Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Integr Neurosci ; 23(2): 32, 2024 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-38419439

RESUMO

The role of growth hormone (GH) in the central nervous system (CNS) involves neuroprotection, neuroregeneration, formation of axonal projections, control of cognition, and regulation of metabolism. As GH induces insulin-like growth factor-1 (IGF-1) expression in many tissues, differentiating the specific functions of GH and IGF-1 in the organism is a significant challenge. The actions of GH and IGF-1 in neurons have been more extensively studied than their functions in nonneuronal cells (e.g., microglial cells). Glial cells are fundamentally important to CNS function. Microglia, astrocytes, oligodendrocytes, and tanycytes are essential to the survival, differentiation, and proliferation of neurons. As the interaction of the GH/IGF-1 axis with glial cells merits further exploration, our objective for this review was to summarize and discuss the available literature regarding the genuine effects of GH on glial cells, seeking to differentiate them from the role played by IGF-1 action whenever possible.


Assuntos
Hormônio do Crescimento , Fator de Crescimento Insulin-Like I , Hormônio do Crescimento/farmacologia , Hormônio do Crescimento/fisiologia , Microglia/metabolismo , Astrócitos/metabolismo , Sistema Nervoso Central/metabolismo
2.
Front Physiol ; 14: 1069485, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36909225

RESUMO

Introduction: Chronic hypertension is accompanied by either blood-brain barrier (BBB) leakage and autonomic dysfunction. There is no consensus on the mechanism determining increased BBB permeability within autonomic areas. While some reports suggested tight junction's breakdown, others indicated the involvement of transcytosis rather than paracellular transport changes. Interestingly, exercise training was able to restore both BBB permeability and autonomic control of the circulation. We sought now to clarify the mechanism(s) governing hypertension- and exercise-induced BBB permeability. Methods: Spontaneously hypertensive rats (SHR) and normotensive controls submitted to 4-week aerobic training (T) or sedentary protocol (S) were chronically cannulated for baseline hemodynamic and autonomic recordings and evaluation of BBB permeability. Brains were harvested for measurement of BBB function (FITC-10 kDa leakage), ultrastructural analysis of BBB constituents (transmission electron microscopy) and caveolin-1 expression (immunofluorescence). Results: In SHR-S the increased pressure, augmented sympathetic vasomotor activity, higher sympathetic and lower parasympathetic modulation of the heart and the reduced baroreflex sensitivity were accompanied by robust FITC-10kDa leakage, large increase in transcytotic vesicles number/capillary, but no change in tight junctions' density within the paraventricular nucleus of the hypothalamus, the nucleus of the solitary tract and the rostral ventrolateral medulla. SHR-T exhibited restored BBB permeability and normalized vesicles counting/capillary simultaneously with a normal autonomic modulation of heart and vessels, resting bradycardia and partial pressure reduction. Caveolin-1 expression ratified the counting of transcellular, not other cytoplasmatic vesicles. Additionally, T caused in both groups significant increases in tight junctions' extension/capillary border. Discussion: Data indicate that transcytosis, not the paracellular transport, is the primary mechanism underlying both hypertension- and exercise-induced BBB permeability changes within autonomic areas. The reduced BBB permeability contributes to normalize the autonomic control of the circulation, which suppresses pressure variability and reduces the occurrence of end-organ damage in the trained SHR. Data also disclose that hypertension does not change but exercise training strengthens the resistance of the paracellular pathway in both strains.

3.
J Endocrinol ; 255(2): 75-90, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-35993424

RESUMO

Recent studies indicated an important role of connexins, gap junction proteins, in the regulation of metabolism. However, most of these studies focused on the glial expression of connexins, whereas the actions of connexins in neurons are still poorly investigated. Thus, the present study had the objective to investigate the possible involvement of gap junctions, and in particular connexin 43 (CX43), for the central regulation of energy homeostasis. Initially, we demonstrated that hypothalamic CX43 expression was suppressed in fasted mice. Using whole-cell patch-clamp recordings, we showed that pharmacological blockade of gap junctions induced hyperpolarization and decreased the frequency of action potentials in 50-70% of agouti-related protein (AgRP)-expressing neurons, depending on the blocker used (carbenoxolone disodium, TAT-Gap19 or Gap 26). When recordings were performed with a biocytin-filled pipette, this intercellular tracer was detected in surrounding cells. Then, an AgRP-specific CX43 knockout (AgRPΔCX43) mouse was generated. AgRPΔCX43 mice exhibited no differences in body weight, adiposity, food intake, energy expenditure and glucose homeostasis. Metabolic responses to 24 h fasting or during refeeding were also not altered in AgRPΔCX43 mice. However, AgRPΔCX43 male, but not female mice, exhibited a partial protection against high-fat diet-induced obesity, even though no significant changes in energy intake or expenditure were detected. In summary, our findings indicate that gap junctions regulate the activity of AgRP neurons, and AgRP-specific CX43 ablation is sufficient to mildly prevent diet-induced obesity specifically in males.


Assuntos
Conexina 43 , Obesidade , Proteína Relacionada com Agouti/genética , Proteína Relacionada com Agouti/metabolismo , Animais , Conexina 43/metabolismo , Conexinas/genética , Conexinas/metabolismo , Dieta Hiperlipídica , Junções Comunicantes/metabolismo , Masculino , Camundongos , Neurônios/metabolismo , Obesidade/etiologia , Obesidade/metabolismo
4.
Brain Struct Funct ; 227(7): 2465-2487, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35869304

RESUMO

Anatomical and functional evidence suggests that the PFC is fairly unique among all cortical regions, as it not only receives input from, but also robustly projects back to mesopontine monoaminergic and cholinergic cell groups. Thus, the PFC is in position to exert a powerful top-down control over several state-setting modulatory transmitter systems that are critically involved in the domains of arousal, motivation, reward/aversion, working memory, mood regulation, and stress processing. Regarding this scenario, the origin of cortical afferents to the ventral tegmental area (VTA), laterodorsal tegmental nucleus (LDTg), and median raphe nucleus (MnR) was here compared in rats, using the retrograde tracer cholera toxin subunit b (CTb). CTb injections into VTA, LDTg, or MnR produced retrograde labeling in the cortical mantle, which was mostly confined to frontal polar, medial, orbital, and lateral PFC subdivisions, along with anterior- and mid-cingulate areas. Remarkably, in all of the three groups, retrograde labeling was densest in layer V pyramidal neurons of the infralimbic, prelimbic, medial/ventral orbital and frontal polar cortex. Moreover, a lambda-shaped region around the apex of the rostral pole of the nucleus accumbens stood out as heavily labeled, mainly after injections into the lateral VTA and LDTg. In general, retrograde PFC labeling was strongest following injections into MnR and weakest following injections into VTA. Altogether, our findings reveal a fairly similar set of prefrontal afferents to VTA, LDTg, and MnR, further supporting an eminent functional role of the PFC as a controller of major state-setting mesopontine modulatory transmitter systems.


Assuntos
Núcleos da Rafe , Área Tegmentar Ventral , Animais , Núcleo Accumbens , Córtex Pré-Frontal , Ratos , Recompensa
5.
J Neurosci Methods ; 371: 109500, 2022 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-35151666

RESUMO

BACKGROUND: The generation of animals expressing reporter proteins (e.g., GFP, mCherry or tdTomato) under the control of genes of interest has become a valuable tool in neuroscience. However, the histological reuse of brain sections of these genetically modified animals in unplanned experiments is often infeasible since the constitutive expression of fluorescent reporter proteins interferes with further fluorescent staining procedures. Thus, expensive or time-demanding experiments frequently need to be repeated using additional experimental animals. NEW METHOD: To improve the reuse of tissues of reporter animals for fluorescent staining procedures, we developed fast, inexpensive and simple methods that induce denaturation of constitutively expressed fluorescent proteins in free-floating brain slices. These procedures consist of incubation of brain sections either in a 1% sodium hydroxide alkaline solution (pH 13.0) for one hour at room temperature or at 95 °C for 10-30 min. RESULTS: The strong fluorescence of tdTomato, mCherry and eGFP was completely eliminated after incubation of brain sections of different reporter mice in a pH 13.0 solution for one hour. hrGFP was resistant to denaturation in an alkaline solution, but incubation of brain sections at 95 °C for 10 min eliminated the fluorescence of hrGFP, as well as of tdTomato, mCherry and eGFP. The denaturing procedures did not prevent the reuse of brain tissues in free-floating immunofluorescence staining using multiple antibodies. Furthermore, the quality of the labeling remained unaffected. Although pretreatment in pH 13.0 solution maintained good tissue integrity, as a side effect, brain sections exhibited increased autofluorescence. However, a rinse in 0.25% Sudan Black B solution was efficient in eliminating the autofluorescence without impairing the immunofluorescence staining or DAPI counterstaining. CONCLUSIONS: The present study provides simple procedures capable of inducing denaturation of fluorescent proteins in free-floating brain slices.


Assuntos
Anticorpos , Encéfalo , Animais , Encéfalo/metabolismo , Corantes/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Coloração e Rotulagem
6.
Neurosci Lett ; 752: 135832, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33746008

RESUMO

During fasting or weight loss, the fall in leptin levels leads to suppression of thyrotropin-releasing hormone (TRH) expression in the paraventricular nucleus of the hypothalamus (PVH) and, consequently, inhibition of the hypothalamic-pituitary-thyroid (HPT) axis. However, differently than rats, just few PVHTRH neurons express the leptin receptor in mice. In the present study, male adult rats and mice were submitted to 48 -h fasting to evaluate the consequences on proTRH peptide expression at the PVH level. Additionally, the proTRH peptide expression was also assessed in the brains of leptin-deficient (Lepob/ob) mice. We observed that approximately 50 % of PVHTRH neurons of leptin-injected rats exhibited phosphorylation of the signal transducer and activator of transcription 3 (pSTAT3), a marker of leptin receptor activation. In contrast, very few PVHTRH neurons of leptin-injected mice exhibited pSTAT3. Rats submitted to 48 -h fasting showed a significant reduction in the number of PVHTRH immunoreactive neurons, as compared to fed rats. On the other hand, no changes in the number of PVHTRH immunoreactive neurons were observed between fasted and fed mice. Next, the number of TRH immunoreactive cells was determined in the PVH, dorsomedial nucleus of the hypothalamus and nucleus raphe pallidus of Lepob/ob and wild-type mice and no significant differences were observed, despite reduced plasma T4 levels in Lepob/ob mice. Taken together, these findings provide additional evidence of the important species-specific differences in the mechanisms used by fasting and/or leptin to regulate the HPT axis.


Assuntos
Jejum/metabolismo , Sistema Hipotálamo-Hipofisário/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Hormônio Liberador de Tireotropina/metabolismo , Animais , Leptina/genética , Leptina/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Modelos Animais , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/citologia , Ratos , Especificidade da Espécie , Tiroxina/metabolismo
7.
Cells ; 10(1)2021 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-33440789

RESUMO

Growth hormone (GH) is secreted by the pituitary gland, and in addition to its classical functions of regulating height, protein synthesis, tissue growth, and cell proliferation, GH exerts profound effects on metabolism. In this regard, GH stimulates lipolysis in white adipose tissue and antagonizes insulin's effects on glycemic control. During the last decade, a wide distribution of GH-responsive neurons were identified in numerous brain areas, especially in hypothalamic nuclei, that control metabolism. The specific role of GH action in different neuronal populations is now starting to be uncovered, and so far, it indicates that the brain is an important target of GH for the regulation of food intake, energy expenditure, and glycemia and neuroendocrine changes, particularly in response to different forms of metabolic stress such as glucoprivation, food restriction, and physical exercise. The objective of the present review is to summarize the current knowledge about the potential role of GH action in the brain for the regulation of different metabolic aspects. The findings gathered here allow us to suggest that GH represents a hormonal factor that conveys homeostatic information to the brain to produce metabolic adjustments in order to promote energy homeostasis.


Assuntos
Hormônio do Crescimento/metabolismo , Metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Glucose/metabolismo , Humanos , Neurônios/metabolismo , Receptores da Somatotropina/metabolismo
8.
Eur J Neurosci ; 53(1): 65-88, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-31833616

RESUMO

The habenula (Hb) is a phylogenetically old epithalamic structure differentiated into two nuclear complexes, the medial (MHb) and lateral habenula (LHb). After decades of search for a great unifying function, interest in the Hb resurged when it was demonstrated that LHb plays a major role in the encoding of aversive stimuli ranging from noxious stimuli to the loss of predicted rewards. Consistent with a role as an anti-reward center, aberrant LHb activity has now been identified as a key factor in the pathogenesis of major depressive disorder. Moreover, both MHb and LHb emerged as new players in the reward circuitry by primarily mediating the aversive properties of distinct drugs of abuse. Anatomically, the Hb serves as a bridge that links basal forebrain structures with monoaminergic nuclei in the mid- and hindbrain. So far, research on Hb has focused on the role of the LHb in regulating midbrain dopamine release. However, LHb/MHb are also interconnected with the dorsal (DR) and median (MnR) raphe nucleus. Hence, it is conceivable that some of the habenular functions are at least partly mediated by the complex network that links MHb/LHb with pontomesencephalic monoaminergic nuclei. Here, we summarize research about the topography and transmitter phenotype of the reciprocal connections between the LHb and ventral tegmental area-nigra complex, as well as those between the LHb and DR/MnR. Indirect MHb outputs via interpeduncular nucleus to state-setting neuromodulatory networks will also be commented. Finally, we discuss the role of specific LHb-VTA and LHb/MHb-raphe circuits in anxiety and depression.


Assuntos
Transtorno Depressivo Maior , Habenula , Animais , Dopamina , Núcleos da Rafe , Ratos , Ratos Wistar
9.
J Comp Neurol ; 529(6): 1228-1239, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-32844436

RESUMO

Multiple neuroendocrine, autonomic and behavioral responses are regulated by the paraventricular nucleus of the hypothalamus (PVH). Previous studies have shown that PVH neurons express the growth hormone (GH) receptor (GHR), although the role of GH signaling on PVH neurons is still unknown. Given the great heterogeneity of cell types located in the PVH, we performed a detailed analysis of the neurochemical identity of GH-responsive cells to understand the possible physiological importance of GH action on PVH neurons. GH-responsive cells were detected via the phosphorylated form of the signal transducer and activator of transcription-5 (pSTAT5) in adult male mice that received an intraperitoneal GH injection. Approximately 51% of GH-responsive cells in the PVH co-localized with the vesicular glutamate transporter 2. Rare co-localizations between pSTAT5 and vesicular GABA transporter or vasopressin were observed, whereas approximately 20% and 38% of oxytocin and tyrosine hydroxylase (TH) cells, respectively, were responsive to GH in the PVH. Approximately 55%, 35% and 63% of somatostatin, thyrotropin-releasing hormone (TRH) and corticotropin-releasing hormone (CRH) neurons expressed GH-induced pSTAT5, respectively. Additionally, 8%, 49% and 75% of neuroendocrine TH, TRH and CRH neurons, and 67%, 32% and 74% of nonneuroendocrine TH, TRH and CRH neurons were responsive to GH in the PVH of Fluoro-Gold-injected mice. Our findings suggest that GH action on PVH neurons is involved in the regulation of the thyroid, somatotropic and adrenal endocrine axes, possibly influencing homeostatic and stress responses.


Assuntos
Hormônio do Crescimento/metabolismo , Núcleo Hipotalâmico Paraventricular/química , Núcleo Hipotalâmico Paraventricular/metabolismo , Fenótipo , Receptores da Somatotropina/metabolismo , Animais , Hormônio do Crescimento/análise , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Núcleo Hipotalâmico Paraventricular/citologia , Receptores da Somatotropina/análise
10.
Neuropharmacology ; 182: 108397, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33188843

RESUMO

The present study aimed to evaluate the effects of pharmacological manipulation of α-adrenergic agonists in the dorsal raphe nucleus (DR) on food intake in satiated rats. Adult male Wistar rats with chronically implanted cannula in the DR were injected with adrenaline (AD) or noradrenaline (NA) (both at doses of 6, 20 and 60 nmol), or α-1 adrenergic agonist phenylephrine (PHE) or α-2 adrenergic agonist clonidine (CLO) (both at doses of 6 and 20 nmol). The injections were followed by the evaluation of ingestive behaviors. Food and water intake were evaluated for 60 min. Administration of AD and NA at 60 nmol and CLO at 20 nmol increased food intake and decreased latency to start consumption in satiated rats. The ingestive behavior was not significantly affected by PHE treatment in the DR. CLO treatment increased Fos expression in the arcuate nucleus (ARC) and paraventricular nucleus of the hypothalamus (PVN) in rats that were allowed to eat during the experimental recording (AF group). However, when food was not offered during the experiment (WAF group), PVN neurons were not activated, whereas, neuronal activity remained high in the ARC when compared to control group. Noteworthy, ARC POMC neurons expressed Fos in the AF group. However, double-labeled POMC/Fos cells were absent in the ARC of the WAF group, although an increase in Fos expression was observed in non-POMC cells after CLO injections in the WAF group. In conclusion, the data from the present study highlight that the pharmacological activation of DR α-adrenoceptors affects food intake in satiated rats. The feeding response evoked by CLO injections into DR was similar to that induced by NA or AD injections, suggesting that the hyperphagia after NA or AD treatment depends on α-2 adrenoceptors activation. Finally, we have demonstrated that CLO injections into DR impact neuronal activity in the ARC, possibly evoking a homeostatic response toward food intake.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/administração & dosagem , Clonidina/administração & dosagem , Núcleo Dorsal da Rafe/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Receptores Adrenérgicos alfa 2 , Saciação/efeitos dos fármacos , Animais , Núcleo Dorsal da Rafe/metabolismo , Relação Dose-Resposta a Droga , Ingestão de Alimentos/fisiologia , Injeções Intraventriculares , Masculino , Ratos , Ratos Wistar , Receptores Adrenérgicos alfa 2/metabolismo , Saciação/fisiologia
11.
Brain Res ; 1751: 147189, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33152340

RESUMO

A growth hormone (GH) injection is able to induce the phosphorylated form of the signal transducer and activator of transcription 5 (pSTAT5) in a large number of cells throughout the mouse brain. The present study had the objective to map the distribution of GH-responsive cells in the brain of rats that received an intracerebroventricular injection of GH and compare it to the pattern found in mice. We observed that rats and mice exhibited a similar distribution of GH-induced pSTAT5 in the majority of areas of the telencephalon, hypothalamus and brainstem. However, rats exhibited a higher density of GH-responsive cells than mice in the horizontal limb of the diagonal band of Broca (HDB), supraoptic and suprachiasmatic nuclei, whereas mice displayed more GH-responsive cells than rats in the hippocampus, lateral hypothalamic area and dorsal motor nucleus of the vagus (DMX). Since both HDB and DMX contain acetylcholine-producing neurons, pSTAT5 was co-localized with choline acetyltransferase in GH-injected animals. We found that 50.0 ± 4.5% of cholinergic neurons in the rat HDB coexpressed GH-induced pSTAT5, whereas very few co-localizations were observed in the mouse HDB. In contrast, rats displayed fewer cholinergic neurons responsive to GH in the DMX at the level of the area postrema. In summary, pSTAT5 can be used as a marker of GH-responsive cells in the rat brain. Although rats and mice exhibit a relatively similar distribution of GH-responsive neurons, some species-specific differences exist, as exemplified for the responsiveness to GH in distinct populations of cholinergic neurons.


Assuntos
Mapeamento Encefálico/métodos , Receptores da Somatotropina/análise , Fator de Transcrição STAT5/análise , Acetilcolina , Animais , Encéfalo/metabolismo , Tronco Encefálico/metabolismo , Colina O-Acetiltransferase/metabolismo , Neurônios Colinérgicos/metabolismo , Hormônio do Crescimento/metabolismo , Hormônio do Crescimento/farmacologia , Hipocampo/metabolismo , Hipotálamo/metabolismo , Infusões Intraventriculares , Masculino , Bulbo/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Ratos , Ratos Long-Evans , Receptores da Somatotropina/metabolismo , Fator de Transcrição STAT5/metabolismo
12.
J Neuroendocrinol ; 32(9): e12895, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32840013

RESUMO

Previous studies indicate that leptin regulates the hypothalamic-pituitary-thyroid (HPT) axis via direct and indirect mechanisms. The indirect mechanism involves leptin action in pro-opiomelanocortin (POMC)- and agouti-related peptide (AgRP)-expressing neurones. These cells innervate the paraventricular nucleus of the hypothalamus (PVH) where they modulate hypophysiotrophic thyrotrophin-releasing hormone (TRH)-producing neurones. The direct mechanism involves the expression of leptin receptor (LepR) in a subpopulation of PVH TRH neurones. However, to our knowledge, the existence of LepR in PVH TRH neurones of mice has not been clearly confirmed. Therefore, we investigated possible species-specific differences between rats and mice with respect to the mechanisms recruited by leptin to regulate the HPT axis. We observed that an acute leptin injection induced phosphorylated signal transducer and activator of transcription 3 (pSTAT3), a marker of leptin-responsive cells, in 46.2 ± 8.0% of PVH proTRH immunoreactive neurones in rats. By contrast, an insignificant number of proTRH positive neurones in the mouse PVH co-expressed leptin-induced pSTAT3 or LepR. Similarly, central leptin injection increased the percentage of PVH proTRH neurones containing cAMP response element-binding protein phosphorylation in rats, but not in mice. We investigated the innervation of AgRP and POMC axons in the PVH and observed that rats exhibited a denser POMC innervation in the PVH compared to mice, whereas rats and mice showed similar density of AgRP axons in the PVH. In conclusion, rats and mice exhibit important species-specific differences in the direct and indirect mechanisms used by leptin to regulate the HPT axis.


Assuntos
Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Leptina/farmacologia , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Glândula Tireoide/efeitos dos fármacos , Animais , Sistema Hipotálamo-Hipofisário/fisiologia , Leptina/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Núcleo Hipotalâmico Paraventricular/metabolismo , Ratos , Ratos Long-Evans , Receptores para Leptina/genética , Receptores para Leptina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Especificidade da Espécie , Glândula Tireoide/fisiologia , Hormônio Liberador de Tireotropina/metabolismo
13.
Radiat Oncol ; 14(1): 156, 2019 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-31477165

RESUMO

BACKGROUND: Adjuvant radiotherapy is the standard treatment after breast-conserving surgery. According to meta-analyses, adjuvant 3d-conventional irradiation reduces the risk of local recurrence and thereby improves long-term survival by 5-10%. However, there is an unintended exposure of organs such as the heart, lungs and contralateral breast. Irradiation of the left breast has been related to long-term effects like increased rates of coronary events as well as second cancer induction. Modern radiotherapy techniques such as tangential intensity modulated radiotherapy (t-IMRT) and tangential volumetric modulated arc therapy (t-VMAT) and particularly deep inspiration breath hold (DIBH) technique have been developed in order to improve coverage of target volume and to reduce dose to normal tissue. The aim of this study was to compare t-IMRT-plans with t-VMAT-plans in DIBH position for left-sided breast irradiation in terms of normal tissue exposure, i.e. of lungs, heart, left anterior descending coronary artery (LADCA), as well as homogeneity (HI) and conformity index (CI) and excess absolute risk (EAR) for second cancer induction for organs at risk (OAR) after irradiation. METHODS: Twenty patients, diagnosed with left-sided breast cancer and treated with breast-preserving surgery, were included in this planning study. For each patient DIBH-t-IMRT plan using 5 to 7 beams and t-VMAT plan using four rotations were generated to achieve 95% dose coverage to 95% of the volume. Data were evaluated on the basis of dose-volume histograms: Cardiac dose and LADCA (mean and maximum dose, D25% and D45%), dose to ipsilateral and contralateral lung (mean, D20%, D30%), dose to contralateral breast (mean dose), total monitor units, V5% of total body and normal tissue integral dose (NTID). In addition, homogeneity index and conformity index, as well as the excess absolute risk (EAR) to estimate the risk of second malignancy were calculated. RESULTS: T-IMRT showed a significant reduction in mean cardiac dose of 26% (p = 0.002) compared to t-VMAT, as well as a significant reduction in the mean dose to LADCA of 20% (p = 0.03). Following t-IMRT, mean dose to the left lung was increased by 5% (p = 0.006), whereas no significant difference was found in the mean dose to the right lung and contralateral breast between the two procedures. Monitor units were 31% (p = 0.000004) lower for t-IMRT than for t-VMAT. T-IMRT technique significantly reduced normal tissue integral dose (NTID) by 19% (p = 0.000005) and the V5% of total body by 24% (p = 0.0007). In contrast, t-VMAT improved CI and HI by 2% (p = 0.001) and 0.4% (p = 0.00001), respectively. EAR with t-IMRT was significantly lower, especially for contralateral lung and contralateral breast (2-5/10,000 person years) but not for ipsilateral lung. CONCLUSION: Compared to t-VMAT, t-IMRT in left-sided breast irradiation significantly reduced dose to organs at risk as well as normal tissue integral dose, and V5% total body. EAR with t-IMRT was significantly lower for contralateral lung and contralateral breast. T-VMAT, however, achieved better homogeneity and conformity. This may be relevant in individual cases where sufficient coverage of medial lymphatic target volumes is warranted.


Assuntos
Carcinoma Ductal de Mama/radioterapia , Carcinoma Intraductal não Infiltrante/radioterapia , Órgãos em Risco/efeitos da radiação , Planejamento da Radioterapia Assistida por Computador/métodos , Radioterapia de Intensidade Modulada/métodos , Neoplasias Unilaterais da Mama/radioterapia , Adulto , Idoso , Carcinoma Ductal de Mama/patologia , Carcinoma Intraductal não Infiltrante/patologia , Feminino , Humanos , Pessoa de Meia-Idade , Invasividade Neoplásica , Prognóstico , Dosagem Radioterapêutica , Estudos Retrospectivos , Neoplasias Unilaterais da Mama/patologia
14.
J Comp Neurol ; 527(18): 3046-3072, 2019 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-31199515

RESUMO

The laterodorsal tegmental nucleus (LDTg) is a hindbrain cholinergic cell group thought to be involved in mechanisms of arousal and the control of midbrain dopamine cells. Nowadays, there is increasing evidence that LDTg is also engaged in mechanisms of anxiety/fear and promotion of emotional arousal under adverse conditions. Interestingly, LDTg appears to be connected with other regulators of aversive motivational states, including the lateral habenula (LHb), medial habenula (MHb), interpeduncular nucleus (IP), and median raphe nucleus (MnR). However, the circuitry between these structures has hitherto not been systematically investigated. Here, we placed injections of retrograde or anterograde tracers into LDTg, LHb, IP, and MnR. We also examined the transmitter phenotype of LDTg afferents to IP by combining retrograde tracing with immunofluorescence and in situ hybridization techniques. We found LHb inputs to LDTg mainly emerging from the medial division of the LHb (LHbM), which also receives axonal input from LDTg. The bidirectional connections between IP and LDTg displayed a lateralized organization, with LDTg inputs to IP being predominantly GABAergic or cholinergic and mainly directed to the contralateral IP. Moreover, we disclosed reciprocal LDTg connections with structures involved in the modulation of hippocampal theta rhythm including MnR, nucleus incertus, and supramammillary nucleus. Our findings indicate that the habenula is linked with LDTg either by direct reciprocal projections from/to LHbM or indirectly via the MHb-IP axis, supporting a functional role of LDTg in the regulation of aversive behaviors, and further characterizing LHb as a master controller of ascending brainstem state-setting modulatory projection systems.


Assuntos
Habenula/fisiologia , Núcleo Interpeduncular/fisiologia , Núcleos da Rafe/fisiologia , Rombencéfalo/fisiologia , Animais , Habenula/química , Núcleo Interpeduncular/química , Masculino , Vias Neurais/química , Vias Neurais/fisiologia , Técnicas de Rastreamento Neuroanatômico/métodos , Núcleos da Rafe/química , Ratos , Ratos Wistar , Rombencéfalo/química
15.
Brain Res ; 1717: 136-146, 2019 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-31009611

RESUMO

The central melanocortin system is composed of neurons that express either the proopiomelanocortin (POMC) or the agouti-related protein (AgRP). POMC is cleaved in bioactive peptides, including the α-melanocyte-stimulating hormone (α-MSH). α-MSH activates the melanocortin-4 receptor (MC4R) inducing satiety, whereas AgRP acts as an inverse agonist of MC4R. However, only limited information is available regarding possible area-specific differences in the interaction between α-MSH and AgRP terminals on MC4R-expressing cells. Therefore, the objective of the present study was to compare the distribution pattern of α-MSH and AgRP terminals on the perikarya of MC4R-expressing neurons. We performed a triple-label immunofluorescence reaction in brain series of MC4R-reporter mice to visualize MC4R-expressing neurons together with AgRP and α-MSH terminals. POMC and AgRP neurons project to areas that contain MC4R-expressing cells, although several brain nuclei exhibit AgRP and α-MSH terminals, but they do no express MC4R, while other brain areas contain MC4R-expressing cells and receive no apparent innervation of AgRP and POMC neurons. AgRP terminals make more presumptive appositions than α-MSH on the soma of MC4R-expressing neurons of the medial preoptic area and paraventricular nucleus of the hypothalamus (Pa). Additionally, a higher percentage of MC4R cells receive at least one presumptive apposition from AgRP terminals in the median preoptic nucleus and Pa, compared to α-MSH appositions. Thus, our study revealed area-specific differences in the interaction between α-MSH and AgRP terminals and the soma of MC4R-expressing neurons. These findings provide new insights about the relationship between first- and second-order neurons of the central melanocortin system.


Assuntos
Proteína Relacionada com Agouti/metabolismo , Receptor Tipo 4 de Melanocortina/metabolismo , alfa-MSH/metabolismo , Animais , Axônios/metabolismo , Encéfalo/metabolismo , Ingestão de Alimentos/fisiologia , Metabolismo Energético/fisiologia , Hipotálamo/metabolismo , Leptina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo
16.
Nat Commun ; 10(1): 662, 2019 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-30737388

RESUMO

Weight loss triggers important metabolic responses to conserve energy, especially via the fall in leptin levels. Consequently, weight loss becomes increasingly difficult with weight regain commonly occurring in most dieters. Here we show that central growth hormone (GH) signaling also promotes neuroendocrine adaptations during food deprivation. GH activates agouti-related protein (AgRP) neurons and GH receptor (GHR) ablation in AgRP cells mitigates highly characteristic hypothalamic and metabolic adaptations induced by weight loss. Thus, the capacity of mice carrying an AgRP-specific GHR ablation to save energy during food deprivation is impaired, leading to increased fat loss. Additionally, administration of a clinically available GHR antagonist (pegvisomant) attenuates the fall of whole-body energy expenditure of food-deprived mice, similarly as seen by leptin treatment. Our findings indicate GH as a starvation signal that alerts the brain about energy deficiency, triggering key adaptive responses to conserve limited fuel stores.


Assuntos
Proteína Relacionada com Agouti/metabolismo , Receptores da Somatotropina/metabolismo , Proteína Relacionada com Agouti/genética , Animais , Peso Corporal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Metabolismo Energético/efeitos dos fármacos , Feminino , Hormônio do Crescimento/metabolismo , Hormônio do Crescimento/farmacologia , Hormônio do Crescimento Humano/análogos & derivados , Hormônio do Crescimento Humano/uso terapêutico , Leptina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores da Somatotropina/genética , Redução de Peso/efeitos dos fármacos
17.
Nat Commun ; 10(1): 980, 2019 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-30804339

RESUMO

The original version of this Article contained an error in the spelling of the author J. Donato Jr, which was incorrectly given as Donato J. Jr. This has now been corrected in both the PDF and HTML versions of the Article.

18.
Neurosci Lett ; 681: 12-16, 2018 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-29772257

RESUMO

A small neuronal subpopulation in the medial nucleus of the amygdala (MeA), expressing the Kiss1 gene, is now considered an important mediator that integrates socio-sexual behavior and odor information in order to modulate the Hypothalamic-Pituitary-Gonadal (HPG) axis. Previous studies demonstrated that exogenous kisspeptin administration or selective activation of Kiss1-expressing neurons in the MeA modulates the onset of puberty, LH secretion and sexual behavior. These functions are supported by the known MeA neuronal connections. In the MeA, as well as in the hypothalamus, Kiss1 mRNA expression mostly depends on sex steroids levels. However, the percentage of Kiss1-expressing cells that co-express estrogen receptor α (ERα) in the MeA is currently unknown. Additionally, whether MeA kisspeptin neurons show Fos expression due to pheromone exposure is still undisclosed. In the present study, we used adult male and female mice that express a reporter protein under the Kiss1 promoters to determine the percentage of Kiss1-expressing neurons that co-express the ERα in the MeA and, whether those cells are activated by olfactory cues. We found a high percentage of Kiss1-expressing neurons in the MeA co-expressing the ERα. The proportion of co-expression was similar between male and female mice in diestrus. Interestingly, a low percentage of Kiss1-expressing neurons in the MeA co-express Fos after conspecific odor exposure, despite a significant increase of Fos positive cells in the MeA. Additionally, odor exposition leads to a sexually dimorphic change in Kiss1 expression in the posterior subdivision of the MeA. Our findings suggest that olfactory signals predominantly activate non-kisspeptin cells in the MeA to modulate responses to pheromones and therefore the HPG axis.


Assuntos
Tonsila do Cerebelo/citologia , Tonsila do Cerebelo/metabolismo , Kisspeptinas/biossíntese , Atrativos Sexuais/administração & dosagem , Tonsila do Cerebelo/efeitos dos fármacos , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/metabolismo , Odorantes
19.
Brain Struct Funct ; 223(5): 2229-2241, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29460051

RESUMO

The signal transducer and activator of transcription 5 (STAT5) is a transcription factor recruited by numerous cytokines. STAT5 is important for several physiological functions, including body and tissue growth, mammary gland development, immune system and lipid metabolism. However, the role of STAT5 signaling for brain functions is still poorly investigated, especially regarding cognitive aspects. Therefore, the objective of the present study was to investigate whether brain STAT5 signaling modulates learning and memory formation. For this purpose, brain-specific STAT5 knockout (STAT5 KO) mice were studied in well-established memory tests. Initially, we confirmed a robust reduction in STAT5a and STAT5b mRNA levels in different brain structures of STAT5 KO mice. STAT5 KO mice showed no significant alterations in metabolism, growth, somatotropic axis and spontaneous locomotor activity. In contrast, brain-specific STAT5 ablation impaired learning and memory formation in the novel object recognition, Barnes maze and contextual fear conditioning tests. To unravel possible mechanisms that might underlie the memory deficits of STAT5 KO mice, we assessed neurogenesis in the hippocampus, but no significant differences were observed between groups. On the other hand, reduced insulin-like growth factor-1 (IGF-1) mRNA expression was found in the hippocampus and hypothalamus of STAT5 KO mice. These findings collectively indicate that brain STAT5 signaling is required to attain normal learning and memory. Therefore, STAT5 is an important downstream cellular mechanism shared by several cytokines to regulate cognitive functions.


Assuntos
Encéfalo/metabolismo , Regulação da Expressão Gênica/genética , Aprendizagem em Labirinto/fisiologia , Reconhecimento Psicológico/fisiologia , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais/fisiologia , Animais , Condicionamento Psicológico , Citocinas/metabolismo , Comportamento Exploratório/fisiologia , Medo/psicologia , Fator de Crescimento Insulin-Like I/metabolismo , Deficiências da Aprendizagem/genética , Camundongos , Camundongos Transgênicos , Nestina/genética , Nestina/metabolismo , Neurogênese/genética , RNA Mensageiro/metabolismo , Tempo de Reação/genética , Fator de Transcrição STAT5/genética
20.
Strahlenther Onkol ; 193(10): 800-811, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28646251

RESUMO

BACKGROUND: Given the reduction in death from breast cancer, as well as improvements in overall survival, adjuvant radiotherapy is considered the standard treatment for breast cancer. However, left-sided breast irradiation was associated with an increased rate of fatal cardiovascular events due to incidental irradiation of the heart. Recently, considerable efforts have been made to minimize cardiac toxicity of left-sided breast irradiation by new treatment methods such as deep-inspiration breath-hold (DIBH) and new radiation techniques, particularly intensity modulated radiotherapy (IMRT) and volumetric modulated arc therapy (VMAT). The primary aim of this study was to evaluate the effect of DIBH irradiation on cardiac dose compared with free-breathing (FB) irradiation, while the secondary objective was to compare the advantages of IMRT versus VMAT plans in both the FB and the DIBH position for left-sided breast cancer. METHODS: In all, 25 consecutive left-sided breast cancer patients underwent CT simulation in the FB and DIBH position. Five patients were excluded with no cardiac displacement following DIBH-CT simulation. The other 20 patients were irradiated in the DIBH position using respiratory gating. Four different treatment plans were generated for each patient, an IMRT and a VMAT plan in the DIBH and in the FB position, respectively. The following parameters were used for plan comparison: dose to the heart, left anterior descending coronary artery (mean dose, maximum dose, D25% and D45%), ipsilateral, contralateral lung (mean dose, D20%, D30%) and contralateral breast (mean dose). The percentage in dose reduction for organs at risk achieved by DIBH for both IMRT and VMAT plans was calculated and compared for each patient by each treatment plan. RESULTS: DIBH irradiation significantly reduced mean dose to the heart and left anterior descending coronary artery (LADCA) using both IMRT (heart -20%; p = 0.0002, LADCA -9%; p = 0.001) and VMAT (heart -23%; p = 0.00003, LADCA -16%; p = 0.01) techniques as compared with FB radiation. There were no significant changes in left lung dose by IMRT; however, with VMAT planning, mean dose to the left lung was reduced by -4% (p = 0.0004). In addition, DIBH significantly increased the mean dose to the contralateral breast with IMRT (+14%, p = 0.002) and significantly reduced the dose to the contralateral breast with VMAT planning (-9%, p = 0.003) compared with the FB position. Additionally, in comparison with VMAT, the IMRT technique reduced mean heart dose both in the FB and the DIBH-position by -30% (p = 0.0004) and -26% (p = 0.002), respectively. Furthermore, IMRT increased the mean dose to the left lung in both the FB and the DIBH position (+5%, p = 0.003, p = 0.006), respectively. There were no significant changes in dose to the right lung and contralateral breast either in the FB or DIBH position between IMRT and VMAT techniques. CONCLUSION: Left-sided breast irradiation is best performed in the DIBH position, since a considerable dose sparing to the heart and LADCA can be achieved by using either IMRT or VMAT techniques. A significant additional decrease in heart and LADCA dose by IMRT in both FB and DIBH irradiation was seen compared with VMAT.


Assuntos
Traumatismos Cardíacos/prevenção & controle , Tratamentos com Preservação do Órgão/métodos , Exposição à Radiação/prevenção & controle , Lesões por Radiação/prevenção & controle , Radioterapia Conformacional/métodos , Radioterapia de Intensidade Modulada/métodos , Neoplasias Unilaterais da Mama/radioterapia , Adulto , Idoso , Suspensão da Respiração , Feminino , Humanos , Pessoa de Meia-Idade , Exposição à Radiação/análise , Dosagem Radioterapêutica , Radioterapia Conformacional/efeitos adversos , Radioterapia de Intensidade Modulada/efeitos adversos , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...