Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 752
Filter
Add more filters

Complementary Medicines
Therapeutic Methods and Therapies TCIM
Country/Region as subject
Publication year range
1.
Science ; 382(6669): 388-394, 2023 10 27.
Article in English | MEDLINE | ID: mdl-37883552

ABSTRACT

The hypothalamus ("hypo" meaning below, and "thalamus" meaning bed) consists of regulatory circuits that support basic life functions that ensure survival. Sitting at the interface between peripheral, environmental, and neural inputs, the hypothalamus integrates these sensory inputs to influence a range of physiologies and behaviors. Unlike the neocortex, in which a stereotyped cytoarchitecture mediates complex functions across a comparatively small number of neuronal fates, the hypothalamus comprises upwards of thousands of distinct cell types that form redundant yet functionally discrete circuits. With single-cell RNA sequencing studies revealing further cellular heterogeneity and modern photonic tools enabling high-resolution dissection of complex circuitry, a new era of hypothalamic mapping has begun. Here, we provide a general overview of mammalian hypothalamic organization, development, and connectivity to help welcome newcomers into this exciting field.


Subject(s)
Hypothalamus , Neurogenesis , Animals , Hypothalamus/physiology , Hypothalamus/ultrastructure , Mammals , Neocortex/cytology , Neocortex/physiology , Neurons/physiology , Thalamus/physiology , Single-Cell Gene Expression Analysis
2.
J Comp Neurol ; 529(3): 553-575, 2021 02.
Article in English | MEDLINE | ID: mdl-32515035

ABSTRACT

Tanycytes are highly specialized ependymal cells that line the bottom and the lateral walls of the third ventricle. In contact with the cerebrospinal fluid through their cell bodies, they send processes into the arcuate nucleus, the ventromedial nucleus, and the dorsomedial nucleus of the hypothalamus. In the present work, we combined transgenic and immunohistochemical approaches to investigate the neuroanatomical associations between tanycytes and neural cells present in the hypothalamic parenchyma, in particular in the arcuate nucleus. The specific expression of tdTomato in tanycytes first allowed the observation of peculiar subcellular protrusions along tanycyte processes and at their endfeet such as spines, swelling, en passant boutons, boutons, or claws. Interestingly, these protrusions contact different neural cells in the brain parenchyma including blood vessels and neurons, and in particular NPY and POMC neurons in the arcuate nucleus. Using both fluorescent and electron microscopy, we finally observed that these tanycyte protrusions contain ribosomes, mitochondria, diverse vesicles, and transporters, suggesting dense tanycyte/neuron and tanycyte/blood vessel communications. Altogether, our results lay the neuroanatomical basis for tanycyte/neural cell interactions, which will be useful to further understand cell-to-cell communications involved in the regulation of neuroendocrine functions.


Subject(s)
Ependymoglial Cells/ultrastructure , Hypothalamus/ultrastructure , Neurons/ultrastructure , Parenchymal Tissue/ultrastructure , Animals , Ependymoglial Cells/chemistry , Guinea Pigs , Humans , Hypothalamus/chemistry , Hypothalamus/cytology , Male , Mice , Mice, Transgenic , Neurons/chemistry , Parenchymal Tissue/chemistry , Parenchymal Tissue/cytology , Rabbits
3.
Cell Rep ; 29(13): 4349-4361.e4, 2019 12 24.
Article in English | MEDLINE | ID: mdl-31875545

ABSTRACT

In addition to their support role in neurotransmitter and ion buffering, astrocytes directly regulate neurotransmission at synapses via local bidirectional signaling with neurons. Here, we reveal a form of neuronal-astrocytic signaling that transmits retrograde dendritic signals to distal upstream neurons in order to activate recurrent synaptic circuits. Norepinephrine activates α1 adrenoreceptors in hypothalamic corticotropin-releasing hormone (CRH) neurons to stimulate dendritic release, which triggers an astrocytic calcium response and release of ATP; ATP stimulates action potentials in upstream glutamate and GABA neurons to activate recurrent excitatory and inhibitory synaptic circuits to the CRH neurons. Thus, norepinephrine activates a retrograde signaling mechanism in CRH neurons that engages astrocytes in order to extend dendritic volume transmission to reach distal presynaptic glutamate and GABA neurons, thereby amplifying volume transmission mediated by dendritic release.


Subject(s)
Adrenergic alpha-Agonists/pharmacology , Astrocytes/drug effects , Dendrites/drug effects , GABAergic Neurons/drug effects , Norepinephrine/pharmacology , Synaptic Transmission/drug effects , Action Potentials/drug effects , Action Potentials/physiology , Animals , Astrocytes/metabolism , Astrocytes/ultrastructure , Cell Communication , Channelrhodopsins/genetics , Channelrhodopsins/metabolism , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Dendrites/metabolism , Dendrites/ultrastructure , GABAergic Neurons/metabolism , GABAergic Neurons/ultrastructure , Gene Expression Regulation , Glutamic Acid/metabolism , Glutamic Acid/pharmacology , Hypothalamus/drug effects , Hypothalamus/metabolism , Hypothalamus/ultrastructure , Male , Mice , Mice, Transgenic , Microtomy , Receptors, Corticotropin/genetics , Receptors, Corticotropin/metabolism , Synapses/drug effects , Synapses/physiology , Synaptic Transmission/physiology , Tissue Culture Techniques , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/pharmacology
4.
Sci Rep ; 9(1): 17373, 2019 11 22.
Article in English | MEDLINE | ID: mdl-31758009

ABSTRACT

Animal studies suggest that obesity-related diets induce structural changes in the hypothalamus, a key brain area involved in energy homeostasis. Whether this translates to humans is however largely unknown. Using a novel multimodal approach with manual segmentation, we here show that a higher body mass index (BMI) selectively predicted higher proton diffusivity within the hypothalamus, indicative of compromised microstructure in the underlying tissue, in a well-characterized population-based cohort (n1 = 338, 48% females, age 21-78 years, BMI 18-43 kg/m²). Results were independent from confounders and confirmed in another independent sample (n2 = 236). In addition, while hypothalamic volume was not associated with obesity, we identified a sexual dimorphism and larger hypothalamic volumes in the left compared to the right hemisphere. Using two large samples of the general population, we showed that a higher BMI specifically relates to altered microstructure in the hypothalamus, independent from confounders such as age, sex and obesity-associated co-morbidities. This points to persisting microstructural changes in a key regulatory area of energy homeostasis occurring with excessive weight. Our findings may help to better understand the pathomechanisms of obesity and other eating-related disorders.


Subject(s)
Body Mass Index , Hypothalamus/diagnostic imaging , Hypothalamus/ultrastructure , Obesity/diagnostic imaging , Adult , Aged , Cohort Studies , Energy Metabolism/physiology , Feeding and Eating Disorders/etiology , Feeding and Eating Disorders/metabolism , Feeding and Eating Disorders/pathology , Female , Humans , Hypothalamus/pathology , Magnetic Resonance Imaging , Male , Middle Aged , Neuroimaging , Obesity/complications , Obesity/pathology , Obesity/psychology , Organ Size , Sex Characteristics , Young Adult
5.
J Therm Biol ; 81: 110-117, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30975407

ABSTRACT

The hypothalamus is crucial to ensure the functionality of the entire organisms, such as body temperature, feed intake and energy regulation. Exposing broilers to high ambient temperature usually induces lower feed intake and energy imbalance. We investigated the molecular mechanisms by which heat stress impairs the appetite via dysfunction in hypothalamus of the broilers. Broilers were allocated to three groups: the normal control (NC) group, and fed ad libitum; heat-stress (HS) group, and fed ad libitum; pair-fed (PF) group, which received the feed intake equal to HS group. Experiment lasted from the age of 28 to 42 d. The results showed that HS increased the head surface temperature of broiler and changed hypothalamic ultrastructure. HS treatment also increased the serum corticosterone in the broilers after 7 days of heat stress, elevated the FT4 and FT3 after 14 days of heat stress. Heat stress of 14 days showed a tendency to increase the leptin. However, the serum corticosterone in the HS group had no significant difference after 14 days of heat stress. In addition, HS treatment decreased the expression of orexigenic gene neuropeptide Y (NPY) after 14 days of heat stress, while HS treatment had no effect on the reactive oxygen species (ROS), as well as the gene expression of AMPKα1 and LKB1 in the hypothalamus. In conclusion, HS increased the surface temperature of head in broiler, and then altered the integrity of hypothalamus. Meanwhile, HS increased the serum corticosterone which may ascribe to the activation of HPA axis in the broilers. In addition, chronic heat stress decreased the expression of orexigenic gene NPY, which may cause the broiler to reduce feed intake.


Subject(s)
Appetite/genetics , Chickens/physiology , Heat-Shock Response , Hypothalamus/pathology , Hypothalamus/physiopathology , Animals , Body Temperature , Chickens/blood , Chickens/genetics , Head/physiology , Hot Temperature , Hypothalamus/ultrastructure , Male , Neurons/pathology , Neurons/ultrastructure , Reactive Oxygen Species/metabolism
6.
Environ Toxicol Pharmacol ; 64: 139-146, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30391875

ABSTRACT

Mancozeb, a dithiocarbamate widely used in agriculture, is considered a developmental hazard in humans; however, more evidences are still needed concerning the consequences of chronic exposure to this pesticide. Mancozeb neurotoxicity in developing mouse hypothalamus was evaluated by subchronic exposure of male Mus musculus mice to low and high doses of mancozeb (30 and 90 mg/kg body weight, respectively) from late neonatal until adolescence. Variations in hypothalamic amino acid neurotransmitter levels and changes in histological as well as cytological characteristics were analyzed in young adult experimental mice and compared with control. A dose-dependent increase in excitation/ inhibition ratio was observed in mancozeb-exposed hypothalamus, indicating an overall state of excitoxicity. Histopathological and ultrastructural studies showed increased apoptosis, neuroinflammation and demyelination, demonstrating mancozeb-induced cytotoxicity in hypothalamic neurosecretory cells. In summary, both neurochemical and morphological data revealed mancozeb-induced alterations during development of hypothalamic circuitry that are critical for maturation of the neuroendocrine system.


Subject(s)
Fungicides, Industrial/toxicity , Hypothalamus/drug effects , Maneb/toxicity , Zineb/toxicity , Animals , Hypothalamus/metabolism , Hypothalamus/pathology , Hypothalamus/ultrastructure , Male , Mice , Microscopy, Electron, Transmission , Neurotransmitter Agents/metabolism
7.
J Intern Med ; 284(6): 568-580, 2018 12.
Article in English | MEDLINE | ID: mdl-30027599

ABSTRACT

The mammalian hypothalamus contains an astounding heterogeneity of neurons to achieve its role in coordinating central responses to virtually any environmental stressor over the life-span of an individual. Therefore, while core features of intrahypothalamic neuronal modalities and wiring patterns are stable during vertebrate evolution, integration of the hypothalamus into hierarchical brain-wide networks evolved to coordinate its output with emotionality, cognition and conscious decision-making. The advent of single-cell technologies represents a recent milestone in the study of hypothalamic organization by allowing the dissection of cellular heterogeneity and establishing causality between opto- and chemogenetic activity modulation of molecularly-resolved neuronal contingents and specific behaviours. Thus, organizational rules to accumulate an unprecedented variety of hierarchical neuroendocrine command networks into a minimal brain volume are being unravelled. Here, we review recent understanding at nanoscale resolution on how neuronal heterogeneity in the mammalian hypothalamus underpins the diversification of hormonal and synaptic output and keeps those sufficiently labile for continuous adaptation to meet environmental demands. Particular emphasis is directed towards the dissection of neuronal circuitry for aggression and food intake. Mechanistic data encompass cell identities, synaptic connectivity within and outside the hypothalamus to link vegetative and conscious levels of innate behaviours, and context- and circadian rhythm-dependent rules of synaptic neurophysiology to distinguish hypothalamic foci that either tune the body's metabolic set-point or specify behaviours. Consequently, novel insights emerge to explain the evolutionary advantages of non-laminar organization for neuroendocrine circuits coincidently using fast neurotransmitters and neuropeptides. These are then accrued into novel therapeutic principles that meet therapeutic criteria for human metabolic diseases.


Subject(s)
Hypothalamus/ultrastructure , Animals , Humans , Hypothalamus/anatomy & histology , Hypothalamus/cytology , Hypothalamus/physiology , Neurons/physiology , Neurons/ultrastructure , Single-Cell Analysis
8.
Mol Cell Endocrinol ; 460: 238-245, 2018 01 15.
Article in English | MEDLINE | ID: mdl-28760600

ABSTRACT

Hypothalamic dysfunction is a common feature of experimental obesity. Studies have identified at least three mechanisms involved in the development of hypothalamic neuronal defects in diet-induced obesity: i, inflammation; ii, endoplasmic reticulum stress; and iii, mitochondrial abnormalities. However, which of these mechanisms is activated earliest in response to the consumption of large portions of dietary fats is currently unknown. Here, we used immunoblot, real-time PCR, mitochondrial respiration assays and transmission electron microscopy to evaluate markers of inflammation, endoplasmic reticulum stress and mitochondrial abnormalities in the hypothalamus of Swiss mice fed a high-fat diet for up to seven days. In the present study we show that the expression of the inflammatory chemokine fractalkine was the earliest event detected. Its hypothalamic expression increased as early as 3 h after the introduction of a high-fat diet and was followed by the increase of cytokines. GPR78, an endoplasmic reticulum chaperone, was increased 6 h after the introduction of a high-fat diet, however the actual triggering of endoplasmic reticulum stress was only detected three days later, when IRE-1α was increased. Mitofusin-2, a protein involved in mitochondrial fusion and tethering of mitochondria to the endoplasmic reticulum, underwent a transient reduction 24 h after the introduction of a high-fat diet and then increased after seven days. There were no changes in hypothalamic mitochondrial respiration during the experimental period, however there were reductions in mitochondria/endoplasmic reticulum contact sites, beginning three days after the introduction of a high-fat diet. The inhibition of TNF-α with infliximab resulted in the normalization of mitofusin-2 levels 24 h after the introduction of the diet. Thus, inflammation is the earliest mechanism activated in the hypothalamus after the introduction of a high-fat diet and may play a mechanistic role in the development of mitochondrial abnormalities in diet-induced obesity.


Subject(s)
Hypothalamus/pathology , Inflammation/pathology , Mitochondria/pathology , Obesity/pathology , Animals , Biomarkers/metabolism , Diet, High-Fat , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , GTP Phosphohydrolases/metabolism , Hypothalamus/ultrastructure , Mice , Mitochondria/ultrastructure , Neutralization Tests , Tumor Necrosis Factor-alpha/metabolism
9.
Neuro Endocrinol Lett ; 38(1): 27-37, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28456145

ABSTRACT

OBJECTIVES: The hypothalamus regulates metabolism and feeding behavior by perceiving the levels of peripheral insulin. However, little is known about the hypothalamic changes after aberrant metabolism. In this study, we investigated the changes of insulin and autophagy relevant signals of hypothalamus under diabetes mellitus. METHODS: C57B/L mice were injected with low-dose streptozotocin (STZ) and fed with high-fat diet to induce type 2 diabetes mellitus. In vitro, PC12 cells were treated with oleic acid to mimic lipotoxicity. RESULTS: Results showed that the cholesterol level in the hypothalamus of the diabetic mice was higher than that of the normal mice. The expression of insulin receptors and insulin receptor substrate-1 were downregulated and the number of Fluoro-Jade C positive cells significantly increased in the hypothalamic arcuate nucleus of the diabetic mice. Furthermore, Upregulation of mammalian target of rapamycin (mTOR) and downregulation of LC 3II were obvious in the hypothalamus of the diabetic mice. In vitro, results showed that high-lipid caused PC12 cell damage and upregulated LC3 II expression. Pretreatment of cells with 3-methyladenine evidently downregulated LC3 II expression and aggravated PC12 cell death under high lipid conditions. By contrast, pretreatment of cells with rapamycin upregulated LC3 II expression and ameliorated PC12 cell death caused by lipotoxicity. CONCLUSION: These results demonstrate that autophagy activation confers protection to neurons under aberrant metabolism and that autophagy dysfunction in the hypothalamus occurs in the chronic metabolic disorder such as T2DM.


Subject(s)
Autophagy , Brain Diseases/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Hypothalamus/metabolism , Neurons/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/metabolism , Arcuate Nucleus of Hypothalamus/ultrastructure , Autophagy/drug effects , Blotting, Western , Cholesterol/metabolism , Diet, High-Fat , Down-Regulation , Glucose Tolerance Test , Hypothalamus/drug effects , Hypothalamus/ultrastructure , Immunosuppressive Agents/pharmacology , In Vitro Techniques , Insulin , Insulin Receptor Substrate Proteins/metabolism , Insulin Resistance , Lipid Metabolism/drug effects , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/drug effects , Microtubule-Associated Proteins/metabolism , Neurons/drug effects , Neurons/ultrastructure , Oleic Acid/pharmacology , PC12 Cells , Rats , Receptor, Insulin/metabolism , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism , Up-Regulation , Ventromedial Hypothalamic Nucleus/drug effects , Ventromedial Hypothalamic Nucleus/metabolism , Ventromedial Hypothalamic Nucleus/ultrastructure
10.
Toxicon ; 130: 19-28, 2017 May.
Article in English | MEDLINE | ID: mdl-28237716

ABSTRACT

Clostridium perfringens epsilon toxin (ETX), the most potent toxin produced by this bacteria, plays a key role in the pathogenesis of enterotoxaemia in ruminants, causing brain edema and encephalomalacia. Studies of animals suffering from ETX intoxication describe severe neurological disorders that are thought to be the result of vasogenic brain edemas and indirect neuronal toxicity, killing oligodendrocytes but not astrocytes, microglia, or neurons in vitro. In this study, by means of intravenous and intracerebroventricular delivery of sub-lethal concentrations of ETX, the histological and ultrastructural changes of the brain were studied in rats and mice. Histological analysis showed degenerative changes in neurons from the cortex, hippocampus, striatum and hypothalamus. Ultrastructurally, necrotic neurons and apoptotic cells were observed in these same areas, among axons with accumulation of neurofilaments and demyelination as well as synaptic stripping. Lesions observed in the brain after sub-lethal exposure to ETX, result in permanent behavioral changes in animals surviving ETX exposure, as observed individually in several animals and assessed in the Inclined Plane Test and the Wire Hang Test. Pharmacological studies showed that dexamethasone and reserpine but not ketamine or riluzole were able to reduce the brain lesions and the lethality of ETX. Cytotoxicity was not observed upon neuronal primary cultures in vitro. Therefore, we hypothesize that ETX can affect the brain of animals independently of death, producing changes on neurons or glia as the result of complex interactions, independently of ETX-BBB interactions.


Subject(s)
Bacterial Toxins/toxicity , Brain/drug effects , Animals , Apoptosis/drug effects , Behavior, Animal/drug effects , Brain/pathology , Brain/ultrastructure , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Cerebral Cortex/ultrastructure , Corpus Striatum/drug effects , Corpus Striatum/pathology , Corpus Striatum/ultrastructure , Demyelinating Diseases/chemically induced , Dexamethasone/therapeutic use , Female , Hippocampus/drug effects , Hippocampus/pathology , Hippocampus/ultrastructure , Hypothalamus/drug effects , Hypothalamus/pathology , Hypothalamus/ultrastructure , Intermediate Filaments/drug effects , Ketamine/therapeutic use , Lethal Dose 50 , Male , Mice , Neurons/drug effects , Neurons/pathology , Neurons/ultrastructure , Neuroprotective Agents/therapeutic use , Rats , Rats, Sprague-Dawley , Reserpine/therapeutic use , Riluzole/therapeutic use , Synapses/drug effects
11.
Sci Rep ; 7: 40768, 2017 01 20.
Article in English | MEDLINE | ID: mdl-28106069

ABSTRACT

The periventricular zone of cerebellum is a germinative niche during the embryonic development, nevertheless its structural organization and functional implications in adult have not been widely studied. Here we disclose the presence of two novel clusters of cells in that area. The first one was named the subventricular cellular cluster (SVCC) and is composed of cells that express glial and neuronal markers. The second was named the ventromedial cord (VMC) and appears as a streak of biciliated cells with microvillosities facing the ventricle, that includes GFAP+ and nestin+ cells organized along the periventricular vasculature. The dorsal limit of the SVCC is associated with myelinated axons of neurons of unknown origin. This paper describes the characteristics and organization of these groups of cells. They can be observed from late embryonic development in the transgenic mouse line GFAP-GFP. The SVCC and VMC expand during early postnatal development but are restricted to the central area of the ventricle in adulthood. We did not find evidence of cell proliferation, cell migration or the presence of fenestrated blood vessels. These findings provide new insights into the knowledge of the cellular composition and structural organization of the periventricular zone of cerebellum.


Subject(s)
Cerebellum/cytology , Cerebellum/metabolism , Hypothalamus/cytology , Hypothalamus/metabolism , Animals , Biomarkers , Cell Proliferation , Cerebellum/physiopathology , Cerebellum/ultrastructure , Electrophysiological Phenomena , Fluorescent Antibody Technique , Genes, Reporter , Hypothalamus/physiopathology , Hypothalamus/ultrastructure , Male , Mice , Mice, Transgenic , Patch-Clamp Techniques
12.
J Neurovirol ; 23(2): 205-215, 2017 04.
Article in English | MEDLINE | ID: mdl-27739033

ABSTRACT

Semliki Forest virus (SFV), a neurotropic virus, has been used to deliver heterologous genes into cells in vitro and in vivo. In this study, we constructed a reporter SFV4-FL-EGFP and found that it can deliver EGFP into neurons located at the injection site without disseminating throughout the brain. Lacking of the capsid gene of SFV4-FL-EGFP does not block its life cycle, while forming replication-competent virus-like particles (VLPs). These VLPs hold subviral genome by using the packaging sequence (PS) located within the nsP2 gene, and can transfer their genome into cells. In addition, we found that the G protein of vesicular stomatitis virus (VSVG) can package SFV subviral genome, which is consistent with the previous reports. The G protein of rabies virus (RVG) could also package SFV subviral genome. These pseudo-typed SFV can deliver EGFP gene into neurons. Taken together, these findings may be used to construct various SFV-based delivery systems for virological studies, gene therapy, and neural circuit labeling.


Subject(s)
Genetic Engineering , Genetic Therapy/methods , Genetic Vectors/metabolism , Hypothalamus/virology , Neurons/virology , Semliki forest virus/genetics , Animals , Cell Line , Cricetulus , Epithelial Cells/ultrastructure , Epithelial Cells/virology , Gene Expression , Genes, Reporter , Genetic Vectors/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hypothalamus/ultrastructure , Injections, Intraventricular , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Neurons/ultrastructure , Primary Cell Culture , Rabies virus/genetics , Rabies virus/metabolism , Semliki forest virus/metabolism , Vesiculovirus/genetics , Vesiculovirus/metabolism , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Virion/genetics , Virion/metabolism , Virus Assembly/genetics
13.
J Biol Regul Homeost Agents ; 30(3): 883-888, 2016.
Article in English | MEDLINE | ID: mdl-27655516

ABSTRACT

The 77 amino prepropeptide apelin has been isolated from bovine stomach tissue and several smaller fragments, including apelin-13, showed high affinity for the orphan APJ receptor. The distribution of apelinergic fibers and receptors in the hypothalamus may suggest a role of apelin-13 on energy balance regulation, albeit the studies reporting the acute effects of apelin on feeding control are inconsistent. Considering the possible involvement of apelinergic system on hypothalamic appetite controlling network, in the present study we evaluated in the rat the effects of intrahypothalamic apelin-13 injection on food intake and the involvement of orexigenic and anorexigenic hypothalamic peptides and neurotransmitters. Eighteen rats (6 for each group of treatment) were injected into the ARC with either vehicle or apelin-13 (1-2 µg/rat). Food intake and hypothalamic peptide and neurotransmitter levels were evaluated 2 and 24 h after injection. Compared to vehicle, apelin-13 administration increased food intake both 2 and 24 h following treatment. This effect could be related to inhibited cocaine- and amphetamine-regulated transcript (CART) gene expression and serotonin (5-hydroxytryptamine, 5-HT) synthesis and release, and increased orexin A gene expression in the hypothalamus.


Subject(s)
Appetite/drug effects , Arcuate Nucleus of Hypothalamus/drug effects , Feeding Behavior/drug effects , Intercellular Signaling Peptides and Proteins/therapeutic use , Animals , Appetite/physiology , Arcuate Nucleus of Hypothalamus/physiology , Electric Stimulation , Feeding Behavior/physiology , Gene Expression Regulation/drug effects , Hypothalamus/metabolism , Hypothalamus/ultrastructure , Injections , Intercellular Signaling Peptides and Proteins/administration & dosage , Male , Motor Activity/drug effects , Neuropeptides/genetics , Neuropeptides/physiology , Neurotransmitter Agents/genetics , Neurotransmitter Agents/physiology , Rats , Rats, Sprague-Dawley , Serotonin/physiology , Synaptosomes/metabolism
14.
Sci Rep ; 6: 23777, 2016 Mar 31.
Article in English | MEDLINE | ID: mdl-27029812

ABSTRACT

Mammalian pituitaries exhibit a high degree of intercellular coordination; this enables them to mount large-scale coordinated responses to various physiological stimuli. This type of communication has not been adequately demonstrated in teleost pituitaries, which exhibit direct hypothalamic innervation and expression of luteinizing hormone (LH) and follicle-stimulating hormone (FSH) in distinct cell types. We found that in two fish species, namely tilapia and zebrafish, LH cells exhibit close cell-cell contacts and form a continuous network throughout the gland. FSH cells were more loosely distributed but maintained some degree of cell-cell contact by virtue of cytoplasmic processes. These anatomical differences also manifest themselves at the functional level as evidenced by the effect of gap-junction uncouplers on gonadotropin release. These substances abolished the LH response to gonadotropin-releasing hormone stimulation but did not affect the FSH response to the same stimuli. Dye transfer between neighboring LH cells provides further evidence for functional coupling. The two gonadotropins were also found to be differently packaged within their corresponding cell types. Our findings highlight the evolutionary origin of pituitary cell networks and demonstrate how the different levels of cell-cell coordination within the LH and FSH cell populations are reflected in their distinct secretion patterns.


Subject(s)
Gap Junctions/metabolism , Gonadotrophs/metabolism , Hypothalamus/metabolism , Tilapia/physiology , Zebrafish/physiology , Animals , Biological Evolution , Cell Communication/drug effects , Female , Fluorescent Dyes/metabolism , Follicle Stimulating Hormone/genetics , Follicle Stimulating Hormone/metabolism , Gap Junctions/drug effects , Gap Junctions/ultrastructure , Gene Expression Regulation , Gonadotrophs/drug effects , Gonadotrophs/ultrastructure , Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/drug effects , Hypothalamus/ultrastructure , Isoquinolines/metabolism , Luteinizing Hormone/genetics , Luteinizing Hormone/metabolism , Meclofenamic Acid/pharmacology , Signal Transduction , Tilapia/anatomy & histology , Zebrafish/anatomy & histology
15.
J Diabetes Res ; 2015: 484721, 2015.
Article in English | MEDLINE | ID: mdl-26199947

ABSTRACT

Currently there has been no effective treatment of diabetic encephalopathy. Radix Polygoni Multiflori, a famous traditional Chinese medicine, is widely used in antiaging treatment, especially in prevention and treatment of Alzheimer's diseases. In this study we tried to explore the effect of Radix Polygoni Multiflori on cognitive function among diabetic rats with demonstrated cognitive impairment. SD rats were divided into group A (control group), group B (diabetes), group C (treated with Radix Polygoni Multiflori at the dose of 2 g/kg/d), and group D (treated with same drug at the dose of 1 g/kg/d). The results showed that 8 weeks of Radix Polygoni Multiflori treatment could improve the cognitive dysfunction of diabetic rats (P < 0.01), recover the ultrastructure of hippocampal neurons, and increase the number of synapses in a dose-dependent manner. Further experiment also suggested that the neuroprotective effect of Radix Polygoni Multiflori was partly achieved by downregulating MLCK expression in hippocampus via ERK signaling.


Subject(s)
Brain Diseases, Metabolic/prevention & control , Diabetes Mellitus, Type 1/drug therapy , Diabetic Neuropathies/prevention & control , Drugs, Chinese Herbal/therapeutic use , Hypothalamus/drug effects , Myosin-Light-Chain Kinase/antagonists & inhibitors , Neuroprotective Agents/therapeutic use , Animals , Brain Diseases, Metabolic/complications , Cognition/drug effects , Cognition Disorders/complications , Cognition Disorders/prevention & control , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Drugs, Chinese Herbal/administration & dosage , Hypothalamus/metabolism , Hypothalamus/ultrastructure , MAP Kinase Signaling System/drug effects , Male , Maze Learning/drug effects , Microscopy, Electron, Transmission , Myosin-Light-Chain Kinase/chemistry , Myosin-Light-Chain Kinase/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/ultrastructure , Neuroprotective Agents/administration & dosage , Nootropic Agents/therapeutic use , Rats, Sprague-Dawley
16.
J Neurophysiol ; 114(2): 1008-21, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26063780

ABSTRACT

Gonadotropin-releasing hormone (GnRH) controls mammalian reproduction via the hypothalamic-pituitary-gonadal (hpg) axis, acting on gonadotrope cells in the pituitary gland that express the GnRH receptor (GnRHR). Cells expressing the GnRHR have also been identified in the brain. However, the mechanism by which GnRH acts on these potential target cells remains poorly understood due to the difficulty of visualizing and identifying living GnRHR neurons in the central nervous system. We have developed a mouse strain in which GnRHR neurons express a fluorescent marker, enabling the reliable identification of these cells independent of the hormonal status of the animal. In this study, we analyze the GnRHR neurons of the periventricular hypothalamic nucleus in acute brain slices prepared from adult female mice. Strikingly, we find that the action potential firing pattern of these neurons alternates in synchrony with the estrous cycle, with pronounced burst firing during the preovulatory period. We demonstrate that GnRH stimulation is sufficient to trigger the conversion from tonic to burst firing in GnRHR neurons. Furthermore, we show that this switch in the firing pattern is reversed by a potent GnRHR antagonist. These data suggest that endogenous GnRH acts on GnRHR neurons and triggers burst firing in these cells during late proestrus and estrus. Our data have important clinical implications in that they indicate a novel mode of action for GnRHR agonists and antagonists in neurons of the central nervous system that are not part of the classical hpg axis.


Subject(s)
Action Potentials/physiology , Estrous Cycle/physiology , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/physiology , Action Potentials/drug effects , Animals , Capillaries/ultrastructure , Estrous Cycle/drug effects , Female , Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/pharmacology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hormone Antagonists/pharmacology , Hypothalamus/blood supply , Hypothalamus/drug effects , Hypothalamus/ultrastructure , Immunohistochemistry , Mice, Transgenic , Microscopy, Confocal , Microscopy, Electron , Neurons/drug effects , Neurons/physiology , Neurons/ultrastructure , Receptors, LHRH/antagonists & inhibitors , Receptors, LHRH/metabolism , Tissue Culture Techniques
17.
Neuropeptides ; 50: 29-33, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25796089

ABSTRACT

Fos immunocytochemistry is a valuable anatomical mapping tool for distinguishing cells within complex tissues that undergo genomic activation, but it is seldom paired with corroborative molecular analytical techniques. Due to preparatory requirements that include protein cross-linking for specimen sectioning, histological tissue sections are regarded as unsuitable for those methods. Our studies show that pharmacological activation of the hindbrain energy sensor AMPK by AICAR elicits estradiol (E)-dependent patterns of Fos immunolabeling of hypothalamic metabolic loci. Here, Western blotting was applied to hypothalamic tissue removed from histological sections of E- versus oil (O)-implanted ovariectomized (OVX) female rat brain to measure levels of metabolic transmitters associated with Fos-positive structures. In both E and O rats, AICAR treatment elicited alterations in pro-opiomelanocortin, neuropeptide Y, SF-1, and orexin-A neuropeptide expression that coincided with patterns of Fos labeling of structures containing neurons that synthesize these neurotransmitters, e.g. arcuate and ventromedial nuclei and lateral hypothalamic area. O, but not E animals also exhibited parallel augmentation of tissue corticotropin-releasing hormone neuropeptide levels and paraventricular nucleus Fos staining. Data demonstrate the utility of immunoblot analysis as a follow-through technique to capitalize on Fos mapping of transactivation sites in the brain. Findings that induction of Fos immunoreactivity coincides with adjustments in hypothalamic metabolic neuropeptide expression affirms that this functional indicator reflects changes in neurotransmission in pathways governing metabolic outflow.


Subject(s)
Blotting, Western/methods , Histological Techniques/methods , Hypothalamus/metabolism , Nerve Tissue Proteins/analysis , Neuropeptides/biosynthesis , Proto-Oncogene Proteins c-fos/analysis , AMP-Activated Protein Kinases/metabolism , Aminoimidazole Carboxamide/administration & dosage , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Corticotropin-Releasing Hormone/analysis , Estradiol/pharmacology , Female , Hypothalamus/ultrastructure , Injections, Intraventricular , Neuropeptides/analysis , Ovariectomy , Rats , Ribonucleotides/administration & dosage , Ribonucleotides/pharmacology , Specimen Handling , Transcriptional Activation
18.
Alcohol Alcohol ; 50(1): 51-5, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25371045

ABSTRACT

AIMS: To clarify the effect of chronic alcohol consumption on the brain histaminergic neurons in rats. METHODS: Male Wistar rats were given 20% ethanol as the only source of drinking during 6 months, control rats had a free access to water. The samples of hypothalamus were prepared for light and electron microscopy accompanied by morphometry to examine the brain histaminergic neurons of E2 group. RESULTS: Chronic ethanol consumption increased the amount of histologically abnormal forms of histaminergic neurons and decreased the whole amount of E2 histaminergic neurons (for 5%). The neuron bodies and nuclei increased in size and sphericity, the nuclear/cytoplasmic ratio decreased by 15%. The ultrastructural changes in histaminergic neurons demonstrate the activation of their nuclear apparatus, both destruction and hypertrophy and hyperplasia of organelles, especially lysosomes. Chronic ethanol consumption induces the disturbances in cytoplasmic enzymes of neurons: increases the activity of type B monoamine oxidase, dehydrogenases of lactate and NADH and, especially, marker enzyme of lysosomes acid phosphatase as well as inhibits the activity of dehydrogenases of succinate and glucose-6-phosphate. CONCLUSION: Chronic alcohol consumption affects significantly the structure and metabolism of the brain histaminergic neurons, demonstrating both the neurotoxic effect of ethanol and processes of adaptation in those neurons, necessary for their survival.


Subject(s)
Alcohol Drinking/adverse effects , Brain/drug effects , Histamine , Hypothalamus/drug effects , Neurons/drug effects , Animals , Brain/cytology , Brain/pathology , Brain/ultrastructure , Histamine/physiology , Hypothalamus/cytology , Hypothalamus/pathology , Hypothalamus/ultrastructure , Male , Microscopy, Electron , Monoamine Oxidase/metabolism , NADH Dehydrogenase/metabolism , Neurons/cytology , Neurons/ultrastructure , Rats , Rats, Wistar
19.
Am J Alzheimers Dis Other Demen ; 30(5): 478-87, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25380804

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disorder, characterized by irreversible decline of mental faculties, emotional and behavioral changes, loss of motor skills, and dysfunction of autonomic nervous system and disruption of circadian rhythms (CRs). We attempted to describe the morphological findings of the hypothalamus in early cases of AD, focusing our study mostly on the suprachiasmatic nucleus (SCN), the supraoptic nucleus (SON), and the paraventricular nucleus (PVN). Samples were processed for electron microscopy and silver impregnation techniques. The hypothalamic nuclei demonstrated a substantial decrease in the neuronal population, which was particularly prominent in the SCN. Marked abbreviation of dendritic arborization, in association with spinal pathology, was also seen. The SON and PVN demonstrated a substantial number of dystrophic axons and abnormal spines. Alzheimer's pathology, such as deposits of amyloid-ß peptide and neurofibrillary degeneration, was minimal. Electron microscopy revealed mitochondrial alterations in the cell body and the dendritic branches. The morphological alterations of the hypothalamic nuclei in early cases of AD may be related to the gradual alteration of CRs and the instability of autonomic regulation.


Subject(s)
Alzheimer Disease/pathology , Neurons/ultrastructure , Paraventricular Hypothalamic Nucleus/ultrastructure , Suprachiasmatic Nucleus/ultrastructure , Supraoptic Nucleus/ultrastructure , Aged , Aged, 80 and over , Case-Control Studies , Dendritic Spines/ultrastructure , Female , Golgi Apparatus/ultrastructure , Humans , Hypothalamus/ultrastructure , Male , Microscopy, Electron , Middle Aged , Mitochondria/ultrastructure , Silver Staining
20.
Med Sci (Paris) ; 30(11): 1034-9, 2014 Nov.
Article in French | MEDLINE | ID: mdl-25388586

ABSTRACT

The primary cilium is a specialized organelle, present at the surface of most eukaryotic cells, whose main function is to detect, integrate and transmit intra- and extra-cellular signals. Its dysfunction usually results in a group of severe clinical manifestations nowadays termed ciliopathies. The latter can be of syndromic nature with multi-organ dysfunctions and can also be associated with a morbid obese phenotype, like it is the case in the iconic ciliopathy, the Bardet Biedl syndrome (BBS). This review will discuss the contribution of the unique context offered by the emblematic BBS for understanding the mechanisms leading to obesity via the involvement of the primary cilium together with identification of novel molecular players and signaling pathways it has helped to highlight. In the current context of translational medicine and system biology, this article will also discuss the potential benefits and challenges posed by these techniques via multi-level approaches to better dissect the underlying mechanisms leading to the complex condition of obesity.


Subject(s)
Bardet-Biedl Syndrome/pathology , Chaperonins/deficiency , Cilia/physiology , Ciliary Motility Disorders/pathology , Microtubule-Associated Proteins/deficiency , Obesity/physiopathology , Adipose Tissue/pathology , Animals , Bardet-Biedl Syndrome/genetics , Bone Marrow/pathology , Chaperonins/genetics , Chaperonins/physiology , Ciliary Motility Disorders/genetics , Disease Models, Animal , Endocrine System/physiopathology , Genes, Recessive , Humans , Hypothalamus/physiopathology , Hypothalamus/ultrastructure , Mice , Mice, Knockout , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/physiology , Models, Biological , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Obesity/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Syndrome , Weight Gain
SELECTION OF CITATIONS
SEARCH DETAIL