Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 295(36): 12605-12617, 2020 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-32647007

RESUMEN

In the heart, the serine carboxypeptidase cathepsin A (CatA) is distributed between lysosomes and the extracellular matrix (ECM). CatA-mediated degradation of extracellular peptides may contribute to ECM remodeling and left ventricular (LV) dysfunction. Here, we aimed to evaluate the effects of CatA overexpression on LV remodeling. A proteomic analysis of the secretome of adult mouse cardiac fibroblasts upon digestion by CatA identified the extracellular antioxidant enzyme superoxide dismutase (EC-SOD) as a novel substrate of CatA, which decreased EC-SOD abundance 5-fold. In vitro, both cardiomyocytes and cardiac fibroblasts expressed and secreted CatA protein, and only cardiac fibroblasts expressed and secreted EC-SOD protein. Cardiomyocyte-specific CatA overexpression and increased CatA activity in the LV of transgenic mice (CatA-TG) reduced EC-SOD protein levels by 43%. Loss of EC-SOD-mediated antioxidative activity resulted in significant accumulation of superoxide radicals (WT, 4.54 µmol/mg tissue/min; CatA-TG, 8.62 µmol/mg tissue/min), increased inflammation, myocyte hypertrophy (WT, 19.8 µm; CatA-TG, 21.9 µm), cellular apoptosis, and elevated mRNA expression of hypertrophy-related and profibrotic marker genes, without affecting intracellular detoxifying proteins. In CatA-TG mice, LV interstitial fibrosis formation was enhanced by 19%, and the type I/type III collagen ratio was shifted toward higher abundance of collagen I fibers. Cardiac remodeling in CatA-TG was accompanied by an increased LV weight/body weight ratio and LV end diastolic volume (WT, 50.8 µl; CatA-TG, 61.9 µl). In conclusion, CatA-mediated EC-SOD reduction in the heart contributes to increased oxidative stress, myocyte hypertrophy, ECM remodeling, and inflammation, implicating CatA as a potential therapeutic target to prevent ventricular remodeling.


Asunto(s)
Catepsina A/metabolismo , Miocitos Cardíacos/metabolismo , Proteolisis , Superóxido Dismutasa/metabolismo , Remodelación Ventricular , Animales , Catepsina A/genética , Masculino , Ratones , Ratones Transgénicos , Miocitos Cardíacos/patología , Superóxido Dismutasa/genética
2.
J Pharmacol Exp Ther ; 376(2): 190-203, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33203659

RESUMEN

As a gut-restricted, nonabsorbed therapy, polymeric bile acid sequestrants (BAS) play an important role in managing hyperlipidemia and hyperglycemia. Similarly, nonabsorbable sequestrants of dietary phosphate have been used for the management of hyperphosphatemia in end-stage renal disease. To evaluate the potential utility of such polymer sequestrants to treat type 2 diabetes (T2D) and its associated renal and cardiovascular complications, we synthesized a novel polymeric sequestrant, SAR442357, possessing optimized bile acid (BA) and phosphate sequestration characteristics. Long-term treatment of T2D obese cZucker fatty/Spontaneously hypertensive heart failure F1 hybrid (ZSF1) with SAR442357 resulted in enhanced sequestration of BAs and phosphate in the gut, improved glycemic control, lowering of serum cholesterol, and attenuation of diabetic kidney disease (DKD) progression. In comparison, colesevelam, a BAS with poor phosphate binding properties, did not prevent DKD progression, whereas losartan, an angiotensin II receptor blocker that is widely used to treat DKD, showed no effect on hyperglycemia. Analysis of hepatic gene expression levels of the animals treated with SAR442357 revealed upregulation of genes responsible for the biosynthesis of cholesterol and BAs, providing clear evidence of target engagement and mode of action of the new sequestrant. Additional hepatic gene expression pathway changes were indicative of an interruption of the enterohepatic BA cycle. Histopathological analysis of ZSF1 rat kidneys treated with SAR442357 further supported its nephroprotective properties. Collectively, these findings reveal the pharmacological benefit of simultaneous sequestration of BAs and phosphate in treating T2D and its associated comorbidities and cardiovascular complications. SIGNIFICANCE STATEMENT: A new nonabsorbed polymeric sequestrant with optimum phosphate and bile salt sequestration properties was developed as a treatment option for DKD. The new polymeric sequestrant offered combined pharmacological benefits including glucose regulation, lipid lowering, and attenuation of DKD progression in a single therapeutic agent.


Asunto(s)
Antihipertensivos/uso terapéutico , Ácidos y Sales Biliares/metabolismo , Nefropatías Diabéticas/tratamiento farmacológico , Hidrogeles/uso terapéutico , Hipertensión/tratamiento farmacológico , Hipoglucemiantes/uso terapéutico , Animales , Antihipertensivos/síntesis química , Colesterol/metabolismo , Hidrogeles/síntesis química , Hipoglucemiantes/síntesis química , Hígado/metabolismo , Fosfatos/metabolismo , Poliaminas/química , Ratas , Ratas Zucker
3.
J Transl Med ; 14(1): 153, 2016 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-27246731

RESUMEN

BACKGROUND: Myocardial infarction (MI) is a major cause of heart failure. The carboxypeptidase cathepsin A is a novel target in the treatment of cardiac failure. We aim to show that recently developed inhibitors of the protease cathepsin A attenuate post-MI heart failure. METHODS: Mice were subjected to permanent left anterior descending artery (LAD) ligation or sham operation. 24 h post-surgery, LAD-ligated animals were treated with daily doses of the cathepsin A inhibitor SAR1 or placebo. After 4 weeks, the three groups (sham, MI-placebo, MI-SAR1) were evaluated. RESULTS: Compared to sham-operated animals, placebo-treated mice showed significantly impaired cardiac function and increased plasma BNP levels. Cathepsin A inhibition prevented the increase of plasma BNP levels and displayed a trend towards improved cardiac functionality. Proteomic profiling was performed for the three groups (sham, MI-placebo, MI-SAR1). More than 100 proteins were significantly altered in placebo-treated LAD ligation compared to the sham operation, including known markers of cardiac failure as well as extracellular/matricellular proteins. This ensemble constitutes a proteome fingerprint of myocardial infarction induced by LAD ligation in mice. Cathepsin A inhibitor treatment normalized the marked increase of the muscle stress marker CA3 as well as of Igγ 2b and fatty acid synthase. For numerous further proteins, cathepsin A inhibition partially dampened the LAD ligation-induced proteome alterations. CONCLUSIONS: Our proteomic and functional data suggest that cathepsin A inhibition has cardioprotective properties and support a beneficial effect of cathepsin A inhibition in the treatment of heart failure after myocardial infarction.


Asunto(s)
Catepsina A/antagonistas & inhibidores , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/etiología , Infarto del Miocardio/complicaciones , Infarto del Miocardio/tratamiento farmacológico , Inhibidores de Proteasas/uso terapéutico , Proteómica/métodos , Animales , Catepsina A/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/patología , Ligadura , Masculino , Ratones Endogámicos C57BL , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Tamaño de los Órganos/efectos de los fármacos , Mapeo Peptídico , Inhibidores de Proteasas/farmacología , Proteoma/metabolismo , Ratas
4.
J Transl Med ; 11: 84, 2013 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-23537041

RESUMEN

BACKGROUND: Lixisenatide is a glucagon-like peptide-1 analog which stimulates insulin secretion and inhibits glucagon secretion and gastric emptying. We investigated cardioprotective effects of lixisenatide in rodent models reflecting the clinical situation. METHODS: The acute cardiac effects of lixisenatide were investigated in isolated rat hearts subjected to brief ischemia and reperfusion. Effects of chronic treatment with lixisenatide on cardiac function were assessed in a modified rat heart failure model after only transient coronary occlusion followed by long-term reperfusion. Freshly isolated cardiomyocytes were used to investigate cell-type specific mechanisms of lixisenatide action. RESULTS: In the acute setting of ischemia-reperfusion, lixisenatide reduced the infarct-size/area at risk by 36% ratio without changes on coronary flow, left-ventricular pressure and heart rate. Treatment with lixisenatide for 10 weeks, starting after cardiac ischemia and reperfusion, improved left ventricular end-diastolic pressure and relaxation time and prevented lung congestion in comparison to placebo. No anti-fibrotic effect was observed. Gene expression analysis revealed a change in remodeling genes comparable to the ACE inhibitor ramipril. In isolated cardiomyocytes lixisenatide reduced apoptosis and increased fractional shortening. Glucagon-like peptide-1 receptor (GLP1R) mRNA expression could not be detected in rat heart samples or isolated cardiomyocytes. Surprisingly, cardiomyocytes isolated from GLP-1 receptor knockout mice still responded to lixisenatide. CONCLUSIONS: In rodent models, lixisenatide reduced in an acute setting infarct-size and improved cardiac function when administered long-term after ischemia-reperfusion injury. GLP-1 receptor independent mechanisms contribute to the described cardioprotective effect of lixisenatide. Based in part on these preclinical findings patients with cardiac dysfunction are currently being recruited for a randomized, double-blind, placebo-controlled, multicenter study with lixisenatide. TRIAL REGISTRATION: (ELIXA, ClinicalTrials.gov Identifier: NCT01147250).


Asunto(s)
Cardiotónicos/farmacología , Daño por Reperfusión Miocárdica/metabolismo , Péptidos/farmacología , Androstadienos/farmacología , Animales , Modelos Animales de Enfermedad , Receptor del Péptido 1 Similar al Glucagón , Insuficiencia Cardíaca/tratamiento farmacológico , Masculino , Ratones , Ratones Noqueados , Contracción Miocárdica/efectos de los fármacos , Miocardio/patología , Miocitos Cardíacos/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Receptores de Glucagón/metabolismo , Daño por Reperfusión/metabolismo , Transducción de Señal , Wortmanina
5.
J Transl Med ; 10: 187, 2012 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-22963383

RESUMEN

BACKGROUND: The additive effects of obesity and metabolic syndrome on left ventricular (LV) maladaptive remodeling and function in hypertension are not characterized. METHODS: We compared an obese spontaneously hypertensive rat model (SHR-ob) with lean spontaneously hypertensive rats (SHR-lean) and normotensive controls (Ctr). LV-function was investigated by cardiac magnetic resonance imaging and invasive LV-pressure measurements. LV-interstitial fibrosis was quantified and protein levels of phospholamban (PLB), Serca2a and glucose transporters (GLUT1 and GLUT4) were determined by immunohistochemistry. RESULTS: Systolic blood pressure was similar in SHR-lean and SHR-ob (252 ± 7 vs. 242 ± 7 mmHg, p = 0.398) but was higher when compared to Ctr (155 ± 2 mmHg, p < 0.01 for both). Compared to SHR-lean and Ctr, SHR-ob showed impaired glucose tolerance and increased body-weight. In SHR-ob, LV-ejection fraction was impaired vs. Ctr (46.2 ± 1.1 vs. 59.6 ± 1.9%, p = 0.007). LV-enddiastolic pressure was more increased in SHR-ob than in SHR-lean (21.5 ± 4.1 vs. 5.9 ± 0.81 mmHg, p = 0.0002) when compared to Ctr (4.3 ± 1.1 mmHg, p < 0.0001 for both), respectively. Increased LV-fibrosis together with increased myocyte diameters and ANF gene expression in SHR-ob were associated with increased GLUT1-protein levels in SHR-ob suggestive for an upregulation of the GLUT1/ANF-axis. Serca2a-protein levels were decreased in SHR-lean but not altered in SHR-ob compared to Ctr. PLB-phosphorylation was not altered. CONCLUSION: In addition to hypertension alone, metabolic syndrome and obesity adds to the myocardial phenotype by aggravating diastolic dysfunction and a progression towards systolic dysfunction. SHR-ob may be a useful model to develop new interventional and pharmacological treatment strategies for hypertensive heart disease and metabolic disorders.


Asunto(s)
Obesidad/complicaciones , Disfunción Ventricular Izquierda/complicaciones , Remodelación Ventricular , Animales , Presión Sanguínea , Calcio/metabolismo , Perfilación de la Expresión Génica , Hemodinámica , Imagen por Resonancia Magnética , Masculino , Obesidad/fisiopatología , Ratas , Ratas Endogámicas SHR , Disfunción Ventricular Izquierda/genética , Disfunción Ventricular Izquierda/fisiopatología , Remodelación Ventricular/genética
6.
Cell Metab ; 34(1): 59-74.e10, 2022 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-34932984

RESUMEN

Unimolecular triple incretins, combining the activity of glucagon-like peptide-1 (GLP-1), glucose-dependent insulinotropic polypeptide (GIP), and glucagon (GCG), have demonstrated reduction in body weight and improved glucose control in rodent models. We developed SAR441255, a synthetic peptide agonist of the GLP-1, GCG, and GIP receptors, structurally based on the exendin-4 sequence. SAR441255 displays high potency with balanced activation of all three target receptors. In animal models, metabolic outcomes were superior to results with a dual GLP-1/GCG receptor agonist. Preclinical in vivo positron emission tomography imaging demonstrated SAR441255 binding to GLP-1 and GCG receptors. In healthy subjects, SAR441255 improved glycemic control during a mixed-meal tolerance test and impacted biomarkers for GCG and GIP receptor activation. Single doses of SAR441255 were well tolerated. The results demonstrate that integrating GIP activity into dual GLP-1 and GCG receptor agonism provides improved effects on weight loss and glycemic control while buffering the diabetogenic risk of chronic GCG receptor agonism.


Asunto(s)
Receptor del Péptido 1 Similar al Glucagón , Control Glucémico , Incretinas , Receptores de la Hormona Gastrointestinal , Receptores de Glucagón , Pérdida de Peso , Animales , Polipéptido Inhibidor Gástrico , Péptido 1 Similar al Glucagón , Receptor del Péptido 1 Similar al Glucagón/agonistas , Humanos , Incretinas/farmacología , Péptidos/farmacología , Receptores de la Hormona Gastrointestinal/agonistas , Receptores de Glucagón/agonistas , Pérdida de Peso/efectos de los fármacos
7.
Cytokine ; 56(3): 739-48, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22004922

RESUMEN

Parthenolide, a sesquiterpene lactone, has been reported to exhibit a variety of anti-inflammatory and immunomodulatory effects. To test the effect of parthenolide on brain inflammatory responses, brain oxidative stress and fever, we treated rats with parthenolide (1 mg/kg), simultaneously or 1 h prior to a systemic (i.p.) challenge with a moderate dose (100 µg/kg) of lipopolysaccharide (LPS). The initial hypothermia was exaggerated; the second phase of the biphasic LPS-induced fever and circulating interleukin-6 (IL-6) and tumor necrosis factor α (TNFα) were significantly attenuated only in parthenolide-pretreated animals. In the hypothalamus, markers of NFκB/NF-IL6 pathway activation (inhibitor κBα, NF-IL6 and the serin/threonin kinase-like protein mRNA expression) and markers of oxidative stress (including nuclear respiratory factor 1) and NFκB immunoreactivity were significantly reduced while NF-IL6 immunoreactivity and suppressor of cytokine signaling 3 mRNA expression remained unaltered, 8 h after LPS-stimulation with parthenolide-pretreatment. Importantly, this response was accompanied by decreased mRNA expression of the rate limiting enzyme in prostaglandin synthesis, cyclooxygenase 2 (COX2), known for its critical role in fever induction pathways. A direct action of parthenolide on brain cells was also confirmed in a primary neuro-glial cell culture of the vascular organ of the lamina terminalis a pivotal brain structure for fever manifestation with a leaky blood-brain barrier. In summary, pretreatment with parthenolide attenuates the febrile response during LPS-induced systemic inflammation by reducing circulating IL-6 and TNFα and decreasing hypothalamic NFκB/NF-IL6 activation, oxidative stress and expression of COX2. Thus parthenolide appears to have the potential to reduce brain inflammation.


Asunto(s)
Citocinas/sangre , Encefalitis/sangre , Encefalitis/tratamiento farmacológico , Fiebre/sangre , Fiebre/tratamiento farmacológico , Sesquiterpenos/uso terapéutico , Animales , Biomarcadores/sangre , Temperatura Corporal/efectos de los fármacos , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Células Cultivadas , Encefalitis/complicaciones , Encefalitis/patología , Fiebre/inducido químicamente , Fiebre/complicaciones , Regulación de la Expresión Génica/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Hipotálamo/patología , Inyecciones Intraperitoneales , Interleucina-6/sangre , Lipopolisacáridos/administración & dosificación , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , FN-kappa B/metabolismo , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Sesquiterpenos/farmacología , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/sangre
8.
JACC Basic Transl Sci ; 4(3): 332-344, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31312757

RESUMEN

After myocardial infarction, remote ventricular remodeling and atrial cardiomyopathy progress despite successful revascularization. In a rat model of ventricular ischemia/reperfusion, pharmacological inhibition of the protease activity of cathepsin A initiated at the time point of reperfusion prevented extracellular matrix remodeling in the atrium and the ventricle remote from the infarcted area. This scenario was associated with preservation of more viable ventricular myocardium and the prevention of an arrhythmogenic and functional substrate for atrial fibrillation. Remote ventricular extracellular matrix remodeling and atrial cardiomyopathy may represent a promising target for pharmacological atrial fibrillation upstream therapy following myocardial infarction.

9.
Sci Rep ; 9(1): 16161, 2019 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-31695063

RESUMEN

The central mechanisms underlying the marked beneficial metabolic effects of bariatric surgery are unclear. Here, we characterized global gene expression in the hypothalamic arcuate nucleus (Arc) in diet-induced obese (DIO) rats following Roux-en-Y gastric bypass (RYGB). 60 days post-RYGB, the Arc was isolated by laser-capture microdissection and global gene expression was assessed by RNA sequencing. RYGB lowered body weight and adiposity as compared to sham-operated DIO rats. Discrete transcriptome changes were observed in the Arc following RYGB, including differential expression of genes associated with inflammation and neuropeptide signaling. RYGB reduced gene expression of glial cell markers, including Gfap, Aif1 and Timp1, confirmed by a lower number of GFAP immunopositive astrocyte profiles in the Arc. Sham-operated weight-matched rats demonstrated a similar glial gene expression signature, suggesting that RYGB and dietary restriction have common effects on hypothalamic gliosis. Considering that RYGB surgery also led to increased orexigenic and decreased anorexigenic gene expression, this may signify increased hunger-associated signaling at the level of the Arc. Hence, induction of counterregulatory molecular mechanisms downstream from the Arc may play an important role in RYGB-induced weight loss.


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Dieta Reductora , Derivación Gástrica , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Gliosis/genética , Adiposidad , Animales , Astrocitos/metabolismo , Biomarcadores , Dieta Alta en Grasa , Ingestión de Alimentos , Proteína Ácida Fibrilar de la Glía/análisis , Péptido 1 Similar al Glucagón/sangre , Inflamación/genética , Captura por Microdisección con Láser , Masculino , Neuropéptidos/biosíntesis , Neuropéptidos/genética , Obesidad/etiología , Obesidad/cirugía , Péptido YY/sangre , Ratas , Ratas Sprague-Dawley , Análisis de Secuencia de ARN , Pérdida de Peso
10.
Peptides ; 118: 170100, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31212005

RESUMEN

OBJECTIVE: Roux-en-Y gastric bypass (RYGB) leads to rapid remission of type 2 diabetes (T2D) and sustained body weight loss, but the underlying molecular mechanisms are still not fully understood. To further elucidate these mechanisms and identify potentially novel preprohormone encoding genes with anti-diabetic and/or anti-obesity properties, we performed a comprehensive analysis of gene expression changes in enteroendocrine cells after RYGB in diet-induced obese (DIO) rats. METHODS: The mRNA expression profiles of enteroendocrine cell enriched samples were characterized at 9, 22 and 60 days after RYGB surgery in a DIO rat model. Enteroendocrine cells were identified by chromogranin A immunohistochemistry and isolated by laser capture microdissection (LCM) from five regions covering the full rostro-caudal extension of the gastrointestinal (GI) tract. RNA sequencing and bioinformatic analyses were subsequently applied to identify differentially expressed preprohormone encoding genes. RESULTS: From the analysis of enteroendocrine cell mRNA expression profiles, a total of 54 preprohormones encoding genes were found to be differentially regulated at one or more time-points following RYGB. These included well-known RYGB associated preprohormone genes (e.g. Gcg, Cck, Gip, Pyy and Sct) and less characterized genes with putative metabolic effects (e.g. Nmu, Guca2a, Guca2b, Npw and Adm), but also 16 predicted novel preprohormone genes. Among the list of gene transcripts, Npw, Apln and Fam3d were further validated using in situ mRNA hybridization and corresponding peptides were characterized for acute effects on food intake and glucose tolerance in mice. CONCLUSION: We present a comprehensive mRNA expression profile of chromogranin A positive enteroendocrine cells following RYGB in rats. The data provides a region-specific characterization of all regulated preprohormone encoding genes in the rat GI tract including 16 not hitherto known. The comprehensive catalogue of preprohormone expression changes may support our understanding of hormone mediated effects of RYGB on diabetes remission and body weight reduction.


Asunto(s)
Células Enteroendocrinas/metabolismo , Derivación Gástrica , Obesidad/genética , Obesidad/metabolismo , Hormonas Peptídicas/genética , Hormonas Peptídicas/metabolismo , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Animales , Colecistoquinina/genética , Colecistoquinina/metabolismo , Biología Computacional , Polipéptido Inhibidor Gástrico/genética , Polipéptido Inhibidor Gástrico/metabolismo , Inmunohistoquímica , Hibridación in Situ , Captura por Microdisección con Láser , Masculino , Ratones , Obesidad/cirugía , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Análisis de Secuencia de ARN , Somatostatina/genética , Somatostatina/metabolismo , Transcriptoma/genética
11.
J Pharmacol Exp Ther ; 325(2): 370-9, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18252813

RESUMEN

Many cardiovascular diseases are associated with reduced levels of bioactive nitric oxide (NO) and an uncoupling of oxygen reduction from NO synthesis in endothelial NO synthase (eNOS uncoupling). In human endothelial EA.hy 926 cells, two small-molecular-weight compounds with related structures, 4-fluoro-N-indan-2-yl-benzamide (CAS no. 291756-32-6; empirical formula C16H14FNO; AVE9488) and 2,2-difluoro-benzo[1,3]dioxole-5-carboxylic acid indan-2-ylamide (CAS no. 450348-85-3; empirical formula C17H13F2NO3; AVE3085), enhanced eNOS promoter activity in a concentration-dependent manner; with the responsible cis-element localized within the proximal 263 base pairs of the promoter region. RNA interference-mediated knockdown of the transcription factor Sp1 significantly reduced the basal activity of eNOS promoter, but it did not prevent the transcription activation by the compounds. Enhanced transcription of eNOS by AVE9488 in primary human umbilical vein endothelial cells was associated with increased levels of eNOS mRNA and protein expression, as well as increased bradykinin-stimulated NO production. In both wild-type C57BL/6J mice and apolipoprotein E-knockout (apoE-KO) mice, treatment with AVE9488 resulted in enhanced vascular eNOS expression. In apoE-KO mice, but not in eNOS-knockout mice, treatment with AVE9488 reduced cuff-induced neointima formation. A 12-week treatment with AVE9488 or AVE3085 reduced atherosclerotic plaque formation in apoE-KO mice, but not in apoE/eNOS-double knockout mice. Aortas from apoE-KO mice showed a significant generation of reactive oxygen species. This was partly prevented by nitric-oxide inhibitor N(omega)-nitro-l-arginine methyl ester, indicating eNOS uncoupling. Treatment of mice with AVE9488 enhanced vascular content of the essential eNOS cofactor (6R)-5,6,7,8-tetrahydro-l-biopterin and reversed eNOS uncoupling. The combination of an up-regulated eNOS expression and a reversal of eNOS uncoupling is probably responsible for the observed vasoprotective properties of this new type of compounds.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Benzamidas/uso terapéutico , Benzodioxoles/uso terapéutico , Indanos/uso terapéutico , Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Sustancias Protectoras/uso terapéutico , Animales , Aorta/efectos de los fármacos , Aorta/metabolismo , Aorta/patología , Apolipoproteínas E/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Línea Celular , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Peso Molecular , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo III/genética , ARN Mensajero/metabolismo
12.
Brain Res ; 1205: 36-46, 2008 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-18353287

RESUMEN

Macrophage-activating lipopeptide-2 (MALP-2) has been identified as the pathogen-associated molecular pattern of Mycoplasma fermentans, which causes stimulation of the innate immune system through the activation of the heterodimeric Toll-like receptors (TLRs) 2 and 6. Based on the reported protective effects of MALP-2 on healing of skin wounds, the central goal of this study was to evaluate the capacity of MALP-2 to induce a localized inflammatory response in an established model of a subcutaneous air pouch. Injections of MALP-2 into the pouch caused fever and some components of sickness behavior in rats. At the subcutaneous site of localized inflammation, a massive formation of tumor necrosis factor-alpha (TNF), interleukin-6 (IL-6), and prostaglandin E2 (PGE2) could be demonstrated in response to injections of MALP-2. Moderate amounts of IL-6 and PGE2 seemed to enter the systemic circulation of MALP-2-treated rats. The IL-6, which appeared in the blood after injection of MALP-2 into the air pouch was sufficient to cause a direct activation of brain cells in areas which lack a complete blood-brain barrier, namely in the sensory circumventricular organs (sCVOs), the organum vasculosum laminae terminalis (OVLT), the subfornical organ (SFO), and the area postrema (AP). The stimulation of cells at these brain sites was revealed by demonstration of a nuclear translocation of the transcription factor STAT3 (signal transducer and activator of transcription 3). Corresponding to the circulating levels of IL-6, the nuclear STAT3 activation of cells within the sCVOs was much less pronounced after local subcutaneous when compared to systemic treatment with MALP-2. In conclusion, cells within the subcutaneous compartment are activated by the TLR2/6 agonist MALP-2. Fever and sickness behavior induced by injection of MALP-2 into subcutaneous tissue may, in part, be mediated by a spillover of IL-6 from the subcutaneous site of inflammation into the blood to cause activation of brain sites which are implicated in the manifestation of these illness responses.


Asunto(s)
Encéfalo/patología , Encéfalo/fisiopatología , Encefalitis/patología , Fiebre/patología , Fiebre/fisiopatología , Oligopéptidos/farmacología , Animales , Conducta Animal , Temperatura Corporal/fisiología , Peso Corporal/fisiología , Citocinas/sangre , Citocinas/metabolismo , Dinoprostona/sangre , Dinoprostona/metabolismo , Ingestión de Líquidos/fisiología , Ingestión de Alimentos/fisiología , Encefalitis/inducido químicamente , Encefalitis/psicología , Fiebre/psicología , Inmunohistoquímica , Interleucina-6/farmacología , Lipopéptidos , Lipopolisacáridos/farmacología , Masculino , Actividad Motora/fisiología , Mycoplasma/química , Ratas , Ratas Wistar , Factor de Transcripción STAT3/metabolismo , Receptor Toll-Like 2/agonistas , Receptor Toll-Like 4/agonistas , Receptor Toll-Like 6/agonistas
13.
Peptides ; 101: 32-43, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29289697

RESUMEN

AIM: To determine whether intestinal expression of guanylate cyclase activator 2A (GUCA2A) and guanylate cyclase activator 2B (GUCA2B) genes is regulated in obese humans following Roux-en-Y gastric bypass (RYGB), and to evaluate the corresponding guanylin (GN) and uroguanylin (UGN) peptides for potentially contributing to the beneficial metabolic effects of RYGB. METHODS: Enteroendocrine cells were harvested peri- and post-RYGB, and GUCA2A/GUCA2B mRNA expression was compared. GN, UGN and their prohormones (proGN, proUGN) were administered subcutaneously in normal-weight mice to evaluate effects on food intake and glucose regulation. The effect of pro-UGN or UGN overexpression, using adeno-associated virus (AAV) vectors, was assessed in diet-induced obese (DIO) mice. Intracerebroventricular administration of GN and UGN was performed in rats for assessment of putative centrally mediated effects on food intake. GN and UGN, as well as their prohormones, were evaluated for effects on glucose-stimulated insulin secretion (GSIS) in rat pancreatic islets and perfused rat pancreas. RESULTS: GUCA2A and GUCA2B mRNA expression was significantly upregulated in enteroendocrine cells after RYGB. Peripheral administration of guanylins or prohormones did not influence food intake, oral glucose tolerance, and GSIS. Central administration of GN and UGN did not affect food intake in rats. Chronic AVV-mediated overexpression of UGN and proUGN had no effect on body weight or glucose homeostasis in DIO mice. CONCLUSION: GN and UGN, as well as their prohormones, do not seem to play a significant role in body weight regulation and glycemic control, suggesting that guanylin-family peptides do not show promise as targets for the treatment of obesity or diabetes.


Asunto(s)
Mantenimiento del Peso Corporal , Células Enteroendocrinas/metabolismo , Derivación Gástrica , Hormonas Gastrointestinales/biosíntesis , Regulación de la Expresión Génica , Péptidos Natriuréticos/biosíntesis , Adulto , Animales , Diabetes Mellitus/metabolismo , Diabetes Mellitus/cirugía , Femenino , Proteínas Activadoras de la Guanilato-Ciclasa/biosíntesis , Humanos , Masculino , Ratones , Persona de Mediana Edad , Obesidad/metabolismo , Obesidad/cirugía
14.
PLoS One ; 13(1): e0189886, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29293525

RESUMEN

Glucagon-like peptide 1 (GLP-1) stimulated insulin secretion has a considerable heritable component as estimated from twin studies, yet few genetic variants influencing this phenotype have been identified. We performed the first genome-wide association study (GWAS) of GLP-1 stimulated insulin secretion in non-diabetic individuals from the Netherlands Twin register (n = 126). This GWAS was enhanced using a tissue-specific protein-protein interaction network approach. We identified a beta-cell protein-protein interaction module that was significantly enriched for low gene scores based on the GWAS P-values and found support at the network level in an independent cohort from Tübingen, Germany (n = 100). Additionally, a polygenic risk score based on SNPs prioritized from the network was associated (P < 0.05) with glucose-stimulated insulin secretion phenotypes in up to 5,318 individuals in MAGIC cohorts. The network contains both known and novel genes in the context of insulin secretion and is enriched for members of the focal adhesion, extracellular-matrix receptor interaction, actin cytoskeleton regulation, Rap1 and PI3K-Akt signaling pathways. Adipose tissue is, like the beta-cell, one of the target tissues of GLP-1 and we thus hypothesized that similar networks might be functional in both tissues. In order to verify peripheral effects of GLP-1 stimulation, we compared the transcriptome profiling of ob/ob mice treated with liraglutide, a clinically used GLP-1 receptor agonist, versus baseline controls. Some of the upstream regulators of differentially expressed genes in the white adipose tissue of ob/ob mice were also detected in the human beta-cell network of genes associated with GLP-1 stimulated insulin secretion. The findings provide biological insight into the mechanisms through which the effects of GLP-1 may be modulated and highlight a potential role of the beta-cell expressed genes RYR2, GDI2, KIAA0232, COL4A1 and COL4A2 in GLP-1 stimulated insulin secretion.


Asunto(s)
Péptido 1 Similar al Glucagón/metabolismo , Insulina/metabolismo , Animales , Humanos , Secreción de Insulina , Ratones
15.
Neurosci Lett ; 417(3): 286-91, 2007 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-17353091

RESUMEN

Leptin binding to its functional receptor stimulates JAK-STAT-signaling pathway, which finally results in activation and nuclear translocation of transcription factors of the signal transducer and activator of transcription (STAT) family, namely of STAT3. Here we report for the first time that systemic treatment with leptin (5 mg/kg; intraperitoneal injection) also increased the number of nuclear STAT5 signals in the hypothalamus. In particular, the entire arcuate nucleus (ARC), the ventral premammilary nucleus (PMV), and the supraoptic nucleus (SO) showed an enhanced nuclear STAT5 translocation in response to leptin when compared to saline, 120 min after the respective injection. Co-localization studies revealed that a high percentage of those STAT5-responsive cells proved to be neurons. In addition, some astrocytes within the ARC showed nuclear STAT5 signals. The functional relevance of leptin-induced nuclear STAT5 activation in hypothalamic cells still has to be determined.


Asunto(s)
Núcleo Celular/metabolismo , Hipotálamo/metabolismo , Leptina/metabolismo , Neuronas/metabolismo , Factor de Transcripción STAT5/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Transporte Activo de Núcleo Celular/fisiología , Animales , Regulación del Apetito/efectos de los fármacos , Regulación del Apetito/fisiología , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Núcleo Arqueado del Hipotálamo/metabolismo , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Peso Corporal/efectos de los fármacos , Peso Corporal/fisiología , Núcleo Celular/efectos de los fármacos , Hipotálamo/anatomía & histología , Hipotálamo/efectos de los fármacos , Inyecciones Intraperitoneales , Leptina/farmacología , Masculino , Neuronas/efectos de los fármacos , Ratas , Ratas Wistar , Factor de Transcripción STAT5/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Núcleo Supraóptico/efectos de los fármacos , Núcleo Supraóptico/metabolismo
16.
Neurosci Lett ; 394(2): 105-10, 2006 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-16289843

RESUMEN

Leptin binding to its functional receptor leads to activation of the JAK-STAT-signaling pathway and especially to the activation of the signal transducer and activator of transcription factor 3 (STAT3). The immunohistochemical detection of nuclear STAT3 translocation is used as a neuroanatomical mapping tool to determine leptin-responsive cells in the rat brain. This study neuroanatomically identifies those brain cell phenotypes showing STAT3 activation after intraperitoneal leptin treatment (5 mg/kg) using immunohistochemical colocalization with neuronal and endothelial cell marker proteins. Leptin treatment induced nuclear STAT3 signals with the strongest response observed 90min after the treatment. The caudobasal hypothalamus showed a particularly pronounced STAT3 response. Leptin-induced nuclear STAT3 signals were additionally determined in the solitary tract nucleus, the choroid plexus and in the brain endothelium. The vast majority of STAT3-responsive cells proved to be neurons located in the caudobasal hypothalamus, however, a marked number of brain endothelial cells distributed throughout the entire brain got activated as well. In conclusion, neurons and non-neuronal brain cells, e.g., endothelial or choroid plexus cells, seem to express functional leptin receptors and might thereby mediate leptin-dependent functions in the rat brain.


Asunto(s)
Encéfalo/citología , Células Endoteliales/metabolismo , Expresión Génica/fisiología , Neuronas/metabolismo , Receptores de Superficie Celular/fisiología , Factor de Transcripción STAT3/metabolismo , Animales , Antígenos/metabolismo , Expresión Génica/efectos de los fármacos , Inmunohistoquímica/métodos , Indoles , Leptina/farmacología , Masculino , Fosfopiruvato Hidratasa/metabolismo , Ratas , Ratas Wistar , Receptores de Superficie Celular/genética , Receptores de Leptina , Factores de Tiempo , Factor de von Willebrand/inmunología
17.
Neurol Clin ; 24(3): 421-39, v, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16877116

RESUMEN

A rise in core temperature during fever usually results from change in the thermocontroller characteristics, resulting in an elevation of the set point of body temperature. Time course and extent of natural fevers are variable, but an upper limit (41 degrees C in humans), at which core temperature is maintained for some time and reduced when the set point of body temperature returns to its normal level, rarely is exceeded. Although any rise in body temperature may result from fever, those rises that are not accompanied by supportive changes in thermoeffector activities are termed hyperthermia.


Asunto(s)
Encéfalo/inmunología , Fiebre/inmunología , Reacción de Fase Aguda/inmunología , Animales , Barrera Hematoencefálica/inmunología , Regulación de la Temperatura Corporal/inmunología , Encéfalo/irrigación sanguínea , Ventrículos Cerebrales/inmunología , Ciclooxigenasa 2/fisiología , Citocinas/sangre , Dinoprostona/fisiología , Modelos Animales de Enfermedad , Endotelio Vascular/inmunología , Humanos , Infecciones/inmunología , Lipopolisacáridos/inmunología , Ratas
18.
Neuropharmacology ; 107: 146-159, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27016016

RESUMEN

During infection-induced inflammation food intake is reduced. Vagal and brainstem pathways are important both in feeding regulation and immune-to-brain communication. Glutamate is released by vagal afferent terminals in the nucleus of the solitary tract and by its neurons projecting to the parabrachial nuclei. We therefore studied the role of brainstem glutamate receptors in spontaneous food intake of healthy animals and during sickness-associated hypophagia after peripheral administration of bacterial lipopolysaccharides or interleukin-1beta. Brainstem group I and II metabotropic, but not ionotropic, glutamate receptor antagonism increased food intake both in saline- and lipopolysaccharide-treated rats. In these animals, expression of the cellular activation marker c-Fos in the lateral parabrachial nuclei and lipopolysaccharide-induced activation of the nucleus of the solitary tract rostral to the area postrema were suppressed. Group I metabotropic glutamate receptors did not colocalize with c-Fos or neurons regulating gastric function in these structures. Group I metabotropic glutamate receptors were, however, found on raphé magnus neurons that were part of the brainstem circuit innervating the stomach and on trigeminal and hypoglossal motor neurons. In conclusion, our findings show that brainstem metabotropic glutamate receptors reduce food intake and activate the lateral parabrachial nuclei as well as the rostral nucleus of the solitary tract after peripheral bacterial lipopolysaccharide administration. They also provide insight into potential group I metabotropic glutamate receptor-dependent brainstem circuits mediating these effects.


Asunto(s)
Tronco Encefálico/metabolismo , Ingestión de Alimentos/fisiología , Conducta de Enfermedad/fisiología , Receptores de Glutamato Metabotrópico/metabolismo , Animales , Peso Corporal/efectos de los fármacos , Peso Corporal/fisiología , Tronco Encefálico/citología , Tronco Encefálico/efectos de los fármacos , Corticosterona/sangre , Modelos Animales de Enfermedad , Ingestión de Alimentos/efectos de los fármacos , Escherichia coli , Conducta de Enfermedad/efectos de los fármacos , Interleucina-1beta/sangre , Lipopolisacáridos , Masculino , Vías Nerviosas/citología , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas Wistar , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Estómago/inervación
19.
J Neurosci ; 24(23): 5370-80, 2004 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-15190110

RESUMEN

Sympathetic premotor neurons directly control sympathetic preganglionic neurons (SPNs) in the intermediolateral cell column (IML) of the thoracic spinal cord, and many of these premotor neurons are localized in the medulla oblongata. The rostral ventrolateral medulla contains premotor neurons controlling the cardiovascular conditions, whereas rostral medullary raphe regions are a candidate source of sympathetic premotor neurons for thermoregulatory functions. Here, we show that these medullary raphe regions contain putative glutamatergic neurons and that these neurons directly control thermoregulatory SPNs. Neurons expressing vesicular glutamate transporter 3 (VGLUT3) were distributed in the rat medullary raphe regions, including the raphe magnus and rostral raphe pallidus nuclei, and mostly lacked serotonin immunoreactivity. These VGLUT3-positive neurons expressed Fos in response to cold exposure or to central administration of prostaglandin E2, a pyrogenic mediator. Transneuronal retrograde labeling after inoculation of pseudorabies virus into the interscapular brown adipose tissue (BAT) or the tail indicated that those VGLUT3-expressing medullary raphe neurons innervated these thermoregulatory effector organs multisynaptically through SPNs of specific thoracic segments, and microinjection of glutamate into the IML of the BAT-controlling segments produced BAT thermogenesis. An anterograde tracing study further showed a direct projection of those VGLUT3-expressing medullary raphe neurons to the dendrites of SPNs. Furthermore, intra-IML application of glutamate receptor antagonists blocked BAT thermogenesis triggered by disinhibition of the medullary raphe regions. The present results suggest that VGLUT3-expressing neurons in the medullary raphe regions constitute excitatory neurons that could be categorized as a novel group of sympathetic premotor neurons for thermoregulatory functions, including fever.


Asunto(s)
Sistemas de Transporte de Aminoácidos Acídicos/metabolismo , Regulación de la Temperatura Corporal , Fiebre/fisiopatología , Proteínas de Transporte de Membrana , Neuronas/metabolismo , Núcleos del Rafe/fisiopatología , Sistema Nervioso Simpático/fisiopatología , Proteínas de Transporte Vesicular , Tejido Adiposo Pardo/fisiopatología , Sistema de Transporte de Aminoácidos X-AG/metabolismo , Animales , Proteínas Portadoras/metabolismo , Dinoprostona/farmacología , Fiebre/metabolismo , Fiebre/virología , Técnica del Anticuerpo Fluorescente , Ácido Glutámico/farmacología , Herpesvirus Suido 1 , Inmunohistoquímica , Masculino , Microinyecciones , Ratas , Ratas Wistar , Virus Sindbis , Sistema Nervioso Simpático/citología , Termogénesis/efectos de los fármacos , Proteína 2 de Transporte Vesicular de Glutamato , Proteínas de Transporte Vesicular de Glutamato
20.
J Comp Neurol ; 491(1): 1-14, 2005 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-16127698

RESUMEN

During systemic inflammation, cytokines are released by immune-competent cells into the circulation, which in turn signal the brain to mediate brain-controlled signs of illness. Cytokine-responsive brain cells can be mapped by histological analysis of cytokine-induced transcription factors or transcription factor-associated molecules revealing different cell phenotypes that respond to activation of the immune system. Critical sites mediating cytokine-dependent immuneffector functions can be divided into two groups, one group of responding cells situated along a tight blood-brain barrier (BBB), and a second cell group in structures with an open BBB, e.g., the sensory circumventricular organs (CVOs). Previous reports from our group suggest that activation of the signal transducer and activator of transcription factor 3 (STAT3) during lipopolysaccharide (LPS)-induced systemic inflammation is mediated by interleukin-6 (IL-6) and occurs in astrocytes of the rat CVOs. Here we show in the guinea pig a time-dependent marked LPS-induced STAT3 activation within astrocytes and endothelial cells of the CVOs, within astrocytes located in brain structures with a functional BBB and within the brain endothelium of the entire brain. In addition, systemic treatment of rats with either rat recombinant IL-6 or LPS induced STAT3 activation in brain endothelial cells in a similar way as observed in the guinea pig brain, stressing the involvement of IL-6 in this phenomenon in a more generalized way. The STAT3-activated brain cells are located in critical target structures mediating cytokine action during LPS-induced inflammation. STAT3-controlled transcriptional activation with yet unknown cell-specific functional consequences seems to be involved in this process.


Asunto(s)
Células Endoteliales/inmunología , Interleucina-6/inmunología , Lipopolisacáridos/inmunología , Sistemas Neurosecretores/inmunología , Factor de Transcripción STAT3/inmunología , Animales , Astrocitos/inmunología , Transporte Biológico/fisiología , Barrera Hematoencefálica/citología , Barrera Hematoencefálica/inmunología , Encéfalo/citología , Encéfalo/inmunología , Núcleo Celular/metabolismo , Ventrículos Cerebrales/inmunología , Endotelio/citología , Endotelio/inmunología , Cobayas , Masculino , Neuroinmunomodulación , Sistemas Neurosecretores/citología , Ratas , Factor de Transcripción STAT3/metabolismo , Distribución Tisular , Translocación Genética/fisiología , Factor de von Willebrand/inmunología , Factor de von Willebrand/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA