with tranilast for 4 weeks significantly attenuated Txnip up-regulation in diabetic rats and this was associated with a reduction in ROS, fibrosis and macrophage infiltration (all P < 0.05). CONCLUSIONS: This is the first study to demonstrate that tranilast not only has anti-inflammatory and anti-fibrotic effects as previously reported but also attenuates the up-regulation of Txnip and oxidative stress in diabetic nephropathy.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Proteínas Portadoras/metabolismo , Nefropatías Diabéticas/tratamiento farmacológico , Nefropatías Diabéticas/metabolismo , Modelos Animales de Enfermedad , Estrés Oxidativo/efectos de los fármacos , ortoaminobenzoatos/farmacología , Albuminuria/etiología , Animales , Antioxidantes/farmacología , Proteínas Portadoras/genética , Proteínas de Ciclo Celular , Colágeno Tipo IV/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Nefropatías Diabéticas/patología , Femenino , Fibrosis/etiología , Fibrosis/patología , Técnicas para Inmunoenzimas , Hibridación in Situ , Luminiscencia , Macrófagos/metabolismo , Nefritis Intersticial/etiología , Nefritis Intersticial/patología , ARN Mensajero/genética , Ratas , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba
17.
Am J Pathol ; 175(5): 1858-67, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19808645

RESUMEN

We demonstrated recently that thioredoxin-interacting protein (Txnip) and the transcription factor Krüppel-like factor 6 (KLF6) were up-regulated in both in vivo and in vitro models of diabetic nephropathy, thus promoting renal injury. Conversely, peroxisome proliferator-activated receptor-gamma (PPAR-gamma) agonists have been shown to be renoprotective. Hence, this study was undertaken to determine whether Txnip expression is regulated by the transcription factors KLF6 and PPAR-gamma. By using siRNAs and overexpressing constructs, the role of KLF6 and PPAR-gamma in Txnip transcriptional regulation was determined in human kidney proximal tubule cells and in streptozocin-induced diabetes mellitus in Sprague-Dawley rats, in vitro and in vivo models of diabetic nephropathy, respectively. KLF6 overexpression increased Txnip expression and promoter activity, which was inhibited by concurrent exposure to PPAR-gamma agonists. In contrast, reduced expression of KLF6 by siRNA or exposure to PPAR-gamma agonists attenuated high glucose-induced Txnip expression and promoter activity. KLF6-Txnip promoter binding was decreased in KLF6-silenced cells, whereas PPAR-gamma agonists increased PPAR-gamma-Txnip promoter binding. Indeed, silencing of KLF6 increased PPAR-gamma expression, suggesting endogenous regulation of PPAR-gamma expression by KLF6. Moreover, renal KLF6 and Txnip expression increased in rats with diabetes mellitus and was inhibited by PPAR-gamma agonist treatment; however, KLF6 expression did not change in HK-2 cells exposed to PPAR-gamma agonists. Hence, Txnip expression and promoter activity are mediated via divergent effects of KLF6 and PPAR-gamma transcriptional regulation.


Asunto(s)
Proteínas Portadoras/metabolismo , Regulación de la Expresión Génica , Glucosa/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , PPAR gamma/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Proteínas Portadoras/genética , Proteínas de Ciclo Celular , Línea Celular , Diabetes Mellitus Experimental , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/metabolismo , Modelos Animales de Enfermedad , Humanos , Factor 6 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Masculino , PPAR gamma/agonistas , PPAR gamma/genética , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley
18.
Front Biosci (Schol Ed) ; 1(1): 132-41, 2009 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-19482689

RESUMEN

Chronic kidney fibrosis is the unifying pathological feature of diverse renal disease leading to a progressive decline in renal function and eventually end-stage kidney failure. Many growth factors are able to induce an imbalance of extracellular matrix production and degradation, leading to excessive matrix and fibrosis in both glomeruli and in the tubulointerstitium. Over the last decade the role of connective tissue growth factor (CTGF) in renal fibrosis has been intensively studied. CTGF participates in cell proliferation, migration, and differentiation and mediates profibrotic activity by acting either directly, or as a co-factor for TGF beta 1, which is well characterised as a key cytokine mediating both the induction and promotion of fibrogenesis. CTGF also has the potential to modulate factors such as VEGF and bone morphogenic proteins, which are integral to both the development and repair process inherent in renal fibrogenesis. This review focuses on the role of CTGF in renal fibrosis and specifically its role in inducing fibrosis by factors integrally involved in the development of diabetic nephropathy, namely high glucose, angiotensin II, TGF beta 1 and AGEs.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/fisiología , Enfermedades Renales/fisiopatología , Enfermedad Crónica , Fibrosis , Humanos , Transducción de Señal
19.
J Am Soc Nephrol ; 20(4): 730-41, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19211714

RESUMEN

Excessive reactive oxygen species play a key role in the pathogenesis of diabetic nephropathy, but to what extent these result from increased generation, impaired antioxidant systems, or both is incompletely understood. Here, we report the expression, localization, and activity of the antioxidant thioredoxin and its endogenous inhibitor thioredoxin interacting protein (TxnIP) in vivo and in vitro. In normal human and rat kidneys, expression of TxnIP mRNA and protein was most abundant in the glomeruli and distal nephron (distal convoluted tubule and collecting ducts). In contrast, thioredoxin mRNA and protein localized to the renal cortex, particularly within the proximal tubules and to a lesser extent in the distal nephron. Induction of diabetes in rats increased expression of TxnIP but not thioredoxin mRNA. Kidneys from patients with diabetic nephropathy had significantly higher levels of TxnIP than control kidneys, but thioredoxin expression did not differ. In vitro, high glucose increased TxnIP expression in mesangial, NRK (proximal tubule), and MDCK (distal tubule/collecting duct) cells, and decreased the expression of thioredoxin in mesangial and MDCK cells. Knockdown of TxnIP with small interference RNA suggested that TxnIP mediates the glucose-induced impairment of thioredoxin activity. Knockdown of TxnIP also abrogated both glucose-induced 3H-proline incorporation (a marker of collagen production) and oxidative stress. Taken together, these findings suggest that impaired thiol reductive capacity contributes to the generation of reactive oxygen species in diabetes in a site- and cell-specific manner.


Asunto(s)
Diabetes Mellitus Experimental/fisiopatología , Nefropatías Diabéticas/fisiopatología , Tiorredoxinas/fisiología , Animales , Línea Celular , Nefropatías Diabéticas/genética , Perros , Femenino , Riñón/fisiología , Túbulos Renales Colectores/fisiología , Túbulos Renales Proximales/fisiología , ARN Mensajero/genética , Ratas , Valores de Referencia , Tiorredoxinas/genética
20.
Methods Mol Biol ; 466: 19-24, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19148604

RESUMEN

Primary cultures of renal proximal tubule cells (PTC) have been widely used to investigate tubule cell function. They provide a model system where confounding influences of renal haemodynamics, cell heterogeneity, and neural activity are eliminated. Additionally they are likely to more closely resemble PTC in vivo than established kidney cell lines, which are often virally immortalised and are of uncertain origin. This chapter describes a method used in our laboratories to isolate and culture pure populations of human PTC. The cortex is dissected away from the medulla and minced finely. Following collagenase digestion, the cells are passed through a sieve and separated on a Percoll density gradient. An almost pure population of tubule fragments form a band at the base of the gradient. Cultured in a hormonally defined serum-free growth media, they form a tightly packed monolayer that retains the differentiated characteristics of PTC for up to three passages.


Asunto(s)
Técnicas de Cultivo de Célula , Células Epiteliales , Túbulos Renales Proximales/citología , Células Cultivadas , Células Epiteliales/química , Células Epiteliales/citología , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA