Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
J Histochem Cytochem ; 71(7): 409-410, 2023 07.
Article in English | MEDLINE | ID: mdl-37415491

ABSTRACT

The fading and quenching of fluorescence intensity has been a major problem in the use of fluorescein isothiocyanate (FITC) for immunofluorescence cytochemical techniques, especially with laser confocal microscopy. The companion article by Longin et al. provided an empirical approach to overcoming this problem. The present commentary highlights the significance of the Longin et al. article when it was published and its continued relevance today.


Subject(s)
Fluorescent Dyes , Isothiocyanates , Fluorescein-5-isothiocyanate , Microscopy, Confocal/methods , Fluorescent Antibody Technique
2.
Front Endocrinol (Lausanne) ; 13: 885909, 2022.
Article in English | MEDLINE | ID: mdl-35880052

ABSTRACT

Stressors of different natures induce activation of the hypothalamic-pituitary-adrenal (HPA) axis at different magnitudes. Moreover, the HPA axis response to repeated exposure is usually distinct from that elicited by a single session. Paradoxical sleep deprivation (PSD) augments ACTH and corticosterone (CORT) levels, but the nature of this stimulus is not yet defined. The purpose of the present study was to qualitatively compare the stress response of animals submitted to PSD to that of rats exposed once or four times to cold, as a physiological stress, movement restraint (RST) as a mixed stressor and predator odour (PRED) as the psychological stressor, whilst animals were submitted for 1 or 4 days to PSD and respective control groups. None of the stressors altered corticotropin releasing factor immunoreactivity in the paraventricular nucleus of the hypothalamus (PVN), median eminence (ME) or central amygdala, compared to control groups, whereas vasopressin immunoreactivity in PSD animals was decreased in the PVN and increased in the ME, indicating augmented activity of this system. ACTH levels were higher after repeated stress or prolonged PSD than after single- or 1 day-exposure and control groups, whereas the CORT response was habituated by repeated stress, but not by 4-days PSD. This dissociation resulted in changes in the CORT : ACTH ratio, with repeated cold and RST decreasing the ratio compared to single exposure, but no change was seen in PRED and PSD groups. Comparing the magnitude and pattern of pituitary-adrenal response to the different stressors, PSD-induced responses were closer to that shown by PRED-exposed rats. In contrast, the hypothalamic response of PSD-exposed rats was unique, inasmuch as this was the only stressor which increased the activity of the vasopressin system. In conclusion, we propose that the pituitary-adrenal response to PSD is similar to that induced by a psychological stressor.


Subject(s)
Pituitary Diseases , Pituitary-Adrenal System , Adrenocorticotropic Hormone/metabolism , Animals , Corticosterone , Hypothalamo-Hypophyseal System/metabolism , Hypothalamus/metabolism , Pituitary-Adrenal System/metabolism , Rats , Sleep Deprivation , Sleep, REM , Stress, Psychological
3.
J Neuroendocrinol ; 34(5): e13115, 2022 05.
Article in English | MEDLINE | ID: mdl-35502534

ABSTRACT

The anatomy and morphology of gonadotropin-releasing hormone (GnRH) neurons makes them both a joy and a challenge to investigate. They are a highly unique population of neurons given their developmental migration into the brain from the olfactory placode, their relatively small number, their largely scattered distribution within the rostral forebrain, and, in some species, their highly varied individual anatomical characteristics. These unique features have posed technological hurdles to overcome and promoted fertile ground for the establishment and use of creative approaches. Historical and more contemporary discoveries defining GnRH neuron anatomy remain critical in shaping and challenging our views of GnRH neuron function in the regulation of reproductive function. We begin this review with a historical overview of anatomical discoveries and developing methodologies that have shaped our understanding of the reproductive axis. We then highlight significant discoveries across specific groups of mammalian species to address some of the important comparative aspects of GnRH neuroanatomy. Lastly, we touch on unresolved questions and opportunities for future neuroanatomical research on this fascinating and important population of neurons.


Subject(s)
Gonadotropin-Releasing Hormone , Neuroanatomy , Animals , Gonadotropin-Releasing Hormone/metabolism , Mammals , Neurons/metabolism , Prosencephalon , Reproduction
4.
Compr Physiol ; 10(2): 549-575, 2020 03 12.
Article in English | MEDLINE | ID: mdl-32163202

ABSTRACT

The scientific community has searched for years for ways of examining neuronal tissue to track neural activity with reliable anatomical markers for stimulated neuronal activity. Existing studies that focused on hypothalamic systems offer a few options but do not always compare approaches or validate them for dependence on cell firing, leaving the reader uncertain of the benefits and limitations of each method. Thus, in this article, potential markers will be presented and, where possible, placed into perspective in terms of when and how these methods pertain to hypothalamic function. An example of each approach is included. In reviewing the approaches, one is guided through how neurons work, the consequences of their stimulation, and then the potential markers that could be applied to hypothalamic systems are discussed. Approaches will use features of neuronal glucose utilization, water/oxygen movement, changes in neuron-glial interactions, receptor translocation, cytoskeletal changes, stimulus-synthesis coupling that includes expression of the heteronuclear or mature mRNA for transmitters or the enzymes that make them, and changes in transcription factors (immediate early gene products, precursor buildup, use of promoter-driven surrogate proteins, and induced expression of added transmitters. This article includes discussion of methodological limitations and the power of combining approaches to understand neuronal function. © 2020 American Physiological Society. Compr Physiol 10:549-575, 2020.


Subject(s)
Hypothalamus/physiology , Neurons/physiology , Animals , Biomarkers/analysis , Humans , Hypothalamus/cytology , Neurons/cytology
5.
Neurobiol Learn Mem ; 165: 106962, 2019 11.
Article in English | MEDLINE | ID: mdl-30502397

ABSTRACT

Rett Syndrome (RTT) is a genetic disorder that is caused by mutations in the x-linked gene coding for methyl-CpG-biding-protein 2 (MECP2) and that mainly affects females. Male and female transgenic mouse models of RTT have been studied extensively, and we have learned a great deal regarding RTT neuropathology and how MeCP2 deficiency may be influencing brain function and maturation. In this manuscript we review what is known concerning structural and coinciding functional and behavioral deficits in RTT and in mouse models of MeCP2 deficiency. We also introduce our own corroborating data regarding behavioral phenotype and morphological alterations in volume of the cortex and striatum and the density of neurons, aberrations in experience-dependent plasticity within the barrel cortex and the impact of MeCP2 loss on glial structure. We conclude that regional structural changes in genetic models of RTT show great similarity to the alterations in brain structure of patients with RTT. These region-specific modifications often coincide with phenotype onset and contribute to larger issues of circuit connectivity, progression, and severity. Although the alterations seen in mouse models of RTT appear to be primarily due to cell-autonomous effects, there are also non-cell autonomous mechanisms including those caused by MeCP2-deficient glia that negatively impact healthy neuronal function. Collectively, this body of work has provided a solid foundation on which to continue to build our understanding of the role of MeCP2 on neuronal and glial structure and function, its greater impact on neural development, and potential new therapeutic avenues.


Subject(s)
Brain/growth & development , Rett Syndrome/etiology , Animals , Basal Ganglia/pathology , Brain/physiopathology , Disease Models, Animal , Hippocampus/pathology , Humans , Methyl-CpG-Binding Protein 2/metabolism , Mice/growth & development , Motor Disorders/etiology , Motor Disorders/physiopathology , Neuronal Plasticity , Rett Syndrome/physiopathology , Rett Syndrome/psychology
6.
Am J Physiol Endocrinol Metab ; 315(5): E1019-E1033, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30040478

ABSTRACT

The effect of estrogen on the differentiation and maintenance of reproductive tissues is mediated by two nuclear estrogen receptors (ERs), ERα, and ERß. Lack of functional ERα and ERß genes in vivo significantly affects reproductive function; however, the target tissues and signaling pathways in the hypothalamus are not clearly defined. Here, we describe the generation and reproductive characterization of a complete-ERß KO (CERßKO) and a GnRH neuron-specific ERßKO (GERßKO) mouse models. Both ERßKO mouse models displayed a delay in vaginal opening and first estrus. Hypothalamic gonadotropin-releasing hormone (GnRH) mRNA expression levels in both ERßKO mice were similar to control mice; however female CERßKO and GERßKO mice had lower basal and surge serum gonadotropin levels. Although a GnRH stimulation test in both female ERßKO models showed preserved gonadotropic function in the same animals, a kisspeptin stimulation test revealed an attenuated response by GnRH neurons, suggesting a role for ERß in normal GnRH neuron function. No alteration in estrogen-negative feedback was observed in either ERßKO mouse models after ovariectomy and estrogen replacement. Further, abnormal development of ovarian follicles with low serum estradiol levels and impairment of fertility were observed in both ERßKO mouse models. In male ERßKO mice, no differences in the timing of pubertal onset or serum luteinizing hormone and follicle-stimulating hormone levels were observed as compared with controls. Taken together, these data provide in vivo evidence for a role of ERß in GnRH neurons in modulating puberty and reproduction, specifically through kisspeptin responsiveness in the female hypothalamic-pituitary-gonadal axis.


Subject(s)
Estrogen Receptor beta/metabolism , Fertility/physiology , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/metabolism , Neurons/metabolism , Sexual Maturation/physiology , Animals , Estradiol/blood , Estrogen Receptor beta/genetics , Feedback, Physiological/physiology , Female , Follicle Stimulating Hormone/blood , Luteinizing Hormone/blood , Mice , Mice, Knockout
7.
Neuroscience ; 369: 1-14, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29126954

ABSTRACT

Kynurenic acid, a metabolite of the kynurenine pathway of tryptophan degradation, acts as an endogenous antagonist of alpha7 nicotinic and NMDA receptors and is implicated in a number of neurophysiological and neuropathological processes including cognition and neurodegenerative events. Therefore, kynurenine aminotransferase II (KAT II/AADAT), the enzyme responsible for the formation of the majority of neuroactive kynurenic acid in the brain, has prompted significant interest. Using immunohistochemistry, this enzyme was localized primarily in astrocytes throughout the adult rat brain, but detailed neuroanatomical studies are lacking. Here, we employed quantitative in situ hybridization to analyze the relative expression of KAT II mRNA in the brain of rats under normal conditions and 6 h after the administration of lipopolysaccharides (LPSs). Specific hybridization signals for KAT II were detected, with the highest expression in the subventricular zone (SVZ), the rostral migratory stream and the floor of the third ventricle followed by the corpus callosum and the hippocampus. This pattern of mRNA expression was paralleled by differential protein expression, determined by serial dilutions of antibodies (up to 1:1 million), and was confirmed to be primarily astrocytic in nature. The mRNA signal in the SVZ and the hippocampus was substantially increased by the LPS treatment without detectable changes elsewhere. These results demonstrate that KAT II is expressed in the rat brain in a region-specific manner and that gene expression is sensitive to inflammatory processes. This suggests an unrecognized role for kynurenic acid in the brain's germinal zones.


Subject(s)
Astrocytes/enzymology , Brain/enzymology , Transaminases/biosynthesis , Aging , Animals , Doublecortin Protein , Female , Male , Rats , Rats, Wistar
8.
PLoS One ; 11(12): e0152252, 2016.
Article in English | MEDLINE | ID: mdl-27997552

ABSTRACT

A competition of neurobehavioral drives of sleep and wakefulness occurs during sleep deprivation. When enforced chronically, subjects must remain awake. This study examines histaminergic neurons of the tuberomammillary nucleus of the posterior hypothalamus in response to enforced wakefulness in rats. We tested the hypothesis that the rate-limiting enzyme for histamine biosynthesis, L-histidine decarboxylase (HDC), would be up-regulated during chronic rapid eye movement sleep deprivation (REM-SD) because histamine plays a major role in maintaining wakefulness. Archived brain tissues of male Sprague Dawley rats from a previous study were used. Rats had been subjected to REM-SD by the flowerpot paradigm for 5, 10, or 15 days. For immunocytochemistry, rats were transcardially perfused with acrolein-paraformaldehyde for immunodetection of L-HDC; separate controls used carbodiimide-paraformaldehyde for immunodetection of histamine. Immunolocalization of histamine within the tuberomammillary nucleus was validated using carbodiimide. Because HDC antiserum has cross-reactivity with other decarboxylases at high antibody concentrations, titrations localized L-HDC to only tuberomammillary nucleus at a dilution of ≥ 1:300,000. REM-SD increased immunoreactive HDC by day 5 and it remained elevated in both dorsal and ventral aspects of the tuberomammillary complex. Our results suggest that up-regulation of L-HDC within the tuberomammillary complex during chronic REM-SD may be responsible for maintaining wakefulness.


Subject(s)
Histamine/metabolism , Histidine Decarboxylase/biosynthesis , Hypothalamic Area, Lateral/enzymology , Sleep Deprivation/enzymology , Up-Regulation , Wakefulness , Animals , Gene Expression Regulation, Enzymologic , Hypothalamic Area, Lateral/pathology , Male , Rats , Rats, Sprague-Dawley , Sleep Deprivation/pathology
9.
Curr Protoc Neurosci ; 76: 2.12.1-2.12.37, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27367962

ABSTRACT

When using immunocytochemistry, investigators may not know how to optimize staining or how to troubleshoot the method when staining fails. Lacking are guides for comparing techniques and applying information derived from one staining method to another. Newer methods amplify signal detection, but will not necessarily work at the same primary antibody concentrations used for less sensitive reactions. Recommendations of optimal titers are often not accurate and are not usually accompanied by information on the method used to test those antibodies or the specifics of the assay. When the staining does not work, the investigators do not know how to determine if the antiserum is bad, the tissue is bad, or the method is inappropriate for their staining. This unit describes detailed procedures for determining optimal staining and applying that information to three common immunofluorescence methods. Lastly, a formula is provided for converting among the different methods. © 2016 by John Wiley & Sons, Inc.


Subject(s)
Antibodies/immunology , Immunoenzyme Techniques , Immunohistochemistry/methods , Animals , Humans , Immune Sera , Immunoenzyme Techniques/methods , Immunologic Tests , Staining and Labeling/methods
10.
Endocrinology ; 155(3): 676-87, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24428532

ABSTRACT

For several decades antibodies raised against specific proteins, peptides, or peptide epitopes have proven to be versatile and very powerful tools to demonstrate molecular identity in cells and tissues. New techniques of immunohistochemistry and immunofluorescence have improved both the optical resolution of such protein identification as well as its sensitivity, particularly through the use of amplification methodology. However, this improved sensitivity has also increased the risks of false-positive and false-negative staining and thereby raised the necessity for proper and adequate controls. In this review, the authors draw on many years of experience to illuminate many of the more common errors and problematic issues in immunohistochemistry, and how these may be avoided. A key factor in all of this is that techniques need to be properly documented and especially antibodies and procedures must be adequately described. Antibodies are a valuable and shared resource within the scientific community; it is essential therefore that mistakes involving antibodies and their controls are not perpetuated through inadequate reporting in the literature.


Subject(s)
Antibodies/immunology , Immunohistochemistry/instrumentation , Immunohistochemistry/methods , Adsorption , Animals , Antibodies/chemistry , Antigens/chemistry , Epitopes/chemistry , Humans , Mice, Knockout , Microscopy, Fluorescence/methods , Staining and Labeling/methods
11.
Sleep ; 36(12): 1839-48, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24293758

ABSTRACT

STUDY OBJECTIVES: The basal forebrain (BF) has been implicated as an important brain region that regulates the sleep-wake cycle of animals. Gamma-aminobutyric acidergic (GABAergic) neurons are the most predominant neuronal population within this region. However, due to the lack of specific molecular tools, the roles of the BF GABAergic neurons have not been fully elucidated. Previously, we have found high expression levels of the Kv2.2 voltage-gated potassium channel on approximately 60% of GABAergic neurons in the magnocellular preoptic area and horizontal limb of the diagonal band of Broca of the BF and therefore proposed it as a potential molecular target to study this neuronal population. In this study, we sought to determine the functional roles of the Kv2.2-expressing neurons in the regulation of the sleep-wake cycle. DESIGN: Sleep analysis between two genotypes and within each genotype before and after sleep deprivation. SETTING: Animal sleep research laboratory. PARTICIPANTS: Adult mice. Wild-type and Kv2.2 knockout mice with C57/BL6 background. INTERVENTIONS: EEG/EMG recordings from the basal state and after sleep-deprivation which was induced by mild agitation for 6 h. RESULTS: Immunostaining of a marker of neuronal activity indicates that these Kv2.2-expressing neurons appear to be preferentially active during the wake state. Therefore, we tested whether Kv2.2-expressing neurons in the BF are involved in arousal using Kv2.2-deficient mice. BF GABAergic neurons exhibited augmented expression of c-Fos. These knockout mice exhibited longer consolidated wake bouts than wild-type littermates, and that phenotype was further exacerbated by sleep deprivation. Moreover, in-depth analyses of their cortical electroencephalogram revealed a significant decrease in the delta-frequency activity during the nonrapid eye movement sleep state. CONCLUSIONS: These results revealed the significance of Kv2.2-expressing neurons in the regulation of the sleep-wake cycle.


Subject(s)
GABAergic Neurons/physiology , Prosencephalon/physiology , Shab Potassium Channels/physiology , Sleep/physiology , Wakefulness/physiology , Animals , Electroencephalography , Electromyography , Genotype , Male , Mice , Mice, Inbred C57BL , Mice, Knockout/genetics , Parvalbumins/physiology , Prosencephalon/cytology , Proto-Oncogene Proteins c-fos/physiology , Shab Potassium Channels/genetics , Sleep/genetics , Sleep Deprivation/physiopathology , Wakefulness/genetics
12.
J Histochem Cytochem ; 60(6): 439-56, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22470123

ABSTRACT

FMR1 premutation (PM) alleles have 55-200 CGG·CCG-repeats in their 5' UTR. PM carriers are at risk of fragile X-associated tremor and ataxia syndrome (FXTAS). Females are also at risk for FX primary ovarian insufficiency (FXPOI). PM pathology is generally attributed to deleterious properties of transcripts with long CGG-tracts. For FXPOI, hormone changes suggest a reduced residual follicle pool. Whether this is due to a smaller than normal original follicle pool or an increased rate of follicle depletion is unclear. A FX-PM mouse the authors generated with 130 CGG·CCG-repeats in the endogenous Fmr1 gene recapitulates features of FXTAS. Here the authors demonstrate that the gross development of the ovary and the establishment of the primordial follicle pool is normal in these mice. However, these animals show a faster loss of follicles of all follicle classes, suggesting that the problem is intrinsic to the ovary. In addition, many oocytes show aberrant nuclear accumulation of FMRP and elevated levels of ubiquitination. Furthermore, PM follicles are smaller and have fewer granulosa cells (GCs) than normal. Thus, these animals have ovarian abnormalities involving both the oocytes and GCs that may shed light on the molecular basis of FXPOI in humans.


Subject(s)
Fragile X Mental Retardation Protein/genetics , Ovarian Follicle/pathology , Primary Ovarian Insufficiency/pathology , Animals , Cell Count , Disease Models, Animal , Female , Follicular Atresia , Fragile X Mental Retardation Protein/metabolism , Granulosa Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Oocytes/metabolism , Organ Specificity , Ovarian Follicle/metabolism , Ovary/metabolism , Ovary/pathology , Primary Ovarian Insufficiency/metabolism , Ubiquitination
13.
Neurobiol Dis ; 42(1): 85-98, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21220020

ABSTRACT

Carriers of FMR1 premutation alleles have 55-200 CGG repeats in the 5' untranslated region of the gene. These individuals are at risk for fragile X associated primary ovarian insufficiency (females) and, in late life, fragile X associated tremor and ataxia syndrome (males, and to a lesser extent, females). Premutation carrier status can also be associated with autism spectrum disorder, attention deficit hyperactivity disorder, and some cognitive deficits. In premutation carriers, FMR1 mRNA levels are often higher than those with normal sized alleles. In contrast, in subjects with full mutation alleles, (>200 repeats) the FMR1 gene is silenced and FMR1 mRNA and its product, FMRP, are absent. We have studied a male knock-in (KI) mouse model of the fragile X premutation (120-140 repeats) during young adulthood. In comparison to wild type, KI mice were hyperactive, exhibited less anxiety in both the open field and the elevated zero maze, were impaired on the passive avoidance test, and showed some subtle deficits on a test of social interaction. Motor learning as assessed by the rotarod test was normal. Dendritic arbors were less complex and spine densities and lengths increased in medial prefrontal cortex, basal lateral amygdala, and hippocampus compared with wild type. Regional rates of cerebral protein synthesis measured in vivo in KI mice were increased. KI mice also had elevated levels of Fmr1 mRNA and decreased levels of FMRP. Our results highlight similarities in phenotype between KI and Fmr1 knockout mice and suggest that the decreased concentration of FMRP contributes to the phenotype in young adult KI mice.


Subject(s)
Cerebral Cortex/metabolism , Dendrites/pathology , Fragile X Mental Retardation Protein/biosynthesis , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Fragile X Syndrome/metabolism , Genetic Predisposition to Disease , Protein Biosynthesis/genetics , Animals , Behavior, Animal/physiology , Cerebral Cortex/abnormalities , Cerebral Cortex/pathology , Dendrites/metabolism , Disease Models, Animal , Female , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/pathology , Gene Knock-In Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
14.
J Neurosci ; 31(1): 184-92, 2011 Jan 05.
Article in English | MEDLINE | ID: mdl-21209203

ABSTRACT

Gonadotropin-releasing hormone (GnRH) neurons represent the final common output of signals from the brain that regulates reproductive function. A wide range of environmental factors impact GnRH neuron activity including disease, stress, nutrition, and seasonal cues, as well as gonadal steroid hormones. The CNS response is thought to be mediated, at least in part, through intermediate signaling molecules that affect GnRH neuronal activity. In vitro, GnRH neuronal cell lines respond to a variety of ligands that activate the Jak (Janus-activated kinase)/STAT (signal transducers and activators of transcription) intracellular signaling pathway. To determine its biological function in reproduction, we used Cre (cAMP response element)/LoxP technology to generate GnRH neuron-specific Jak2 conditional knock-out (Jak2 G(-/-)) mice. GnRH mRNA levels were reduced in Jak2 G(-/-) mice when compared with controls, while the number of GnRH neurons was equivalent, indicating a reduction in GnRH gene expression. Secretion of GnRH is also reduced as basal serum luteinizing hormone (LH) levels were significantly lower in female Jak2 G(-/-) mice while the pituitary responded normally to exogenous GnRH. Preovulatory LH surge levels were blunted in Jak2 G(-/-) mice, which was correlated with reduced GnRH neuronal activation as assessed by c-Fos. However, the activation of GnRH neurons following release from estrogen-negative feedback is retained. Female Jak2 G(-/-) mice exhibited significantly delayed puberty and first estrus, abnormal estrous cyclicity, and impaired fertility. These results demonstrate an essential role for Jak2 signaling in GnRH neurons for normal reproductive development and fertility in female mice.


Subject(s)
Down-Regulation/physiology , Gonadotropin-Releasing Hormone/metabolism , Janus Kinase 2/physiology , Reproduction/physiology , Animals , Cell Count/methods , Down-Regulation/drug effects , Down-Regulation/genetics , Estrous Cycle/genetics , Exons/genetics , Female , Fertility/genetics , Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/pharmacology , Green Fluorescent Proteins/genetics , Hypothalamus/cytology , Janus Kinase 2/deficiency , Luteinizing Hormone/blood , Mice , Mice, Knockout , Neurons/metabolism , Ovariectomy , Ovary/pathology , Proto-Oncogene Proteins c-fos/metabolism , Puberty, Delayed/genetics , RNA, Messenger/metabolism , Reproduction/drug effects , Reproduction/genetics
15.
J Mol Neurosci ; 45(2): 110-8, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21125428

ABSTRACT

Dopamine (DA) and enkephalin (ENK) release from the tuberoinfundibular dopaminergic neurons (TIDA) into the hypophysial portal circulation is fundamentally different under non-lactating and lactating conditions. The aim of this experiment was to compare the effect of a brief interruption then resumption of suckling on the temporal program of tyrosine hydroxylase (TH; rate-limiting enzyme of dopamine synthesis) and ENK regulation in dams. On post partum day 10, pups were removed for a 4-h period from a group of the dams then returned for 4- and 24-h periods. It was examined whether such a brief interruption of suckling provokes full up-regulation of TH and down-regulation of ENK, and whether reinitiation of suckling limits the extent to which TH up- and ENK down-regulate. At the end of experiment, the animals were decapitated. In situ hybridization was used to examine the expression of TH and ENK mRNA in the arcuate nucleus where TIDA neurons reside. The results showed that, on one hand, the removal of pups induced TH up-regulation, on the other hand, ENK expression also increased 8 h after removal of pups and then started to slowly decline. In dams whose sucklings were reinitiated both TH and ENK mRNAs were up-regulated at least for a day. ENK expression responded more slowly to the removal of pups than expression of TH, and after reinitiation of suckling, the temporal program of regulation of both TH and ENK expressions ran parallel in the first 24 h.


Subject(s)
Animals, Suckling/metabolism , Arcuate Nucleus of Hypothalamus/cytology , Dopaminergic Neurons/metabolism , Enkephalins/metabolism , Lactation/physiology , Tyrosine 3-Monooxygenase/metabolism , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Dopaminergic Neurons/cytology , Enkephalins/genetics , Female , Pregnancy , RNA, Messenger/metabolism , Rats , Tyrosine 3-Monooxygenase/genetics
16.
Endocrinology ; 152(1): 214-22, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21047947

ABSTRACT

We tested the working hypothesis that Fos will identify the critical population of kisspeptin neurons that accompanies the LHRH surge using a synchronized follicular phase model in intact cycling ewes. The model generates an LH surge that starts within a defined 2-h window in a 20-d synchronized cycle. With a modified push-pull cannula in vivo LHRH release from the median eminence was sampled in luteal phase ewes, ewes undergoing an LH surge for 2-4 h, and postsurge animals whose LH surge peaked 10-12 h earlier. In vivo release of LHRH was lower in the luteal and follicular phases than in animals undergoing an LH surge (P < 0.01); it fell to presurge levels after the LH surge. Ewes killed 2-4 h after the surge started, expressed Fos in a large portion of preoptic area (POA) kisspeptin (53.90 ± 4.69%, P < 0.01) and LHRH neurons (48.20 ± 4.49%, P < 0.0001) compared with animals euthanized at any of the other times tested (under <5% of the cells activated). Little Fos activation (under 5%) was observed during any of the times sampled in arcuate (Arc) kisspeptin neurons. The relationship between the number of LHRH neurons and the POA kisspeptin neurons stimulated showed a striking positive correlation with r(2) = 0.68, P = 0.0003, reinforcing the evidence that POA kisspeptin neurons actively participate in the stimulation of LHRH surges.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Luteinizing Hormone/metabolism , Median Eminence/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Sheep/physiology , Tumor Suppressor Proteins/metabolism , Animals , Gene Expression Regulation/physiology , Neurons/physiology , Preoptic Area , Tumor Suppressor Proteins/genetics
17.
Endocrine ; 37(1): 62-70, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20963557

ABSTRACT

The neuronal pathways, through which prolactin secretion is regulated during lactation, have still not been fully explored. Studies indicate that the suckling stimulus travels through the spinal cord, the brain stem, and then reaches the hypothalamus. The focus of this present experiment is to further explore the neuronal connections between the brain stem and the arcuate nucleus that may be involved in suckling-induced prolactin release. Ante- and retrograde tracing techniques were used. To chemically characterize the explored neurons neuropeptide immunohistochemistry was applied. Previous studies have indicated that the peripeduncular nucleus is a relay of the suckling stimulus in the midbrain, conveying the information to the hypothalamus. In our experiments, we have found an additional cell group in the subparafascicular parvocellular nucleus located just behind the posterior thalamus that projects to the arcuate neurons. The injection of the retrograde tracer into the ventrolateral part of the arcuate nucleus labeled cells in the lateral subdivision of the subparafascicular parvocellular nucleus. Anterograde tracing from the subparafascicular parvocellular nucleus resulted in fiber labeling in the arcuate nucleus in close apposition with dynorphin immunopositive neurons. Double labeling revealed that a subpopulations of the subparafascicular parvocellular neurons projecting to the arcuate nucleus contained tuberoinfundibular peptide of 39 residues or calcitonin gene-related peptide. The presented findings suggest that the ascending fibers from the subparafascicular parvocellular nucleus might be in the pathway involved in the suckling-induced prolactin release.


Subject(s)
Arcuate Nucleus of Hypothalamus/physiology , Hypothalamus, Posterior/physiology , Neural Pathways/physiology , Neuropeptides/metabolism , Prolactin/metabolism , Animals , Arcuate Nucleus of Hypothalamus/anatomy & histology , Biotin/analogs & derivatives , Calcitonin Gene-Related Peptide/metabolism , Dextrans , Dynorphins/metabolism , Female , Galanin/metabolism , Hypothalamus, Posterior/anatomy & histology , Immunohistochemistry , Neuroanatomical Tract-Tracing Techniques , Neuronal Tract-Tracers , Neurons/classification , Neurons/physiology , Protein Precursors/metabolism , Rats , Rats, Sprague-Dawley , Stilbamidines , Tyrosine 3-Monooxygenase/metabolism
18.
Endocrinology ; 151(7): 3247-57, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20410200

ABSTRACT

Prolactin (PRL) is tonically inhibited by dopamine (DA) released from neurons in the arcuate and periventricular nuclei. Kisspeptin plays a pivotal role in LH regulation. In rodents, kisspeptin neurons are found mostly in the anteroventral periventricular and arcuate nuclei, but the physiology of arcuate kisspeptin neurons is not completely understood. We investigated the role of kisspeptin in the control of hypothalamic DA and pituitary PRL secretion in adult rats. Intracerebroventricular kisspeptin-10 (Kp-10) elicited PRL release in a dose-dependent manner in estradiol (E2)-treated ovariectomized rats (OVX+E2), whereas no effect was found in oil-treated ovariectomized rats (OVX). Kp-10 increased PRL release in males and proestrous but not diestrous females. Associated with the increase in PRL release, intracerebroventricular Kp-10 reduced Fos-related antigen expression in tyrosine hydroxylase-immunoreactive (ir) neurons of arcuate and periventricular nuclei in OVX+E2 rats, with no effect in OVX rats. Kp-10 also decreased 3,4-dihydroxyphenylacetic acid concentration and 3,4-dihydroxyphenylacetic acid-DA ratio in the median eminence but not striatum in OVX+E2 rats. Double-label immunofluorescence combined with confocal microscopy revealed kisspeptin-ir fibers in close apposition to and in contact with tyrosine hydroxylase-ir perikarya in the arcuate. In addition, Kp-10 was not found to alter PRL release from anterior pituitary cell cultures regardless of E2 treatment. We provide herein evidence that kisspeptin regulates PRL release through inhibition of hypothalamic dopaminergic neurons, and that this mechanism is E2 dependent in females. These findings suggest a new role for central kisspeptin with possible implications for reproductive physiology.


Subject(s)
Dopamine/metabolism , Hypothalamus/cytology , Neurons/drug effects , Neurons/metabolism , Oligopeptides/pharmacology , Prolactin/metabolism , Animals , Cells, Cultured , Chromatography, High Pressure Liquid , Dinoprostone/pharmacology , Female , Immunohistochemistry , Kisspeptins , Male , Radioimmunoassay , Rats , Rats, Sprague-Dawley , Rats, Wistar
19.
Neurosci Lett ; 452(2): 146-50, 2009 Mar 13.
Article in English | MEDLINE | ID: mdl-19383429

ABSTRACT

Loss of appetite occurs in the cecal ligation and puncture (CLP) model of sepsis in conjunction with the activation of central neural stress pathways. Neuropeptide Y (NPY) in the arcuate nucleus of the hypothalamus is upregulated by several stressors and is stimulatory to feeding. To examine the response of NPY messenger RNA in the arcuate nucleus to sepsis, we used biotinylated RNA probes and a quantitative non-isotopic in situ hybridization approach in cryo-preserved sections from rats made septic by CLP. The mRNA in arcuate neurons was upregulated from the first day after CLP. By the afternoon of the third day through the morning of the fourth day, the average grey level of NPY mRNA clusters was 30% greater after CLP than after sham surgery (P<0.05), and the integrated optical density based on both the grey level and the amount of area with detectable mRNA was 60% greater after CLP than after sham surgery (P<0.03). Both the average grey level and area with detectable staining were positively correlated to plasma ACTH (r=0.953 and 0.917, respectively, n=10 and P<0.01 in each case). Thus sepsis increases the expression of the mRNA for NPY in the arcuate nucleus in proportion to the magnitude of the stress response. However, the suppression of feeding behavior in the CLP model suggests that sepsis activates additional mechanisms that negate the orexigenic contribution of the neuronal increase in NPY mRNA.


Subject(s)
Adrenocorticotropic Hormone/blood , Arcuate Nucleus of Hypothalamus/metabolism , Neuropeptide Y/genetics , Sepsis/blood , Stress, Physiological/physiology , Adrenal Cortex/metabolism , Adrenal Cortex Hormones/metabolism , Adrenocorticotropic Hormone/metabolism , Animals , Appetite/physiology , Arcuate Nucleus of Hypothalamus/cytology , Disease Models, Animal , Feeding Behavior/physiology , Male , Neurons/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Sepsis/genetics , Sepsis/physiopathology , Up-Regulation/physiology
20.
Curr Protoc Neurosci ; Chapter 2: Unit 2.12, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18972376

ABSTRACT

When using immunocytochemistry, investigators may not know how to optimize staining or how to troubleshoot the method when staining fails. Lacking are guides for comparing techniques and applying information derived from one staining method to another. Newer methods amplify signal detection, but will not necessarily work at the same primary antibody concentrations used for less sensitive reactions. Recommendations of optimal titers are often not accurate and are not usually accompanied by information on the method used to test those antibodies or the specifics of the assay. When the staining does not work, the investigators do not know how to determine if the antiserum is bad, the tissue is bad, or the method is inappropriate for their staining. This unit describes detailed procedures for determining optimal staining and applying that information to three common immunofluorescence methods. Lastly, a formula is provided for converting among the different methods.


Subject(s)
Antibodies/analysis , Antibodies/chemistry , Immunohistochemistry/methods , Animals , Antibodies/immunology , Antigens/immunology , Fluorescent Antibody Technique/methods , Fluorescent Dyes/chemistry , Immunohistochemistry/standards , Nervous System/chemistry , Nervous System/cytology , Nervous System/immunology , Rats , Staining and Labeling/methods , Staining and Labeling/standards , Tissue Fixation/methods , Titrimetry
SELECTION OF CITATIONS
SEARCH DETAIL
...