Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Annu Rev Neurosci ; 42: 1-26, 2019 07 08.
Article in English | MEDLINE | ID: mdl-30735460

ABSTRACT

Peripheral endocrine output relies on either direct or feed-forward multi-order command from the hypothalamus. Efficient coding of endocrine responses is made possible by the many neuronal cell types that coexist in intercalated hypothalamic nuclei and communicate through extensive synaptic connectivity. Although general anatomical and neurochemical features of hypothalamic neurons were described during the past decades, they have yet to be reconciled with recently discovered molecular classifiers and neurogenetic function determination. By interrogating magnocellular as well as parvocellular dopamine, GABA, glutamate, and phenotypically mixed neurons, we integrate available information at the molecular, cellular, network, and endocrine output levels to propose a framework for the comprehensive classification of hypothalamic neurons. Simultaneously, we single out putative neuronal subclasses for which future research can fill in existing gaps of knowledge to rationalize cellular diversity through function-determinant molecular marks in the hypothalamus.


Subject(s)
Hypothalamus/cytology , Neurons/classification , Animals , Connectome , Humans , Hypothalamic Hormones/analysis , Nerve Net/ultrastructure , Neurons/cytology , Neurons/metabolism , Neurotransmitter Agents/analysis , Peptide Hormones/analysis , Single-Cell Analysis
2.
Biochem Biophys Res Commun ; 528(4): 628-635, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32505354

ABSTRACT

Phoenixin (PNX) is a newly discovered peptide produced by proteolytic cleavage of a small integral membrane protein 20 (Smim20), which acts as an important regulator of energy homeostasis and reproduction. Since dysfunction of reproduction is characteristic in polycystic ovarian syndrome (PCOS), the role of PNX in pathogenesis of PCOS needs further investigation. The objective of this study was to determine expression of Smim20, PNX-14 and its receptor GRP173 in the hypothalamus, ovary and periovarian adipose tissue (PAT) of letrozole induced PCOS rats. Phosphorylation of extracellular signal-regulated kinase (ERK1/2), protein kinases A (PKA) and B (Akt) were also estimated. We observed that PCOS rats had high weight gain and a number of ovarian cyst, high levels of testosterone, luteinizing hormone and PNX-14, while low estradiol. Smim20 mRNA expression was higher in the ovary and PAT, while PNX-14 peptide production was higher only in the ovary of PCOS rat. Moreover, in PCOS rats Gpr173 level was lower in PAT but at the protein level increased only in the ovary. Depending on the tissues, kinases phosphorylation were significantly differ in PCOS rats. Our results showed higher levels of PNX-14 in PCOS rats and indicated some novel findings regarding the mechanisms of PCOS pathophysiology.


Subject(s)
Adipose Tissue/pathology , Hypothalamic Hormones/analysis , Hypothalamus/pathology , Ovary/pathology , Peptide Hormones/analysis , Polycystic Ovary Syndrome/pathology , Receptors, G-Protein-Coupled/analysis , Animals , Female , Rats , Rats, Wistar
3.
Biochem Biophys Res Commun ; 485(2): 342-348, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28216160

ABSTRACT

Hypothalamic peptides, gonadotropin-releasing hormone (GnRH) and gonadotropin inhibitory hormone (GnIH), play pivotal roles in the control of reproduction and gonadal maturation in fish. In the present study we tested the possibility that stress-mediated reproductive dysfunction in teleost may involve changes in GnRH and GnIH activity. We studied expression of brain GnIH, GnIH-R, seabream GnRH (sbGnRH), as well as circulating levels of follicle stimulating hormone (FSH), and luteinizing hormone (LH) in the cinnamon clownfish, Amphiprion melanopus. Treatment with cortisol increased GnIH mRNA level, but reduced sbGnRH mRNA and circulating levels of LH and FSH in cinnamon clownfish. Using double immunofluorescence staining, we found expression of both GnIH and GnRH in the diencephalon region of cinnamon clownfish brain. These findings support the hypothesis that cortisol, an indicator of stress, affects reproduction, in part, by increasing GnIH in cinnamon clownfish which contributes to hypothalamic suppression of reproductive function in A. melanopus, a protandrous hermaphroditic fish.


Subject(s)
Gonadotropin-Releasing Hormone/genetics , Hydrocortisone/metabolism , Hypothalamic Hormones/genetics , Perciformes/physiology , Animals , Brain/physiology , Follicle Stimulating Hormone/blood , Gene Expression Regulation, Developmental , Gonadotropin-Releasing Hormone/analysis , Hypothalamic Hormones/analysis , Luteinizing Hormone/blood , Perciformes/blood , Perciformes/genetics , RNA, Messenger/genetics , Reproduction , Stress, Physiological
4.
Biochem Biophys Res Commun ; 475(2): 189-93, 2016 06 24.
Article in English | MEDLINE | ID: mdl-27208779

ABSTRACT

The present study aimed to determine the relationship between melatonin and gonadotropin-inhibitory hormone (GnIH) and their effect on reproduction in cinnamon clownfish, Amphiprion melanopus. Accordingly, we investigated the expression pattern of GnIH, GnIH receptor (GnIH-R), and melatonin receptor (MT-R1) mRNA and protein, as well as the plasma levels of melatonin, during sex change in cinnamon clownfish. We found that GnIH and MT-R1 mRNA and melatonin activity were higher in fish with mature brain than in fish with developing gonads, and using double immunofluorescence staining, we found that both GnIH and MT-R1 proteins were co-expressed in the hypothalamus of cinnamon clownfish. These findings support the hypothesis that melatonin plays an important role in the negative regulation of maturation and GnIH regulation during reproduction.


Subject(s)
Fish Proteins/metabolism , Hypothalamic Hormones/metabolism , Melatonin/metabolism , Perciformes/growth & development , Receptors, Melatonin/metabolism , Sexual Behavior, Animal , Animals , Female , Fish Proteins/analysis , Fish Proteins/genetics , Gene Expression Regulation, Developmental , Hypothalamic Hormones/analysis , Hypothalamic Hormones/genetics , Hypothalamus/growth & development , Hypothalamus/metabolism , Male , Melatonin/analysis , Melatonin/blood , Melatonin/genetics , Perciformes/blood , Perciformes/metabolism , RNA, Messenger/genetics , Receptors, Melatonin/analysis , Receptors, Melatonin/genetics , Sexual Development
5.
Anal Bioanal Chem ; 407(2): 497-507, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25358911

ABSTRACT

Gonadotropin-inhibitory hormones (GnIH) and gonadotropin-releasing hormones (GnRH) are neuropeptides essential for the regulation of reproduction in all vertebrate animals examined. Determination of neuropeptides in the biological sample is highly challenging due to their complex matrix and weak stability. The wide variety of peptides or protein degradation products often interferes with the determination of the target peptide. This study aims to develop a specific ultra-high performance liquid chromatography-tandem mass spectrometry method for simultaneous determination of nine critical neuropeptides in biological samples. A separation method by ultra-performance liquid chromatography coupled to a multiple reaction monitoring (MRM) by tandem mass spectrometry allows the selective determination of the neuropeptides in brain and plasma matrices after solid-phase extraction. Specific MSMS transitions were optimized using MRM of multiple-charged peptides generated by electrospray ionization in positive mode. The resulting analytical method was fully validated with thorough evaluation of stability, recovery, matrix effect, and intra- and interday accuracy and precision in sea lamprey brain and plasma. The optimized method shows linearity in a wide range of concentrations with limit of quantification ranging from 0.1 to 0.75 ng/mL. With slight modification, this method can be applied to other biological samples.


Subject(s)
Chromatography, High Pressure Liquid/methods , Gonadotropin-Releasing Hormone/analysis , Tandem Mass Spectrometry/methods , Amino Acid Sequence , Animals , Brain Chemistry , Female , Gonadotropin-Releasing Hormone/blood , Hypothalamic Hormones/analysis , Lampreys/blood , Limit of Detection , Male , Molecular Sequence Data , Neuropeptides/analysis , Neuropeptides/blood , Reproducibility of Results , Sensitivity and Specificity , Solid Phase Extraction , Spectrometry, Mass, Electrospray Ionization/methods
6.
J Comp Neurol ; 529(5): 929-956, 2021 04 01.
Article in English | MEDLINE | ID: mdl-32678476

ABSTRACT

As stressful environment is a potent modulator of feeding, we seek in the present work to decipher the neuroanatomical basis for an interplay between stress and feeding behaviors. For this, we combined anterograde and retrograde tracing with immunohistochemical approaches to investigate the patterns of projections between the dorsomedial division of the bed nucleus of the stria terminalis (BNST), well connected to the amygdala, and hypothalamic structures such as the paraventricular (PVH) and dorsomedial (DMH), the arcuate (ARH) nuclei and the lateral hypothalamic areas (LHA) known to control feeding and motivated behaviors. We particularly focused our study on afferences to proopiomelanocortin (POMC), agouti-related peptide (AgRP), melanin-concentrating-hormone (MCH) and orexin (ORX) neurons characteristics of the ARH and the LHA, respectively. We found light to intense innervation of all these hypothalamic nuclei. We particularly showed an innervation of POMC, AgRP, MCH and ORX neurons by the dorsomedial and dorsolateral divisions of the BNST. Therefore, these results lay the foundation for a better understanding of the neuroanatomical basis of the stress-related feeding behaviors.


Subject(s)
Amygdala/anatomy & histology , Hypothalamus/anatomy & histology , Mice/anatomy & histology , Neural Pathways/anatomy & histology , Septal Nuclei/anatomy & histology , Agouti-Related Protein/analysis , Animals , Axonal Transport , Feeding Behavior/physiology , Feeding Behavior/psychology , Hypothalamic Hormones/analysis , Luminescent Proteins/analysis , Male , Melanins/analysis , Mice, Inbred C57BL , Nerve Tissue Proteins/analysis , Neurons/chemistry , Neurons/classification , Neurons/ultrastructure , Orexins/analysis , Phytohemagglutinins/analysis , Pituitary Hormones/analysis , Proprotein Convertases/analysis , Rabies virus , Species Specificity , Tyrosine 3-Monooxygenase/analysis , Red Fluorescent Protein
7.
Neuropharmacology ; 178: 108270, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32795460

ABSTRACT

Melanin-concentrating hormone (MCH) is an orexigenic neuropeptide produced in the lateral hypothalamus and zona incerta that increases food intake. The neuronal pathways and behavioral mechanisms mediating the orexigenic effects of MCH are poorly understood, as is the extent to which MCH-mediated feeding outcomes are sex-dependent. Here we investigate the hypothesis that MCH-producing neurons act in the nucleus accumbens shell (ACBsh) to promote feeding behavior and motivation for palatable food in a sex-dependent manner. We utilized ACBsh MCH receptor (MCH1R)-directed pharmacology as well as a dual virus chemogenetic approach to selectively activate MCH neurons that project to the ACBsh. Results reveal that both ACBsh MCH1R activation and activating ACBsh-projecting MCH neurons increase consumption of standard chow and palatable sucrose in male rats without affecting motivated operant responding for sucrose, general activity levels, or anxiety-like behavior. In contrast, food intake was not affected in female rats by either ACBsh MCH1R activation or ACBsh-projecting MCH neuron activation. To determine a mechanism for this sexual dimorphism, we investigated whether the orexigenic effect of ACBsh MCH1R activation is reduced by endogenous estradiol signaling. In ovariectomized female rats on a cyclic regimen of either estradiol (EB) or oil vehicle, ACBsh MCH1R activation increased feeding only in oil-treated rats, suggesting that EB attenuates the ability of ACBsh MCH signaling to promote food intake. Collective results show that MCH ACBsh signaling promotes feeding in an estrogen- and sex-dependent manner, thus identifying novel neurobiological mechanisms through which MCH and female sex hormones interact to influence food intake.


Subject(s)
Feeding Behavior/physiology , Hypothalamic Hormones/metabolism , Melanins/metabolism , Nucleus Accumbens/metabolism , Pituitary Hormones/metabolism , Sex Characteristics , Signal Transduction/physiology , Animals , Feeding Behavior/psychology , Female , Hypothalamic Hormones/analysis , Male , Melanins/analysis , Neural Pathways/chemistry , Neural Pathways/metabolism , Nucleus Accumbens/chemistry , Pituitary Hormones/analysis , Rats , Rats, Sprague-Dawley
8.
Brain Res ; 1741: 146872, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32360868

ABSTRACT

Melanin-concentrating hormone (MCH) is a neuropeptide primarily transcribed in the lateral hypothalamus (LH), with vast projections to many areas throughout the central nervous system that play an important role in motivated behaviors and drug use. Anatomical, pharmacological and genetic studies implicate MCH in mediating the intake and reinforcement of commonly abused substances, acting by influencing several systems including the mesolimbic dopaminergic system, glutamatergic as well as GABAergic signaling and being modulated by inflammatory neuroimmune pathways. Further support for the role of MCH in controlling behavior related to drug use will be discussed as it relates to cerebral ventricular volume transmission and intracellular molecules including cocaine- and amphetamine-regulated transcript peptide, dopamine- and cAMP-regulated phosphoprotein 32 kDa. The primary goal of this review is to introduce and summarize current literature surrounding the role of MCH in mediating the intake and reinforcement of commonly abused drugs, such as alcohol, cocaine, amphetamine, nicotine and opiates.


Subject(s)
Brain/metabolism , Hypothalamic Hormones/metabolism , Melanins/metabolism , Neuroimmunomodulation/physiology , Pituitary Hormones/metabolism , Substance-Related Disorders/metabolism , Substance-Related Disorders/psychology , Animals , Brain Chemistry , Humans , Hypothalamic Hormones/analysis , Melanins/analysis , Neural Pathways/chemistry , Neural Pathways/metabolism , Neurons/chemistry , Neurons/metabolism , Neuropeptides/analysis , Neuropeptides/metabolism , Pituitary Hormones/analysis
9.
Neuroimmunomodulation ; 16(5): 272-83, 2009.
Article in English | MEDLINE | ID: mdl-19571588

ABSTRACT

The hypothalamic-pituitary-adrenal (HPA) axis plays an important role in the maintenance of basal and stress-related homeostasis. The hypothalamus controls the secretion of adrenocorticotropic hormone (ACTH) from the anterior pituitary, which in turn stimulates the secretion of glucocorticoids from the adrenal cortex. Glucocorticoids, the final effectors of the HPA axis, regulate a broad spectrum of physiologic functions essential for life and exert their effects through their ubiquitously distributed intracellular receptors. Alterations in the activity of the HPA axis may present with symptoms and signs of glucocorticoid deficiency or excess. Detailed endocrinologic evaluation is of primary importance in determining the diagnosis and/or etiology of the underlying condition. We review the most common endocrinologic investigations used in the evaluation of the HPA axis integrity and function.


Subject(s)
Diagnostic Techniques, Endocrine/trends , Endocrine System Diseases/diagnosis , Glucocorticoids/metabolism , Hypothalamo-Hypophyseal System/physiopathology , Neurosecretory Systems/physiopathology , Pituitary-Adrenal System/physiopathology , Adolescent , Child , Clinical Laboratory Techniques/standards , Clinical Laboratory Techniques/trends , Diagnostic Techniques, Endocrine/standards , Endocrine System Diseases/physiopathology , Glucocorticoids/analysis , Humans , Hypothalamic Hormones/analysis , Hypothalamic Hormones/metabolism , Hypothalamo-Hypophyseal System/growth & development , Neurosecretory Systems/growth & development , Pituitary-Adrenal Function Tests/methods , Pituitary-Adrenal Function Tests/standards , Pituitary-Adrenal Function Tests/trends , Pituitary-Adrenal System/growth & development
10.
J Comp Neurol ; 527(18): 2973-3001, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31152440

ABSTRACT

Melanin-concentrating hormone (MCH) is a conserved neuropeptide, predominantly located in the diencephalon of vertebrates, and associated with a wide range of functions. While functional studies have focused on the use of the traditional mouse laboratory model, critical gaps exist in our understanding of the morphology of the MCH system in this species. Even less is known about the nontraditional animal model Neotomodon alstoni (Mexican volcano mouse). A comparative morphological study among these rodents may, therefore, contribute to a better understanding of the evolution of the MCH peptidergic system. To this end, we employed diverse immunohistochemical protocols to identify key aspects of the MCH system, including its spatial relationship to another neurochemical population of the tuberal hypothalamus, the orexins. Three-dimensional (3D) reconstructions were also employed to convey a better sense of spatial distribution to these neurons. Our results show that the distribution of MCH neurons in all rodents studied follows a basic plan, but individual characteristics are found for each species, such as the preeminence of a periventricular group only in the rat, the lack of posterior groups in the mouse, and the extensive presence of MCH neurons in the anterior hypothalamic area of Neotomodon. Taken together, these data suggest a strong anatomical substrate for previously described functions of the MCH system, and that particular neurochemical and morphological features may have been determinant to species-specific phenotypes in rodent evolution.


Subject(s)
Hypothalamic Hormones/metabolism , Hypothalamus/cytology , Hypothalamus/metabolism , Melanins/metabolism , Melanophores/metabolism , Pituitary Hormones/metabolism , Animals , Female , Hypothalamic Hormones/analysis , Hypothalamus/chemistry , Male , Melanins/analysis , Mice , Mice, Inbred C57BL , Phylogeny , Pituitary Hormones/analysis , Rats , Rats, Sprague-Dawley , Species Specificity
11.
Brain ; 130(Pt 6): 1586-95, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17491094

ABSTRACT

It has recently been reported that Parkinson's disease (PD) is preceded and accompanied by daytime sleep attacks, nocturnal insomnia, REM sleep behaviour disorder, hallucinations and depression, symptoms which are frequently as troublesome as the motor symptoms of PD. All these symptoms are present in narcolepsy, which is linked to a selective loss of hypocretin (Hcrt) neurons. In this study, the Hcrt system was examined to determine if Hcrt cells are damaged in PD. The hypothalamus of 11 PD (mean age 79 +/- 4) and 5 normal (mean age 77 +/- 3) brains was examined. Sections were immunostained for Hcrt-1, melanin concentrating hormone (MCH) and alpha synuclein and glial fibrillary acidic protein (GFAP). The substantia nigra of 10 PD brains and 7 normal brains were used for a study of neuromelanin pigmented cell loss. The severity of PD was assessed using the Hoehn and Yahr scale and the level of neuropathology was assessed using the Braak staging criteria. Cell number, distribution and size were determined with stereologic techniques on a one in eight series. We found an increasing loss of hypocretin cells with disease progression. Similarly, there was an increased loss of MCH cells with disease severity. Hcrt and MCH cells were lost throughout the anterior to posterior extent of their hypothalamic distributions. The percentage loss of Hcrt cells was minimal in stage I (23%) and was maximal in stage V (62%). Similarly, the percentage loss of MCH cells was lowest in stage I (12%) and was highest in stage V (74%). There was a significant increase (P = 0.0006, t = 4.25, df = 15) in the size of neuromelanin containing cells in PD patients, but no difference in the size of surviving Hcrt (P = 0.18, t = 1.39, df = 14) and MCH (P = 0.28, t = 1.39, df = 14) cells relative to controls. In summary, we found that PD is characterized by a massive loss of Hcrt neurons. Thus, the loss of Hcrt cells may be a cause of the narcolepsy-like symptoms of PD and may be ameliorated by treatments aimed at reversing the Hcrt deficit. We also saw a substantial loss of hypothalamic MCH neurons. The losses of Hcrt and MCH neurons are significantly correlated with the clinical stage of PD, not disease duration, whereas the loss of neuromelanin cells is significantly correlated only with disease duration. The significant correlations that we found between the loss of Hcrt and MCH neurons and the clinical stage of PD, in contrast to the lack of a relationship of similar strength between loss of neuromelanin containing cells and the clinical symptoms of PD, suggests a previously unappreciated relationship between hypothalamic dysfunction and the time course of the overall clinical picture of PD.


Subject(s)
Intracellular Signaling Peptides and Proteins/analysis , Neuropeptides/analysis , Parkinson Disease/metabolism , Aged , Aged, 80 and over , Cell Count , Disease Progression , Female , Glial Fibrillary Acidic Protein/analysis , Humans , Hypothalamic Hormones/analysis , Hypothalamus/chemistry , Hypothalamus/pathology , Immunoenzyme Techniques , Intracellular Signaling Peptides and Proteins/deficiency , Male , Melanins/analysis , Middle Aged , Neurons/chemistry , Neuropeptides/deficiency , Orexins , Parkinson Disease/pathology , Pituitary Hormones/analysis , Severity of Illness Index , Substantia Nigra/chemistry , alpha-Synuclein/analysis
12.
Microsc Res Tech ; 81(12): 1374-1382, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30431686

ABSTRACT

Fast free of acrylamide clearing tissue (FACT) is a modified sodium dodecyl sulfate-based clearing protocol for the chemical clearing of lipids that completely preserves fluorescent signals in the cleared tissues. The FACT protocol was optimized to image translucent immunostained brain and non-nervous tissues. For this purpose adult male Chukar partridge (Alectoris chukar) was used as a model. After clearing the tissues, 1 or 2 mm-thickness sections of tissues were immunolabeled. The paraventricular nucleus in the hypothalamus (2-mm section) was cleared with FACT, and then was stained with gonadotropin-inhibitory hormone (GnIH) antibody and Hoechst. Simultaneously, immunohistochemical (IHC) staining of cryosectioned brain (30 µm) was done by GnIH-antibody. The FACT protocol and staining of cell nuclei of nine other tissues were done by a z-stack motorized fluorescent microscope. GnIH-immunoreactive neurons were found by FACT and IHC during the breeding season in male partridges. Deep imaging of the kidney, duodenum, jejunum, lung, pancreas, esophagus, skeletal muscle, trachea, and testis were also done. The FACT protocol can be used for the three-dimensional imaging of various tissues and immunostained evaluation of protein markers. RESEARCH HIGHLIGHT: The FACT is a simple and cheap method for whole tissue clearing. The FACT-cleared tissues can be imaged with simple fluorescent microscopes. For the first time, using the FACT, three-dimensional imaging of various tissues was done.


Subject(s)
Brain Chemistry , Histocytological Preparation Techniques/methods , Acrylamide/chemistry , Animals , Brain/metabolism , Cryoultramicrotomy , Galliformes , Hypothalamic Hormones/analysis , Hypothalamic Hormones/metabolism , Imaging, Three-Dimensional , Immunohistochemistry , Male , Microtomy
13.
Acta Biochim Pol ; 65(2): 251-258, 2018.
Article in English | MEDLINE | ID: mdl-29850655

ABSTRACT

We aimed to quantify the gene expression changes of the potent orexigenic melanin-concentrating hormone (MCH) in chicken (Gallus gallus) hypothalamus with quantitative real-time polymerase chain reaction (qPCR), and for the first time determine peptide concentrations with a novel radioimmunoassay (RIA) under different feeding status. Three different experimental conditions, namely ad libitum feeding; fasting for 24 h; fasting for 24 h and then refeeding for 2 h, were applied to study changes of the aforementioned target and its receptor (MCHR4) gene expression under different nutritional status. The relative changes of MCH and MCHR4 were also studied from 7 to 35 days of age. Expression of PMCH and MCHR4 along the gastrointestinal tract (GIT) was also investigated. We found that expression of both targets was significant in the hypothalamus, while only weak expression was detected along the GIT. Different nutritional states did not affect the PMCH and MCHR4 mRNA levels. However, fasting for 24 h had significantly increased the MCH-like immunoreactivity by 25.65%. Fasting for 24 h and then refeeding for 2 h had further significantly increased the MCH peptide concentration by 32.51%, as compared to the ad libitum state. A decreasing trend with age was observable for both, the PMCH and MCHR4 mRNA levels, and also for the MCH-like immunoreactivity. Correlation analysis did not result in a significant correlation between MCH peptide concentration and abdominal fat mass in ad libitum fed birds. In conclusion, MCH peptide concentration altered in response to 24 h fasting, which indicated that this peptide may take part in feed intake regulation of broiler chickens.


Subject(s)
Feeding Behavior , Hypothalamic Hormones/metabolism , Hypothalamus/metabolism , Melanins/metabolism , Pituitary Hormones/metabolism , Animals , Chickens , Fasting , Hypothalamic Hormones/analysis , Melanins/analysis , Pituitary Hormones/analysis , Polymerase Chain Reaction , RNA, Messenger/metabolism , Receptors, Somatostatin
14.
J Neurosci ; 26(2): 389-97, 2006 Jan 11.
Article in English | MEDLINE | ID: mdl-16407534

ABSTRACT

The observation that loss of orexin (hypocretin) neurons causes human narcolepsy raises the possibility that other acquired disorders might also result from loss of hypothalamic neurons. To test this possibility for body weight, mice with selective loss of melanin concentrating hormone (MCH) neurons were generated. MCH was chosen to test because induced mutations of the MCH gene in mice cause hypophagia and leanness. Mice with ablation of MCH neurons were generated using toxin (ataxin-3)-mediated ablation strategy. The mice appeared normal but, after 7 weeks, developed reduced body weight, body length, fat mass, lean mass, and leptin levels. Leanness was characterized by hypophagia and increased energy expenditure. To study the role of MCH neurons on obesity secondary to leptin deficiency, we generated mice deficient in both ob gene product (leptin) and MCH neurons. Absence of MCH neurons in ob/ob mice improved obesity, diabetes, and hepatic steatosis, suggesting that MCH neurons are important mediators of the response to leptin deficiency. These data show that loss of MCH neurons can lead to an acquired leanness. This has implications for the pathogenesis of acquired changes of body weight and might be considered in clinical settings characterized by substantial weight changes later in life.


Subject(s)
Hypothalamic Hormones/physiology , Hypothalamus/physiopathology , Leptin/deficiency , Melanins/physiology , Nerve Tissue Proteins/physiology , Neurons/physiology , Pituitary Hormones/physiology , Thinness/etiology , Adipose Tissue/pathology , Animals , Ataxin-3 , Blood Glucose/analysis , Cell Death , Crosses, Genetic , Diabetes Mellitus, Experimental/physiopathology , Energy Intake , Energy Metabolism , Fasting/blood , Fatty Liver/physiopathology , Female , Gene Expression , Hypothalamic Hormones/analysis , Hypothalamus/pathology , Insulin/blood , Leptin/genetics , Leptin/therapeutic use , Machado-Joseph Disease/genetics , Male , Melanins/analysis , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neuropeptides/metabolism , Nuclear Proteins , Obesity/genetics , Obesity/physiopathology , Pituitary Hormones/analysis , Recombinant Fusion Proteins/physiology , Repressor Proteins , Transcription Factors , Weight Loss
15.
J Endocrinol ; 193(1): 157-69, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17400813

ABSTRACT

We have investigated expression of molecular elements of the hypothalamic-pituitary-adrenal (HPA) axis in the human retinal pigment epithelium (RPE) cells. The presence of corticotropin-releasing factor (CRF); urocortins I, II and III; CRF receptor type 1 (CRFR1); POMC and prohormone convertases 1 and 2 (PC1 and PC2) mRNAs were shown by RT-PCR; the protein products were detected by ELISA, western blot or immunocytochemical methods in an ARPE-19 cell line derived from an adult human donor. CRFR2 was below the level of detectability. The CRFR1 was functional as evidenced by CRF stimulation of cAMP and inositol triphosphate production as well as by ligand induction of transcriptional activity of inducible cis-elements cAMP responsive element (CRE), activator protein 1 responsive element (AP-1) and POMC promoter) in ARPE-19 using luciferase reporter assay. Immunoreactivities representative of CRF, pre-urocortin, CRFR1 receptor and ACTH were also detected in mouse retina by in situ immunocytochemistry. Finally, using RT-PCR, we detected expression of genes encoding four key enzymes participating in steroids synthesis (CYP11A1, CYP11B1, CYP17 and CYP21A2) and showed transformation of progesterone into cortisol-immunoreactivity in cultured ARPE-19 cells. Therefore, we suggest that ocular tissue expresses CRF-driven signalling system that follows organisational structure of the HPA axis.


Subject(s)
Adrenal Cortex Hormones/analysis , Hypothalamic Hormones/analysis , Pigment Epithelium of Eye/metabolism , Pituitary Hormones/analysis , Adrenal Cortex Hormones/genetics , Adrenocorticotropic Hormone/metabolism , Adult , Animals , Cell Line , Cholesterol Side-Chain Cleavage Enzyme/genetics , Cholesterol Side-Chain Cleavage Enzyme/metabolism , Corticotropin-Releasing Hormone/analysis , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/pharmacology , Cyclic AMP/biosynthesis , DNA Primers/genetics , Enzyme-Linked Immunosorbent Assay/methods , Gene Expression , Humans , Hydrocortisone/analysis , Hydrocortisone/genetics , Hydrocortisone/metabolism , Hypothalamic Hormones/genetics , Immunohistochemistry , Inositol 1,4,5-Trisphosphate/biosynthesis , Mice , Pigment Epithelium of Eye/chemistry , Pituitary Hormones/genetics , Pro-Opiomelanocortin/analysis , Pro-Opiomelanocortin/genetics , Progesterone/metabolism , Proprotein Convertase 1/genetics , Proprotein Convertase 2/genetics , Receptors, Corticotropin-Releasing Hormone/analysis , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Steroid 11-beta-Hydroxylase/genetics , Steroid 11-beta-Hydroxylase/metabolism , Steroid 17-alpha-Hydroxylase/genetics , Steroid 17-alpha-Hydroxylase/metabolism , Urocortins
16.
Crit Care Clin ; 23(3): 467-90, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17900481

ABSTRACT

The evaluation of hormonal status in critically ill patients is challenging and has many pitfalls. This article reviews proper assessment of glycemic status AND adrenal and thyroid function during critical care.


Subject(s)
Critical Care/standards , Endocrine Glands/physiopathology , Intensive Care Units/standards , Monitoring, Physiologic/standards , Point-of-Care Systems , Adrenal Glands/physiopathology , Adrenal Insufficiency , Blood Glucose/analysis , Humans , Hyperglycemia , Hypoglycemia , Hypothalamic Hormones/analysis , Pituitary Gland/physiopathology , Shock, Septic , Thyroid Gland/physiopathology
17.
Life Sci ; 148: 241-6, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26874026

ABSTRACT

AIMS: Melanin-concentrating hormone (MCH) is implicated in the control of food intake, body weight regulation and energy homeostasis. Lactation is an important physiological model to study the hypothalamic integration of peripheral sensory signals, such as suckling stimuli and those related to energy balance. MCH can be detected in the medial preoptic area (MPOA), especially around the 19th day of lactation, when this hormone is described as displaying a peak synthesis followed by a decrease after weaning. The physiological significance of this phenomenon is unclear. Therefore, we aimed to investigate hypothalamic changes associated to sensory stimulation by the litter, in special its influence over MCH synthesis. MAIN METHODS: Female Wistar rats (n=56) were euthanized everyday from lactation days 15-21, with or without suckling stimulus (WS and NS groups, respectively). MCH and Fos immunoreactivity were evaluated in the MPOA and lateral and incerto-hypothalamic areas (LHA and IHy). KEY FINDINGS: Suckling stimulus induced Fos synthesis in all regions studied. An increase on the number of suckling-induced Fos-ir neurons could be detected in the LHA after the 18th day. Conversely, the amount of MCH decreased in the MPOA from days 15-21, independent of suckling stimulation. No colocalization between MCH and Fos could be detected in any region analyzed. SIGNIFICANCE: Suckling stimulus is capable of stimulating hypothalamic regions not linked to maternal behavior, possibly to mediate energy balance aspects of lactation. Although dams are hyperphagic before weaning, this behavioral change does not appear to be mediated by MCH.


Subject(s)
Hypothalamic Hormones/biosynthesis , Hypothalamus/metabolism , Lactation/metabolism , Melanins/biosynthesis , Melanophores/metabolism , Pituitary Hormones/biosynthesis , Proto-Oncogene Proteins c-fos/biosynthesis , Animals , Animals, Suckling , Female , Hypothalamic Hormones/analysis , Melanins/analysis , Pituitary Hormones/analysis , Proto-Oncogene Proteins c-fos/analysis , Rats , Rats, Wistar
18.
J Comp Neurol ; 524(14): 2753-75, 2016 10 01.
Article in English | MEDLINE | ID: mdl-26917324

ABSTRACT

In vertebrates, gonadotropin-releasing hormone (GnRH) and gonadotropin-inhibitory hormone (GnIH), respectively, regulate reproduction in positive and negative manners. GnIH belongs to the LPXRFa family of peptides previously identified in mammalian and nonmammalian vertebrates. Studying the detailed distribution of LPXRFa as well as its receptor (LPXRFa-R) in the brain and pituitary is important for understanding their multiple action sites and potential functions. However, the distribution of LPXRFa and LPXRFa-R has not been studied in teleost species, partially because of the lack of fish-specific antibodies. Therefore, in the present study, we generated specific antibodies against LPXRFa and its receptor from Nile tilapia (Oreochromis niloticus), and examined their distributions in the brain and pituitary by immunohistochemistry. Tilapia LPXRFa-immunoreactive neurons lie in the posterior ventricular nucleus of the caudal preoptic area, whereas LPXRFa-R-immunoreactive cells are distributed widely. Double immunofluorescence showed that neither LPXRFa-immunoreactive fibers nor LPXRFa-R is closely associated or coexpressed with GnRH1, GnRH3, or kisspeptin (Kiss2) neurons. In the pituitary, LPXRFa fibers are closely associated with gonadotropic endocrine cells [expressing luteinizing hormone (LH) and follicle-stimulating hormone (FSH)], with adrenocorticomelanotropic cells [corticotropin (ACTH) and α-melanotropin (α-MSH)], and with somatolactin endocrine cells. In contrast, LPXRFa-R are expressed only in LH, ACTH, and α-MSH cells. These results suggest that LPXRFa and LPXRFa-R signaling acts directly on the pituitary cells independent from GnRH or kisspeptin and could play multiple roles in reproductive and nonreproductive functions in teleosts. J. Comp. Neurol. 524:2753-2775, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Brain Chemistry , Gonadotropin-Releasing Hormone/analysis , Hypothalamic Hormones/analysis , Pituitary Gland/chemistry , Receptors, Gonadotropin/analysis , Receptors, LHRH/analysis , Animals , Brain/metabolism , Brain Chemistry/physiology , Gonadotropin-Releasing Hormone/biosynthesis , Hypothalamic Hormones/biosynthesis , Male , Pituitary Gland/metabolism , Receptors, Gonadotropin/biosynthesis , Receptors, LHRH/biosynthesis , Tilapia
19.
J Neuroendocrinol ; 28(5)2016 05.
Article in English | MEDLINE | ID: mdl-26919074

ABSTRACT

The role of gonadotrophin-inhibitory hormone (GnIH) in the inhibition of the reproductive axis has been well-established in birds and mammals. However, its role in other vertebrates, such as the teleost fish, remains controversial. In this context, the present study aimed to evaluate whether GnIH modulates the release of gonadotrophins and growth hormone (GH) in the cichlid fish Cichlasoma dimerus. First, we partially sequenced the precursor polypeptide for GnIH and identified three putative GnIH peptides. Next, we analysed the expression of this precursor polypeptide via a polymerase chain reaction in the reproductive axis of both sexes. We found a high expression of the polypeptide in the hypothalamus and gonads of males. Immunocytochemistry allowed the observation of GnIH-immunoreactive somata in the nucleus posterioris periventricularis and the nucleus olfacto-retinalis, with no differences between the sexes. GnIH-immunoreactive fibres were present in all brain regions, with a high density in the nucleus lateralis tuberis and at both sides of the third ventricle. Finally, we performed in vitro studies on intact pituitary cultures to evaluate the effect of two doses (10(-6)  m and 10(-8)  m) of synthetic C. dimerus (cd-) LPQRFa-1 and LPQRFa-2 on the release of gonadotrophins and GH. We observed that cd-LPQRFa-1 decreased ß-luteinising hormone (LH) and ß-follicle-stimulating hormone (FSH) and also increased GH release to the culture medium. The release of ß-FSH was increased only when it was stimulated with the higher cd-LPQRFa-2 dose. The results of the present study indicate that cd-LPQRFa-1, the cichlid fish GnIH, inhibits ß-LH and ß-FSH release and stimulates GH release in intact pituitary cultures of C. dimerus. The results also show that cd-LPQRF-2 could act as an ß-FSH-releasing factor in this fish species.


Subject(s)
Cichlids/metabolism , Fish Proteins/metabolism , Gonadotropins/metabolism , Growth Hormone/metabolism , Hypothalamic Hormones/metabolism , Animals , Cichlids/genetics , Female , Follicle Stimulating Hormone, beta Subunit/metabolism , Hypothalamic Hormones/analysis , Hypothalamic Hormones/genetics , Male , Peptide Hormones/administration & dosage , Pituitary Gland/drug effects , Pituitary Gland/metabolism , RNA, Messenger/analysis , RNA, Messenger/metabolism
20.
Endocrinology ; 140(12): 5736-45, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10579339

ABSTRACT

A recently identified neuropeptide with PRL-releasing capabilities binds to and activates a previously known orphan G protein-coupled receptor, GPR10. We initiated a study to define the pharmacology of the peptide/receptor interaction and to identify the distribution of the peptide and its receptor in the central nervous system to elucidate sites of action of the peptide. The PRL-releasing peptide (PrRP) is a C-terminally amidated, 31-amino acid peptide derived from a 98-amino acid precursor. Radioiodinated PrRP-(1-31) binds to its receptor with high affinity (1 nM) and stimulates calcium mobilization in CHOK1 cells stably transfected with the receptor. A series of N-terminal deletions reveals that the PrRP-(12-31) amino acid is equipotent to PrRP-(1-31). Further N-terminal deletions reduce the affinity of the ligand considerably, although PrRP-(25-31) is still able to compete for binding and behaves as an agonist. The arginine residues at position 26 and 30 are critical for binding, as substitution with either lysine or citrulline reduces the affinity substantially. In situ hybridization reveals a distinct tissue distribution for both the peptide and receptor messenger RNAs. The receptor is expressed abundantly in the reticular thalamic nucleus, periventricular hypothalamus, dorsomedial hypothalamus, nucleus of the solitary tract, area postrema, anterior pituitary, and adrenal medulla. The peptide messenger RNA is expressed in the dorsomedial hypothalamus, nucleus of the solitary tract, ventrolateral reticular nucleus, and intestine. This tissue distribution suggests an alternative function of PrRP than its purported hypophysiotropic function, such as a potential role for PrRP in the central feedback control of neuroendocrine and autonomic homeostasis. Further work using selective agonists and antagonists should help define additional physiological roles of this novel mammalian neuropeptide.


Subject(s)
Central Nervous System/chemistry , Hypothalamic Hormones/analysis , Neuropeptides/analysis , Receptors, Neuropeptide/analysis , Animals , Brain/metabolism , Brain Chemistry , CHO Cells , Cricetinae , Gene Expression , Hypothalamic Hormones/genetics , Hypothalamic Hormones/metabolism , In Situ Hybridization , Iodine Radioisotopes , Male , Neuropeptides/genetics , Neuropeptides/metabolism , Prolactin-Releasing Hormone , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism , Thalamic Nuclei/chemistry , Thyrotropin-Releasing Hormone/genetics , Tissue Distribution , Transfection , Tyrosine 3-Monooxygenase/analysis
SELECTION OF CITATIONS
SEARCH DETAIL