Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
J Comp Physiol B ; 187(4): 649-676, 2017 May.
Article in English | MEDLINE | ID: mdl-27987017

ABSTRACT

Grizzly bears (Ursus arctos horribilis) have evolved remarkable metabolic adaptations including enormous fat accumulation during the active season followed by fasting during hibernation. However, these fluctuations in body mass do not cause the same harmful effects associated with obesity in humans. To better understand these seasonal transitions, we performed insulin and glucose tolerance tests in captive grizzly bears, characterized the annual profiles of circulating adipokines, and tested the anorectic effects of centrally administered leptin at different times of the year. We also used bear gluteal adipocyte cultures to test insulin and beta-adrenergic sensitivity in vitro. Bears were insulin resistant during hibernation but were sensitive during the spring and fall active periods. Hibernating bears remained euglycemic, possibly due to hyperinsulinemia and hyperglucagonemia. Adipokine concentrations were relatively low throughout the active season but peaked in mid-October prior to hibernation when fat content was greatest. Serum glycerol was highest during hibernation, indicating ongoing lipolysis. Centrally administered leptin reduced food intake in October, but not in August, revealing seasonal variation in the brain's sensitivity to its anorectic effects. This was supported by strong phosphorylated signal transducer and activator of transcription 3 labeling within the hypothalamus of hibernating bears; labeling virtually disappeared in active bears. Adipocytes collected during hibernation were insulin resistant when cultured with hibernation serum but became sensitive when cultured with active season serum. Heat treatment of active serum blocked much of this action. Clarifying the cellular mechanisms responsible for the physiology of hibernating bears may inform new treatments for metabolic disorders.


Subject(s)
Adipose Tissue/metabolism , Hibernation/physiology , Insulin Resistance/physiology , Ursidae/physiology , Adipokines/blood , Animals , Brain/metabolism , Eating , Female , Glucose/metabolism , Glucose Tolerance Test , Leptin/blood , Leptin/pharmacology , Lipogenesis/physiology , Lipolysis/physiology , Male , Proteins/metabolism , Seasons
2.
Cytotechnology ; 68(5): 2177-91, 2016 Oct.
Article in English | MEDLINE | ID: mdl-26856588

ABSTRACT

Brown bears (Ursus arctos) exhibit hyperphagia each fall and can become obese in preparation for hibernation. They do this without displaying the physiological problems typically seen in obese humans, such as Type 2 diabetes and heart disease. The study of brown bear hibernation biology could therefore aid in the development of novel methods for combating metabolic diseases. To this end, we isolated mesenchymal stem cells from subcutaneous fat biopsies, and culture methods were developed to differentiate these into the adipogenic lineage. Biopsies were taken from 8 captive male (N = 6) and female (N = 2) brown bears, ages 2-12 years. Plastic adherent, fibroblast-like cells were proliferated and subsequently cryopreserved or differentiated. Differentiation conditions were optimized with respect to fetal bovine serum content and time spent in differentiation medium. Cultures were characterized through immunostaining, RT-qPCR, and Oil red O staining to quantify lipid accumulation. Adiponectin, leptin, and glycerol medium concentrations were also determined over the course of differentiation. The culturing protocol succeeded in generating hormone-sensitive lipase-expressing, lipid-producing white-type adipocytes (UCP1 negative). Serum concentration and time of exposure to differentiation medium were both positively related to lipid production. Cells cultured to low passage numbers retained similar lipid production and expression of lipid markers PLIN2 and FABP4. Ultimately, the protocols described here may be useful to biologists in the field investigating the health of wild bear populations and could potentially increase our understanding of metabolic disorders in humans.

3.
Eur J Pharm Sci ; 70: 29-36, 2015 Apr 05.
Article in English | MEDLINE | ID: mdl-25603031

ABSTRACT

Human organic anion-transporting polypeptide 1B1 (OATP1B1) and OATP1B3 are important hepatic uptake transporters. Early assessment of OATP1B1/1B3-mediated drug-drug interactions (DDIs) is therefore important for successful drug development. A promising approach for early screening and prediction of DDIs is computational modeling. In this study we aimed to generate a rapid, single Bayesian prediction model for OATP1B1, OATP1B1∗15 and OATP1B3 inhibition. Besides our previously generated HEK-OATP1B1 and HEK-OATP1B1∗15 cells, we now generated and characterized HEK-OATP1B3 cells. Using these cell lines we investigated the inhibitory potential of 640 FDA-approved drugs from a commercial library (10µM) on the uptake of [(3)H]-estradiol-17ß-d-glucuronide (1µM) by OATP1B1, OATP1B1∗15, and OATP1B3. Using a cut-off of ⩾60% inhibition, 8% and 7% of the 640 drugs were potent OATP1B1 and OATP1B1∗15 inhibitors, respectively. Only 1% of the tested drugs significantly inhibited OATP1B3, which was not sufficient for Bayesian modeling. Modeling of OATP1B1 and OATP1B1∗15 inhibition revealed that presence of conjugated systems and (hetero)cycles with acceptor/donor atoms in- or outside the ring enhance the probability of a molecule binding these transporters. The overall performance of the model for OATP1B1 and OATP1B1∗15 was ⩾80%, including evaluation with a true external test set. Our Bayesian classification model thus represents a fast, inexpensive and robust means of assessing potential binding of new chemical entities to OATP1B1 and OATP1B1∗15. As such, this model may be used to rank compounds early in the drug development process, helping to avoid adverse effects in a later stage due to inhibition of OATP1B1 and/or OATP1B1∗15.


Subject(s)
Drug Evaluation, Preclinical/methods , Models, Biological , Organic Anion Transporters, Sodium-Independent/physiology , Organic Anion Transporters/physiology , Pharmaceutical Preparations/metabolism , Bayes Theorem , Drug Interactions/physiology , Forecasting , HEK293 Cells , Humans , Liver-Specific Organic Anion Transporter 1 , Solute Carrier Organic Anion Transporter Family Member 1B3
4.
J Pharmacol Exp Ther ; 347(3): 635-44, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24049060

ABSTRACT

Correct prediction of human pharmacokinetics (PK) and the safety and efficacy of novel compounds based on preclinical data, is essential but often fails. In the current study, we aimed to improve the predictive value of ApoE*3Leiden (E3L) transgenic mice regarding the cholesterol-lowering efficacy of various statins in humans by combining pharmacokinetic with efficacy data. The efficacy of five currently marketed statins (atorvastatin, simvastatin, lovastatin, pravastatin, and rosuvastatin) in hypercholesterolemic patients (low-density lipoprotein ≥ 160 mg/dl) was ranked based on meta-analysis of published human trials. Additionally, a preclinical combined PK efficacy data set for these five statins was established in E3L mice that were fed a high-cholesterol diet for 4 weeks, followed by 6 weeks of drug intervention in which statins were supplemented to the diet. Plasma and tissue levels of the statins were determined on administration of (radiolabeled) drugs (10 mg/kg p.o.). As expected, all statins reduced plasma cholesterol in the preclinical model, but a direct correlation between cholesterol lowering efficacy of the different statins in mice and in humans did not reach statistical significance (R(2) = 0.11, P < 0.57). It is noteworthy that, when murine data were corrected for effective liver uptake of the different statins, the correlation markedly increased (R(2) = 0.89, P < 0.05). Here we show for the first time that hepatic uptake of statins is related to their cholesterol-lowering efficacy and provide evidence that combined PK and efficacy studies can substantially improve the translational value of the E3L mouse model in the case of statin treatment. This strategy may also be applicable for other classes of drugs and other preclinical models.


Subject(s)
Anticholesteremic Agents/pharmacokinetics , Anticholesteremic Agents/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hypercholesterolemia/drug therapy , Translational Research, Biomedical/methods , Animals , Apolipoproteins E/metabolism , Body Weight/drug effects , Cholesterol/blood , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Eating/physiology , Female , Hypercholesterolemia/blood , Lipids/blood , Mice , Mice, Transgenic
5.
Neuroscience ; 176: 284-95, 2011 Mar 10.
Article in English | MEDLINE | ID: mdl-21185915

ABSTRACT

The rewarding influence of drugs of abuse varies with time of day and appears to involve interactions between the circadian and the mesocorticolimbic dopamine systems. The circadian system is also intimately involved in measuring daylength. Thus, the present study examined the impact of changing daylength (photoperiod) on cocaine-seeking behaviors. Male Sprague-Dawley rats were trained and tested on a 12L:12D light:dark schedule for cocaine-induced reinstatement of conditioned place preference (CPP) at three times of day (Zeitgeber time (ZT): 4, 12, and 20) to determine a preference score. Rats were then shifted to either shorter (6L:18D) or longer (18L:6D) photoperiods and then to constant conditions, re-tested for cocaine-induced reinstatement under each different condition, and then returned to their original photoperiod (12L:12D) and tested once more. Rats exhibited a circadian profile of preference score in constant darkness with a peak at 12 h after lights-off. At both ZT4 and ZT20, but not at ZT12, shorter photoperiods profoundly suppressed cocaine reinstatement, which did not recover even after switching back to 12L:12D. In contrast, longer photoperiods did not alter reinstatement. Separate studies showed that the suppression of cocaine reinstatement was not due to repeated testing. In an additional experiment, we examined the photoperiodic regulation of tyrosine hydroxylase (TH) and dopamine transporter (DAT) proteins in drug-naive rats. These results revealed photoperiodic modulation of proteins in the prefrontal cortex and dorsal striatum, but not in the nucleus accumbens or ventral tegmental area. Together, these findings add further support to the circadian genesis of cocaine-seeking behaviors and demonstrate that drug-induced reinstatement is modulated by photoperiod. Furthermore, the results suggest that photoperiod partly contributes to the seasonal expression of certain drug-related behaviors in humans living at different latitudes and thus our findings may have implications for novel targeting of circadian rhythms in the treatment of addiction.


Subject(s)
Behavior, Addictive/physiopathology , Brain/physiology , Circadian Rhythm/physiology , Cocaine-Related Disorders/physiopathology , Drug-Seeking Behavior/physiology , Animals , Behavior, Animal , Blotting, Western , Brain/drug effects , Conditioning, Psychological , Dopamine Plasma Membrane Transport Proteins/biosynthesis , Male , Photoperiod , Rats , Rats, Sprague-Dawley , Tyrosine 3-Monooxygenase/biosynthesis
6.
J Neuroendocrinol ; 22(7): 674-81, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20456601

ABSTRACT

Seasonal reproduction in ewes is caused by a dramatic increase in response to oestradiol (E(2)) negative feedback during the nonbreeding (anoestrous) season. Considerable evidence supports the hypothesis that A15 dopaminergic neurones in the retrochiasmatic area (RCh) play a key role in these seasonal changes. These A15 neurones are stimulated by E(2) and inhibit gonadotrophin-releasing hormone (GnRH) secretion in anoestrus, but not the breeding season. Because A15 neurones do not contain oestrogen receptors-alpha (ER alpha), it is likely that E(2)-responsive afferents stimulate their activity when circulating E(2) levels increase during anoestrus. Retrograde tract tracing studies identified a limited set of ER alpha-containing afferents primarily found in four areas [ventromedial preoptic area, RCh, ventromedial and arcuate (ARC) nuclei]. Pharmacological and anatomical data are consistent with GABA- and glutamate-containing afferents controlling A15 activity in anoestrus, with E(2) inhibiting GABA and stimulating glutamate release at this time of year. Tract tracing demonstrated that A15 efferents project posteriorly to the median eminence and the ARC, suggesting possible direct actions on GnRH terminals or indirect actions via kisspeptin neurones in the ARC to inhibit GnRH in anoestrus. Identification of this neural circuitry sets the stage for the development of specific hypotheses for morphological or transmitter/receptor expression changes that would account for seasonal breeding in ewes.


Subject(s)
Breeding , Neurons/physiology , Reproduction/physiology , Seasons , Sheep/physiology , Animals , Brain/anatomy & histology , Brain/metabolism , Dopamine/metabolism , Estradiol/metabolism , Female , Nerve Net/anatomy & histology , Nerve Net/physiology , Neurons/cytology , gamma-Aminobutyric Acid/metabolism
7.
Virology ; 303(2): 364-73, 2002 Nov 25.
Article in English | MEDLINE | ID: mdl-12490397

ABSTRACT

Arteriviruses are enveloped, positive-strand RNA viruses for which the two major envelope proteins GP(5) and M occur as disulfide-linked heterodimers. These were assumed to serve the viral targeting functions, but recent ectodomain swapping studies with equine arteritis virus (EAV) indicate that the GP(5) protein does not determine arteriviral tropism. Here, we focused on the short, 13- to 18-residue ectodomain of the M protein. Using an infectious cDNA clone of the Lelystad virus isolate of porcine reproductive and respiratory syndrome virus (PRRSV), we substituted the genomic sequence encoding the M ectodomain by that of murine lactate dehydrogenase-elevating virus, EAV, and the US PRRSV-isolate, VR2332. Viable viruses with a chimeric M protein were obtained in all three cases, but for the latter two only after removal of the genomic overlap between the M and GP(5) genes. Characterization of the chimeric viruses revealed that they could be distinguished immunologically from wild-type virus, that they were genetically stable in vitro, but that they were impaired in their growth, reaching lower titers than the parental virus. The latter appeared to be due to an increased particle-to-infectivity ratio of the chimeric virus particles. Interestingly, the chimeric viruses had retained their ability to infect porcine cells and had not acquired tropism for cells susceptible to the viruses from which the foreign ectodomains were derived. We conclude that the surface structures composed by the arterivirus M and GP(5) ectodomains do not determine viral tropism.


Subject(s)
Arterivirus/physiology , Recombinant Fusion Proteins/physiology , Viral Matrix Proteins/physiology , Amino Acid Sequence , Animals , Arterivirus/genetics , Arterivirus/immunology , Base Sequence , Equartevirus/physiology , Lactate dehydrogenase-elevating virus/physiology , Molecular Sequence Data , Open Reading Frames , Porcine respiratory and reproductive syndrome virus/physiology , Swine , Transfection , Viral Envelope Proteins/physiology , Viral Matrix Proteins/chemistry
8.
Biol Reprod ; 65(3): 866-72, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11514352

ABSTRACT

Ovarian growth and development are critically dependent upon the influence of endogenous estrogens, and both are highly regulated during the reproductive cycle. The observation that estrogen-receptor-alpha-deficient mice still exhibit follicular growth and development, together with other evidence, suggests that responsiveness of the ovary to estradiol occurs predominantly through the second estrogen receptor, ERbeta. We characterized the physiological regulation of ERbeta expression in ovarian follicles during the follicular phase of sheep that were synchronized for estrus during the breeding season with intravaginal progesterone implants (controlled internal drug release [CIDR] device; InterAg, Hamilton, New Zealand). Ovaries were removed at times corresponding to the early (EF) and late follicular phases (LF) of the ovine estrous cycle (12 h [n = 5] and 32 h [n = 5] after CIDR device removal, respectively). Sections of ovary were then hybridized with a cRNA probe corresponding to the 5' region of ovine ERbeta. ERbeta mRNA expression within the granulosa layer of different size follicles (size classes: < or =3 mm, 3.1-4.0 mm, 4.1-5.0 mm, >5 mm) was quantified. ERbeta mRNA expression varied both with follicle size (P < 0.01) and with cycle stage (P < 0.01). In EF ewes, the highest levels of ERbeta mRNA expression were found in follicles < or = 3 mm in size. ERbeta mRNA expression declined progressively thereafter among the different size classes with lowest levels expressed in >5-mm follicles. By contrast, expression of ERbeta mRNA in the 3.1- to 4.0-mm follicles of LF group was significantly higher than in the < or =3-mm size follicles and declined thereafter progressively to the >5-mm size levels as in the EF group. Furthermore, expression of ERbeta mRNA in < or =3-mm size follicles of LF group was significantly lower than the corresponding size class in the EF group. Lower expression of ERbeta mRNA in >5-mm follicle is suggestive of a down-regulation by the local estrogen milieu.


Subject(s)
Gene Expression Regulation , Ovulation , RNA, Messenger/analysis , Receptors, Estrogen/genetics , Sheep/physiology , Animals , Base Sequence , DNA, Complementary/chemistry , Estrogen Receptor beta , Female , Follicular Phase , Humans , In Situ Hybridization , Luteinizing Hormone/metabolism , Molecular Sequence Data , Sequence Alignment , Sequence Analysis, DNA
9.
Endocrinology ; 142(3): 1317-24, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11181550

ABSTRACT

The GnRH neurosecretory system undergoes marked structural and functional changes throughout life. The initial goal of this study was to examine the neuroanatomical relationship between GnRH neurons and a glycoprotein implicated in neuroplasticity, the polysialylated form of neural cell adhesion molecule (PSA-NCAM). Using dual label immunocytochemistry in conjunction with confocal microscopy, we determined that fibers, terminals, and perikarya of GnRH neurons in adult ovariectomized ewes are intimately associated with PSA-NCAM. In the preoptic area, intense PSA-NCAM immunoreactivity was evident around the periphery of GnRH cell bodies. The second goal of this study was to determine whether PSA-NCAM expression associated with GnRH neurons varies in conjunction with seasonal changes in the activity of the GnRH neurosecretory system in ovariectomized ewes treated with constant release implants of estradiol. During the breeding season when reproductive neuroendocrine activity was enhanced, the expression of PSA-NCAM immunoreactivity associated with GnRH neurons was significantly greater than that during anestrus when GnRH secretion was reduced. This difference, which occurred despite an unchanging ovarian steroid milieu, was not observed in preoptic area structures devoid of GnRH immunoreactivity, suggesting that the seasonal change is at least partially specific to the GnRH system. The close association between PSA-NCAM and GnRH neurons and the change in this relationship in conjunction with seasonal alterations in GnRH secretion provide anatomical evidence that this molecule may contribute to seasonal remodeling of the GnRH neurosecretory system of the adult.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Neural Cell Adhesion Molecule L1 , Neural Cell Adhesion Molecules/physiology , Neuronal Plasticity/physiology , Neurosecretory Systems/physiology , Sialic Acids/physiology , Animals , Drug Implants , Estradiol/pharmacology , Female , Luteinizing Hormone/blood , Neural Cell Adhesion Molecules/metabolism , Neurons/metabolism , Ovariectomy , Reproduction/physiology , Seasons , Sheep , Sialic Acids/metabolism , Staining and Labeling
10.
Brain Res Mol Brain Res ; 86(1-2): 145-52, 2001 Jan 31.
Article in English | MEDLINE | ID: mdl-11165381

ABSTRACT

Stress-induced analgesia is a well-documented phenomenon that occurs in all mammalian species. Forced cold water swim produces a type of stress-induced analgesia that is independent of mu opioid receptors. The neuropeptide neurotensin (NT) has been implicated in mu opioid-independent analgesia (MOIA), but the circuitry of this system is largely unknown. The medial preoptic area (MPO) and lateral hypothalamus (LH) are two regions that are known to modulate pain processing. These two regions also contain neurotensinergic projections to the periaqueductal gray, a region that has been shown to produce MOIA upon injection of NT. The goal of this study was to determine if cold water swim (CWS) stress, which produces MOIA, activates the NT-ergic systems in these two regions. In situ hybridization results indicate that CWS increases the level of NT mRNA within neurons in the MPO and LH, suggesting that these two regions are activated during this process.


Subject(s)
Cold Temperature , Hypothalamic Area, Lateral/physiology , Neurotensin/genetics , Preoptic Area/physiology , Stress, Physiological/physiopathology , Animals , Gene Expression/physiology , Hot Temperature , In Situ Hybridization , Male , Pain Threshold/physiology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Swimming/physiology
11.
Mol Vis ; 6: 101-8, 2000 Jun 24.
Article in English | MEDLINE | ID: mdl-10869099

ABSTRACT

PURPOSE: To examine the effect of loss of cone photoreceptor cells on retinal degeneration. METHODS: We previously identified a cone photoreceptor cell-specific promoter of human cone transducin a-subunit (GNAT2) gene. In this report, a minigene, Trc-Tox176, that contains the GNAT2 promoter, an attenuated diphtheria toxin A-chain gene, and an enhancer element from human interphotoreceptor retinoid-binding protein (IRBP) was used to generate coneless transgenic mice. Transgenic mice were identified by PCR and the copy number of the transgene was determined by Southern hybridization, and examined by histology. RESULTS: The results of immunostaining with anti-mouse GNAT2 antibodies and reverse transcription-PCR (RT-PCR) analysis with mRNA from the retinas of transgenic mice showed that cone photoreceptor cells were ablated in one of four transgenic mouse lines. The ablation of cone cells began at postnatal day 8, at the same time as the expression of endogenous GNAT2. An age-related rod degeneration was also found in this cone-ablated mouse line, beginning at postnatal day 9, proceeding from the central retina to the peripheral retina. CONCLUSIONS: Cone photoreceptor cells may play an important role in the survival of rod photoreceptor cells during mouse retina development.


Subject(s)
Retinal Cone Photoreceptor Cells/pathology , Retinal Degeneration/genetics , Animals , Blotting, Southern , Diphtheria Toxin/genetics , Female , Humans , Immunohistochemistry , Male , Mice , Mice, Transgenic , Promoter Regions, Genetic , Retina/growth & development , Retina/metabolism , Retina/pathology , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinal Rod Photoreceptor Cells/pathology , Reverse Transcriptase Polymerase Chain Reaction , Transducin/genetics , Transducin/metabolism
12.
Endocrinology ; 141(1): 190-6, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10614639

ABSTRACT

In the ewe, seasonal anestrus results from a change in the hypothalamic responsiveness to estradiol (E2) negative feedback. Considerable evidence has implicated a specific group of dopaminergic neurons (the A15 group) in this seasonally dependent E2 effect, but these neurons do not appear to contain estrogen receptor-alpha (ERalpha). This apparent discrepancy raises the possibility that at least one other neural system is also involved in mediating E2 inhibition. The purpose of this study was to determine whether ERalpha-containing neurons are activated by the negative feedback action of E2 in anestrus. In Exp 1, we examined the effects of E2 on expression of the immediate early gene products, Fos and Fos-related antigens, in ERalpha-positive cells in anestrous ewes. ERalpha and Fos/Fos-related antigens were colocalized using a dual immunofluorescence procedure in sections throughout the hypothalamus from ovariectomized and E2-treated ovariectomized anestrous ewes. A low dose E2 treatment that inhibited LH pulse frequency and induced Fos in A15 dopaminergic neurons in a previous study significantly increased the percentage of ERalpha-containing neurons expressing Fos (17.8% vs. 1.7%) in the medial preoptic area, but not in other hypothalamic areas. In Exp 2, we determined whether there was a seasonal difference in the effect of E2 on Fos/ERalpha colocalization in this region. E2 treatment produced a 3-fold increase in the percentage of ERalpha-positive cells expressing Fos (15.1% vs. 3.4%) in anestrus, but failed to increase ERalpha/Fos colocalization (1.8% vs. 3.5%) during the breeding season. These data raise the possibility that a subset of ERalpha-containing neurons in the medial preoptic area plays a role in the seasonal change in response to E2 negative feedback in the ewe.


Subject(s)
Anestrus/physiology , Estradiol/pharmacology , Gene Expression Regulation/drug effects , Genes, fos/drug effects , Neurons/metabolism , Preoptic Area/metabolism , Receptors, Estrogen/biosynthesis , Animals , Cell Count , Female , Fluorescent Antibody Technique, Indirect , Luteinizing Hormone/metabolism , Neurons/drug effects , Preoptic Area/cytology , Preoptic Area/drug effects , Radioimmunoassay , Receptors, Estrogen/genetics , Seasons , Sheep , Tyrosine 3-Monooxygenase/biosynthesis , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
13.
Neuroreport ; 10(15): 3165-74, 1999 Oct 19.
Article in English | MEDLINE | ID: mdl-10574554

ABSTRACT

It is well established that the mammalian circadian system consists of pacemaker cells in the suprachiasmatic nuclei (SCN). The mouse has become increasingly important in understanding the circadian timing system, due to the availability of mutant animals with abnormal circadian rhythms. In the present paper, we describe the organization of the mouse SCN, comparing the wild type and Clock mutant animal, with a special focus on those peptides bearing an upstream E-box element (vasopressin, vasoactive intestinal peptide, cholecystokinin and substance P). To this end, we describe the distribution of the foregoing SCN peptidergic cell types as well as gastrin-related peptide, calretinin, calbindin, somatostatin, neurotensin and retinal input to the SCN (determined by both tract tracing and fos-immunoreactivity in response to a light pulse). The Clock mutant mouse has decreased expression of vasopressin mRNA and protein in the SCN, with normal patterns of expression elsewhere in the brain. No other differences were detected between the Clock mutant and the wild type mouse. The results are consistent with the hypothesis that there are multiple regulatory elements of clock-controlled genes in the SCN.


Subject(s)
Circadian Rhythm/physiology , Neuropeptides/metabolism , Suprachiasmatic Nucleus/metabolism , Suprachiasmatic Nucleus/physiology , Animals , Antibody Specificity , Brain Mapping , Calbindin 2 , Calbindins , Cholecystokinin/analysis , Gastrin-Releasing Peptide/analysis , Gene Expression Regulation , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mutation/genetics , Neuropeptide Y/analysis , Neuropeptides/genetics , Neurotensin/analysis , RNA, Messenger/analysis , Retina/cytology , S100 Calcium Binding Protein G/analysis , Somatostatin/analysis , Substance P/analysis , Vasoactive Intestinal Peptide/analysis , Vasopressins/analysis , Visual Pathways/cytology
14.
J Neurosci Methods ; 91(1-2): 1-8, 1999 Sep 15.
Article in English | MEDLINE | ID: mdl-10522819

ABSTRACT

In studying reciprocally connected brain networks, it is advantageous to use techniques that allow simultaneous visualization of both efferent and afferent connections from a single injection site. We report on a new technique to achieve this using pressure injections of a mixture of biotinylated dextran amine (BDA) and the beta subunit of cholera toxin (Ctb). Adult male hamsters (n = 12) received 20-30-nl injections of either a 1:1 mixture of BDA (Sigma, 10%) and Ctb (List Biological, 0.5%), or each tracer by itself, into the medial amygdala. Adult female sheep (n = 4) received 200-300 nl of the combined tracer into the A15 region of the hypothalamus. After 1 (hamster) or 2 weeks' (sheep) survival, animals were perfused with 4% paraformaldehyde. Sections were double-labeled, first for BDA histochemistry using nickel-enhanced DAB, then for Ctb using a PAP technique and unenhanced DAB. In all animals, combined injections resulted in clear and consistent patterns of both anterograde and retrograde labeling. Ctb immunoreactivity was distinct and easily distinguished from BDA labeling. There was no evidence for loss of sensitivity of either tracer due to the combined delivery; no differences were seen between combined or single tracer injections in numbers of retrogradely-labeled cells or in the distribution of anterogradely-labeled fibers. In summary, the combined delivery of BDA and Ctb is an easy and reliable technique for simultaneous afferent and efferent tract tracing in both small and large animals; it could potentially be combined with immunocytochemistry to determine the neurochemical content of labeled cells or fibers.


Subject(s)
Biotin/analogs & derivatives , Brain/cytology , Cholera Toxin , Dextrans , Fluorescent Dyes , Neural Pathways/cytology , Animals , Cricetinae , Histocytochemistry/methods , Male , Sheep
15.
Neuroendocrinology ; 69(6): 430-7, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10364695

ABSTRACT

The mechanism whereby undernutrition enhances the ability of estradiol (E) to inhibit reproductive activity is unknown. This study aimed to determine the effect of feed restriction on E receptor (ER)-containing cell numbers in the female sheep hypothalamus. Ovariectomized lambs at 7 months of age received either ad libitum (AL; n = 5) or restricted (FR; n = 10) levels of feed intake. Lambs were weighted weekly and FR lambs fed to lose approximately 15% of their initial body weights over 7 weeks, at the end of which jugular blood samples were collected at 10-min intervals for 5 h to assess the patterns of LH release. After blood collection, lambs were euthanized and hypothalami collected for immunocytochemical detection of ER. Based on LH secretory profiles, FR lambs were subdivided into two groups. The first group (FR + LH; n = 5) exhibited patterns of LH release similar to AL controls. LH secretion in the second group (FR-LH; n = 5) was obviously suppressed. Numbers of ER-containing cells did not differ significantly (p > 0.10) among treatment groups in the bed nucleus stria terminalis, anterior hypothalamic area and arcuate nucleus. ER-containing cell numbers were greater (p < 0.05) in the preoptic area (POA) but less (p < 0.05) in the ventromedial/ventrolateral hypothalamus (VMH/VLH) for FR-LH lambs compared to AL animals. Notably, for both the POA and VMH/VLH, ER-containing cell numbers in the FR + LH animals were intermediate and did not differ (p > 0.10) from either FR-LH or AL lambs. These results suggest that feed restriction differentially alters ER-containing cell numbers in specific regions of the ovine hypothalamus (numbers increased in the POA but decreased in the VMH/VLH). These changes may, at least in part, represent a mechanism whereby undernutrition enhances the ability of E to inhibit reproduction.


Subject(s)
Eating/physiology , Hypothalamus/metabolism , Receptors, Estrogen/metabolism , Animals , Body Weight/physiology , Cell Count , Estrogens/metabolism , Female , Hypothalamus/cytology , Immunohistochemistry , Luteinizing Hormone/metabolism , Ovariectomy , Sheep
16.
Brain Res Bull ; 45(1): 27-39, 1998.
Article in English | MEDLINE | ID: mdl-9434199

ABSTRACT

Pheromonal stimuli elicit rapid behavioral and reproductive endocrine changes in the ewe. The neural pathways responsible for these effects in sheep are unknown, in part, because the olfactory bulb projections have not been examined in this species. Using the anterograde and retrograde neuronal tracer, wheat germ agglutinin-conjugated horseradish peroxidase (WGA-HRP), we describe the afferent and efferent olfactory bulb connections of the Suffolk ewe. Injections of WGA-HRP limited to the main olfactory bulb resulted in retrograde labeling of cells in numerous telencephalic, diencephalic, and metencephalic regions. Terminal labeling was limited to layer la of ipsilateral cortical structures extending rostrally from the anterior olfactory nucleus (AON), piriform cortex, anterior-, and posterolateral-cortical amygdaloid nuclei to lateral entorhinal cortex caudally. Injections involving the accessory olfactory bulb and AON produced additional labeling of cells within the bed nucleus of the stria terminalis (BNST), medial nucleus of the amygdala, and a few cells in the posteromedial cortical nucleus of the amygdala. Terminal labeling included a small dorsomedial quadrant of BNST and also extended to the far lateral portions of the supraoptic nucleus. A clearly defined accessory olfactory tract and nucleus was not evident, perhaps due to limitations in the sensitivity of the method. With this possible exception, the afferent and efferent olfactory connections in the sheep appear similar to those reported for other species.


Subject(s)
Brain/physiology , Efferent Pathways/physiology , Olfactory Bulb/anatomy & histology , Olfactory Bulb/physiology , Olfactory Pathways/anatomy & histology , Olfactory Pathways/physiology , Amygdala/anatomy & histology , Amygdala/physiology , Animals , Axonal Transport , Brain/anatomy & histology , Diencephalon/anatomy & histology , Diencephalon/physiology , Efferent Pathways/anatomy & histology , Female , Nerve Endings/physiology , Nerve Endings/ultrastructure , Pons/anatomy & histology , Pons/physiology , Sheep , Telencephalon/anatomy & histology , Telencephalon/physiology , Wheat Germ Agglutinin-Horseradish Peroxidase Conjugate
17.
Endocrinology ; 138(11): 5039-47, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9348236

ABSTRACT

Thyroid hormones appear to play an important role in the seasonal reproductive transitions of a number of mammalian and avian species. These seasonal transitions as well as the effects of thyroid hormones on the reproductive neuroendocrine axis are mediated by the GnRH system. How thyroid hormones affect the GnRH system is unclear. Double label immunocytochemistry was used to examine GnRH- and other neurotransmitter/neuropeptide-containing neurons for thyroid hormone receptor (alphaTHR) colocalization in two seasonal breeders, the golden hamster and the sheep. AlphaTHR was identified in hamster and sheep brain by Western blot analysis. Furthermore, alphaTHR immunoreactivity was widely distributed in brain and was colocalized in identified populations: GnRH neurons (hamster, 28%; sheep, 46%); dopaminergic neurons of the A14 (hypothalamic) and A16 (olfactory bulb) cell groups, but not in the hypothalamic A13 cell group; and neurophysin-immunoreactive neurons of the supraoptic and paraventricular nuclei. The finding of alphaTHR in GnRH and A14 dopamine neurons provides an anatomical substrate for direct thyroid hormone action on the reproductive neuroendocrine system of these two seasonally breeding species. It remains to be determined whether the GnRH gene itself or the gene of another constituent within the same GnRH neuron is responsive to thyroid hormones.


Subject(s)
Brain/metabolism , Cricetinae/metabolism , Gonadotropin-Releasing Hormone/metabolism , Neurons/metabolism , Receptors, Thyroid Hormone/metabolism , Sheep/metabolism , Animals , Blotting, Western , Brain/cytology , Dopamine/metabolism , Female , Immunohistochemistry , Male , Neurophysins/metabolism , Tissue Distribution , Tyrosine 3-Monooxygenase/metabolism
18.
J Exp Med ; 185(8): 1517-22, 1997 Apr 21.
Article in English | MEDLINE | ID: mdl-9126934

ABSTRACT

The mu opioid receptor is thought to be the cellular target of opioid narcotics such as morphine and heroin, mediating their effects in both pain relief and euphoria. Its involvement is also implicated in a range of diverse biological processes. Using a mouse model in which the receptor gene was disrupted by targeted homologous recombination, we explored the involvement of this receptor in a number of physiological functions. Mice homozygous for the disrupted gene developed normally, but their motor function was altered. Drug-naive homozygotes displayed reduced locomotor activity, and morphine did not induce changes in locomotor activity observed in wild-type mice. Unexpectedly, lack of a functional receptor resulted in changes in both the host defense system and the reproductive system. We observed increased proliferation of granulocyte-macrophage, erythroid, and multipotential progenitor cells in both bone marrow and spleen, indicating a link between hematopoiesis and the opioid system, both of which are stress-responsive systems. Unexpected changes in sexual function in male homozygotes were also observed, as shown by reduced mating activity, a decrease in sperm count and motility, and smaller litter size. Taken together, these results suggest a novel role of the mu opioid receptor in hematopoiesis and reproductive physiology, in addition to its known involvement in pain relief.


Subject(s)
Behavior, Animal/physiology , Hematopoiesis , Receptors, Opioid, mu/deficiency , Animals , Female , Male , Mice , Mice, Knockout , Motor Activity/physiology , Sexual Behavior, Animal/physiology , Sperm Motility
19.
Biol Reprod ; 56(3): 655-62, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9047010

ABSTRACT

The final common pathway controlling reproductive function in vertebrates is the GnRH neuron and its projection to the median eminence (ME), site of peptide release into the pituitary portal system. GnRH neurons are widely distributed; therefore we sought to test the hypothesis that those projecting to the ME are located in specific regions. We used as a model the sheep, a species in which a great deal of information regarding the physiology of GnRH secretion is known. To identify cells projecting to the ME (i.e., neuroendocrine neurons), ewes (n = 10) received injections into the ME of neuronal tract-tracing compounds: cholera toxin-beta subunit (CT-beta) or one of two fluorescent compounds (rhodamine isothiocyanate or fluorescein-conjugated dextran). Forty-eight h later, animals were perfused intracranially and their brains were processed for immunocytochemical localization of GnRH and CT-beta using a dual-immunofluorescent procedure or by single-label immunofluorescent visualization of GnRH combined with direct visualization of fluorescent tracers. Small, well-circumscribed injections into the ME were made successfully in 6 of 10 animals, and these overlapped the location of GnRH terminals and fibers. Neuroendocrine GnRH neurons (those GnRH neurons containing retrogradely transported tracer) were identified throughout their previously reported range: within the diagonal band of the Broca/medial septal region, medial preoptic area (MPOA), anterior hypothalamic area, and medial basal hypothalamus. Although the absolute number of neuroendocrine GnRH neurons varied by region, the percentage of the total GnRH population within each of these areas that was retrogradely labeled did not differ (p > 0.05). Injections placed unilaterally within the ME labeled a similar proportion of GnRH cells both ipsilateral and contralateral to the injection site in all areas except the MPOA, where ipsilaterally labeled cells were approximately twice as numerous as those labeled contralaterally. Injections that missed the ME and were placed either into the third ventricle or into the arcuate nucleus labeled only 0.5% and 4-11% of GnRH neurons, respectively. These results do not support the hypothesis that in the ewe, GnRH neurons projecting to the ME are localized to specific regions. Thus, we postulate that GnRH release into the hypophyseal portal system reflects the output of GnRH neurons located in multiple areas.


Subject(s)
Brain/cytology , Gonadotropin-Releasing Hormone/physiology , Neurons/physiology , Neurosecretory Systems/physiology , Animals , Brain Chemistry/physiology , Cholera Toxin , Female , Fluorescent Antibody Technique, Direct , Frontal Lobe/cytology , Frontal Lobe/metabolism , Hypothalamus/cytology , Hypothalamus/metabolism , Immunohistochemistry , Injections, Intraventricular , Neural Pathways/cytology , Neural Pathways/physiology , Sheep , Tissue Fixation
20.
Brain Res Bull ; 44(4): 445-57, 1997.
Article in English | MEDLINE | ID: mdl-9370210

ABSTRACT

Seasonal breeders, such as sheep and hamsters, by virtue of their annual cycles of reproduction, represent valuable models for the study of plasticity in the adult mammalian neuroendocrine brain. A major factor responsible for the occurrence of seasonal reproductive transitions is a striking change in the responsiveness of gonadotropin-releasing hormone (GnRH) neurons to the inhibitory effects of gonadal steroids. However, the neural circuitry mediating these seasonal changes is still relatively unexplored. In this article, we review recent findings that have begun to define that circuitry and its plasticity in a well-studied seasonal breeder, the ewe. Tract tracing studies and immunocytochemical analyses using Fos and FRAs as markers of activation point to a subset of neuroendocrine GnRH neurons in the MBH as potential mediators of pulsatile GnRH secretion. Because the vast majority of GnRH neurons lack estrogen receptors, seasonal changes in responsiveness to estradiol are most probably conveyed by afferents. Two possible mediators of this influence are dopaminergic cells in the A14/A15 cell groups of the hypothalamus, and estrogen receptor-containing cells in the arcuate nucleus that project to the median eminence. The importance of GnRH afferents in the regulation of season breeding is underscored by observations of seasonal changes in the density of synaptic inputs onto GnRH neurons. Thyroid hormones may participate in this remodeling, because they are important in seasonal reproduction, influence the morphology of other brain systems, and thyroid hormone receptors are expressed within GnRH neurons. Finally, in the hamster, neonatal hypothyroidism affects the number of caudally placed GnRH neurons in the adult brain, suggesting that thyroid hormones may influence development of the GnRH system as well as its reproductive functions in the adult brain.


Subject(s)
Brain/physiology , Gonadotropin-Releasing Hormone/physiology , Neurons/physiology , Neurosecretory Systems/physiology , Reproduction/physiology , Seasons , Animals , Brain/anatomy & histology , Cricetinae , Estradiol/physiology , Female , Luteinizing Hormone/metabolism , Male , Mammals , Models, Neurological , Neuronal Plasticity , Neurosecretory Systems/anatomy & histology , Sheep
SELECTION OF CITATIONS
SEARCH DETAIL
...