Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Eur J Nutr ; 63(3): 751-762, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38157050

RESUMEN

PURPOSE: The liver-expressed antimicrobial peptide 2 (LEAP2) is a newly recognized peptide hormone that acts via the growth hormone secretagogue receptor (GHSR) blunting the effects of ghrelin and displaying ghrelin-independent actions. Since the implications of LEAP2 are beginning to be elucidated, we investigated if plasma LEAP2 concentration varies with feeding status or sex and whether it is associated with glucose metabolism and appetite sensations. METHODS: We performed a single test meal study, in which plasma concentrations of LEAP2, ghrelin, insulin and glucose as well as visual analogue scales for hunger, desire to eat, prospective food consumption, fullness were assessed before and 60 min after breakfast in 44 participants (n = 21 females) with normal weight (NW) or overweight/obesity (OW/OB). RESULTS: Pre-prandial plasma LEAP2 concentration was ~ 1.6-fold higher whereas ghrelin was ~ 2.0-fold lower in individuals with OW/OB (p < 0.001) independently of sex. After adjusting for body mass index (BMI) and sex, pre-prandial plasma LEAP2 concentration displayed a direct relationship with BMI (ß: 0.09; 95%CI: 0.05, 0.13; p < 0.001), fat mass (ß: 0.05; 95%CI: 0.01, 0.09; p = 0.010) and glycemia (ß: 0.24; 95%CI: 0.05, 0.43; p = 0.021), whereas plasma ghrelin concentration displayed an inverse relationship with BMI and fat mass but not with glycemia. Postprandial plasma LEAP2 concentration increased ~ 58% in females with OW/OB (p = 0.045) but not in females with NW or in males. Pre-prandial plasma LEAP2 concentration displayed an inverse relationship with hunger score (ß: - 11.16; 95% CI: - 18.52, - 3.79; p = 0.004), in a BMI-, sex- and ghrelin-independent manner. CONCLUSIONS: LEAP2 emerges as a key hormone implicated in the regulation of metabolism and appetite in humans. TRIAL REGISTRATION: The study was retrospectively registered in clinicaltrials.gov (April 2023). CLINICALTRIALS: gov Identifier: NCT05815641.


Asunto(s)
Ghrelina , Hambre , Masculino , Femenino , Humanos , Hambre/fisiología , Hepcidinas , Apetito , Obesidad , Sensación
2.
Colloids Surf B Biointerfaces ; 230: 113508, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37562121

RESUMEN

Stimuli-responsive nanocarriers are being widely applied in the development of new strategies for the diagnosis and treatment of diseases. An inherent difficulty in general drug therapy is the lack of precision with respect to a specific pathological site, which can lead to toxicity, excessive drug consumption, or premature degradation. In this work, the controlled drug delivery is achieved by using magnetite nanoparticles coated with mesoporous silica with core-shell structure (MMS) and grafted with the thermoresponsive polymer poly [N-isopropylacrylamide-co-3-(trimethoxysilyl)propyl methacrylate] (MMS-P). The efficiency of MMS-P as a temperature-controlled drug delivery system was evaluated by in vitro release experiments using ibuprofen (IBU) in various mammalian cell models. Further, the effects of IBU as a photoprotectant in cells exposed to photodynamic therapy (PDT) in a carbaryl-induced neurodegenerative model were evaluated. The results showed that MMS-P nanocarriers do not exhibit cytotoxicity in HepG2 cells at high doses such as 7600 µg mL-1. Pre-incubation of MMS-P charged with IBU showed no effect on the PDT in N2A cells; however, it produced a further decrease in the viability of HepG2 cells, leading to a reduction to PDT resistance. On the other hand, a cytoprotective effect against carbaryl toxicity in N2A cells was observed in IBU administrated by MMS-P, which confirms the effective intracellular IBU uptake by means of MMS-P. These results encourage the potential application of MMS-P as a drug delivery system and confirm the effect of IBU as a cytoprotective agent in a neurodegenerative model.


Asunto(s)
Ibuprofeno , Nanopartículas , Ibuprofeno/química , Carbaril , Sistemas de Liberación de Medicamentos , Polímeros/química , Fenómenos Magnéticos , Dióxido de Silicio/química , Nanopartículas/química
3.
Pediatr Obes ; 18(11): e13072, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37650320

RESUMEN

BACKGROUND: Molecular biomarkers of maternal leptin resistance associated with infant weight are needed. OBJECTIVES: To evaluate gene expression of leptin receptor (LEPR), suppressor of cytokine signalling 3 (SOCS3) and insulin receptor in peripheral blood mononuclear cells (PBMCs) of lactating women and their relationship with infant body weight and adiposity. METHODS: At day 10 postpartum, maternal gene expression in PBMCs as well as leptin and insulin concentrations in plasma and milk were assessed (n = 68). Infant weight and BMI z-scores, skinfolds and arm circumference were obtained at 10 days and/or at 3 months old. RESULTS: In mothers with pre-pregnancy overweight or obesity (OW/OB), LEPR expression was reduced (p = 0.013) whereas plasma and milk leptin and milk insulin concentrations were elevated. LEPR expression was positively related with infant weight z-score (Beta (95% CI): 0.40 (0.17, 0.63), p = 0.001) but not with leptin concentrations. SOCS3 expression was positively related with infant weight z-score (Beta (95% CI): 0.28 (0.04, 0.51), p = 0.024) and arm circumference (Beta (95% CI): 0.57 (0.32, 0.82), p < 0.001). Relationships remained significant after adjusting for maternal and infant confounders. CONCLUSIONS: LEPR and SOCS3 gene expression in PBMCs are novel maternal molecular biomarkers that reflect leptin resistance and are associated with infant body weight and adiposity.


Asunto(s)
Leptina , Receptores de Leptina , Embarazo , Lactante , Femenino , Humanos , Recién Nacido , Índice de Masa Corporal , Lactancia , Leche Humana/metabolismo , Leucocitos Mononucleares/metabolismo , Obesidad/metabolismo , Insulina , Biomarcadores/metabolismo
4.
Life Sci ; 322: 121681, 2023 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-37040814

RESUMEN

AIM: Glucocorticoids (GCs) play a crucial role in energy homeostasis including white adipose tissue function; however, chronic GC excess is detrimental to mammals' health. White hypertrophic adiposity is a main factor for neuroendocrine-metabolic dysfunctions in monosodium L-glutamate (MSG)-damaged hypercorticosteronemic rat. Nevertheless, little is known about the receptor path in endogenous GC impact on white adipose tissue-resident precursor cells to bring them into beige lineage. Thus, our aim was to explore whether transient/chronic endogenous hypercorticosteronemia affects browning capacity in white adipose tissue pads from MSG rats during development. MAIN METHODS: Control and MSG male rats aged 30 and 90 days were 7-day exposed to cold conditions in order to stimulate wet white epidydimal adipose tissue (wEAT) beiging capacity. This procedure was also replicated in adrenalectomized rats. KEY FINDINGS: Data indicated that whereas epidydimal white adipose tissue pads from prepubertal hypercorticosteronemic rats retained full expression of GR/MR genes resulting in a drastic reduction in wEAT beiging capacity, conversely, chronic hypercorticosteronemic adult MSG rats developed down-regulation of corticoid genes (and reduced GR cytosolic mediators) in wEAT pads and consequently partially restored local beiging capacity. Finally, wEAT pads from adrenalectomized rats revealed up-regulation of GR gene accompanied by full local beiging capacity. SIGNIFICANCE: This study strongly supports a GR-dependent inhibitory effect of GC excess on white adipose tissue browning, an issue strongly supporting a key role of GR in the non-shivering thermogenic process. As a consequence, normalizing the GC milieu could be a relevant factor to handle dysmetabolism in white hyperadipose phenotypes.


Asunto(s)
Tejido Adiposo Blanco , Receptores de Glucocorticoides , Animales , Masculino , Ratas , Adipocitos Blancos/metabolismo , Tejido Adiposo/metabolismo , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Adiposidad , Metabolismo Energético , Glucocorticoides/metabolismo , Mamíferos/metabolismo , Obesidad/metabolismo , Receptores de Glucocorticoides/metabolismo , Termogénesis
5.
Life Sci ; 313: 121305, 2023 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-36543283

RESUMEN

AIMS: Since plasma ghrelin can undergo des-acylation and proteolysis, the aim of this study was to investigate the extent to which an enhancement of these reactions is associated to the decrease of ghrelin in plasma after food intake or in individuals with obesity. MAIN METHODS: we performed an intervention cross-sectional study, in which levels of ghrelin, desacyl-ghrelin (DAG), glucose, insulin, ghrelin des-acylation and ghrelin proteolysis were assessed in plasma before and after a test meal in 40 people (n = 21 males) with normal weight (NW, n = 20) or overweight/obesity (OW/OB, n = 20). KEY FINDINGS: Preprandial ghrelin and DAG levels were lower, whereas preprandial ghrelin proteolysis was ∼4.6-fold higher in plasma of males with OW/OB. In males, ghrelin proteolysis positively correlated with glycemia. Ghrelin and DAG levels were also lower in females with OW/OB, but preprandial ghrelin proteolysis was not different between females with NW or OW/OB. Ghrelin and DAG levels decreased postprandially in males and females, independently of BMI, and ghrelin proteolysis increased postprandially ∼2 folds only in individuals with NW. Ghrelin des-acylation remained unaffected by BMI or feeding status in both sexes. SIGNIFICANCE: Current study shows that ghrelin proteolysis increases in males with obesity as well as after meal in lean individuals. Therefore, ghrelin proteolysis may be an important checkpoint and, consequently, a putative pharmacological target to control circulating ghrelin levels in humans.


Asunto(s)
Ghrelina , Obesidad , Caracteres Sexuales , Femenino , Humanos , Masculino , Estudios Transversales , Ghrelina/sangre , Ghrelina/metabolismo , Insulina , Obesidad/metabolismo , Sobrepeso
6.
Cell Mol Life Sci ; 79(5): 277, 2022 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-35504998

RESUMEN

OBJECTIVE: Prolonged fasting is a major challenge for living organisms. An appropriate metabolic response to food deprivation requires the activation of the corticotropin-releasing factor-producing neurons of the hypothalamic paraventricular nucleus (PVHCRF neurons), which are a part of the hypothalamic-pituitary-adrenal axis (HPA), as well as the growth hormone secretagogue receptor (GHSR) signaling, whose activity is up- or down-regulated, respectively, by the hormones ghrelin and the liver-expressed antimicrobial peptide 2 (LEAP2). Since ghrelin treatment potently up-regulates the HPA axis, we studied the role of GHSR in mediating food deprivation-induced activation of the PVHCRF neurons in mice. METHODS: We estimated the activation of the PVHCRF neurons, using immuno-staining against CRF and the marker of neuronal activation c-Fos in brain sections, and assessed plasma levels of corticosterone and glucose in different pharmacologically or genetically manipulated mouse models exposed, or not, to a 2-day food deprivation protocol. In particular, we investigated ad libitum fed or food-deprived male mice that: (1) lacked GHSR gene expression, (2) had genetic deletion of the ghrelin gene, (3) displayed neurotoxic ablation of the hypothalamic arcuate nucleus, (4) were centrally treated with an anti-ghrelin antibody to block central ghrelin action, (5) were centrally treated with a GHSR ligand that blocks ghrelin-evoked and constitutive GHSR activities, or (6) received a continuous systemic infusion of LEAP2(1-12). RESULTS: We found that food deprivation results in the activation of the PVHCRF neurons and in a rise of the ghrelin/LEAP2 molar ratio. Food deprivation-induced activation of PVHCRF neurons required the presence and the signaling of GHSR at hypothalamic level, but not of ghrelin. Finally, we found that preventing the food deprivation-induced fall of LEAP2 reverses the activation of the PVHCRF neurons in food-deprived mice, although it has no effect on body weight or blood glucose. CONCLUSION: Food deprivation-induced activation of the PVHCRF neurons involves ghrelin-independent actions of GHSR at hypothalamic level and requires a decrease of plasma LEAP2 levels. We propose that the up-regulation of the actions of GHSR associated to the fall of plasma LEAP2 level are physiologically relevant neuroendocrine signals during a prolonged fasting.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/metabolismo , Privación de Alimentos , Núcleo Hipotalámico Paraventricular , Receptores de Ghrelina/metabolismo , Animales , Hormona Liberadora de Corticotropina/metabolismo , Hormona Liberadora de Corticotropina/farmacología , Ingestión de Alimentos , Ghrelina/metabolismo , Ghrelina/farmacología , Sistema Hipotálamo-Hipofisario/metabolismo , Masculino , Ratones , Neuronas/metabolismo , Núcleo Hipotalámico Paraventricular/citología , Núcleo Hipotalámico Paraventricular/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Receptores de Ghrelina/genética
7.
Mol Cell Endocrinol ; 538: 111449, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34478806

RESUMEN

Ghrelin is a peptide hormone mainly secreted from gastrointestinal tract that acts via the growth hormone secretagogue receptor (GHSR), which is highly expressed in the brain. Strikingly, the accessibility of ghrelin to the brain seems to be limited and restricted to few brain areas. Previous studies in mice have shown that ghrelin can access the brain via the blood-cerebrospinal fluid (CSF) barrier, an interface constituted by the choroid plexus and the hypothalamic tanycytes. Here, we performed a variety of in vivo and in vitro studies to test the hypothesis that the transport of ghrelin across the blood-CSF barrier occurs in a GHSR-dependent manner. In vivo, we found that the uptake of systemically administered fluorescent ghrelin in the choroid plexus epithelial (CPE) cells and in hypothalamic tanycytes depends on the presence of GHSR. Also, we detected lower levels of CSF ghrelin after a systemic ghrelin injection in GHSR-deficient mice, as compared to WT mice. In vitro, the internalization of fluorescent ghrelin was reduced in explants of choroid plexus from GHSR-deficient mice, and unaffected in primary cultures of hypothalamic tanycytes derived from GHSR-deficient mice. Finally, we found that the GHSR mRNA is detected in a pool of CPE cells, but is nearly undetectable in hypothalamic tanycytes with current approaches. Thus, our results suggest that circulating ghrelin crosses the blood-CSF barrier mainly by a mechanism that involves the GHSR, and also possibly via a GHSR-independent mechanism.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Ghrelina/sangre , Ghrelina/líquido cefalorraquídeo , Receptores de Ghrelina/metabolismo , Animales , Células Cultivadas , Plexo Coroideo/metabolismo , Células Ependimogliales/citología , Células Ependimogliales/metabolismo , Ghrelina/genética , Ratones , Cultivo Primario de Células , Transducción de Señal
8.
ACS Omega ; 6(19): 12567-12576, 2021 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-34056407

RESUMEN

Plasmonic metal nanoparticles (NPs) can be used as enhancers of the efficiency of standard photosensitizers (PSs) in photodynamic therapy (PDT). Protein corona, the adsorption layer that forms spontaneously around NPs once in contact with biological fluids, determines to a great extent the efficiency of PDT. In this work, we explore the possibility that pectin-coated Au NPs (Au@Pec NPs) could act as adjuvants in riboflavin (Rf)-based PDT by comparing the photodamage in HeLa cells cultured in the presence and in the absence of the NPs. Moreover, we investigate the impact that the preincubation of Rf and Au@Pec NPs (or Ag@Pec NPs) at two very different serum concentrations could have on cell's photodamage. Because reactive oxygen species (ROS) precursors are the excited states of the PS, the effect of proteins on the photophysics of Rf and Rf/plasmonic NPs was studied by transient absorption experiments. The beneficial effect of Au@Pec NPs in Rf-based PDT on HeLa cells cultured under standard serum conditions was demonstrated for the first time. However, the preincubation of Rf and Au@Pec NPs (or Ag@Pec NPs) with serum has undesirable results regarding the enhancement of Rf-based PDT. In this sense, we also verified that more concentrated protein conditions result in lower amounts of the triplet excited state of Rf and thus an expected lower production of ROS, which are the key elements for PDT's efficacy. These findings point out the relevance of serum concentration in the design of in vitro cell culture experiments carried out to determine the best way to combine and use potential sensitizers with plasmonic NPs to develop more effective PDTs.

9.
Eur J Endocrinol ; 182(2): 165-175, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31770106

RESUMEN

OBJECTIVE: The octanoylated peptide hormone ghrelin regulates appetite and glycaemic control. Des-acyl ghrelin abolishes some effects of ghrelin, but does not bind to ghrelin receptor. LEAP2 is a novel ligand for ghrelin receptor that blocks the effects of ghrelin. Some evidences show that plasma levels of these peptides are altered in adults with obesity, but their levels in childhood obesity remain poorly studied. Therefore, the objective of this study was to assess fasting plasma levels of ghrelin, des-acyl ghrelin and LEAP2 in children with normoweight, overweight/obesity and their association with different anthropometric and metabolic variables. DESIGN: A total of 42 females and 40 males, ages 3-12 years old were enrolled as a cross-sectional cohort. RESULTS: Plasma levels of des-acyl ghrelin and LEAP2 (but not ghrelin) were lower and ghrelin/des-acyl ghrelin ratio was higher in children with overweight/obesity. Des-acyl ghrelin negatively correlated with age, BMI z-score, insulin and HOMA index, and the correlations were stronger in children with overweight/obesity. LEAP2 levels negatively correlated with BMI z-score. No gender differences were found. CONCLUSIONS: Our findings suggest that ghrelin tone is increased in childhood obesity, due to a decrease on plasma levels of des-acyl ghrelin and LEAP2, and that des-acyl ghrelin is associated to insulin resistance, particularly in children with overweight/obesity.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/sangre , Ghrelina/sangre , Obesidad/sangre , Factores de Edad , Proteínas Sanguíneas , Niño , Preescolar , Estudios Transversales , Humanos , Obesidad/fisiopatología , Factores Sexuales
10.
J Neuroendocrinol ; 31(10): e12785, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31469195

RESUMEN

The growth hormone secretagogue receptor (GHSR) is a G protein-coupled receptor that is highly expressed in the central nervous system. GHSR acts as a receptor for ghrelin and for liver-expressed antimicrobial peptide 2 (LEAP2), which blocks ghrelin-evoked activity. GHSR also displays ligand-independent activity, including a high constitutive activity that signals in the absence of ghrelin and is reduced by LEAP2. GHSR activity modulates a variety of food intake-related behaviours, including binge eating. Previously, we reported that GHSR-deficient mice daily and time-limited exposed to a high-fat (HF) diet display an attenuated binge-like HF intake compared to wild-type mice. In the present study, we aimed to determine whether ligand-independent GHSR activity affects binge-like HF intake in a 4-day binge-like eating protocol. We found that plasma levels of ghrelin and LEAP2 were not modified in mice exposed to this binge-like eating protocol. Moreover, systemic administration of ghrelin or LEAP2 did not alter HF intake in our experimental conditions. Interestingly, we found that central administration of LEAP2 or K-(D-1-Nal)-FwLL-NH2 , which are both blockers of constitutive GHSR activity, reduced binge-like HF intake, whereas central administration of ghrelin or the ghrelin-evoked GHSR activity blockers [D-Lys3]-GHRP-6 and JMV2959 did not modify binge-like HF intake. Taken together, current data indicate that GHSR activity in the brain affects binge-like HF intake in mice independently of plasma levels of ghrelin and LEAP2.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/fisiología , Bulimia/fisiopatología , Ghrelina/fisiología , Receptores de Ghrelina/agonistas , Receptores de Ghrelina/fisiología , Animales , Péptidos Catiónicos Antimicrobianos/administración & dosificación , Péptidos Catiónicos Antimicrobianos/sangre , Péptidos Catiónicos Antimicrobianos/farmacología , Bulimia/prevención & control , Dieta Alta en Grasa , Ghrelina/administración & dosificación , Ghrelina/sangre , Ghrelina/farmacología , Glicina/análogos & derivados , Glicina/farmacología , Infusiones Intraventriculares , Masculino , Ratones , Oligopéptidos/farmacología , Receptores de Ghrelina/antagonistas & inhibidores , Factores de Tiempo , Triazoles/farmacología
11.
Mol Neurobiol ; 56(6): 4120-4134, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30276663

RESUMEN

The stomach-derived hormone ghrelin mainly acts in the brain. Studies in mice have shown that the accessibility of ghrelin into the brain is limited and that it mainly takes place in some circumventricular organs, such as the median eminence. Notably, some known brain targets of ghrelin are distantly located from the circumventricular organs. Thus, we hypothesized that ghrelin could also access the brain via the blood-cerebrospinal fluid (CSF) barrier, which consists of the choroid plexus and the hypothalamic tanycytes. Using systemic injection of ghrelin or fluorescent-ghrelin in mice, we found that cells of the blood-CSF barrier internalize these molecules. In time-response studies, we found that peripherally injected fluorescent-ghrelin quickly reaches hypothalamic regions located in apposition to the median eminence and more slowly reaches the periventricular hypothalamic parenchyma, adjacent to the dorsal part of the third ventricle. Additionally, we found that CSF ghrelin levels increase after the systemic administration of ghrelin, and that central infusions of either an anti-ghrelin antibody, which immuno-neutralizes CSF ghrelin, or a scrambled version of ghrelin, which is also internalized by cells of the blood-CSF barrier, partially impair the orexigenic effect of peripherally injected ghrelin. Thus, current evidence suggests that the blood-CSF barrier can transport circulating ghrelin into the brain, and that the access of ghrelin into the CSF is required for its full orexigenic effect.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Líquido Cefalorraquídeo/metabolismo , Ghrelina/sangre , Animales , Anticuerpos/metabolismo , Ventrículos Cerebrales/metabolismo , Ghrelina/administración & dosificación , Ghrelina/líquido cefalorraquídeo , Masculino , Ratones Endogámicos C57BL , Orexinas/metabolismo
13.
Photochem Photobiol ; 94(6): 1159-1166, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29978491

RESUMEN

Riboflavin (Rf) is an endogenous photosensitizer, which can participate in Type I and Type II processes. We have recently shown that the yield of the triplet excited states of Rf is enhanced in the presence of pectin-coated silver nanoparticles (Pec@AgNP) due to formation of a complex between Rf and Pec@AgNP (Rf-Pec@AgNP). Consequently, under aerobic conditions, the amounts of singlet molecular oxygen and superoxide radical anion generated are also larger in the presence of the nanoparticles. This result made us suspect that the nanoparticles could have a beneficial effect in Rf-based PDT. To prove this hypothesis, we here compared the photodamage in HeLa cells incubated with Rf in the presence and in the absence of Pec@AgNP applying several optical assays. We used fluorescence imaging of irradiated HeLa cells incubated with Annexin V and propidium iodide to evaluate the occurrence of apoptosis/necrosis, the reduction of the tetrazolium dye MTT to formazan and neutral red uptake to prove cell viability, as well as synchrotron infrared microscopy of single cells to evaluate possible structural changes of DNA and nuclear proteins. The enhanced photodamage observed in the presence of Pec@AgNP seems to indicate that Rf enters into the cells complexed with the nanoparticles.


Asunto(s)
Apoptosis/efectos de los fármacos , Nanopartículas del Metal/química , Fármacos Fotosensibilizantes/farmacología , Riboflavina/farmacología , Plata/química , Apoptosis/efectos de la radiación , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Células HeLa , Humanos , Cinética , Luz , Nanopartículas del Metal/ultraestructura , Oxígeno/química , Oxígeno/metabolismo , Pectinas/química , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/química , Riboflavina/química , Análisis de la Célula Individual , Oxígeno Singlete/química , Oxígeno Singlete/metabolismo , Superóxidos/química , Superóxidos/metabolismo
14.
Acta bioquím. clín. latinoam ; 52(1): 23-32, mar. 2018. ilus, graf, tab
Artículo en Español | LILACS | ID: biblio-886157

RESUMEN

Las mujeres embarazadas con insulino-sensibilidad disminuida están en riesgo de desarrollar trastornos hipertensivos. Utilizando el corte HOMA-IR en 2,64 la población en estudio fue dividida en dos grupos: (n=154 mujeres embarazadas), las que arrojaron un HOMA-IR basal (HOMA-0) <2,64 (no-insulinorresistentes; n=113) y aquellas con HOMA-0>2,64 (insulinorresistentes, n=41). Se analizaron: a) las concentraciones circulantes de glucosa e insulina durante una prueba de tolerancia oral a 75 g de glucosa (PTOG), y b) las relaciones entre varios parámetros de insulino-sensibilidad y la predicción del desarrollo de trastornos hipertensivos. A las mujeres embarazadas (semana 24-28) se les cuantificaron las concentraciones plasmáticas de glucosa e insulina a ambos tiempos de la PTOG. Se calcularon los valores de HOMA-IR y las relaciones glucosa a insulina (G:I) y se registraron parámetros antropométricos y resultados del embarazo. Las mujeres con HOMA-0 >2,64, aunque con glucemias en ayunas normales, mostraron mayores niveles de insulinemia y de HOMA-IR, y menores valores G:I en ambos tiempos de la PTOG. Estas mujeres embarazadas fueron las que tuvieron un mayor riesgo de desarrollar trastornos hipertensivos y mayores parámetros de morbilidad durante el período estudiado al compararlas con aquellas cuyo HOMA-0 fue <2,64.


Pregnant women with impaired insulin sensitivity are at risk for developing hypertensive disorders. By using a cut-off at 2.64 of the homeostasis model assessment (HOMA-IR) in basal condition (HOMA-0), the population under study (n=154 pregnant women) was split into two groups: 1) with basal HOMA- 0 <2.64 (non-insulin resistant; n=113) and 2) with basal HOMA-0 >2.64 (insulin resistant; n=41). Glucose and insulin circulating levels were analyzed throughout a 2-h oral 75 g glucose tolerance test (OGTT). The relationship between several parameters related to insulin resistance and the prevalence of pregnancy-induced hypertensive disorders was analyzed. Pregnant women (on week 24-28) were submitted to an OGTT, and glucose and insulin plasma concentrations were measured throughout the test. These peripheral metabolites levels and the values of the HOMA-IR and the glucose to insulin ratio (G:I) were analyzed. Anthropometric parameters and pregnancy outcome were recorded. Women with HOMA-0 >2.64 but normal fasting glycemia showed higher insulinemias, G:I values and HOMA-IR values at both times of the OGTT. The latter were at greater risk for developing late pregnancy-induced hypertension compared to women with HOMA-0 ≤2.64.


As mulheres grávidas com diminuição da sensibilidade à insulina correm o risco de desenvolver distúrbios hipertensivos. Usando o corte HOMA-IR 2,64, a população em estudo foi dividida em dois grupos: (n=154 mulheres grávidas), que deram um HOMA-IR basal (HOMA-0) ≤2,64 (não resistentes à insulina; n=113) e aquelas com HOMA-0 >2,64 (resistentes à insulina, n=41). Foram analisadas: a) as concentrações circulantes de glicose e insulina durante uma prova de tolerância oral a 75 g. de glicose (PTOG), e b) as relações entre diversos parâmetros de sensibilidade à insulina e a predição de desenvolver distúrbios de hipertensão. Foram quantificadas nas mulheres grávidas (24-28 semanas) as concentrações plasmáticas de glicose e insulina a ambos os tempos da PTOG. Valores de HOMA-IR foram calculados e as relações glicose a insulina (G:I) e se registraram parâmetros antropométricos e os resultados da gravidez. Mulheres com HOMA-0 >2,64, mas com glicemias em jejuns normais, mostraram níveis mais elevados de insulinemia e de HOMA-IR, e menores valores G:I em ambos os tempos da PTOG. Essas mulheres grávidas foram aquelas que tiveram maior risco de desenvolver distúrbios de hipertensão e maiores parâmetros de morbidade durante o período estudado em comparação com as mulheres cujo HOMA-0 foi ≤2,64.


Asunto(s)
Humanos , Femenino , Embarazo , Adulto , Glucemia , Resistencia a la Insulina , Embarazo , Hipertensión Inducida en el Embarazo , Argentina , Prevención Primaria , Antropometría , Salud Pública , Encuestas y Cuestionarios , Diabetes Gestacional , Embarazo de Alto Riesgo , Diabetes Mellitus Tipo 2 , Factor de Crecimiento Placentario , Insulina
15.
Cell Tissue Res ; 369(2): 369-380, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28413862

RESUMEN

Hypothalamic tanycytes are specialized bipolar ependymal cells that line the floor of the third ventricle. Given their strategic location, tanycytes are believed to play several key functions including being a selective barrier and controlling the amount of hypothalamic-derived factors reaching the anterior pituitary. The in vitro culture of these cells has proved to be difficult. Here, we report an improved method for the generation of primary cultures of rat hypothalamic tanycytes. Ependymal cultures were derived from tissue dissected out of the median eminence region of 10-day-old rats and cultured in a chemically defined medium containing DMEM:F12, serum albumin, insulin, transferrin and the antibiotic gentamycin. After 7 days in vitro, ∼30% of the cultured cells exhibited morphological features of tanycytes as observed by phase contrast or scanning electron microscopy. Tanycyte-like cells were strongly immuno-reactive for vimentin and dopamine-cAMP-regulated phospho-protein (DARPP-32) and weakly immune-reactive for glial fibrillary acidic protein. Tanycyte-like cells displayed a stable negative resting plasma membrane potential and failed to show spiking properties in response to current injections. When exposed to fluorescent beads in the culture medium, tanycyte-like cells exhibited a robust endocytosis. Thus, the present method effectively yields cultures containing tanycyte-like cells that resemble in vivo tanycytes in terms of morphologic features and molecular markers as well as electrical and endocytic activity. To our knowledge, this is the first protocol that allows the culturing of tanycyte-like cells that can be individually identified and that conserve the morphology of tanycytes in their natural physiological environment.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Forma de la Célula , Células Ependimogliales/citología , Hipotálamo/citología , Animales , Células Cultivadas , Fenómenos Electrofisiológicos , Endocitosis , Inmunohistoquímica , Ratas Sprague-Dawley
16.
Int J Endocrinol ; 2016: 7838290, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27597864

RESUMEN

We investigated the impact of chronic hypercorticosteronemia (due to neonatal monosodium L-glutamate, MSG, and treatment) on liver oxidative stress (OS), inflammation, and carbohydrate/lipid metabolism in adult male rats. We evaluated the peripheral concentrations of several metabolic and OS markers and insulin resistance indexes. In liver we assessed (a) OS (GSH and protein carbonyl groups) and inflammatory (IL-1b, TNFa, and PAI-1) biomarkers and (b) carbohydrate and lipid metabolisms. MSG rats displayed degenerated optic nerves, hypophagia, low body and liver weights, and enlarged adipose tissue mass; higher peripheral levels of glucose, triglycerides, insulin, uric acid, leptin, corticosterone, transaminases and TBARS, and peripheral and liver insulin resistance; elevated liver OS, inflammation markers, and glucokinase (mRNA/activity) and fructokinase (mRNA). Additionally, MSG liver phosphofructokinase-2, glucose-6-phosphatase (mRNA and activity) and glucose-6-phosphate dehydrogenase, Chrebp, Srebp1c, fatty acid synthase, and glycerol-3-phosphate (mRNAs) were increased. In conclusion adult MSG rats developed an insulin-resistant state and increased OS and serious hepatic dysfunction characterized by inflammation and metabolic signs suggesting increased lipogenesis. These features, shared by both metabolic and Cushing's syndrome human phenotypes, support that a chronic glucocorticoid-rich endogenous environment mainly impacts on hepatic glucose cycle, displacing local metabolism to lipogenesis. Whether correcting the glucocorticoid-rich environment ameliorates such dysfunctions requires further investigation.

17.
Nutrients ; 8(7)2016 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-27384583

RESUMEN

Adipose tissue (AT) expansion is the result of two processes: hyperplasia and hypertrophy; and both, directly or indirectly, depend on the adipogenic potential of adipocyte precursor cells (APCs). Glucocorticoids (GCs) have a potent stimulatory effect on terminal adipogenesis; while their effects on early stages of adipogenesis are largely unknown. In the present work, we study, in a model of high GC levels, the adipogenic potential of APCs from retroperitoneal AT (RPAT) and its relationship with RPAT mass expansion. We employed a model of hyper-adiposity (30- and 60-day-old rats) due to high endogenous GC levels induced by neonatal treatment with l-monosodium glutamate (MSG). We found that the RPAT APCs from 30-day-old MSG rats showed an increased adipogenic capacity, depending on the APCs' competency, but not in their number. Analyses of RPAT adipocyte diameter revealed an increase in cell size, regardless of the rat age, indicating the prevalence of a hypertrophic process. Moreover, functional RPAT alterations worsened in 60-day-old rats, suggesting that the hyperplastic AT expansion found in 30-day-old animals might have a protective role. We conclude that GCs chronic excess affects APCs' adipogenic capacity, modifying their competency. This change would modulate the hyperplastic/hypertrophic balance determining healthy or unhealthy RPAT expansion and, therefore, its functionality.


Asunto(s)
Glucocorticoides/sangre , Grasa Intraabdominal/metabolismo , Obesidad/sangre , Adipocitos/metabolismo , Adipogénesis/fisiología , Adiposidad/fisiología , Animales , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Células Cultivadas , Corticosterona/sangre , Modelos Animales de Enfermedad , Hiperplasia/sangre , Hiperplasia/complicaciones , Hipertrofia/sangre , Hipertrofia/complicaciones , Insulina/sangre , Leptina/sangre , Masculino , Malonatos/efectos adversos , Ratas , Ratas Sprague-Dawley
18.
Artículo en Inglés | MEDLINE | ID: mdl-26653979

RESUMEN

We analyzed chromosomal aberrations involving telomeres in the progeny of mammalian cells exposed to the methylating agent and antineoplastic/diabetogenic drug streptozotocin (STZ), to test whether it induces long-term telomere instability (by chromosome end loss and/or telomere dysfunction). Rat cells (ADIPO-P2 cell line, derived from Sprague-Dawley rat adipose cells) were treated with a single concentration of STZ (2mM). Chromosomal aberrations were analyzed 18h, 10 days, and 15 days after treatment, using PNA-FISH with a pan-telomeric probe [Cy3-(CCCTAA)3] to detect (TTAGGG)n repeats. Cytogenetic analysis revealed a higher frequency of chromosomal aberrations in STZ-exposed cultures vs. untreated cultures at each time point analyzed. The yield of induced aberrations was very similar at each time point. Induction of aberrations not involving telomere dysfunction was only observed 18h and 15 days after treatment, whereas induction of telomere dysfunction-related aberrations by STZ (mainly in the form of telomere FISH signal loss and duplications, most of them chromatid-type aberrations) was observed at each time point. Our results show that STZ induces persistent telomere instability in mammalian cells, cytogenetically manifested as telomere dysfunction-related chromosomal aberrations. Neither telomere length nor telomerase activity is related to the telomere dysfunction.


Asunto(s)
Aberraciones Cromosómicas/inducido químicamente , Estreptozocina/efectos adversos , Telómero/efectos de los fármacos , Tejido Adiposo/citología , Tejido Adiposo/efectos de los fármacos , Animales , Línea Celular , Análisis Citogenético , Inestabilidad Genómica/efectos de los fármacos , Humanos , Hibridación Fluorescente in Situ , Células Jurkat , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Telómero/patología
19.
J Gen Physiol ; 146(3): 205-19, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26283199

RESUMEN

The growth hormone secretagogue receptor type 1a (GHSR1a) has the highest known constitutive activity of any G protein-coupled receptor (GPCR). GHSR1a mediates the action of the hormone ghrelin, and its activation increases transcriptional and electrical activity in hypothalamic neurons. Although GHSR1a is present at GABAergic presynaptic terminals, its effect on neurotransmitter release remains unclear. The activities of the voltage-gated calcium channels, CaV2.1 and CaV2.2, which mediate neurotransmitter release at presynaptic terminals, are modulated by many GPCRs. Here, we show that both constitutive and agonist-dependent GHSR1a activity elicit a strong impairment of CaV2.1 and CaV2.2 currents in rat and mouse hypothalamic neurons and in a heterologous expression system. Constitutive GHSR1a activity reduces CaV2 currents by a Gi/o-dependent mechanism that involves persistent reduction in channel density at the plasma membrane, whereas ghrelin-dependent GHSR1a inhibition is reversible and involves altered CaV2 gating via a Gq-dependent pathway. Thus, GHSR1a differentially inhibits CaV2 channels by Gi/o or Gq protein pathways depending on its mode of activation. Moreover, we present evidence suggesting that GHSR1a-mediated inhibition of CaV2 attenuates GABA release in hypothalamic neurons, a mechanism that could contribute to neuronal activation through the disinhibition of postsynaptic neurons.


Asunto(s)
Canales de Calcio Tipo N/metabolismo , Ghrelina/metabolismo , Hipotálamo/fisiología , Neuronas/fisiología , Receptores de Ghrelina/metabolismo , Animales , Secuencia de Bases , Calcio/metabolismo , Señalización del Calcio/fisiología , Células Cultivadas , Células HEK293 , Humanos , Activación del Canal Iónico/fisiología , Ratones , Datos de Secuencia Molecular , Ratas , Ratas Sprague-Dawley , Receptores de Ghrelina/genética
20.
Int Sch Res Notices ; 2015: 284042, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-27347514

RESUMEN

Rats neonatally treated with monosodium L-glutamate (MSG) are deeply dysfunctional in adulthood. We explored the effect of an oral low dose of metformin treatment in male MSG rats on adipoinsular axis and visceral adipose tissue (VAT) dysfunctions, in both basal (nonfasting) and endotoxemia conditions. MSG rats, treated or not treated with metformin (30 days prior to experimentation), and control litter-mates (CTR) were studied at 90 days of age. Peripheral concentrations of glucose, lipids, and hormones were determined in basal and post-lipopolysaccharide (LPS) treatment conditions. Food intake and body weight (BW) were recorded and VAT mass and leptin mRNA levels were evaluated. Data indicated that MSG rats were lighter and displayed hypercorticosteronemia, hypophagia, adipoinsular axis hyperactivity, and enhanced VAT mass associated with an increased leptin gene expression. Interestingly, metformin-treated MSG rats corrected BW catch-up and counteracted VAT (mass and leptin mRNA level) and adipoinsular axis (basal and post-LPS) dysfunctions. Thus metformin treatment in MSG rats is able to correct several VAT and metabolic-endocrine dysfunctions. Our study suggests that a low-dose metformintherapy is effective to correct, at least in part, adipoinsular axis dysfunction in hypertrophic obese phenotypes, such as that of the human Cushing syndrome.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...