Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Luana Lopes Souza is active.

Publication


Featured researches published by Luana Lopes Souza.


Journal of Endocrinology | 2013

Non-classic thyroid hormone signalling involved in hepatic lipid metabolism

Aline Cordeiro; Luana Lopes Souza; Marcelo Einicker-Lamas; Carmen Cabanelas Pazos-Moura

Thyroid hormones are important modulators of lipid metabolism because the liver is a primary hormonal target. The hypolipidaemic effects of thyroid hormones result from the balance between direct and indirect actions resulting in stimulation of lipid synthesis and lipid oxidation, which favours degradation pathways. Originally, it was believed that thyroid hormone activity was only transduced by alteration of gene transcription mediated by the nuclear receptor thyroid hormone receptors, comprising the classic action of thyroid hormone. However, the discovery of other effects independent of this classic mechanism characterised a new model of thyroid hormone action, the non-classic mechanism that involves other signalling pathways. To date, this mechanism and its relevance have been intensively described. Considering the increasing evidence for non-classic signalling of thyroid hormones and the major influence of these hormones in the regulation of lipid metabolism, we reviewed the role of thyroid hormone in cytosolic signalling cascades, focusing on the regulation of second messengers, and the activity of effector proteins and the implication of these mechanisms on the control of hepatic lipid metabolism.


The Journal of Physiology | 2010

Female mice target deleted for the neuromedin B receptor have partial resistance to diet‐induced obesity

Gabriela Silva Monteiro Paula; Luana Lopes Souza; Adriana Cabanelas; Flavia Fonseca Bloise; Valeria de Mello-Coelho; Etsuko Wada; Tania M. Ortiga-Carvalho; Karen Jesus Oliveira; Carmen C. Pazos-Moura

Previous studies have proposed a role for neuromedin B (NB), a bombesin‐like peptide, in the control of body weight homeostasis. However, the nature of this role is unclear. The actions of NB are mediated preferentially by NB‐preferring receptors (NBRs). Here we examined the consequences of targeted deletion of NBRs in female mice on body weight homeostasis in mice fed a normolipid diet (ND) or a high‐fat diet (HFD) for 13 weeks. Body weight and food ingestion of neuromedin B receptor knockout (NBR‐KO) mice fed a normolipid diet showed no difference in relation to wild‐type (WT). However, the high‐fat diet induced an 8.9‐ and 4.8‐fold increase in body weight of WT and NBR‐KO, respectively, compared to their controls maintained with a normolipid diet, even though the mice ingested the same amount of calories, regardless of genotype. Comparing mice fed the high‐fat diet, NBR‐KO mice accumulated approximately 45% less fat depot mass than WT, exhibited a lower percentage of fat in their carcasses (19.2 vs. 31.3%), and their adipocytes were less hypertrophied. Serum leptin and leptin mRNA in inguinal and perigonadal fat were lower in HFD NBR‐KO than HFD WT, and serum adiponectin was similar among HFD groups and unaltered in comparison to ND‐fed mice. HFD‐fed WT mice developed glucose intolerance but not the HFD‐fed NBR‐KO mice, although they had similar glycaemia and insulinaemia. NBR‐KO and WT mice on the normolipid diet showed no differences in any parameters, except for a trend to lower insulin levels. Therefore, disruption of the neuromedin B receptor pathway did not change body weight homeostasis in female mice fed a normolipid diet; however, it did result in partial resistance to diet‐induced obesity.


Journal of Endocrinology | 2011

Thyroid hormone contributes to the hypolipidemic effect of polyunsaturated fatty acids from fish oil: in vivo evidence for cross talking mechanisms

Luana Lopes Souza; Aline Cordeiro; Lorraine Soares Oliveira; Gabriela Silva Monteiro Paula; Larissa Costa Faustino; Tania M. Ortiga-Carvalho; Karen Jesus Oliveira; Carmen Cabanelas Pazos-Moura

n-3 polyunsaturated fatty acids (n-3 PUFA) from fish oil (FO) exert important lipid-lowering effects, an effect also ascribed to thyroid hormones (TH) and TH receptor β1 (TRβ1)-specific agonists. n-3 PUFA effects are mediated by nuclear receptors, such as peroxisome proliferator-activated receptors (PPAR) and others. In this study, we investigated a role for TH signaling in n-3 PUFA effects. Euthyroid and hypothyroid adult rats (methimazole-treated for 5 weeks) received FO or soybean oil (control) by oral administration for 3 weeks. In euthyroid rats, FO treatment reduced serum triglycerides and cholesterol, diminished body fat, and increased protein content of the animals. In addition, FO-treated rats exhibited higher liver expression of TRβ1 and mitochondrial α-glycerophosphate dehydrogenase (mGPD), at protein and mRNA levels, but no alteration of glutathione S-transferase or type 1 deiodinase. In hypothyroid condition, FO induced reduction in serum cholesterol and increase in body protein content, but lost the ability to reduce triglycerides and body fat, and to induce TRβ1 and mGDP expression. FO did not change PPARα liver abundance regardless of thyroid state; however, hypothyroidism led to a marked increase in PPARα liver content but did not alter TRβ1 or TRα expression. The data suggest that part of the effect of n-3 PUFA from FO on lipid metabolism is dependent on TH signaling in specific steps and together with the marked upregulation of PPARα in liver of hypothyroid rats suggest important in vivo consequences of the cross-talking between those fatty acids and TH pathways in liver metabolism.


Journal of Endocrinology | 2010

Resveratrol reduces lipid peroxidation and increases sirtuin 1 expression in adult animals programed by neonatal protein restriction

Juliana Gastão Franco; Egberto Gaspar de Moura; Josely C. Koury; Paula Affonso Trotta; Aline Cordeiro; Luana Lopes Souza; Norma Aparecida dos Santos Almeida; Natália da Silva Lima; Carmen Cabanelas Pazos-Moura; Patricia Cristina Lisboa; Magna Cottini Fonseca Passos

Resveratrol (Res) has been associated with protective effects against oxidative stress. This study evaluated the effect of Res over lipid peroxidation, antioxidant defense, hepatic sirtuin 1 (SIRT1), which up-regulates antioxidant enzymes, and copper/zinc superoxide dismutase (Cu/Zn SOD) in adult offspring whose mothers were protein restricted during lactation. Lactating Wistar rats were divided into control (C) group, which were fed a normal diet (23% protein), and low-protein and high-carbohydrate (LPHC) group, which were fed a diet containing 8% protein. After weaning (21 days), C and LPHC offspring were fed a normal diet until they were 180 days old. At the 160th day, animals were separated into four groups as follows: control, control+Res, LPHC, and LPHC+Res. Resveratrol was given for 20 days (30  mg/kg per day by gavage). LPHC animals showed a higher total antioxidant capacity (TAC) without change in lipid peroxidation and SIRT1 expression. The treatment with Res increased TAC only in the control group without effect on lipid peroxidation and SIRT1. LPHC animals treated with Res had lower lipid peroxidation and higher protein and mRNA expression of SIRT1 without any further increase in TAC. No significant difference in liver Cu/Zn SOD expression was observed among the groups. In conclusion, maternal protein restriction during lactation programs the offspring for a higher antioxidant capacity, and these animals seem to respond to Res treatment with a lower lipid peroxidation and higher hepatic SIRT1 expression that we did not observe in the Res-treated controls. It is probable that the protective effect can be attributed to Res activating SIRT1, only in the LPHC-programmed group.


Journal of Endocrinology | 2013

Thyroid hormone regulation of Sirtuin 1 expression and implications to integrated responses in fasted mice

Aline Cordeiro; Luana Lopes Souza; Lorraine Soares Oliveira; Larissa Costa Faustino; Leticia Aragao Santiago; Flavia Fonseca Bloise; Tania M. Ortiga-Carvalho; Norma Aparecida dos Santos Almeida; Carmen C. Pazos-Moura

Sirtuin 1 (SIRT1), a NAD(+)-dependent deacetylase, has been connected to beneficial effects elicited by calorie restriction. Physiological adaptation to starvation requires higher activity of SIRT1 and also the suppression of thyroid hormone (TH) action to achieve energy conservation. Here, we tested the hypothesis that those two events are correlated and that TH may be a regulator of SIRT1 expression. Forty-eight-hour fasting mice exhibited reduced serum TH and increased SIRT1 protein content in liver and brown adipose tissue (BAT), and physiological thyroxine replacement prevented or attenuated the increment of SIRT1 in liver and BAT of fasted mice. Hypothyroid mice exhibited increased liver SIRT1 protein, while hyperthyroid ones showed decreased SIRT1 in liver and BAT. In the liver, decreased protein is accompanied by reduced SIRT1 activity and no alteration in its mRNA. Hyperthyroid and hypothyroid mice exhibited increases and decreases in food intake and body weight gain respectively. Food-restricted hyperthyroid animals (pair-fed to euthyroid group) exhibited liver and BAT SIRT1 protein levels intermediary between euthyroid and hyperthyroid mice fed ad libitum. Mice with TH resistance at the liver presented increased hepatic SIRT1 protein and activity, with no alteration in Sirt1 mRNA. These results suggest that TH decreases SIRT1 protein, directly and indirectly, via food ingestion control and, in the liver, this reduction involves TRβ. The SIRT1 reduction induced by TH has important implication to integrated metabolic responses to fasting, as the increase in SIRT1 protein requires the fasting-associated suppression of TH serum levels.


Endocrinology | 2009

Connexin40 messenger ribonucleic acid is positively regulated by thyroid hormone (TH) acting in cardiac atria via the TH receptor.

Norma A. S. Almeida; Aline Cordeiro; Danielle S. Machado; Luana Lopes Souza; Tânia M. Ortiga-Carvalho; Antonio C. Campos-de-Carvalho; Fredric E. Wondisford; Carmen C. Pazos-Moura

Thyroid hormone (TH) regulates many cardiac genes via nuclear thyroid receptors, and hyperthyroidism is frequently associated with atrial fibrillation. Electrical activity propagation in myocardium depends on the transfer of current at gap junctions, and connexins (Cxs) 40 and 43 are the predominant junction proteins. In mice, Cx40, the main Cx involved in atrial conduction, is restricted to the atria and fibers of the conduction system, which also express Cx43. We studied cardiac expression of Cx40 and Cx43 in conjunction with electrocardiogram studies in mice overexpressing the dominant negative mutant thyroid hormone receptor-beta Delta337T exclusively in cardiomyocytes [myosin heavy chain (MHC-mutant)]. These mice develop the cardiac hypothyroid phenotype in the presence of normal serum TH. Expression was also examined in wild-type mice rendered hypothyroid or hyperthyroid by pharmacological treatment. Atrial Cx40 mRNA and protein levels were decreased (85 and 55%, respectively; P < 0.001) in MHC-mt mice. Atrial and ventricular Cx43 mRNA levels were not significantly changed. Hypothyroid and hyperthyroid animals showed a 25% decrease and 40% increase, respectively, in Cx40 mRNA abundance. However, MHC-mt mice presented very low Cx40 mRNA expression regardless of whether they were made hypothyroid or hyperthyroid. Atrial depolarization velocity, as represented by P wave duration in electrocardiograms of unanesthetized mice, was extremely reduced in MHC-mt mice, and to a lesser extent also in hypothyroid mice (90 and 30% increase in P wave duration). In contrast, this measure was increased in hyperthyroid mice (19% decrease in P wave duration). Therefore, this study reveals for the first time that Cx40 mRNA is up-regulated by TH acting in cardiac atria via the TH receptor and that this may be one of the mechanisms contributing to atrial conduction alterations in thyroid dysfunctions.


Experimental Physiology | 2011

Liver glutathione S-transferase α expression is decreased by 3,5,3 -triiodothyronine in hypothyroid but not in euthyroid mice

Larissa Costa Faustino; Rachel M. Pires; Ana Claudia Lima; Aline Cordeiro; Luana Lopes Souza; Tânia Maria Ortiga-Carvalho

As previously reported, the activity of liver glutathione S‐transferases, an important family of enzymes for detoxification processes, is regulated by thyroid hormone levels. Here, we specifically studied glutathione S‐transferase α (Gsta) gene expression in livers of mice. First, in wild‐type (WT) mice, hypothyroidism was induced by 5 weeks of a diet containing 5‐propyl‐2‐thiouracil plus water containing metimazole, whereas hyperthyroidism was induced by daily injections of 50 μg (100 g body weight)−1 of 3,3′, 5‐triiodo‐L‐thyronine (L‐T3) for 15 days. Importantly, hypothyroidism induced liver Gsta mRNA (>500%) and protein levels (70%; P < 0.01), indicating an important role of baseline thyroid hormone levels to repress this gene; however, surprisingly, no differences were seen in hyperthyroid mice. To further investigate Gsta repression by T3, we used animals expressing a naturally occurring mutation of the gene for thyroid hormone receptor (TR)‐β (Δ337T), which prevents T3 binding and causes a general resistance to thyroid hormone. At baseline, homozygous animals showed increased Gsta levels (mRNA 3.5 times, protein 1.3 times) similar to those found in hypothyroid animals. After a T3 suppression test, we found a blunted response of liver Gsta after the lower doses of T3 in homozygous animals, as expected. However, after the highest dose of T3, we observed a decrease in Gsta expression (80%), similar to normal animals, explained by a higher expression of TR‐α1 (60%; P < 0.01) and a lower expression of Src1 (steroid coactivator receptor) in the mutant animals (50% decrease). In summary, a decrease in Gsta expression caused by T3 was observed only in the hypothyroid state. In addition, an essential role of TR‐β1 is to mediate Gsta suppression in response to T3 and, in the absence of a functional TR‐β, there is a compensatory action of TR‐α1 that depends on low levels of Src1.


Regulatory Peptides | 2011

Central NPY-Y5 receptors activation plays a major role in fasting-induced pituitary-thyroid axis suppression in adult rat.

Ricardo Henrique Costa-e-Sousa; Luana Lopes Souza; Camila Calviño; Adriana Cabanelas; Norma Aparecida dos Santos Almeida; Karen Jesus Oliveira; Carmen C. Pazos-Moura

Neuropeptide Y (NPY) inhibits TRH neurons in fed state, and hypothalamic NPY higher expression during fasting has been proposed to be involved in fasting-induced suppression of the hypothalamus-pituitary-thyroid (HPT) axis. We investigated the role of central Y5 receptors in the control of thyrotropin (TSH) and thyroid hormone (TH) secretion. Fed and fasting rats received twice daily central injections (3rd ventricle) of Y5 receptor antagonist (CGP71683; 15nmol/rat) for 72h. Fasted rats also received a single central injection of CGP71683 (15nmol/rat) at the end of 72h of fasting. In fed rats, Y5 receptor blockade reduced total food intake by 32% and body mass by almost 10% (p<0.01), corroborating the role of this receptor in food intake control. 72h-fasted rats exhibited a 4-fold increase in serum TSH (p<0.001), 1h after a single injection of Y5 antagonist. Also with multiple injections during 72h of fasting, Y5 blockade resulted in activation of thyroid axis, as demonstrated by a 3-times rise in serum T4 (p<0.001), accompanied by unchanged TSH and T3. In fed rats, the chronic central administration of CGP71683 resulted in reduced total serum T4 without changes in free T4 and TSH. Serum leptin and PYY were not altered by the NPY central blockade in both fed and fasted rats, suggesting no role of these hormones in the alterations observed. Therefore, the inhibition of central Y5 neurotransmission resulted in activation of thyroid axis during fasting suggesting that NPY-Y5 receptors contribute to fasting-induced TSH and TH suppression.


Journal of Endocrinology | 2008

Acute effects of endocannabinoid anandamide and CB1 receptor antagonist, AM251 in the regulation of thyrotropin secretion

Marco Aurélio Liberato Costa da Veiga; Flavia Fonseca Bloise; Ricardo Henrique Costa-e-Sousa; Luana Lopes Souza; Norma Aparecida dos Santos Almeida; Karen Jesus Oliveira; Carmen Cabanelas Pazos-Moura

We examined the acute effects of endocannabinoid, anandamide, and of synthetic cannabinoid receptor antagonist, AM251[N-(piperidin-1-yl)-1-(2,4-dichlorophenyl)-5-(4-chlorophenyl)-4-methyl-1H-pyrazole-3-carboxamide], on TSH, thyroxine (T(4)), and triiodothyronine (T(3)) secretions. Euthyroid male rats showed a 42% decrease in serum TSH, 2 h after a single i.p. injection of 0.02, but not 0.2 mg/kg body weight (BW), anandamide, accompanied by a 39% reduction in serum T(4), without alteration in serum T(3). At 0.5 and 1 h, these serum hormones showed no significant change. Hypothyroid rats showed a 35% reduction in serum TSH (P<0.01), 2 h after anandamide injection, which had no effect on hyperthyroid rats. In both thyroid states, no modification of serum thyroid hormones was observed. Intraperitoneal injection of 0.17 or 1.7 mg/kg BW AM251 in euthyroid rats caused, 1.5 h later, 1.7-fold or 4.3-fold increase in serum TSH respectively, without changing thyroid hormones. Stimulatory effect of 0.17 mg/kg BW AM251 and inhibitory effect of anandamide was abolished in the group injected with AM251 followed by an anandamide injection, 30 min later. Intracerebroventricular injection of 20 ng (but not 200 ng) anandamide induced a decrease in serum TSH at 60 min after injection, which tended to disappear at 120 min. Anterior pituitary explants presented significant reduction in TSH release in the presence of 10(-7) M anandamide in incubation medium, which was blocked by 10(-7) M AM251. In conclusion, anandamide has the ability to acutely inhibit TSH release in eu- and hypothyroid rats, acting at the hypothalamus-pituitary axis. Since, in addition, the cannabinoid receptor antagonist AM251 increased TSH release, we suggest that endocannabinoid system has a role as negative regulator of TSH secretion.


Molecular Nutrition & Food Research | 2016

Perinatal maternal high‐fat diet promotes alterations in hepatic lipid metabolism and resistance to the hypolipidemic effect of fish oil in adolescent rat offspring

Lorraine Soares Oliveira; Luana Lopes Souza; Aline F. P. Souza; Aline Cordeiro; George Eduardo Gabriel Kluck; Georgia C. Atella; Isis Hara Trevenzoli; Carmen C. Pazos-Moura

SCOPE Maternal high-fat diet (HFD) promotes obesity and metabolic disturbances in offspring at weaning and adult life. We investigated metabolic consequences of maternal HFD in adolescent rat offspring and the potential benefic effects of fish oil (FO) (n-3 polyunsaturated fatty acid source). METHODS AND RESULTS Female rats received isocaloric, standard diet (STD: 9% fat) or HFD (28.6%) before mating, and throughout pregnancy and lactation. After weaning, male offspring received standard diet and, from 25th to 45th day, received oral administration of soybean oil (SO) or FO. HFD offspring showed higher body weight and adiposity, which was not attenuated by FO. In STD offspring, FO reduced serum triglyceride and cholesterol, as expected, but not in HFD offspring. Liver of HFD offspring groups showed increased free cholesterol and FO-treated HFD group showed lower expression of Abcg8, suggesting decreased cholesterol biliary excretion. HFD offspring presented higher hepatic expression of lipogenic markers, Srebf1 mRNA and acetyl CoA carboxylase (ACC). Serum n-3 PUFA were decreased in FO-treated HFD compared to FO-treated STD offspring, which may explain the reduced hypolipidemic FO effect. CONCLUSION Maternal HFD impaired the ability of FO to reduce adiposity and serum lipids in adolescent offspring, suggesting a potential predisposition to future development of metabolic disorders.

Collaboration


Dive into the Luana Lopes Souza's collaboration.

Top Co-Authors

Avatar

Carmen C. Pazos-Moura

Federal University of Rio de Janeiro

View shared research outputs
Top Co-Authors

Avatar

Karen Jesus Oliveira

Federal University of Rio de Janeiro

View shared research outputs
Top Co-Authors

Avatar

Aline Cordeiro

Federal University of Rio de Janeiro

View shared research outputs
Top Co-Authors

Avatar

Gabriela Silva Monteiro Paula

Federal University of Rio de Janeiro

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Norma Aparecida dos Santos Almeida

Universidade Federal Rural do Rio de Janeiro

View shared research outputs
Top Co-Authors

Avatar

Tania M. Ortiga-Carvalho

Federal University of Rio de Janeiro

View shared research outputs
Top Co-Authors

Avatar

Adriana Cabanelas

Federal University of Rio de Janeiro

View shared research outputs
Top Co-Authors

Avatar

Ricardo Henrique Costa-e-Sousa

Federal University of Rio de Janeiro

View shared research outputs
Top Co-Authors

Avatar

Flavia Fonseca Bloise

Federal University of Rio de Janeiro

View shared research outputs
Researchain Logo
Decentralizing Knowledge