Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Circ Res ; 127(7): 877-892, 2020 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-32564710

RESUMEN

RATIONALE: Treatment efficacy for diabetes mellitus is largely determined by assessment of HbA1c (glycated hemoglobin A1c) levels, which poorly reflects direct glucose variation. People with prediabetes and diabetes mellitus spend >50% of their time outside the optimal glucose range. These glucose variations, termed transient intermittent hyperglycemia (TIH), appear to be an independent risk factor for cardiovascular disease, but the pathological basis for this association is unclear. OBJECTIVE: To determine whether TIH per se promotes myelopoiesis to produce more monocytes and consequently adversely affects atherosclerosis. METHODS AND RESULTS: To create a mouse model of TIH, we administered 4 bolus doses of glucose at 2-hour intervals intraperitoneally once to WT (wild type) or once weekly to atherosclerotic prone mice. TIH accelerated atherogenesis without an increase in plasma cholesterol, seen in traditional models of diabetes mellitus. TIH promoted myelopoiesis in the bone marrow, resulting in increased circulating monocytes, particularly the inflammatory Ly6-Chi subset, and neutrophils. Hematopoietic-restricted deletion of S100a9, S100a8, or its cognate receptor Rage prevented monocytosis. Mechanistically, glucose uptake via GLUT (glucose transporter)-1 and enhanced glycolysis in neutrophils promoted the production of S100A8/A9. Myeloid-restricted deletion of Slc2a1 (GLUT-1) or pharmacological inhibition of S100A8/A9 reduced TIH-induced myelopoiesis and atherosclerosis. CONCLUSIONS: Together, these data provide a mechanism as to how TIH, prevalent in people with impaired glucose metabolism, contributes to cardiovascular disease. These findings provide a rationale for continual glucose control in these patients and may also suggest that strategies aimed at targeting the S100A8/A9-RAGE (receptor for advanced glycation end products) axis could represent a viable approach to protect the vulnerable blood vessels in diabetes mellitus. Graphic Abstract: A graphic abstract is available for this article.


Asunto(s)
Aterosclerosis/etiología , Glucemia/metabolismo , Hiperglucemia/complicaciones , Monocitos/metabolismo , Mielopoyesis , Neutrófilos/metabolismo , Animales , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Biomarcadores/sangre , Calgranulina A/genética , Calgranulina A/metabolismo , Calgranulina B/genética , Calgranulina B/metabolismo , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Transportador de Glucosa de Tipo 1/genética , Transportador de Glucosa de Tipo 1/metabolismo , Glucólisis , Hiperglucemia/sangre , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Monocitos/patología , Neutrófilos/patología , Placa Aterosclerótica , Receptor para Productos Finales de Glicación Avanzada/genética , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Transducción de Señal
2.
J Am Soc Nephrol ; 29(5): 1437-1448, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29490938

RESUMEN

Background The failure of spontaneous resolution underlies chronic inflammatory conditions, including microvascular complications of diabetes such as diabetic kidney disease. The identification of endogenously generated molecules that promote the physiologic resolution of inflammation suggests that these bioactions may have therapeutic potential in the context of chronic inflammation. Lipoxins (LXs) are lipid mediators that promote the resolution of inflammation.Methods We investigated the potential of LXA4 and a synthetic LX analog (Benzo-LXA4) as therapeutics in a murine model of diabetic kidney disease, ApoE-/- mice treated with streptozotocin.Results Intraperitoneal injection of LXs attenuated the development of diabetes-induced albuminuria, mesangial expansion, and collagen deposition. Notably, LXs administered 10 weeks after disease onset also attenuated established kidney disease, with evidence of preserved kidney function. Kidney transcriptome profiling defined a diabetic signature (725 genes; false discovery rate P≤0.05). Comparison of this murine gene signature with that of human diabetic kidney disease identified shared renal proinflammatory/profibrotic signals (TNF-α, IL-1ß, NF-κB). In diabetic mice, we identified 20 and 51 transcripts regulated by LXA4 and Benzo-LXA4, respectively, and pathway analysis identified established (TGF-ß1, PDGF, TNF-α, NF-κB) and novel (early growth response-1 [EGR-1]) networks activated in diabetes and regulated by LXs. In cultured human renal epithelial cells, treatment with LXs attenuated TNF-α-driven Egr-1 activation, and Egr-1 depletion prevented cellular responses to TGF-ß1 and TNF-αConclusions These data demonstrate that LXs can reverse established diabetic complications and support a therapeutic paradigm to promote the resolution of inflammation.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Nefropatías Diabéticas/tratamiento farmacológico , Nefropatías Diabéticas/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Lipoxinas/uso terapéutico , Albuminuria/etiología , Animales , Antiinflamatorios no Esteroideos/farmacología , Colágeno/metabolismo , Diabetes Mellitus Experimental , Nefropatías Diabéticas/complicaciones , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Mesangio Glomerular/patología , Humanos , Inyecciones Intraperitoneales , Lipoxinas/farmacología , Masculino , Ratones Noqueados para ApoE , FN-kappa B/genética , Factor de Crecimiento Derivado de Plaquetas/genética , Transcriptoma , Factor de Crecimiento Transformador beta1/genética , Factor de Necrosis Tumoral alfa/genética
3.
Clin Sci (Lond) ; 123(9): 547-55, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22616837

RESUMEN

TRAIL [TNF (tumour necrosis factor)-related apoptosis-inducing ligand] has recently been shown to ameliorate the natural history of DM (diabetes mellitus). It has not been determined yet whether systemic TRAIL delivery would prevent the metabolic abnormalities due to an HFD [HF (high-fat) diet]. For this purpose, 27 male C57bl6 mice aged 8 weeks were randomly fed on a standard diet, HFD or HFD+TRAIL for 12 weeks. TRAIL was delivered weekly by intraperitoneal injection. Body composition was evaluated; indirect calorimetry studies, GTT (glucose tolerance test) and ITT (insulin tolerance test) were performed. Pro-inflammatory cytokines, together with adipose tissue gene expression and apoptosis, were measured. TRAIL treatment reduced significantly the increased adiposity associated with an HFD. Moreover, it reduced significantly hyperglycaemia and hyperinsulinaemia during a GTT and it improved significantly the peripheral response to insulin. TRAIL reversed the changes in substrate utilization induced by the HFD and ameliorated skeletal muscle non-esterified fatty acids oxidation rate. This was associated with a significant reduction of pro-inflammatory cytokines together with a modulation of adipose tissue gene expression and apoptosis. These findings shed light on the possible anti-adipogenic and anti-inflammatory effects of TRAIL and open new therapeutic possibilities against obesity, systemic inflammation and T2DM (Type 2 DM).


Asunto(s)
Adiposidad , Grasas de la Dieta/administración & dosificación , Inflamación/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Animales , Apoptosis , Calorimetría , Citocinas/genética , Ingestión de Energía , Prueba de Tolerancia a la Glucosa , Hiperglucemia/prevención & control , Hiperinsulinismo/prevención & control , Inflamación/prevención & control , Mediadores de Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Oxidación-Reducción , Ácido Palmítico/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
4.
Clin Sci (Lond) ; 123(1): 29-37, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22288735

RESUMEN

Deficiency of ACE2 (angiotensin-converting enzyme 2), which degrades Ang (angiotensin) II, promotes the development of glomerular lesions. However, the mechanisms explaining why the reduction in ACE2 is associated with the development of glomerular lesions have still to be fully clarified. We hypothesized that ACE2 may regulate the renoprotective actions of ANP (atrial natriuretic peptide). The aim of the present study was to investigate the effect of ACE2 deficiency on the renal production of ANP. We evaluated molecular and structural abnormalities, as well as the expression of ANP in the kidneys of ACE2-deficient mice and C57BL/6 mice. We also exposed renal tubular cells to AngII and Ang-(1-7) in the presence and absence of inhibitors and agonists of RAS (renin-angiotensin system) signalling. ACE2 deficiency resulted in increased oxidative stress, as well as pro-inflammatory and profibrotic changes. This was associated with a down-regulation of the gene and protein expression on the renal production of ANP. Consistent with a role for the ACE2 pathway in modulating ANP, exposing cells to either Ang-(1-7) or ACE2 or the Mas receptor agonist up-regulated ANP gene expression. This work demonstrates that ACE2 regulates renal ANP via the generation of Ang-(1-7). This is a new mechanism whereby ACE2 counterbalances the renal effects of AngII and which explains why targeting ACE2 may be a promising strategy against kidney diseases, including diabetic nephropathy.


Asunto(s)
Angiotensina I/metabolismo , Factor Natriurético Atrial/metabolismo , Riñón/metabolismo , Fragmentos de Péptidos/metabolismo , Peptidil-Dipeptidasa A/deficiencia , Angiotensina II/metabolismo , Enzima Convertidora de Angiotensina 2 , Animales , Diabetes Mellitus/metabolismo , Riñón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Peptidil-Dipeptidasa A/metabolismo , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Sistema Renina-Angiotensina/fisiología
5.
Nephrol Dial Transplant ; 27(5): 1793-800, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22036945

RESUMEN

BACKGROUND: Angiotensin II (AngII) contributes to salt-driven kidney damage. In this study, we aimed at investigating whether and how the renal damage associated with a high-salt diet could result from changes in the ratio between angiotensin-converting enzyme (ACE) and angiotensin-converting enzyme 2 (ACE2). METHODS: Forty-eight rats randomly allocated to three different dietary contents of salt were studied for 4 weeks after undergoing a left uninephrectomy. We focussed on kidney functional, structural and molecular changes. At the same time, we studied kidney molecular changes in 20 weeks old Ace2-knockout mice (Ace2KO), with and without ACE inhibition. RESULTS: A high salt content diet significantly increased the glomerular ACE/ACE2 ratio. This was associated with increased oxidative stress. To assess whether these events were related, we measured renal oxidative stress in Ace2KO, and found that the absence of ACE2 promoted oxidative stress, which could be prevented by ACE inhibition. CONCLUSION: One of the mechanisms by which a high-salt diet leads to renal damage seems to be the modulation of the ACE/ACE2 ratio which in turn is critical for the cause of oxidative stress, through AngII.


Asunto(s)
Enfermedades Renales/metabolismo , Glomérulos Renales/efectos de los fármacos , Glomérulos Renales/metabolismo , Estrés Oxidativo/efectos de los fármacos , Peptidil-Dipeptidasa A/metabolismo , Cloruro de Sodio Dietético/farmacología , Angiotensina II/metabolismo , Enzima Convertidora de Angiotensina 2 , Animales , Modelos Animales de Enfermedad , Enfermedades Renales/inducido químicamente , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nefrectomía , Peptidil-Dipeptidasa A/deficiencia , Peptidil-Dipeptidasa A/genética , Ratas , Ratas Wistar , Sistema Renina-Angiotensina/fisiología , Cloruro de Sodio Dietético/efectos adversos
6.
J Clin Invest ; 129(1): 406-421, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30530993

RESUMEN

Activation of the type 1 angiotensin II receptor (AT1) triggers proinflammatory signaling through pathways independent of classical Gq signaling that regulate vascular homeostasis. Here, we report that the AT1 receptor preformed a heteromeric complex with the receptor for advanced glycation endproducts (RAGE). Activation of the AT1 receptor by angiotensin II (Ang II) triggered transactivation of the cytosolic tail of RAGE and NF-κB-driven proinflammatory gene expression independently of the liberation of RAGE ligands or the ligand-binding ectodomain of RAGE. The importance of this transactivation pathway was demonstrated by our finding that adverse proinflammatory signaling events induced by AT1 receptor activation were attenuated when RAGE was deleted or transactivation of its cytosolic tail was inhibited. At the same time, classical homeostatic Gq signaling pathways were unaffected by RAGE deletion or inhibition. These data position RAGE transactivation by the AT1 receptor as a target for vasculoprotective interventions. As proof of concept, we showed that treatment with the mutant RAGE peptide S391A-RAGE362-404 was able to inhibit transactivation of RAGE and attenuate Ang II-dependent inflammation and atherogenesis. Furthermore, treatment with WT RAGE362-404 restored Ang II-dependent atherogenesis in Ager/Apoe-KO mice, without restoring ligand-mediated signaling via RAGE, suggesting that the major effector of RAGE activation was its transactivation.


Asunto(s)
Aterosclerosis/metabolismo , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Transducción de Señal , Activación Transcripcional , Animales , Aterosclerosis/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Eliminación de Gen , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Noqueados para ApoE , Dominios Proteicos , Receptor de Angiotensina Tipo 1/genética
7.
ScientificWorldJournal ; 8: 434-45, 2008 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-18454253

RESUMEN

Hypertension is now recognized as a key contributory factor to the development and progression of kidney disease in both type 1 and type 2 diabetes. The renin angiotensin system (RAS) and its effector molecule angiotensin II, in particular, have a range of hemodynamic and nonhemodynamic effects that contribute not only to the development of hypertension, but also to renal disease. As a result, therapeutic inhibition of the RAS with angiotensin-converting enzyme inhibitors and/or selective angiotensin II type 1 receptor blockers has been proposed as a key strategy for reducing kidney damage beyond the expected effects one would observe with blood pressure reduction per se. Although the relationship between the RAS and the progression of diabetic renal disease has been known for many decades, recent advances have revealed a more complex paradigm with the discovery of a number of new components. Thus, further understanding of these new components of the renin angiotensin aldosterone system (RAAS), such as the angiotensin type 2 receptor subtype, angiotensin converting enzyme 2, and the recently cloned renin receptor, is likely to have therapeutic implications for disorders such as diabetic nephropathy, where interruption of the RAAS is widely used.


Asunto(s)
Angiotensina II/metabolismo , Nefropatías Diabéticas/fisiopatología , Modelos Biológicos , Sistema Renina-Angiotensina , Animales , Humanos
8.
J Am Heart Assoc ; 7(13)2018 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-29960985

RESUMEN

BACKGROUND: 8-Oxo-2'-deoxyguanosine (8-oxo-2'-dG) is a biomarker of oxidative DNA damage that is associated with cardiovascular disease and premature mortality in the general population. Although oxidative stress has a proven role in cardiovascular complications in diabetes mellitus, evidence for a relationship between plasma 8-oxo-2'-dG and major cardiovascular outcomes in diabetes mellitus is weak. METHODS AND RESULTS: A case-cohort study was performed in 3766 participants with prevalent diabetes mellitus in the ADVANCE (Action in Diabetes and Vascular Disease: Preterax and Diamicron Modified Release Controlled Evaluation) trial (ClinicalTrials.gov number NCT00145925). The hazard ratios for mortality and major acute cardiovascular events were derived using Cox regression models. During a median of 5 years of follow-up, 695 (18.4%) participants in this enriched cohort died (including 354 deaths from cardiovascular disease). Individuals with higher levels of 8-oxo-2'-dG were more likely to die. After adjusting for cardiovascular disease risk factors, the hazard ratio for a 1-SD increase in plasma 8-oxo-2'-dG was 1.10 (95% confidence interval, 1.01-1.20; P=0.03). This was driven by an independent association between plasma 8-oxo-2'-dG and cardiovascular death (hazard ratio, 1.23; 95% confidence interval, 1.10-1.37 [P<0.001]). By contrast, no association was seen between 8-oxo-2'-dG and noncardiovascular disease death (of which cancer was the major single cause). 8-Oxo-2'-dG was also not significantly associated with either nonfatal myocardial infarction or nonfatal stroke. CONCLUSIONS: In adults with type 2 diabetes mellitus, increased levels of 8-oxo-2'-dG are independently associated with all-cause mortality and cardiovascular mortality in adults with longstanding type 2 diabetes mellitus who participated in the ADVANCE trial, consistent with the role of oxidative damage in the development and progression of cardiovascular decompensation in diabetes mellitus. CLINICAL TRIAL REGISTRATION: URL: http://www.clinicaltrials.gov. Unique identifier: NCT00145925.


Asunto(s)
Enfermedades Cardiovasculares/sangre , Daño del ADN , Desoxiguanosina/análogos & derivados , Diabetes Mellitus Tipo 2/sangre , Estrés Oxidativo , 8-Hidroxi-2'-Desoxicoguanosina , Anciano , Biomarcadores/sangre , Enfermedades Cardiovasculares/diagnóstico , Enfermedades Cardiovasculares/epidemiología , Causas de Muerte , Desoxiguanosina/sangre , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/epidemiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Prospectivos , Ensayos Clínicos Controlados Aleatorios como Asunto , Medición de Riesgo , Factores de Riesgo , Factores de Tiempo , Regulación hacia Arriba
9.
Diabetes ; 67(12): 2657-2667, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30213823

RESUMEN

Increasing evidence points to the fact that defects in the resolution of inflammatory pathways predisposes individuals to the development of chronic inflammatory diseases, including diabetic complications such as accelerated atherosclerosis. The resolution of inflammation is dynamically regulated by the production of endogenous modulators of inflammation, including lipoxin A4 (LXA4). Here, we explored the therapeutic potential of LXA4 and a synthetic LX analog (Benzo-LXA4) to modulate diabetic complications in the streptozotocin-induced diabetic ApoE-/- mouse and in human carotid plaque tissue ex vivo. The development of diabetes-induced aortic plaques and inflammatory responses of aortic tissue, including the expression of vcam-1, mcp-1, il-6, and il-1ß, was significantly attenuated by both LXA4 and Benzo-LXA4 in diabetic ApoE-/- mice. Importantly, in mice with established atherosclerosis, treatment with LXs for a 6-week period, initiated 10 weeks after diabetes onset, led to a significant reduction in aortic arch plaque development (19.22 ± 2.01% [diabetic]; 12.67 ± 1.68% [diabetic + LXA4]; 13.19 ± 1.97% [diabetic + Benzo-LXA4]). Secretome profiling of human carotid plaque explants treated with LXs indicated changes to proinflammatory cytokine release, including tumor necrosis factor-α and interleukin-1ß. LXs also inhibited platelet-derived growth factor-stimulated vascular smooth muscle cell proliferation and transmigration and endothelial cell inflammation. These data suggest that LXs may have therapeutic potential in the context of diabetes-associated vascular complications.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Aorta/efectos de los fármacos , Aterosclerosis/tratamiento farmacológico , Diabetes Mellitus Experimental/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Lipoxinas/uso terapéutico , Animales , Antiinflamatorios no Esteroideos/farmacología , Aterosclerosis/etiología , Quimiocina CCL2/metabolismo , Citocinas/metabolismo , Diabetes Mellitus Experimental/complicaciones , Humanos , Inflamación/etiología , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Lipoxinas/farmacología , Ratones , Molécula 1 de Adhesión Celular Vascular/metabolismo
10.
Circ Res ; 92(7): 785-92, 2003 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-12623881

RESUMEN

The formation of advanced glycation end products (AGEs) on extracellular matrix components leads to accelerated increases in collagen cross linking that contributes to myocardial stiffness in diabetes. This study determined the effect of the crosslink breaker, ALT-711 on diabetes-induced cardiac disease. Streptozotocin diabetes was induced in Sprague-Dawley rats for 32 weeks. Treatment with ALT-711 (10 mg/kg) was initiated at week 16. Diabetic hearts were characterized by increased left ventricular (LV) mass and brain natriuretic peptide (BNP) expression, decreased LV collagen solubility, and increased collagen III gene and protein expression. Diabetic hearts had significant increases in AGEs and increased expression of the AGE receptors, RAGE and AGE-R3, in association with increases in gene and protein expression of connective tissue growth factor (CTGF). ALT-711 treatment restored LV collagen solubility and cardiac BNP in association with reduced cardiac AGE levels and abrogated the increase in RAGE, AGE-R3, CTGF, and collagen III expression. The present study suggests that AGEs play a central role in many of the alterations observed in the diabetic heart and that cleavage of preformed AGE crosslinks with ALT-711 leads to attenuation of diabetes-associated cardiac abnormalities in rats. This provides a potential new therapeutic approach for cardiovascular disease in human diabetes.


Asunto(s)
Diabetes Mellitus Experimental/prevención & control , Productos Finales de Glicación Avanzada/antagonistas & inhibidores , Hipertrofia Ventricular Izquierda/prevención & control , Tiazoles/farmacología , Animales , Presión Sanguínea/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Colágeno/genética , Colágeno/metabolismo , Citocinas/genética , Citocinas/metabolismo , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Productos Finales de Glicación Avanzada/metabolismo , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Hipertrofia Ventricular Izquierda/etiología , Hipertrofia Ventricular Izquierda/metabolismo , Inmunohistoquímica , Masculino , Miocardio/metabolismo , Miocardio/patología , Péptido Natriurético Encefálico/genética , Tamaño de los Órganos/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo
11.
Diabetes ; 53(4): 989-97, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15047614

RESUMEN

The renin-angiotensin system (RAS) has an important role in the endocrine pancreas. Although angiotensin II has significant effects on cell proliferation and apoptosis, the contribution of the RAS to changes in islet structure and function associated with type 2 diabetes is yet to be defined. This study examined the specific effects of RAS blockade on islet structure and function in diabetes. Thirty-six male Zucker diabetic fatty (ZDF) rats, 10 weeks of age, were randomized to receive the angiotensin-converting enzyme inhibitor perindopril (8 mg/l in drinking water; n = 12), irbesartan (15 mg/kg via gavage; n = 12), or no treatment (n = 12) for 10 weeks. Results were compared with lean littermates (ZL) (n = 12) studied concurrently. ZDF rats had increased intra-islet expression of components of the RAS correlating with increased intraislet fibrosis, apoptosis, and oxidative stress. Disordered islet architecture, seen in ZDF rats, was attenuated after treatment with perindopril or irbesartan. Islet fibrogenesis was also diminished, as measured by picrosirius staining and expression of collagens I and IV. Gene expression of transforming growth factor-beta1 was increased in the ZDF pancreas (ZL, 1.0 +/- 0.1; ZDF, 2.0 +/- 0.3; P < 0.05) and reduced after blockade of the RAS (ZDF + P, 1.3 +/- 0.2; ZDF + I, 1.5 +/- 0.1; vs. ZDF, both P < 0.05). Improvements in structural parameters were also associated with functional improvements in first-phase insulin secretion. These findings provide a possible mechanism for the reduced incidence of new-onset diabetes that has been observed in clinical trials of RAS blockade.


Asunto(s)
Islotes Pancreáticos/metabolismo , Sistema Renina-Angiotensina/fisiología , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Apoptosis , Compuestos de Bifenilo/farmacología , Glucemia/metabolismo , Presión Sanguínea , Peso Corporal , División Celular , Ayuno , Prueba de Tolerancia a la Glucosa , Irbesartán , Islotes Pancreáticos/efectos de los fármacos , Masculino , Perindopril/farmacología , Ratas , Ratas Zucker , Sistema Renina-Angiotensina/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tetrazoles/farmacología
12.
J Hypertens ; 23(3): 463-73, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15716683

RESUMEN

Recent trials have suggested that inhibitors of the renin-angiotensin system (RAS), such as angiotensin-converting enzyme (ACE) inhibitors and angiotensin II receptor blockers (ARBs), may reduce the incidence of new-onset diabetes in patients with or without hypertension and at high risk of developing diabetes. In this review, we critically evaluate the evidence from recent clinical trials for such a potential preventive effect of ACE inhibitors and ARBs, including a meta-analysis of these recent trials. The reduced incidence of diabetes in patients at high risk of developing diabetes by ACE inhibitors or ARBs has been explained by haemodynamic effects, such as improved delivery of insulin and glucose to the peripheral skeletal muscle, and non-haemodynamic effects, including direct effects on glucose transport and insulin signalling pathways, all of which decrease insulin resistance. There is now evidence that the pancreas may contain an in situ active RAS, which appears to be upregulated in an animal model of type 2 diabetes. Thus, ACE inhibitors and ARBs may act by attenuating the deleterious effect of angiotensin II on vasoconstriction, fibrosis, inflammation, apoptosis and beta-cell death in the pancreas, thereby protecting a critical beta-cell mass essential for insulin production. New evidence is presented that ACE inhibitors and ARBs may delay or prevent the development of insulin resistance and diabetes, for which novel mechanisms are suggested. The actions of agents that interrupt the RAS on insulin resistance, obesity and diabetes warrant further investigation in other animal models. Prospective clinical studies with the primary endpoint of the prevention of diabetes are now indicated to (i) further explore whether the inhibitors of the RAS are superior compared to other antihypertensive agents such as calcium channel blockers (CCBs) and (ii) to evaluate the potential beneficial effects of combination antihypertensive regimens on the development of diabetes.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/uso terapéutico , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Diabetes Mellitus Tipo 2/prevención & control , Hipertensión/tratamiento farmacológico , Sistema Renina-Angiotensina/efectos de los fármacos , Diabetes Mellitus Tipo 2/epidemiología , Humanos , Hipertensión/epidemiología , Incidencia
13.
Trends Endocrinol Metab ; 15(4): 166-9, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15109615

RESUMEN

Angiotensin-converting enzyme (ACE) is a zinc metalloproteinase and a key regulator of the renin-angiotensin system (RAS). ACE2 is a newly described enzyme identified in rodents and humans with a more restricted distribution than ACE, and is found mainly in heart and kidney. ACE2 cleaves a single residue from angiotensin I (Ang I) to generate Ang 1-9, and degrades Ang II, the main effector of the RAS, to the vasodilator Ang 1-7. The importance of ACE2 in normal physiology and pathophysiological states is largely unknown. ACE2 might act in a counter-regulatory manner to ACE, modulating the balance between vasoconstrictors and vasodilators within the heart and kidney, and playing a significant role in regulating cardiovascular and renal function.


Asunto(s)
Carboxipeptidasas/fisiología , Peptidil-Dipeptidasa A/fisiología , Sistema Renina-Angiotensina/fisiología , Enzima Convertidora de Angiotensina 2 , Animales , Carboxipeptidasas/metabolismo , Corazón/fisiología , Humanos , Hipertensión/fisiopatología , Riñón/enzimología , Riñón/fisiología , Miocardio/enzimología , Peptidil-Dipeptidasa A/metabolismo , Distribución Tisular
14.
Metabolism ; 64(3): 406-15, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25484288

RESUMEN

BACKGROUND: This study aimed at investigating the effects of genetic angiotensin-converting enzyme (ACE) 2 deficiency on glucose homeostasis in the pancreas and skeletal muscle and their reversibility following ACE inhibition. PROCEDURES: ACE2-knockout and C57bl6J mice were placed on a standard diet (SD) or a high-fat diet (HFD) for 12 weeks. An additional group of ACE2-knockout mice was fed a SD and treated with the ACE inhibitor, perindopril (2 mg kg(-1)day(-1)). Glucose and insulin tolerance tests, indirect calorimetry measurements and EchoMRI were performed. Non-esterfied 'free' fatty acid oxidation rate in skeletal muscle was calculated by measuring the palmitate oxidation rate. ß-cell mass was determined by immunostaining. Insulin, collectrin, glucose transporter protein, and peroxisome proliferator-activated receptor-γ expression were analysed by RT-PCR. Markers of mithocondrial biogenesis/content were also evaluated. MAIN FINDINGS: ACE2-knockout mice showed a ß-cell defect associated with low insulin and collectrin levels and reduced compensatory hypertrophy in response to a HFD, which were not reversed by perindopril. On the other hand, ACE2 deficiency shifted energy metabolism towards glucose utilization, as it increased the respiratory exchange ratio, reduced palmitate oxidation and PCG-1α expression in the skeletal muscle, where it up-regulated glucose transport proteins. Treatment of ACE2-knockout mice with perindopril reversed the skeletal muscle changes, suggesting that these were dependent on Angiotensin II (Ang II). PRINCIPAL CONCLUSIONS: ACE2-knockout mice display a ß-cell defect, which does not seem to be dependent on Ang II but may reflect the collectrin-like action of ACE2. This defect seemed to be compensated by the fact that ACE2-knockout mice shifted their energy consumption towards glucose utilisation via Ang II.


Asunto(s)
Metabolismo Energético/genética , Glucosa/metabolismo , Peptidil-Dipeptidasa A/deficiencia , Enzima Convertidora de Angiotensina 2 , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Animales , Composición Corporal/genética , Dieta Alta en Grasa , Homeostasis , Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/metabolismo , Páncreas/metabolismo , Peptidil-Dipeptidasa A/genética , Perindopril/uso terapéutico
15.
Endocrinology ; 145(2): 860-6, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14592956

RESUMEN

Connective tissue growth factor (CTGF) has been postulated to have prosclerotic and angiogenic properties. The aim of this present study was to characterize retinal CTGF expression in the absence and presence of diabetes and in the context of treatment with the angiotensin-converting enzyme (ACE) inhibitor, perindopril. Retinas were obtained from control, diabetic, and diabetic plus perindopril-treated (3 mg/d) rats. CTGF gene expression was quantitated by RT-PCR and localized by in situ hybridization. CTGF protein expression was analyzed by Western blotting and localized by immunohistochemistry. Diabetes was associated with a greater than 2-fold increase in CTGF mRNA levels, which was attenuated by perindopril treatment. CTGF immunoreactivity was increased almost 2-fold in diabetes and was ameliorated by the ACE inhibitor perindopril. By in situ hybridization and immunohistochemistry, the major site of CTGF gene expression in the retina of diabetic rats was the ganglion cell layer. Based on the known in vivo effects of CTGF, it is postulated that this growth factor plays a pivotal role in mediating diabetes-associated retinal pathology. Furthermore, the protective effects of ACE inhibitors on retinal pathology may partly be mediated via effects on retinal CTGF expression.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Diabetes Mellitus Experimental/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas Inmediatas-Precoces/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Retina/química , Animales , Western Blotting , Factor de Crecimiento del Tejido Conjuntivo , Retinopatía Diabética/metabolismo , Proteínas Inmediatas-Precoces/análisis , Inmunohistoquímica , Hibridación in Situ , Péptidos y Proteínas de Señalización Intercelular/análisis , Masculino , Perindopril/farmacología , ARN Mensajero/análisis , Ratas , Ratas Sprague-Dawley , Células Ganglionares de la Retina/química , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Distribución Tisular
16.
J Comp Neurol ; 456(1): 29-38, 2003 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-12508311

RESUMEN

In this study, the expression of receptors for calcitonin (CTR), the CTR C1a and C1b isoforms, was investigated during development of the fetal rat central nervous system (CNS) by using in situ hybridization and immunohistochemistry. Coincident expression with both techniques was evident. Immunohistochemical evidence for the expression of the C1a isoform alone was found. Expression was first observed at embryonic day 12/13 (E12/E13) within and adjacent to the ventricular zones known to include primary matrices of proliferation, in regions of the preoptic area, anterior and posterior hypothalamus, anterior and posterior pons, medulla, and spinal cord. At later times, with the decline in the density of immunoreactivity at these loci (E15), expression in primary matrices was found later at distinct loci within the ventricular zones of cerebellum (E17), and at E19, the tectum, lateral ventricle, and cortical subplate. By E19, the density of staining had increased and was widespread throughout the expanding CNS. In the rostral domains, moderate to high density was found in the external plexiform layer; the medial preoptic area and nucleus; the ventromedial, dorsomedial, and arcuate hypothalamic nuclei; and the lateral and posterior hypothalamic areas. In the midbrain, similar levels of expression were noted in the central nucleus of raphe; the deep mesencephalic, dorsal raphe, and laterodorsal tegmental nuclei; and the ventral periaqueductal gray. In the pons, positive loci included the locus coeruleus and the gigantocellular and pontine reticular nuclei. In the medulla, high expression was evident in the gigantocellular, intermediate, magnocellular, and medullary reticular, spinal trigeminal and cuneate nuclei; and the nucleus tractus solitarius. In the spinal cord, moderate to high density of staining was found in the ventral, dorsal, and lateral horns, and in the ventral, dorsal, and cuneate funiculi. On the other hand, transitory expression was found in the diagonal band, bed nucleus of the stria terminalis, amygdala, and the lateral mamillary and anterobasal nuclei of the hypothalamus. These studies indicate a role for CTR in the activation of some premigratory neuroblasts in the CNS as well as a possible role later in an undefined function associated with mature neurons of particular nuclei.


Asunto(s)
Sistema Nervioso Central/embriología , Sistema Nervioso Central/crecimiento & desarrollo , Receptores de Calcitonina/metabolismo , Animales , Inmunohistoquímica , Hibridación in Situ , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Calcitonina/genética
17.
ScientificWorldJournal ; 3: 163-75, 2003 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-12806128

RESUMEN

Amylin (islet amyloid polypeptide) is a peptide synthesized principally in the beta-cells of the pancreatic islets together with insulin and has actions as a hormone, growth factor, and modifier of behavior. As a hormone, amylin acts to modify gastric motility, renal resorption, and has metabolic actions. It is postulated that the principal function of amylin as a hormone is the activation of physiological processes associated with feeding. As a growth factor, amylin acts on bone cells, renal proximal tubular cells, and islet beta-cells. Amylin has important targets in the brain that mediate its actions in the modification of behavior, including thirst and satiety. In man, amylin can form islet amyloid deposits, an event linked to the reduction of b-cell mass and loss of signal-secretion coupling. Recent evidence has defined a new role for monomeric amylin as a growth factor and regulator of beta-cell mass that is postulated to be a key factor in pathophysiological processes that result in overt diabetes.


Asunto(s)
Amiloide/fisiología , Animales , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos
18.
Mol Cell Endocrinol ; 394(1-2): 13-20, 2014 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-24998520

RESUMEN

BACKGROUND: Inflammation is believed to link obesity to insulin resistance, as in the setting of metabolic syndrome (MetS). Osteoprotegerin (OPG) is a soluble protein that seems to exert proatherogenic and prodiabetogenic effects. This study aims at determining OPG levels in MetS and whether OPG might contribute to MetS development and progression. METHODOLOGY/PRINCIPAL FINDINGS: Circulating OPG was measured in 46 patients with MetS and 63 controls, and was found significantly elevated in those with MetS. In addition, circulating and tissue OPG was significantly increased in high-fat diet (HFD) fed C57BL6 mice, which is one of the animal models for the study of MetS. To evaluate the consequences of OPG elevation, we delivered this protein to C57BL6 mice, finding that it promoted systemic and adipose tissue proinflammatory changes in association with metabolic abnormalities. CONCLUSIONS/SIGNIFICANCE: These data suggest that OPG may trigger adipose tissue proinflammatory changes in MetS/HFD-induced obesity.


Asunto(s)
Tejido Adiposo/metabolismo , Síndrome Metabólico/sangre , Obesidad/sangre , Osteoprotegerina/sangre , Tejido Adiposo/patología , Adulto , Animales , Glucemia/metabolismo , Índice de Masa Corporal , Proteína C-Reactiva/metabolismo , Estudios de Casos y Controles , HDL-Colesterol/sangre , LDL-Colesterol/sangre , Dieta Alta en Grasa/efectos adversos , Femenino , Humanos , Inflamación/sangre , Inflamación/etiología , Inflamación/patología , Insulina/sangre , Resistencia a la Insulina , Masculino , Síndrome Metabólico/etiología , Síndrome Metabólico/patología , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Obesidad/etiología , Obesidad/patología , Osteoprotegerina/administración & dosificación , Triglicéridos/sangre
19.
Diabetes ; 61(8): 2105-13, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22698914

RESUMEN

Advanced glycation end products (AGEs) are important mediators of diabetic nephropathy that act through the receptor for AGEs (RAGE), as well as other mechanisms, to promote renal inflammation and glomerulosclerosis. The relative contribution of RAGE-dependent and RAGE-independent signaling pathways has not been previously studied in vivo. In this study, diabetic RAGE apoE double-knockout (KO) mice with streptozotocin-induced diabetes were treated with the AGE inhibitor, alagebrium (1 mg/kg/day), or the ACE inhibitor, quinapril (30 mg/kg/day), for 20 weeks, and renal parameters were assessed. RAGE deletion attenuated mesangial expansion, glomerular matrix accumulation, and renal oxidative stress associated with 20 weeks of diabetes. By contrast, inflammation and AGE accumulation associated with diabetes was not prevented. However, treatment with alagebrium in diabetic RAGE apoE KO mice reduced renal AGE levels and further reduced glomerular matrix accumulation. In addition, even in the absence of RAGE expression, alagebrium attenuated cortical inflammation, as denoted by the reduced expression of monocyte chemoattractant protein-1, intracellular adhesion molecule-1, and the macrophage marker cluster of differentiation molecule 11b. These novel findings confirm the presence of important RAGE-independent as well as RAGE-dependent signaling pathways that may be activated in the kidney by AGEs. This has important implications for the design of optimal therapeutic strategies for the prevention of diabetic nephropathy.


Asunto(s)
Apolipoproteínas E/genética , Productos Finales de Glicación Avanzada/metabolismo , Receptores Inmunológicos/metabolismo , Tiazoles/farmacología , Albuminuria , Animales , Diabetes Mellitus Experimental , Fibrosis/etiología , Mesangio Glomerular/efectos de los fármacos , Glomerulonefritis/etiología , Masculino , Ratones , Ratones Noqueados , Nefritis/etiología , Estrés Oxidativo/fisiología , Quinapril , Receptor para Productos Finales de Glicación Avanzada , Tetrahidroisoquinolinas/farmacología
20.
Diabetes ; 58(5): 1229-36, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19208907

RESUMEN

OBJECTIVE: Results from the Diabetes Control Complications Trial (DCCT) and the subsequent Epidemiology of Diabetes Interventions and Complications (EDIC) Study and more recently from the U.K. Prospective Diabetes Study (UKPDS) have revealed that the deleterious end-organ effects that occurred in both conventional and more aggressively treated subjects continued to operate >5 years after the patients had returned to usual glycemic control and is interpreted as a legacy of past glycemia known as "hyperglycemic memory." We have hypothesized that transient hyperglycemia mediates persistent gene-activating events attributed to changes in epigenetic information. RESEARCH DESIGN AND METHODS: Models of transient hyperglycemia were used to link NFkappaB-p65 gene expression with H3K4 and H3K9 modifications mediated by the histone methyltransferases (Set7 and SuV39h1) and the lysine-specific demethylase (LSD1) by the immunopurification of soluble NFkappaB-p65 chromatin. RESULTS: The sustained upregulation of the NFkappaB-p65 gene as a result of ambient or prior hyperglycemia was associated with increased H3K4m1 but not H3K4m2 or H3K4m3. Furthermore, glucose was shown to have other epigenetic effects, including the suppression of H3K9m2 and H3K9m3 methylation on the p65 promoter. Finally, there was increased recruitment of the recently identified histone demethylase LSD1 to the p65 promoter as a result of prior hyperglycemia. CONCLUSIONS: These studies indicate that the active transcriptional state of the NFkappaB-p65 gene is linked with persisting epigenetic marks such as enhanced H3K4 and reduced H3K9 methylation, which appear to occur as a result of effects of the methyl-writing and methyl-erasing histone enzymes.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Hiperglucemia/enzimología , Hiperglucemia/genética , Lisina/metabolismo , Animales , Bovinos , Metilación de ADN , Cartilla de ADN , ADN Complementario/genética , Angiopatías Diabéticas/genética , Endotelio Vascular/fisiología , Predisposición Genética a la Enfermedad , Histona Metiltransferasas , Humanos , Hiperglucemia/complicaciones , Hiperglucemia/patología , ARN/genética , ARN/aislamiento & purificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA