Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 114(9): 2413-2418, 2017 02 28.
Article in English | MEDLINE | ID: mdl-28196880

ABSTRACT

Mammalian reproductive function depends upon a neuroendocrine circuit that evokes the pulsatile release of gonadotropin hormones (luteinizing hormone and follicle-stimulating hormone) from the pituitary. This reproductive circuit is sensitive to metabolic perturbations. When challenged with starvation, insufficient energy reserves attenuate gonadotropin release, leading to infertility. The reproductive neuroendocrine circuit is well established, composed of two populations of kisspeptin-expressing neurons (located in the anteroventral periventricular hypothalamus, Kiss1AVPV, and arcuate hypothalamus, Kiss1ARH), which drive the pulsatile activity of gonadotropin-releasing hormone (GnRH) neurons. The reproductive axis is primarily regulated by gonadal steroid and circadian cues, but the starvation-sensitive input that inhibits this circuit during negative energy balance remains controversial. Agouti-related peptide (AgRP)-expressing neurons are activated during starvation and have been implicated in leptin-associated infertility. To test whether these neurons relay information to the reproductive circuit, we used AgRP-neuron ablation and optogenetics to explore connectivity in acute slice preparations. Stimulation of AgRP fibers revealed direct, inhibitory synaptic connections with Kiss1ARH and Kiss1AVPV neurons. In agreement with this finding, Kiss1ARH neurons received less presynaptic inhibition in the absence of AgRP neurons (neonatal toxin-induced ablation). To determine whether enhancing the activity of AgRP neurons is sufficient to attenuate fertility in vivo, we artificially activated them over a sustained period and monitored fertility. Chemogenetic activation with clozapine N-oxide resulted in delayed estrous cycles and decreased fertility. These findings are consistent with the idea that, during metabolic deficiency, AgRP signaling contributes to infertility by inhibiting Kiss1 neurons.


Subject(s)
Agouti-Related Protein/genetics , Fertility/genetics , Hypothalamus/metabolism , Kisspeptins/genetics , Neurons/metabolism , Starvation/genetics , Agouti-Related Protein/deficiency , Animals , Circadian Clocks/drug effects , Circadian Clocks/physiology , Clozapine/analogs & derivatives , Clozapine/pharmacology , Estrous Cycle/drug effects , Estrous Cycle/physiology , Female , Fertility/drug effects , Gene Expression Regulation , Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/drug effects , Kisspeptins/metabolism , Leptin/genetics , Leptin/metabolism , Luteinizing Hormone/genetics , Luteinizing Hormone/metabolism , Male , Mice , Mice, Transgenic , Neurons/cytology , Neurons/drug effects , Optogenetics , Reproduction/drug effects , Reproduction/genetics , Signal Transduction , Stereotaxic Techniques
2.
Cell Metab ; 19(4): 682-93, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24703699

ABSTRACT

Proopiomelanocortin (POMC) neurons within the hypothalamic arcuate nucleus are vital anorexigenic neurons. Although both the leptin and insulin receptors are coupled to the activation of phosphatidylinositide 3 kinase (PI3K) in POMC neurons, they are thought to have disparate actions on POMC excitability. Using whole-cell recording and selective pharmacological tools, we have found that, similar to leptin, purified insulin depolarized POMC and adjacent kisspeptin neurons via activation of TRPC5 channels, which are highly expressed in these neurons. In contrast, insulin hyperpolarized and inhibited NPY/AgRP neurons via activation of KATP channels. Moreover, Zn(2+), which is found in insulin formulations at nanomolar concentrations, inhibited POMC neurons via activation of KATP channels. Finally, as predicted, insulin given intracerebroventrically robustly inhibited food intake and activated c-fos expression in arcuate POMC neurons. Our results show that purified insulin excites POMC neurons in the arcuate nucleus, which we propose is a major mechanism by which insulin regulates energy homeostasis.


Subject(s)
Eating/drug effects , Insulin/pharmacology , Kisspeptins/metabolism , Neurons/metabolism , Pro-Opiomelanocortin/metabolism , Signal Transduction/physiology , Transient Receptor Potential Channels/metabolism , Agouti-Related Protein/metabolism , Animals , Guinea Pigs , KATP Channels/drug effects , Mice , Models, Biological , Neurons/drug effects , Neuropeptide Y/metabolism , Patch-Clamp Techniques , Proto-Oncogene Proteins c-fos/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Zinc/pharmacology
3.
J Neurosci ; 31(33): 11825-35, 2011 Aug 17.
Article in English | MEDLINE | ID: mdl-21849543

ABSTRACT

Multiple K(+) conductances are targets for many peripheral and central signals involved in the control of energy homeostasis. Potential K(+) channel targets are the KCNQ subunits that form the channels underlying the M-current, a subthreshold, non-inactivating K(+) current that is a common target for G-protein-coupled receptors. Whole-cell recordings were made from GFP (Renilla)-tagged neuropeptide Y (NPY) neurons from the arcuate nucleus of the hypothalamus using protocols to isolate and characterize the M-current in these orexigenic neurons. We recorded robust K(+) currents in the voltage range of the M-current, which were inhibited by the selective KCNQ channel blocker 10,10-bis(4-pyridinylmethyl)-9(10H)-anthracenone dihydrochloride (XE991) (40 µm), in both intact males and ovariectomized, 17ß-estradiol (E2)-treated females. Since NPY neurons are orexigenic and are active during fasting, the M-current was measured in fed and fasted male mice. Fasting attenuated the XE991-sensitive current by threefold, which correlated with decreased expression of the KCNQ2 and KCNQ3 subunits as measured with quantitative real-time PCR. Furthermore, E2 treatment augmented the XE991-sensitive M-current by threefold in ovariectomized (vs oil-treated) female mice. E2 treatment increased the expression of the KCNQ5 subunit in females but not KCNQ2 or KCNQ3 subunits. Fasting in females abrogated the effects of E2 on M-current activity, at least in part, by decreasing KCNQ2 and KCNQ3 expression. In summary, these data suggest that the M-current plays a pivotal role in the modulation of NPY neuronal excitability and may be an important cellular target for neurotransmitter and hormonal signals in the control of energy homeostasis in both males and females.


Subject(s)
Action Potentials/physiology , Estradiol/physiology , Fasting/physiology , KCNQ Potassium Channels/physiology , Neurons/physiology , Neuropeptide Y/physiology , Action Potentials/drug effects , Animals , Energy Metabolism/physiology , Estradiol/pharmacology , Female , Male , Mice , Mice, Transgenic , Neurons/drug effects , Receptors, G-Protein-Coupled/physiology
4.
J Parasitol ; 95(1): 191-7, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19245280

ABSTRACT

During the winter of 2004, 48 fecal samples were collected from live-trapped fox squirrels (Sciurus niger) from central Wyoming (Natrona County) and examined for species of Eimeria. Two species, Eimeria lancasterensis (prevalence, 65%) and Eimeria ontarioensis (prevalence, 27%), were identified. Genomic DNA sequences ITS1 and ITS2 were amplified, cloned, and sequenced. Additional sequences from E. lancasterensis isolated from a Delmarva fox squirrel (Sciurus niger cinereus) collected on Chincoteague Island, Virginia, were also identified. Comparison of pairwise distances suggests that E. lancasterensis from Wyoming and Virginia are conspecific. Maximum Parsimony tree construction identified 2 lineages, one E. ontarioensis and one E. lancasterensis; and both lineages had a strong bootstrap support (100%). The Maximum Parsimony analysis was unable to resolve the Wyoming and Virginia strains.


Subject(s)
Coccidiosis/veterinary , DNA, Protozoan/chemistry , Eimeria/classification , Rodent Diseases/parasitology , Sciuridae/parasitology , Animals , Coccidiosis/epidemiology , Coccidiosis/parasitology , DNA, Protozoan/isolation & purification , DNA, Ribosomal/chemistry , DNA, Ribosomal Spacer/genetics , Eimeria/genetics , Eimeria/isolation & purification , Feces/parasitology , Oocysts , Polymerase Chain Reaction/veterinary , RNA, Ribosomal, 5.8S/genetics , Rodent Diseases/epidemiology , Sequence Alignment/veterinary , Virginia/epidemiology , Wyoming/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL
...