Skip to main content
Endocrinology logoLink to Endocrinology
. 2012 Apr 16;153(6):2800–2812. doi: 10.1210/en.2012-1045

Arcuate Kisspeptin/Neurokinin B/Dynorphin (KNDy) Neurons Mediate the Estrogen Suppression of Gonadotropin Secretion and Body Weight

Melinda A Mittelman-Smith 1, Hemalini Williams 1, Sally J Krajewski-Hall 1, Josephine Lai 1, Philippe Ciofi 1, Nathaniel T McMullen 1, Naomi E Rance 1,
PMCID: PMC3359616  PMID: 22508514

Abstract

Estrogen withdrawal increases gonadotropin secretion and body weight, but the critical cell populations mediating these effects are not well understood. Recent studies have focused on a subpopulation of hypothalamic arcuate neurons that coexpress estrogen receptor α, neurokinin 3 receptor (NK3R), kisspeptin, neurokinin B, and dynorphin for the regulation of reproduction. To investigate the function of kisspeptin/neurokinin B/dynorphin (KNDy) neurons, a novel method was developed to ablate these cells using a selective NK3R agonist conjugated to the ribosome-inactivating toxin, saporin (NK3-SAP). Stereotaxic injections of NK3-SAP in the arcuate nucleus ablated KNDy neurons, as demonstrated by the near-complete loss of NK3R, NKB, and kisspeptin-immunoreactive (ir) neurons and depletion of the majority of arcuate dynorphin-ir neurons. Selectivity was demonstrated by the preservation of proopiomelanocortin, neuropeptide Y, and GnRH-ir elements in the arcuate nucleus and median eminence. In control rats, ovariectomy (OVX) markedly increased serum LH, FSH, and body weight, and these parameters were subsequently decreased by treatment with 17β-estradiol. KNDy neuron ablation prevented the rise in serum LH after OVX and attenuated the rise in serum FSH. KNDy neuron ablation did not completely block the suppressive effects of E2 on gonadotropin secretion, a finding consistent with redundant pathways for estrogen negative feedback. However, regardless of estrogen status, KNDy-ablated rats had lower levels of serum gonadotropins compared with controls. Surprisingly, KNDy neuron ablation prevented the dramatic effects of OVX and 17β-estradiol (E2) replacement on body weight and abdominal girth. These data provide evidence that arcuate KNDy neurons are essential for tonic gonadotropin secretion, the rise in LH after removal of E2, and the E2 modulation of body weight.


Estradiol suppression of gonadotropin secretion, known as estrogen negative feedback, is an essential component of the female reproductive cycle. Estrogens also act to suppress body weight by altering food intake and metabolism (1, 2). In the human, neurons in the infundibular (arcuate) nucleus that coexpress estrogen receptor (ER)α, neurokinin B (NKB), kisspeptin, and dynorphin have been proposed to mediate estrogen negative feedback on gonadotropin secretion (37). A homologous group of kisspeptin/NKB/dynorphin (KNDy) neurons resides in the arcuate nucleus of a variety of other species, including mice, rats, sheep, goats, and monkeys (815). Mutations in the genes encoding kisspeptin, NKB, or their receptors are associated with low serum gonadotropins, infertility, and absence of pubertal maturation in both men and women (1620). Transgenic mouse models also illustrate the importance of kisspeptin and NKB signaling in reproductive regulation (16, 21). Because kisspeptin and NKB peptides are located in numerous brain regions, it is not known whether KNDy neurons represent the essential cell type for reproduction.

To investigate the role of KNDy neurons, we developed a method to selectively ablate these neurons based on their expression of the neurokinin 3 receptor (NK3R) (10, 12, 22). Within the arcuate nucleus, the expression of NK3R protein or mRNA is restricted to a small subpopulation of neurons (10, 23). Arcuate neuroendocrine neurons (24), proopiomelanocortin (POMC) neurons, or tyrosine hydroxylase-immunoreactive (ir) neurons do not express NK3R (Krajewski-Hall, S. J., and N. E. Rance, unpublished observations). Moreover, NK3R mRNA is exclusively expressed in kisspeptin neurons in the mouse arcuate nucleus (12). Based on these data, we reasoned that destruction of NK3R-expressing cells in the arcuate nucleus should selectively ablate KNDy neurons.

Saporin (SAP) is a ribosome-inactivating toxin that can be conjugated to selective receptor ligands to produce targeted cell ablation (25). Because NK3R internalizes after ligand binding, conjugation of SAP to a selective NK3R agonist (NK3-SAP) provides a selective toxin for NK3R-expressing neurons. In the present study, we characterized the morphological and physiological effects of injecting NK3-SAP into the rat arcuate nucleus to ablate KNDy neurons. Our primary goal was to determine the effects of KNDy neuron ablation on the changes in gonadotropin secretion in response to ovariectomy (OVX) and estradiol replacement. We were surprised to find that the 17β-estradiol (E2) modulation of body weight was also strikingly altered in KNDy-ablated animals.

Materials and Methods

Young adult, female Sprague Dawley rats (∼12 wk old, 200–250 g; Harlan Laboratories, Indianapolis, IN) were individually housed in a quiet, temperature-controlled room (21.1–22.5 C) in the University of Arizona Animal Care Facility with a 12-h light, 12-h dark cycle (lights on at 0700 h). Rats had ad libitum access to water and a low-phytoestrogen diet (Harlan Teklad 2014; Harlan Laboratories). All protocols were approved by the University of Arizona Animal Care and Use Committee and conformed to National Institutes of Health guidelines.

Validation of NK3-SAP for selective ablation of NK3R-expressing neurons in rat brain

[MePhe7]NKB is a modified NKB peptide that has been established to be a potent and selective agonist for NK3R (26, 27). A custom conjugate of SAP and [MePhe7]NKB (NK3-SAP) was obtained from Advanced Targeting Systems (San Diego, CA). NK3-SAP stock [40 ng/100 nl in 0.1 m PBS (pH 7.4)] was stored at −80 C until use. Blank-SAP, an 11-amino acid peptide conjugated to SAP (Advanced Targeting Systems), was injected as a control. The amino acid sequence of the peptide in Blank-SAP (scrambled αMSH) has no significant homology to known G protein-coupled receptor ligands.

To determine whether NK3-SAP retains high affinity for NK3R, competitive binding assays of [MePhe7]NKB (Calbiochem, Rokland, MA) or NK3-SAP against the selective NK3R radioligand, [3H]senktide (PerkinElmer, Waltham, MA), were conducted using freshly prepared crude membranes from rat brain neocortex. Competitive binding was carried out in a total assay volume of 0.5 ml using 10 nm [3H]senktide (50.6 Ci/mmol) and various concentrations, in duplicates, of [MePhe7]NKB or NK3-SAP for 2 h at 22 C in a shaking water bath. Total binding of [3H]senktide was defined by the radioactivity bound in the absence of competing ligand, and nonspecific binding of [3H]senktide was defined as the radioactivity bound in the presence of 10 μm [MePhe7]NKB. Incubation was terminated by rapid filtration of the samples through polyethyleneimine-presoaked GF/B filters. Radioactivity on the filters was determined by liquid scintillation counting. Data were analyzed by nonlinear least squares analysis using GraphPad Prism software (Prism Software Corp., Irvine, CA). Data were based on two independent experiments.

Best-fit analysis of the data based on a two-site competition analysis (GraphPad Prism) showed that [MePhe7]NKB had a high-affinity IC50 value of 11 nm (Log IC50 = −7.96 ± 0.75, n = 2), and the NK3-SAP had an IC50 value of 6.4 nm (Log IC50 = −8.20 ± 1.5, n = 2) in a parallel assay. Thus, both [MePhe7]NKB and NK3-SAP exhibited comparable, high-affinity binding for the NK3R receptor. These data show that conjugation of [MePhe7]NKB to SAP did not significantly alter its affinity for NK3R. This high-affinity interaction of NK3-SAP with NK3R appeared well suited for selective ablation of NK3R-expressing cells via ligand-mediated uptake.

Pilot experiments were conducted to determine the optimum dose of NK3-SAP for selective ablation of NK3R neurons. The medial septum was studied because NK3R in this area is restricted to neurons that exhibit a distinctive magnocellular appearance (24, 28). Under general anesthesia, NK3-SAP (2, 10, or 40 ng diluted in 100 nl of PBS) was injected unilaterally into the medial septum (0.0 mm posterior to Bregma, 0.2 mm lateral, and 7.5 mm below the skull surface; n = 2–4 rats/group). The rats were killed 1 wk later, and the brains were processed as described below. Inspection of Nissl-stained sections revealed that the 10-ng NK3-SAP injections produced unilateral reduction of the magnocellular neurons in the medial septal nucleus, with preservation of small neurons, and no other discernible damage to brain parenchyma other than the small injection tract. Immunohistochemistry confirmed that unilateral injection of 10 ng produced unilateral loss of magnocellular NK3R neurons in the medial septum. The 2-ng injections produced barely perceptible NK3R cell loss, and there was unacceptable nonspecific damage with the highest dose of 40 ng. Therefore, 10 ng of NK3-SAP (in 100 nl of PBS) was used for all further studies.

Experiment 1. Effects of arcuate NK3-SAP injections on the E2 modulation of gonadotropin secretion and body weight

Experimental day 0, OVX, and injection of NK3-SAP or Blank-SAP in the arcuate nucleus

Twenty-four rats were anesthetized with a cocktail (1.0 ml/kg im) containing ketamine (33.3 mg/ml), xylazine (10.7 mg/ml), and acepromazine (1.3 mg/ml) and ovariectomized using a dorsal surgical approach. A DSI PhysioTel transmitter (TA10-F40; Data Sciences International, St. Paul, MN) was inserted into the peritoneal cavity for measurements of core temperature. Stereotaxic surgery was used to make four injections of 10 ng/100 nl PBS of either NK3-SAP (n = 14 rats) or Blank-SAP (n = 10 rats). Injections were made bilaterally (two sites per side) to target the dorsolateral border of the arcuate nucleus (rostral coordinates: 2.4 mm posterior to bregma, ±0.5 mm lateral, and 9.8 mm ventral to skull surface; caudal coordinates: 3.5 mm posterior to bregma, ±0.5 mm lateral, and 9.7 mm ventral to skull surface). Microinjections were made using a NanoFil 10-μl syringe with a 33-gauge beveled tip needle (World Precision Instruments, Sarasota, FL) inserted into an UltraMicroPump with a Micro 4 controller (UMP3; World Precision Instruments). Each injection was made over a period of 5 min, and the needle was left in place for an additional 5 min before removal. The incision was closed, and animals were given postoperative analgesia (0.03–0.05 mg/kg sc of buprenorphine).

Estradiol replacement

Twenty to 23 d after the initial surgery, under isofluorane anesthesia, rats were implanted with two sc capsules (each 20 mm effective length, 1.57 mm inner diameter, and 3.18 mm outer diameter; Dow Corning, Midland, MI) containing 17β-estradiol dissolved in sesame oil (E2, 360 μg/ml).

Blood collection and body weight measurements

Blood samples and body weight measurements were taken at three time points: immediately before OVX (d 0, intact), 20–23 d after OVX, and 11 d after E2 treatment (OVX + E2). Girth (circumference at the widest point of the torso) was measured in the OVX and OVX + E2 conditions while animals were anesthetized and in the prone position. Blood samples were collected between 0700 and 1100 h. The first two blood samples (∼50 μl each) were collected from a saphenous vein puncture using a 5-mm lancet (Braintree Scientific, Braintree, MA) and a Microvette capillary tube (Sarstedt, Newton, NC) while the rats were anesthetized for the initial surgery or the capsule implantation. The final sample was taken at the time of killing via cardiac puncture. Blood from all samples was left to clot for 90 min and centrifuged. Serum was collected and stored at −20 C until assay.

Brain tissue collection for histology

After an overdose injection of sodium pentobarbital (100 mg/kg ip), animals were perfused through the ascending aorta with 200 ml of heparinized, 0.1 m PBS followed by 400 ml of 4% paraformaldehyde in phosphate buffer (pH 7.4). Brains were extracted, postfixed in the same fixative for 1 h, and cryoprotected in ascending solutions of sucrose (10, 20, and 30%) in PBS. Brains were blocked using a rat brain matrix (Braintree Scientific). Blocks containing the hypothalamus were frozen and sectioned (40-μm thickness) in the coronal plane with a sliding microtome. Every tenth section was Nissl stained, and the remaining tissue was stored in a cryoprotectant solution at −20 C until use (29).

Hormone assays

Serum samples were assayed by the Ligand Assay and Analysis Core Laboratory at the University of Virginia Center for Research in Reproduction. (Charlottesville, VA). Serum LH and FSH (10-μl sample, in singlet) were assayed using a multiplex panel assay (Milliplex MAP for Luminex xMAP Technology, RPT-86K-02). The LH and FSH assays had an intraassay variability of 6.9 and 6.7% and a sensitivity of 0.24 and 2.4 ng/ml, respectively. LH values that fell below the sensitivity of the assay (rats with arcuate NK3-SAP injections, three OVX and five OVX + E2 values) were assigned the minimal detectable value of 0.24 ng/ml.

Terminal serum samples (n = 12) were also assayed for estradiol (60 μl each, in duplicate, DSL Ultrasensitive E2 rat-RIA). The estradiol RIA had an intraassay variability of 6.7%. The serum E2 in these terminal samples (11 d after capsule implantation) was at physiological levels of 36.6 ± 14.9 pg/ml (30). There was no difference in serum E2 between Blank-SAP and NK3-SAP injected rats.

Immunohistochemical studies

Antibodies

For detailed information, see Table 1.

Table 1.

Antibodies used in immunohistochemical procedures

Primary antibody (host, dilution) Immunogen Source, lot Secondary antibody
NK3R (polyclonal rabbit, 1:20,000) Residues 443–452 of rat NK3R Phillipe Ciofi IS-7/7, bleed 040595 Biotinylated goat antirabbit (Vector Laboratories, CABA-1000, lot W0117)
pro-NKB (polyclonal rabbit, 1:15,000) Residues 50–79 of mouse pro-NKB Novus Biologicals NB300-201, lot A2 Biotinylated goat antirabbit
Kisspeptin (polyclonal rabbit, 1:20,000) Residues 43–52 of mouse kisspeptin Millipore (Temecula, CA) AB9754, lot NMM1669485 Biotinylated goat antirabbit
Prodynorphin (polyclonal guinea pig, 1:1000) Residues 235–248 of rat prodynorphin Neuromics GP10110, lot Q10016 Goat antiguinea pig + Alexa Fluor 488 (Invitrogen A11073, lot 752761)
POMC (polyclonal rabbit, 1:50,000) Residues 27–52 of porcine POMC precursor Phoenix Pharmaceuticals (Burlingame, CA) H-029-30, lot 01181-3 Biotinylated goat antirabbit
NPY (polyclonal rabbit,1:100,000) 36 amino acid porcine NPY peptide Peninsula Laboratories (Torrance, CA) IHC-7172, lot 991025 Goat antirabbit + Alexa Fluor 568 (Invitrogen A11036, lot 84C1-1)
GnRH/LHRH (monocloncal mouse, 1:5000) LHRH decapeptide Henryk Urbanski clone HU4U Highly cross-adsorbed goat antimouse + Alexa Fluor 488 (Invitrogen A11029, lot 727756)

Neurokinin 3 receptor

The NK3R antiserum showed staining comparable with previous reports of the distribution of NK3R mRNA (23) and protein (24, 31). Labeling was abolished by preadsorption with a synthetic peptide corresponding to amino acids 443–452 of the C-terminal region of rat NK3R protein (Bio-Synthesis, Lewisville, TX).

Neurokinin B

The distribution of pro-NKB immunoreactivity was similar to previous reports with this (10, 24) and other pro-NKB antibodies (3234). Preadsorption with the synthetic peptide used for immunization (Novus Biologicals, Littleton, CO) prevented immunolabeling.

Kisspeptin

The pattern of staining using this antibody was similar to previous reports of kisspeptin immunoreactivity (35, 36). The characterization of this antibody has been previously described (35).

Dynorphin

Labeling with the prodynorphin antibody matched the distribution of dynorphin immunoreactivity using this (10) and other (37) prodynorphin antibodies. Staining was not observed in dynorphin knockout mice (38) or after preadsorption with the immunizing antigen (Neuromics, Edina, MN).

Proopiomelanocortin

The POMC antiserum revealed the expected distribution of labeling based on previous immunocytochemical (39) and in situ hybridization studies (8). Using this antibody, colocalization of POMC and green fluorescent protein was observed in enhanced green fluorescent protein-POMC mice (40).

Neuropeptide Y (NPY)

Within the arcuate nucleus, the NPY antiserum labeled predominantly fibers. The labeling was in agreement with previous descriptions of NPY immunoreactivity in the arcuate nucleus and median eminence (41). This antibody has been previously characterized, including preadsorption experiments (42).

Gonadotropin-releasing hormone

This extensively characterized antibody (43) labeled cells with the classic appearance of GnRH neurons (24). Labeling was prevented by preadsorption with the GnRH peptide (43).

NK3R immunohistochemistry was performed on every tenth section from the level of the paraventricular nucleus to the mammillary bodies. For all other antibodies, sections were matched to middle (plate 56) and posterior (plate 64) levels of the arcuate nucleus using a rat brain atlas (44). NK3R immunohistochemistry used an antigen retrieval step consisting of incubation in 15 mm sodium citrate for 30 min at 80 C and then cooling to room temperature (45). Kisspeptin, pro-NKB, and POMC immunohistochemistry used the same protocol but without the antigen retrieval step. Sections were rinsed in PBS, incubated in 0.3% H2O2 for the removal of endogenous peroxidases, rinsed, and blocked in 3% normal goat serum (NGS) + 0.4% Triton X-100 in PBS. Sections were incubated with the antibody in blocking solution at 4 C for 48 h (see Table 1 for antibody dilutions). Sections were then rinsed and incubated in blocking solution containing a biotinylated goat antirabbit antibody (1:600), followed by rinsing and incubation in an Avidin-Biotin Complex solution (Vector Laboratories, Burlingame, CA). After rinsing with PBS, then 0.175 m sodium acetate, filtered nickel-intensified 3,3′ diaminobenzidine (DAB) solution was used for visualization (1.25 g of nickel ammonium sulfate, 10 mg of DAB in 50 ml of sodium acetate solution, and 41.5 μl of 30% H2O2). Sections were mounted on subbed slides, dehydrated, and coverslipped.

Prodynorphin, NPY, and GnRH were visualized using fluorescent immunohistochemistry. Sections were rinsed in PBS, incubated in blocking solution for 1 h, and then incubated with a primary antibody diluted in blocking solution for 48 h at 4 C (see Table 1 for antibody dilutions). After PBS rinses, sections were incubated in the appropriate secondary antibody (1:500) for 4 h (Table 1). After final rinses in PBS, sections were mounted, allowed to dry, and coverslipped using ProLong Gold Antifade (Invitrogen, Carlsbad, CA).

Cell counts and fiber density measurements

Morphologic analysis was performed by an investigator blind to the treatment condition. For the nickel-DAB-labeled sections (NK3R, NKB, kisspeptin, and POMC), an image-combining computer microscope (Nikon, Tokyo, Japan) equipped with a Ludl motorized stage (Ludl Electronic Products, Hawthorne, NY), a Lucivid miniature CRT, and Neurolucida software (Microbrightfield, Williston, VT) was used to count cells. Outlines of the sections and regions of interest were drawn bilaterally using a ×4 Plan objective. Nissl stains were used to facilitate identification of landmarks. Cells were then mapped and systematically counted using a ×20 Plan apochromat objective (numerical aperture 0.50).

Immunofluorescence images (prodynorphin, NPY, and GnRH) were captured using a Nikon E1000 microscope equipped with a VFM epifluorescent attachment, a LUDL motorized stage, a Uniblitz model VMM-D1 shutter driver (Vincent Associates, Rochester, NY) and a Photometrics Coolsnap FX camera (Roper Scientific, Trenton, NJ). Digital images of the sections were taken in systematic step-wise fashion using a ×20 Nikon Plan Fluor objective (N.A. 0.50) and the appropriate filter set: Alexa Fluor 488 (excitation, 480/30 nm; diachromatic mirror; 505 nm; and emission filter 535/40 nm) or Alexa Fluor 568 (excitation, 560/40; DM, 595; and emission filter 630/60). Images were assembled into montages using MetaMorph imaging software (Molecular Devices, Sunnyvale, CA). Prodynorphin neurons were counted within the arcuate nucleus using a ×40 Nikon Plan Fluor objective (numerical aperture 0.75). Because NPY immunoreactivity in arcuate cell bodies is not visible without previous colchicine injection, we evaluated the status of NPY neurons by measuring the density of their local projections. NPY fiber densities were evaluated by drawing a 250-μm2 square within the arcuate nucleus. GnRH fiber densities were evaluated within a boundary drawn to include the bilateral arcuate nucleus and adjacent median eminence. The total area analyzed did not differ between groups. Thresholds were established so that the labeled fibers in focus were above threshold. The fiber density value consisted of the area (in pixels) covered by labeled fibers divided by total area (in pixels) within the boundaries.

Experiment 2. Effects of NK3-SAP injections in the lateral hypothalamus on gonadotropin secretion and body weight

Injections of NK3-SAP into the arcuate nucleus resulted in variable loss of large NK3R neurons in the lateral hypothalamus and zona incerta. Because loss of these extraarcuate NK3R neurons complicated the interpretation of the first experiment, a second experiment was designed to replicate the pattern of NK3R neuron loss in the lateral hypothalamus and zona incerta, while preserving KNDy neurons in the arcuate nucleus. NK3-SAP (10 ng/100 nl PBS) was injected bilaterally in the lateral hypothalamus (n = 4 rats; coordinates: 3 mm posterior to bregma, ±1.8 mm lateral to midline, and 7.0 mm ventral to skull surface). Control rats (n = 4) received bilateral injections of Blank-SAP (10 ng/100 nl PBS). The protocol for this experiment and the immunohistochemical procedures were identical to that described above.

To evaluate the amount of cell degeneration after lateral hypothalamic injections, tissue was evaluated using Nissl histology and NK3R immunohistochemistry. NK3R neurons were counted and mapped throughout the lateral hypothalamus and zona incerta using the image combining computer microscope described above. Counts were made at three levels corresponding to plates 56, 60, and 64 of a rat brain atlas (44). NKB and NK3R immunohistochemistry was used to verify preservation of KNDy neurons in the arcuate nucleus.

Data analysis

Three rats with preservation of KNDy neurons after arcuate NK3-SAP injections were considered to have missed injections and excluded from the study. The numbers of NK3R, pro-NKB, kisspeptin, prodynorphin, and POMC neurons per unilateral arcuate section were calculated for each animal to determine group means. Fiber staining density (NPY and GnRH) was also averaged for each animal, and these values were used to obtain group means. To determine the percent loss of NK3R neurons in the lateral hypothalamus/zona incerta, the total number of neurons counted in the three sections was subtracted from the average number counted in Blank-SAP controls, then divided by the average number counted in Blank-SAP controls. Groups were compared using a t test. Serum gonadotropins, body weight, and girth measurements were compared using a two-way ANOVA followed by Tukey's post hoc analysis (α = 0.05).

Results

Arcuate NK3-SAP injections ablate NK3R-expressing KNDy neurons, with preservation of POMC, NPY, and GnRH neuronal elements

The morphological appearance of Nissl-stained sections at the level of the arcuate nucleus was similar between NK3-SAP and Blank-SAP rats (Fig. 1A). Other than the small needle tracts, destruction of brain parenchyma was not observed. These data are consistent with the small percentage of arcuate neurons expressing either the NK3R protein (10) or its mRNA (23). In contrast, immunohistochemistry revealed near-complete loss of NK3R, NKB, and kisspeptin cell bodies in the arcuate nucleus of NK3-SAP rats (Fig. 1, B–D). This change was accompanied by the disappearance of virtually all NKB and kisspeptin fibers in the arcuate nucleus and adjacent median eminence, as well as fibers extending along the third ventricle in the periventricular pathway (46). Quantitative analysis confirmed loss of KNDy neurons: the number of NK3R, NKB, and kisspeptin-ir neurons in anterior and posterior levels of the arcuate nucleus was reduced by 93–98% in NK3-SAP rats, compared with Blank-SAP controls (Table 2). In addition, the number of prodynorphin cell bodies in the arcuate nucleus was reduced by 77% in NK3-SAP rats (Fig. 1E and Table 2). Preservation of a small number of arcuate dynorphin neurons is consistent with previous studies showing that there is a subgroup of dynorphin neurons in the rat arcuate nucleus that do not express NK3R (10) and are not KNDy neurons (47). Dynorphin neurons were also preserved in the adjacent ventromedial nucleus.

Fig. 1.

Fig. 1.

Representative photomicrographs of sections from rats with injections of Blank-SAP (left) or NK3-SAP (right) in the arcuate nucleus, stained with Cresyl-violet (A) or immunohistochemistry (B–H). A, Cresyl violet-stained sections show preservation of the Nissl architecture in NK3-SAP rats. B–D, NK3-SAP rats exhibited near total depletion of NK3R, NKB, and kisspeptin-ir neurons and fibers in the arcuate nucleus and median eminence. E, The majority of dynorphin-ir neurons and fibers in the arcuate nucleus and median eminence were ablated in NK3-SAP rats. F and G, There was no difference in the number of POMC neurons or NPY fiber density between Blank-SAP and NK3-SAP rats. H, GnRH fibers in the median eminence were preserved in NK3-SAP rats. 3V, Third ventricle; ARC, arcuate nucleus; ME, median eminence; pro-DYN, prodynorphin; VMN, ventromedial nucleus. Scale bar, 100 μm.

Table 2.

Quantitative analysis of immunoreactive cell bodies or fiber densities in rats injected with either Blank-SAP or NK3-SAP in the arcuate nucleus

Immunohistochemical phenotype Mid-level arcuate
Posterior arcuate
Total percent loss
Blank-SAP NK3-SAP Blank-SAP NK3-SAP
NK3R 7.9 ± 0.7 0.3 ± 0.2a 13.4 ± 1.9 0.2 ± 0.1a 98.1 ± 0.8
NKB 10.4 ± 1.1 0.6 ± 0.5a 50.8 ± 3.6 3.1 ± 1.1a 92.6 ± 2.2
Kisspeptin 6.4 ± 1.0 0.1 ± 0.1a 31.4 ± 3.6 1.4 ± 0.6a 94.8 ± 1.6
Dynorphin 11.0 ± 1.5 3.6 ± 0.5a 24.0 ± 5.7 4.1 ± 1.0a 76.5 ± 2.9
POMC 33.7 ± 3.8 37.2 ± 3.2 29.7 ± 2.1 36.0 ± 3.0 0
NPY 0.07 ± 0.01 0.06 ± 0.02 0.03 ± 0.003 0.03 ± 0.004 0
GnRH 0.04 ± 0.004 0.04 ± 0.008 NA NA 0

Values represent mean ± sem NK3R, NKB, kisspeptin, dynorphin, and POMC values represent the average number of labeled neurons/unilateral arcuate section. NPY and GnRH values reflect average fiber density values (see Materials and Methods). NA, Not analyzed.

a

P < 0.01, Significantly different, Blank-SAP vs. NK3-SAP-treated animals, n = 5–11 rats/group.

The selectivity of NK3-SAP for ablation of NK3R-expressing KNDy neurons was demonstrated by preservation of POMC, NPY, and GnRH-ir elements. NK3-SAP had no qualitative or quantitative effect on the number of POMC neurons or NPY fibers within the arcuate nucleus (Fig. 1, F and G, and Table 2). Despite a previous demonstration of punctate NK3R-staining on GnRH fibers (24), there were no qualitative differences in the distribution of GnRH fibers in the median eminence and no change in GnRH fiber density between NK3-SAP and Blank-SAP rats (Fig. 1H and Table 2). These data agree with reports that SAP-peptide conjugates do not produce retrograde neuronal degeneration (48).

In Blank-SAP rats, large NK3R neurons with intense dendritic labeling were identified in lateral hypothalamus (including the perifornical region) and zona incerta (see Ref. 24 for photomicrographs). No NK3R neurons were identified in the ventral premamillary nucleus or the ventromedial nucleus, in agreement with previous reports (23, 24, 31, 49). In NK3-SAP rats, there was variable and incomplete loss of the large NK3R neurons in the lateral hypothalamus and zona incerta. NK3R neurons in the supraoptic and paraventricular nuclei were intact, indicative of the limited diffusion of the toxin.

Arcuate KNDy neuron ablation prevents the rise in LH secretion after OVX and lowers serum gonadotropins

Blank-SAP rats exhibited a 13-fold increase in serum LH after OVX that returned to intact levels 11 d after E2 treatment (Fig. 2A). Serum LH was not significantly elevated above intact levels in OVX rats that received injections of NK3-SAP in the arcuate nucleus (Fig. 2A). E2 treatment of OVX NK3-SAP rats resulted in a small but significant decrease of serum LH. In both the OVX and OVX + E2 conditions, serum LH was significantly lower in NK3-SAP rats compared with Blank-SAP controls.

Fig. 2.

Fig. 2.

Effects of KNDy neuron ablation on serum LH (A) and FSH (B). Rats were ovariectomized and injected with NK3-SAP or Blank-SAP in the arcuate nucleus on d 0, given E2 capsules 20–23 d later (shown as d 22), and killed after 11 d of E2. Serum LH and FSH were markedly increased by OVX and reduced by E2 treatment in Blank-SAP animals. In contrast, serum LH after OVX was not significantly different from intact values in NK3-SAP rats, and the rise in FSH was attenuated (mean ± sem, n = 6–11 rats/group for both A and B). Serum LH and FSH were significantly lower in KNDy-ablated rats than Blank-SAP controls in both OVX and OVX + E2-treated groups. #, Significantly different, NK3-SAP vs. Blank-SAP, P ≤ 0.05; +, significantly different from intact values on d 0, P ≤ 0.01; *, significantly different after E2 (vs. OVX), P ≤ 0.05.

In Blank-SAP control rats, serum FSH was increased after OVX and decreased by E2 treatment (Fig. 2B). In arcuate NK3-SAP rats, serum FSH was also increased by OVX and reduced by E2 treatment, but FSH levels were significantly lower after NK3-SAP injections, compared with the control animals (Fig. 2B).

Arcuate KNDy neuron ablation prevents the E2 modulation of body weight and girth observed in Blank-SAP controls

Three weeks after OVX, Blank-SAP rats were 25% heavier relative to their initial body weight (Fig. 3A), and E2 treatment of these rats induced significant weight loss. In contrast, the body weight of arcuate NK3-SAP rats was only 5% higher after OVX, and these values were not significantly different compared with those obtained on experimental d 0. In addition, E2 treatment did not reduce the body weight of OVX NK3-SAP rats. Overall, arcuate NK3-SAP rats gained weight throughout the experiment in amounts expected from the standard growth charts of intact female Sprague Dawley rats (Growth Curve, Harlan Sprague Dawley).

Fig. 3.

Fig. 3.

Effects of KNDy neuron ablation on body weight and abdominal girth. Rats were ovariectomized and injected with NK3-SAP or Blank-SAP in the arcuate nucleus on d 0, given E2 capsules 20–23 d later (shown as d 22), and killed after 11 d of E2. A, After OVX, Blank-SAP animals were 25% heavier relative to their initial body weight and lost weight after E2 treatment. Rats with arcuate NK3-SAP injections gained small amounts of weight throughout the study, regardless of E2 status (mean ± sem, n = 9–11 rats/group). B, After OVX, the girth of NK3-SAP rats was significantly smaller than Blank-SAP rats. Girth was significantly decreased by E2 in Blank-SAP but not NK3-SAP rats (mean ± sem, n = 3–4 rats/group). #, Significantly different, Blank-SAP vs. NK3-SAP, P < 0.01; +, significantly different from intact values on d 0, P < 0.05; *, significantly different after E2 (vs. OVX), P < 0.01.

Three weeks after OVX, the girth of Blank-SAP rats was significantly larger than in NK3-SAP rats. E2 treatment significantly decreased girth in Blank-SAP animals but had no effect on girth in NK3-SAP rats (Fig. 3B).

Loss of NK3R neurons in the lateral hypothalamus and zona incerta does not alter the E2 modulation of gonadotropin secretion or body weight

NK3-SAP injections in the lateral hypothalamus produced incomplete NK3R neuron degeneration in lateral hypothalamus (including the perifornical area) and zona incerta (Fig. 4). The amount of neuronal loss was similar to that seen in experiment 1, but KNDy neurons were spared. Cell counts revealed that lateral hypothalamic NK3-SAP injections depleted 80.6 ± 2.5% of the large NK3R neurons in the lateral hypothalamus and zona incerta, compared with 71.3 ± 2.9% in rats with arcuate NK3-SAP injections.

Fig. 4.

Fig. 4.

Representative maps of NK3R neurons from a Blank-SAP-injected (A), arcuate NK3-SAP-injected (B) or lateral hypothalamic NK3-SAP-injected (C) rat. A, Map from Blank-SAP rat showing the location of NK3R neurons in the arcuate nucleus, lateral hypothalamus, supraoptic nucleus, and zona incerta. B, Map from a rat with an arcuate NK3-SAP injection with KNDy neuron ablation and incomplete loss of NK3R neurons in the lateral hypothalamus and zona incerta. C, Map from rat with a lateral hypothalamic NK3-SAP injection showing NK3R neurons in the arcuate nucleus and incomplete loss of NK3R cell bodies in the lateral hypothalamus and zona incerta. Each dot represents one labeled neuron. 3V, Third ventricle; ARC, arcuate nucleus; LHA, lateral hypothalamic area; ot, optic tract; SON, supraoptic nucleus; VMN, ventromedial nucleus; ZI, zona incerta. Scale bar, 250 μm.

Despite the extensive loss of the large NK3R cells, lateral hypothalamic NK3-SAP injections did not alter the E2 modulation of gonadotropin secretion or body weight (Fig. 5). In both groups, serum LH and FSH levels were increased after OVX and decreased after E2 treatment (Fig. 5, A and B). Moreover, rats with NK3-SAP injections in the lateral hypothalamus gained a large amount of weight after OVX, and E2 treatment resulted in significant weight loss (Fig. 5C).

Fig. 5.

Fig. 5.

Effects of NK3-SAP injections in the lateral hypothalamus on serum LH (A), FSH (B), and body weight (C). Rats were ovariectomized and injected with NK3-SAP or Blank-SAP on d 0, given E2 capsules 20–23 d later (shown as d 22), and killed after 11 d of E2. A–C, There were no significant differences between rats with lateral hypothalamic NK3-SAP and Blank-SAP injections (mean ± sem, n = 4 values/group, except n = 2 for Blank-SAP gonadotropin levels). +, Significantly different from intact values on d 0, P < 0.05; *, significantly different after E2 (vs. OVX), P < 0.05.

Discussion

To evaluate the role of NK3R-expressing KNDy neurons in the E2 modulation of gonadotropin secretion and body weight, we ablated these neurons using NK3-SAP, a novel conjugate of a highly selective NK3R agonist and SAP. Immunohistochemical studies documented the selectivity of NK3-SAP injections for KNDy neurons in arcuate nucleus. In contrast to the marked rise in serum LH in Blank-SAP controls, serum LH was not significantly increased after OVX in KNDy-ablated rats. Moreover, LH and FSH were significantly lower in KNDy-ablated rats than controls, regardless of E2 status. Surprisingly, KNDy neuron ablation blocked the pronounced effects of OVX and E2 replacement on body weight and abdominal girth. Our data indicate an essential role for KNDy neurons in the regulation of gonadotropin secretion and the E2 modulation of body weight.

The classic reciprocal relationship between the ovaries and anterior pituitary gland hormone secretion was demonstrated in Blank-SAP controls (50). Three weeks after OVX, serum LH and FSH were markedly increased in control rats. Replacement of physiological levels of E2 suppressed LH to intact levels, consistent with numerous studies showing that that the rise in LH after OVX is largely due to removal of E2 (51). Serum FSH was also decreased by E2 replacement in control rats, although not to intact levels. These data reflect the dual ovarian control of FSH secretion, which includes protein hormones such as inhibin, as well as E2 feedback (51).

Remarkably, in rats with KNDy neuron ablation, serum LH was not significantly increased 3 wk after OVX. These data indicate that the suppressive effects of E2 on serum LH in OVX rats are predominantly mediated through KNDy neurons. E2 treatment of KNDy-ablated rats still produced a small decrease in LH. Although these findings are consistent with redundant sites of E2 negative feedback (52, 53), we cannot exclude E2 effects on the few remaining KNDy neurons. Serum FSH was significantly modulated by OVX and E2 replacement in KNDy-ablated rats but lower than Blank-SAP controls, regardless of E2 status. Similarly, serum LH was lower in KNDy-ablated rats than in Blank-SAP rats in both the OVX and OVX + E2 conditions. Thus, KNDy neurons play a role in stimulating the high levels of serum gonadotropins after withdrawal of E2 as well as tonic stimulation of gonadotropin secretion in OVX E2-treated rats.

Previous studies show that OVX markedly increases NKB and kisspeptin gene expression in the rodent arcuate nucleus, and these changes are reversed by E2 replacement (8, 54). KNDy neurons express ERα (10, 54), the isoform required for E2 suppression of GnRH mRNA, serum LH (55), and NKB and kisspeptin gene expression (56, 57). KNDy neurons do not directly modulate the anterior pituitary gland, because they do not project to fenestrated capillaries in the median eminence (24, 58), but they could influence LH secretion by projections to GnRH terminals in the median eminence (24, 46, 5962). Pharmacological studies show that kisspeptin stimulates GnRH secretion (62, 63) and activates GnRH neurons in tissue slice preparations (64). Moreover, similar to the effects of KNDy neuron ablation, kisspeptin receptor knockout mice do not exhibit a rise in serum LH after OVX (65). Pharmacological studies have also shown NKB and dynorphin receptor activation to alter levels of serum gonadotropins, in addition to changing the frequency of LH pulses (13, 15, 6668). Although the relative contribution of different hypothalamic cell groups in the regulation of GnRH secretion has been controversial, our data clearly underscore the importance of KNDy neurons in this circuitry.

In many mammalian species, ovarian hormone withdrawal causes obesity that can be reversed by E2 treatment (1, 69, 70). E2 decreases food intake and increases energy expenditure in OVX animals via an ERα-dependent mechanism (2, 71, 72). In our study, control rats gained a large amount of weight after OVX and lost weight after E2 replacement. Based on abdominal girth measurements, the suppressive effect of E2 on body weight was due (at least in part) to loss of visceral adiposity.

In contrast to the controls, KNDy-ablated rats did not gain a significant amount of weight 3 wk after OVX, and E2 treatment did not cause weight loss or changes in abdominal girth. The KNDy-lesioned rats were not compromised in their ability to maintain body weight. Rather, they gained weight throughout the experiment at rates nearly identical to OVX + E2-replaced rats under similar experimental conditions (73) and intact female rats (1). In addition, we have recently observed no effect of NK3-SAP (vs. Blank-SAP) injections on the body weight of ovary-intact female rats within 4 wk after injection (Mittelman-Smith, M. A., and N. E. Rance, unpublished observations). Overall, the body weight regulation of KNDy-ablated rats was characterized by insensitivity to the effects of OVX and E2 replacement.

Our data provide evidence that increased KNDy neuron signaling promotes visceral fat storage in parallel with the increase in serum LH after the removal of E2. Further studies are necessary to determine whether the effects of KNDy neuron ablation are secondary to changes in food intake and/or energy expenditure. KNDy neurons have been proposed to relay metabolic signals to influence reproductive function (7477), but there are no previous reports of a role for KNDy neurons in the E2 modulation of body weight. Based on several studies, it is possible that KNDy neurons influence body weight via local projections to NPY and POMC neurons within the arcuate nucleus (7880). However, because KNDy neurons project to multiple hypothalamic centers (46, 58), there are many potential pathways for these neurons to influence energy homeostasis.

The present study used targeted cell ablation to study the function of a subpopulation of neurons, rather than knockout of a specific gene. Kisspeptin cell ablation has also been accomplished using a transgenic mouse strategy, in which the cells are made selectively vulnerable to diphtheria toxin (81). Consistent with our findings, ablation of kisspeptin cells in mice on postnatal d 20 (and in adult mice) produced disrupted estrous cycles and infertility (81). However, ip administration of diphtheria toxin would have systemic effects on kisspeptin cells, producing ablation in the ovary, pituitary, arcuate nucleus, amygdala, and the anteroventral periventricular nucleus (AVPV), which has numerous kisspeptin mRNA-expressing neurons by postnatal d 18 (82). In contrast, our study used stereotaxic injections of NK3-SAP in very small volumes (100 nl) to target NK3R-expressing KNDy neurons. Limited diffusion of the toxin from the injection site was documented by sparing of the NK3R-ir neurons in the supraoptic and parventicular nuclei. Thus, the NK3-SAP could not have diffused in a high enough concentration (through tissue or the ventricle) to directly affect rostral areas, such as the AVPV, the medial preoptic area, or median preoptic nucleus. In addition, the AVPV kisspeptin neurons would not be vulnerable to NK3-SAP because AVPV neurons do not express NK3R (23, 49). We did observe variable and incomplete loss of large NK3R cells in the lateral hypothalamus and zona incerta adjacent to the injection site. This was a potential confounding factor, because some of these cells contain melanin-concentrating hormone (31), a peptide involved in the regulation of body weight (83). However, a second experiment designed to replicate the extraarcuate NK3R cell loss did not produce the same effects as the arcuate injections. Thus, the alterations in the E2 modulation of gonadotropin secretion and body weight observed after injections of NK3-SAP in the arcuate nucleus were not secondary to degeneration of NK3R-expressing neurons in the lateral hypothalamus and zona incerta.

In postmenopausal women, KNDy neurons exhibit somatic hypertrophy and increased NKB and kisspeptin gene expression (4, 5). Experiments in cynomolgus monkeys indicate that the changes in postmenopausal women are due to loss of ovarian estrogen and not aging per se (5, 7, 9, 84). Based on these studies, we have hypothesized that increased activity of KNDy neurons stimulates the elevation in gonadotropin secretion that occurs in response to the ovarian failure of menopause (4, 5, 7). The present studies strongly support this hypothesis, because the rise in LH secretion after OVX was prevented in rats with KNDy neuron ablation. Dysfunction of KNDy neurons may also explain the low serum gonadotropins in patients with inactivating mutations in the genes encoding NKB, kisspeptin, or their receptors (1620). A role of KNDy neurons in the E2 modulation of body weight and abdominal girth is also relevant to women, because polymorphisms in the gene encoding ERα are associated with increased visceral adiposity and body mass index (85, 86). Moreover, postmenopausal women exhibit a change in body weight distribution characterized by an increased waist to hip ratio indicative of visceral adiposity (87). This is important, because increased visceral adiposity, independent of body mass index, is associated with hypertension, glucose intolerance, and increased risk for cardiovascular disease in women (88, 89).

In summary, we have demonstrated that arcuate KNDy neurons are essential for the rise in LH secretion after removal of ovarian E2, supporting a role for these neurons in E2 negative feedback. KNDy neurons are also required for the maintenance of tonic gonadotropin secretion and the E2 modulation of body weight. Recent studies have implicated NK3R signaling in the regulation of body temperature (90), and we are currently examining whether KNDy neuron ablation alters hypothalamic thermoregulation. Through integration of sex-steroid signals with various physiological systems, KNDy neurons could play a role in optimizing the internal environment for reproduction. Based on their location in the arcuate nucleus, expression of ERα and diverse projections to multiple homeostatic and neuroendocrine control centers (46, 58), KNDy neurons are ideally positioned for this task.

Acknowledgments

We thank Dr. Michelle Burke for expert technical assistance, Dr. Robert Sloviter for advice on the use of saporin-conjugates, and Dr. Penny Dacks and Marina Cholanian for insightful comments on the manuscript. Dr. Douglas Lappi designed the conjugation of saporin with an NK3 receptor agonist.

This work was supported by the National Institutes of Health, National Institute on Aging Grant R01 AG032315. The hormone assays were performed at the Ligand Assay and Analysis Core, University of Virginia Center for Research in Reproduction supported by the National Institute of Child Health and Human Development (Specialized Cooperative Centers Program in Reproduction Research) Grant U54-HD28934.

Disclosure Summary: The authors have nothing to disclose.

Footnotes

Abbreviations:
AVPV
Anteroventral periventricular nucleus
DAB
3,3′ diaminobenzidine
E2
17β-estradiol
ER
estrogen receptor
ir
immunoreactive
KNDy
kisspeptin/NKB/dynorphin
NKB
neurokinin B
NK3R
neurokinin 3 receptor
NPY
neuropeptide Y
OVX
ovariectomy
POMC
proopiomelanocortin
SAP
saporin.

References

  • 1. Tarttelin MF, Gorski RA. 1973. The effects of ovarian steroids on food and water intake and body weight in the female rat. Acta Endocrinol 72:551–568 [DOI] [PubMed] [Google Scholar]
  • 2. Xu Y, Nedungadi TP, Zhu L, Sobhani N, Irani BG, Davis KE, Zhang X, Zou F, Gent LM, Hahner LD, Khan SA, Elias CF, Elmquist JK, Clegg DJ. 2011. Distinct hypothalamic neurons mediate estrogenic effects on energy homeostasis and reproduction. Cell Metab 14:453–465 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Rance NE, McMullen NT, Smialek JE, Price DL, Young WS., 3rd 1990. Postmenopausal hypertrophy of neurons expressing the estrogen receptor gene in the human hypothalamus. J Clin Endocrinol Metab 71:79–85 [DOI] [PubMed] [Google Scholar]
  • 4. Rance NE, Young WS., 3rd 1991. Hypertrophy and increased gene expression of neurons containing neurokinin-B and substance-P messenger ribonucleic acids in the hypothalami of postmenopausal women. Endocrinology 128:2239–2247 [DOI] [PubMed] [Google Scholar]
  • 5. Rometo AM, Krajewski SJ, Voytko ML, Rance NE. 2007. Hypertrophy and increased kisspeptin gene expression in the hypothalamic infundibular nucleus of postmenopausal women and ovariectomized monkeys. J Clin Endocrinol Metab 92:2744–2750 [DOI] [PubMed] [Google Scholar]
  • 6. Rometo AM, Rance NE. 2008. Changes in prodynorphin gene expression and neuronal morphology in the hypothalamus of postmenopausal women. J Neuroendocrinol 20:1376–1381 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Rance NE. 2009. Menopause and the human hypothalamus: evidence for the role of kisspeptin/neurokinin B neurons in the regulation of estrogen negative feedback. Peptides 30:111–122 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Rance NE, Bruce TR. 1994. Neurokinin B gene expression is increased in the arcuate nucleus of ovariectomized rats. Neuroendocrinology 60:337–345 [DOI] [PubMed] [Google Scholar]
  • 9. Sandoval-Guzmán T, Stalcup ST, Krajewski SJ, Voytko ML, Rance NE. 2004. Effects of ovariectomy on the neuroendocrine axes regulating reproduction and energy balance in young cynomolgus macaques. J Neuroendocrinol 16:146–153 [DOI] [PubMed] [Google Scholar]
  • 10. Burke MC, Letts PA, Krajewski SJ, Rance NE. 2006. Coexpression of dynorphin and neurokinin B immunoreactivity in the rat hypothalamus: morphologic evidence of interrelated function within the arcuate nucleus. J Comp Neurol 498:712–726 [DOI] [PubMed] [Google Scholar]
  • 11. Goodman RL, Lehman MN, Smith JT, Coolen LM, de Oliveira CV, Jafarzadehshirazi MR, Pereira A, Iqbal J, Caraty A, Ciofi P, Clarke IJ. 2007. Kisspeptin neurons in the arcuate nucleus of the ewe express both dynorphin A and neurokinin B. Endocrinology 148:5752–5760 [DOI] [PubMed] [Google Scholar]
  • 12. Navarro VM, Gottsch ML, Chavkin C, Okamura H, Clifton DK, Steiner RA. 2009. Regulation of gonadotropin-releasing hormone secretion by kisspeptin/dynorphin/neurokinin B neurons in the arcuate nucleus of the mouse. J Neurosci 29:11859–11866 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Wakabayashi Y, Nakada T, Murata K, Ohkura S, Mogi K, Navarro VM, Clifton DK, Mori Y, Tsukamura H, Maeda K, Steiner RA, Okamura H. 2010. Neurokinin B and dynorphin A in kisspeptin neurons of the arcuate nucleus participate in generation of periodic oscillation of neural activity driving pulsatile gonadotropin-releasing hormone secretion in the goat. J Neurosci 30:3124–3132 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Ramaswamy S, Seminara SB, Ali B, Ciofi P, Amin NA, Plant TM. 2010. Neurokinin B stimulates GnRH release in the male monkey (Macaca mulatta) and is colocalized with kisspeptin in the arcuate nucleus. Endocrinology 151:4494–4503 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Lehman MN, Coolen LM, Goodman RL. 2010. Minireview: kisspeptin/neurokinin B/dynorphin (KNDy) cells of the arucate nucleus: a central node in the control of gonadotropin-releasing hormone secretion. Endocrinology 151:3479–3489 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Seminara SB, Messager S, Chatzidaki EE, Thresher RR, Acierno JS, Jr, Shagoury JK, Bo-Abbas Y, Kuohung W, Schwinof KM, Hendrick AG, Zahn D, Dixon J, Kaiser UB, Slaugenhaupt SA, Gusella JF, O'Rahilly S, Carlton MB, Crowley WF, Jr, Aparicio SA, Colledge WH. 2003. The GPR54 gene as a regulator of puberty. N Engl J Med 349:1614–1627 [DOI] [PubMed] [Google Scholar]
  • 17. de Roux N, Genin E, Carel JC, Matsuda F, Chaussain JL, Milgrom E. 2003. Hypogonadotropic hypogonadism due to loss of function of the KiSS1-derived peptide receptor GPR54. Proc Natl Acad Sci USA 100:10972–10976 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Topaloglu AK, Reimann F, Guclu M, Yalin AS, Kotan LD, Porter KM, Serin A, Mungan NO, Cook JR, Ozbek MN, Imamoglu S, Akalin NS, Yuksel B, O'Rahilly S, Semple RK. 2009. TAC3 and TACR3 mutations in familial hypogonadotropic hypogonadism reveal a key role for Neurokinin B in the central control of reproduction. Nat Genet 41:354–358 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Chan YM, Broder-Fingert S, Paraschos S, Lapatto R, Au M, Hughes V, Bianco SD, Min L, Plummer L, Cerrato F, De Guillebon A, Wu IH, Wahab F, Dwyer A, Kirsch S, Quinton R, Cheetham T, Ozata M, Ten S, Chanoine JP, Pitteloud N, Martin KA, Schiffmann R, Van der Kamp HJ, Nader S, Hall JE, Kaiser UB, Seminara SB. 2011. GnRH-deficient phenotypes in humans and mice with heterozygous variants in KISS1/Kiss1. J Clin Endocrinol Metab 96:E1771–E1781 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Topaloglu AK, Tello JA, Kotan LD, Ozbek MN, Yilmaz MB, Erdogan S, Gurbuz F, Temiz F, Millar RP, Yuksel B. 2012. Inactivating KISS1 mutation and hypogonadotropic hypogonadism. N Engl J Med 366:629–635 [DOI] [PubMed] [Google Scholar]
  • 21. Yang JJ, Caligioni CS, Chan YM, Seminara SB. 2012. Uncovering novel reproductive deficits in neurokinin B receptor null mice: closing the gap between mice and men. Endocrinology 153:1498–1508 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Amstalden M, Coolen LM, Hemmerle AM, Billings HJ, Connors JM, Goodman RL, Lehman MN. 2010. Neurokinin 3 receptor immunoreactivity in the septal region, preoptic area and hypothalamus of the female sheep: colocalisation in neurokinin B cells of the arcuate nucleus but not in gonadotrophin-releasing hormone neurones. J Neuroendocrinol 22:1–12 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Shughrue PJ, Lane MV, Merchenthaler I. 1996. In situ hybridization analysis of the distribution of neurokinin-3 mRNA in the rat central nervous system. J Comp Neurol 372:395–414 [DOI] [PubMed] [Google Scholar]
  • 24. Krajewski SJ, Anderson MJ, Iles-Shih L, Chen KJ, Urbanski HF, Rance NE. 2005. Morphologic evidence that neurokinin B modulates gonadotropin-releasing hormone secretion via neurokinin 3 receptors in the rat median eminence. J Comp Neurol 489:372–386 [DOI] [PubMed] [Google Scholar]
  • 25. Lappi DA, Wiley RG. 2000. Entering through the doors of perception: characterization of a highly selective substance P receptor-targeted toxin. Neuropeptides 34:323–328 [DOI] [PubMed] [Google Scholar]
  • 26. Drapeau G, d'Orléans-Juste P, Dion S, Rhaleb NE, Regoli D. 1987. Specific agonists for neurokinin B receptors. Eur J Pharmacol 136:401–403 [DOI] [PubMed] [Google Scholar]
  • 27. Sadowski S, Huang RR, Fong TM, Marko O, Cascieri MA. 1993. Characterization of the binding of [125I-Iodo-histidyl, Methyl-Phe7] neurokinin B to the neurokinin-3 receptor. Neuropeptides 24:317–319 [DOI] [PubMed] [Google Scholar]
  • 28. Mileusnic D, Lee JM, Magnuson DJ, Hejna MJ, Krause JE, Lorens JB, Lorens SA. 1999. Neurokinin-3 receptor distribution in rat and human brain: an immunohistochemical study. Neuroscience 89:1269–1290 [DOI] [PubMed] [Google Scholar]
  • 29. Watson RE, Jr, Wiegand SJ, Clough RW, Hoffman GE. 1986. Use of cryoprotectant to maintain long-term peptide immunoreactivity and tissue morphology. Peptides 7:155–159 [DOI] [PubMed] [Google Scholar]
  • 30. Ström JO, Theodorsson A, Theodorsson E. 2008. Substantial discrepancies in 17β-oestradiol concentrations obtained with three different commercial direct radioimmunoassay kits in rat sera. Scand J Clin Lab Invest 68:806–813 [DOI] [PubMed] [Google Scholar]
  • 31. Griffond B, Ciofi P, Bayer L, Jacquemard C, Fellmann D. 1997. Immunocytochemical detection of the neurokinin B receptor (NK3) on melanin-concentrating hormone (MCH) neurons in rat brain. J Chem Neuroanat 12:183–189 [DOI] [PubMed] [Google Scholar]
  • 32. Lucas LR, Hurley DL, Krause JE, Harlan RE. 1992. Localization of the tachykinin neurokinin B precursor peptide in rat brain by immunocytochemistry and in situ hybridization. Neuroscience 51:317–345 [DOI] [PubMed] [Google Scholar]
  • 33. Marksteiner J, Sperk G, Krause JE. 1992. Distribution of neurons expressing neurokinin B in the rat brain: immunohistochemistry and in situ hybridization. J Comp Neurol 317:341–356 [DOI] [PubMed] [Google Scholar]
  • 34. Ciofi P, Krause JE, Prins GS, Mazzuca M. 1994. Presence of nuclear androgen receptor-like immunoreactivity in neurokinin B-containing neurons of the hypothalamic arcuate nucleus of the adult male rat. Neurosci Lett 182:193–196 [DOI] [PubMed] [Google Scholar]
  • 35. Desroziers E, Mikkelsen J, Simonneaux V, Keller M, Tillet Y, Caraty A, Franceschini I. 2010. Mapping of kisspeptin fibres in the brain of the pro-oestrous rat. J Neuroendocrinol 22:1101–1112 [DOI] [PubMed] [Google Scholar]
  • 36. Franceschini I, Lomet D, Cateau M, Delsol G, Tillet Y, Caraty A. 2006. Kisspeptin immunoreactive cells of the ovine preoptic area and arcuate nucleus co-express estrogen receptor α. Neurosci Lett 401:225–230 [DOI] [PubMed] [Google Scholar]
  • 37. Fallon JH, Leslie FM. 1986. Distribution of dynorphin and enkephalin peptides in the rat brain. J Comp Neurol 249:293–336 [DOI] [PubMed] [Google Scholar]
  • 38. Wang Z, Gardell LR, Ossipov MH, Vanderah TW, Brennan MB, Hochgeschwender U, Hruby VJ, Malan TP, Jr, Lai J, Porreca F. 2001. Pronociceptive actions of dynorphin maintain chronic neuropathic pain. J Neurosci 21:1779–1786 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Mezey E, Kiss JZ, Mueller GP, Eskay R, O'Donohue TL, Palkovits M. 1985. Distribution of the pro-opiomelanocortin derived peptides, adrenocorticotrope hormone, a-melanocyte-stimulating hormone and β-endorphin (ACTH, a-MSH, b-END) in the rat hypothalamus. Brain Res 328:341–347 [DOI] [PubMed] [Google Scholar]
  • 40. Yee CL, Wang Y, Anderson S, Ekker M, Rubenstein JLR. 2009. Arcuate nucleus expression of NKX2.1 and DLX and lineages expressing these transcription factors in neuropeptide Y+, proopiomelanocortin+, and tyrosine hydroxylase+ neurons in neonatal and adult mice. J Comp Neurol 517:37–50 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Li C, Chen P, Smith MS. 1999. Morphological evidence for direct interaction between arcuate nucleus neuropeptide Y (NPY) neurons and gonadotropin-releasing hormone neurons and the possible involvement of NPY Y1 receptors. Endocrinology 140:5382–5390 [DOI] [PubMed] [Google Scholar]
  • 42. Kienzler F, Norwood BA, Sloviter RS. 2009. Hippocampal injury, atrophy, synaptic reorganization, and epileptogenesis after perforant pathway stimulation-induced status epilepticus in the mouse. J Comp Neurol 515:181–196 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Urbanski HF. 1991. Monoclonal antibodies to luteinizing hormone-releasing hormone: production, characterization, and immunocytochemical application. Biol Reprod 44:681–686 [DOI] [PubMed] [Google Scholar]
  • 44. Paxinos G, Watson C. 2007. The rat brain in stereotaxic coordinates. Burlington, MA: Elsevier, Inc; [DOI] [PubMed] [Google Scholar]
  • 45. Jiao Y, Sun Z, Lee T, Fusco FR, Kimble TD, Meade CA, Cuthbertson S, Reiner A. 1999. A simple and sensitive antigen retrieval method for free-floating and slide-mounted tissue sections. J Neurosci Methods 93:149–162 [DOI] [PubMed] [Google Scholar]
  • 46. Krajewski SJ, Burke MC, Anderson MJ, McMullen NT, Rance NE. 2010. Forebrain projections of arcuate neurokinin B neurons demonstrated by anterograde tract-tracing and monosodium glutamate lesions in the rat. Neuroscience 166:680–697 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Maolood N, Meister B. 2008. Dynorphin in pro-opiomelanocortin neurons of the hypothalamic arcuate nucleus. Neuroscience 154:1121–1131 [DOI] [PubMed] [Google Scholar]
  • 48. Bugarith K, Dinh TT, Li AJ, Speth RC, Ritter S. 2005. Basomedial hypothalamic injections of neuropeptide Y conjugated to saporin selectively disrupt hypothalamic controls of food intake. Endocrinology 146:1179–1191 [DOI] [PubMed] [Google Scholar]
  • 49. Ding YQ, Shigemoto R, Takada M, Ohishi H, Nakanishi S, Mizuno N. 1996. Localization of the neuromedin K receptor (NK3) in the central nervous system of the rat. J Comp Neurol 364:290–310 [DOI] [PubMed] [Google Scholar]
  • 50. Moore CR, Price D. 1932. Gonad hormone functions, and the reciprocal influence between gonads and hypophysis with its bearing on the problem of sex hormone antagonism. Am J Anat 50:13–71 [Google Scholar]
  • 51. Freeman ME. 2006. The neuroendocrine control of the estrous cycle of the rat. In: Neill JD, Plant TM, Pfaff DW, Challis JRG, de Kretser DM, Richards JS, Wassarman PM, eds. Knobil and Neill's physiology of reproduction. 3rd ed San Diego: Elsevier; 2327–2388 [Google Scholar]
  • 52. Herbison AE. 1998. Multimodal influence of estrogen upon gonadotropin-releasing hormone neurons. Endocr Rev 19:302–330 [DOI] [PubMed] [Google Scholar]
  • 53. Singh SP, Wolfe A, Ng Y, DiVall SA, Buggs C, Levine JE, Wondisford FE, Radovick S. 2009. Impaired estrogen feedback and infertility in female mice with pituitary-specific deletion of estrogen receptor α (ESR1). Biol Reprod 81:488–496 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Smith JT, Cunningham MJ, Rissman EF, Clifton DK, Steiner RA. 2005. Regulation of Kiss1 gene expression in the brain of the female mouse. Endocrinology 146:3686–3692 [DOI] [PubMed] [Google Scholar]
  • 55. Dorling AA, Todman MG, Korach KS, Herbison AE. 2003. Critical role for estrogen receptor α in negative feedback regulation of gonadotropin-releasing hormone mRNA expression in the female mouse. Neuroendocrinology 78:204–209 [DOI] [PubMed] [Google Scholar]
  • 56. Dellovade TL, Merchenthaler I. 2004. Estrogen regulation of neurokinin B gene expression in the mouse arcuate nucleus is mediated by estrogen receptor a. Endocrinology 145:736–742 [DOI] [PubMed] [Google Scholar]
  • 57. Mayer C, Acosta-Martinez M, Dubois SL, Wolfe A, Radovick S, Boehm U, Levine JE. 2010. Timing and completion of puberty in female mice depend on estrogen receptor a-signaling in kisspeptin neurons. Proc Natl Acad Sci USA 107:22693–22698 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Yeo SH, Herbison AE. 2011. Projections of arcuate nucleus and rostral periventricular kisspeptin neurons in the adult female mouse brain. Endocrinology 152:2387–2399 [DOI] [PubMed] [Google Scholar]
  • 59. Ciofi P, Leroy D, Tramu G. 2006. Sexual dimorphism in the organization of the rat hypothalamic infundibular area. Neuroscience 141:1731–1745 [DOI] [PubMed] [Google Scholar]
  • 60. Ramaswamy S, Guerriero KA, Gibbs RB, Plant TM. 2008. Structural interactions between kisspeptin and GnRH neurons in the mediobasal hypothalamus of the male rhesus monkey (Macaca mulatta) as revealed by double immunofluorescence and confocal microscopy. Endocrinology 149:4387–4395 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Matsuyama S, Ohkura S, Mogi K, Wakabayashi Y, Mori Y, Tsukamura H, Maeda K, Ichikawa M, Okamura H. 2011. Morphological evidence for direct interaction between kisspeptin and gonadotropin-releasing hormone neurons at the median eminence of the male goat: an immunoelectron microscopic study. Neuroendocrinology 94:323–332 [DOI] [PubMed] [Google Scholar]
  • 62. Smith JT, Li Q, Yap KS, Shahab M, Roseweir AK, Millar RP, Clarke IJ. 2011. Kisspeptin is essential for the full preovulatory LH surge and stimulates GnRH release from the isolated ovine median eminence. Endocrinology 152:1001–1012 [DOI] [PubMed] [Google Scholar]
  • 63. Keen KL, Wegner FH, Bloom SR, Ghatei MA, Terasawa E. 2008. An increase in kisspeptin-54 release occurs with the pubertal increase in luteinizing hormone-releasing hormone-1 release in the stalk-median eminence of female rhesus monkeys in vivo. Endocrinology 149:4151–4157 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Han SK, Gottsch ML, Lee KJ, Popa SM, Smith JT, Jakawich SK, Clifton DK, Steiner RA, Herbison AE. 2005. Activation of gonadotropin-releasing hormone neurons by kisspeptin as a neuroendocrine switch for the onset of puberty. J Neurosci 25:11349–11356 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Dungan HM, Gottsch ML, Zeng H, Gragerov A, Bergmann JE, Vassilatis DK, Clifton DK, Steiner RA. 2007. The role of kisspeptin-GPR54 signaling in the tonic regulation and surge release of gonadotropin-releasing hormone/luteinizing hormone. J Neurosci 27:12088–12095 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Goodman RL, Coolen LM, Anderson GM, Hardy SL, Valent M, Connors JM, Fitzgerald ME, Lehman MN. 2004. Evidence that dynorphin plays a major role in mediating progesterone negative feedback on gonadotropin-releasing hormone neurons in sheep. Endocrinology 145:2959–2967 [DOI] [PubMed] [Google Scholar]
  • 67. Rance NE, Krajewski SJ, Smith MA, Cholanian M, Dacks PA. 2010. Neurokinin B and the hypothalamic regulation of reproduction. Brain Res 1364:116–128 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Kinsey-Jones JS, Grachev P, Li XF, Lin YS, Milligan SR, Lightman SL, O'Byrne KT. 2012. The inhibitory effects of neurokinin B on GnRH pulse generator frequency in the female rat. Endocrinology 153:307–315 [DOI] [PubMed] [Google Scholar]
  • 69. Bogart R, Lasley JF, Mayer DT. 1944. Influence of reproductive hormones upon growth in ovariectomized and normal female rats. Endocrinology 35:173–181 [Google Scholar]
  • 70. Blaustein JD, Gentry RT, Roy EJ, Wade GN. 1976. Effects of ovariectomy and estradiol on body weight and food intake in gold thioglucose-treated mice. Physiol Behav 17:1027–1030 [DOI] [PubMed] [Google Scholar]
  • 71. Heine PA, Taylor JA, Iwamoto GA, Lubahn DB, Cooke PS. 2000. Increased adipose tissue in male and female estrogen receptor-α knockout mice. Proc Natl Acad Sci USA 97:12729–12734 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Gao Q, Mezei G, Nie Y, Rao Y, Choi CS, Bechmann I, Leranth C, Toran-Allerand D, Priest CA, Roberts JL, Gao XB, Mobbs C, Shulman GI, Diano S, Horvath TL. 2007. Anorectic estrogen mimics leptin's effect on the rewiring of melanocortin cells and Stat3 signaling in obese animals. Nat Med 13:89–94 [DOI] [PubMed] [Google Scholar]
  • 73. Williams H, Dacks PA, Rance NE. 2010. An improved method for recording tail skin temperature in the rat reveals changes during the estrous cycle and effects of ovarian steroids. Endocrinology 151:5389–5394 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Tena-Sempere M. 2006. KiSS-1 and reproduction: focus on its role in the metabolic regulation of fertility. Neuroendocrinology 83:275–281 [DOI] [PubMed] [Google Scholar]
  • 75. Kalamatianos T, Grimshaw SE, Poorun R, Hahn JD, Coen CW. 2008. Fasting reduces KiSS-1 expression in the anteroventral periventricular nucleus (AVPV): effects of fasting on the expression of KiSS-1 and neuropeptide Y in the AVPV or arcuate nucleus of female rats. J Neuroendocrinol 20:1089–1097 [DOI] [PubMed] [Google Scholar]
  • 76. Xu J, Kirigiti MA, Grove KL, Smith MS. 2009. Regulation of food intake and gonadotropin-releasing hormone/luteinizing hormone during lactation: role of insulin and leptin. Endocrinology 150:4231–4240 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Quennell JH, Howell CS, Roa J, Augustine RA, Grattan DR, Anderson GM. 2011. Leptin deficiency and diet-induced obesity reduce hypothalamic kisspeptin expression in mice. Endocrinology 152:1541–1550 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Backholer K, Smith JT, Rao A, Pereira A, Iqbal J, Ogawa S, Li Q, Clarke IJ. 2010. Kisspeptin cells in the ewe brain respond to leptin and communicate with neuropeptide Y and proopiomelanocortin cells. Endocrinology 151:2233–2243 [DOI] [PubMed] [Google Scholar]
  • 79. Kim GL, Dhillon SS, Belsham DD. 2010. Kisspeptin directly regulates neuropeptide Y synthesis and secretion via the ERK1/2 and p38 mitogen-activated protein kinase signaling pathways in NPY-secreting hypothalamic neurons. Endocrinology 151:5038–5047 [DOI] [PubMed] [Google Scholar]
  • 80. Fu LY, van den Pol AN. 2010. Kisspeptin directly excites anorexigenic proopiomelanocortin neurons but inhibits orexigenic neuropeptide Y cells by an indirect synaptic mechanism. J Neurosci 30:10205–10219 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Mayer C, Boehm U. 2011. Female reproductive maturation in the absence of kisspeptin/GPR54 signaling. Nat Neurosci 14:704–710 [DOI] [PubMed] [Google Scholar]
  • 82. Kauffman AS, Navarro VM, Kim J, Clifton DK, Steiner RA. 2009. Sex differences in the regulation of Kiss1/NKB neurons in juvenile mice: implications for the timing of puberty. Am J Physiol Endocrinol Metab 297:E1212–E1221 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Shimada M, Tritos NA, Lowell BB, Flier JS, Maratos-Flier E. 1998. Mice lacking melanin-concentrating hormone are hypophagic and lean. Nature 396:670–674 [DOI] [PubMed] [Google Scholar]
  • 84. Abel TW, Voytko ML, Rance NE. 1999. The effects of hormone replacement therapy on hypothalamic neuropeptide gene expression in a primate model of menopause. J Clin Endocrinol Metab 84:2111–2118 [DOI] [PubMed] [Google Scholar]
  • 85. Deng HW, Li J, Li JL, Dowd R, Davies KM, Johnson M, Gong G, Deng H, Recker RR. 2000. Association of estrogen receptor-a genotypes with body mass index in normal healthy postmenopausal caucasian women. J Clin Endocrinol Metab 85:2748–2751 [DOI] [PubMed] [Google Scholar]
  • 86. Okura T, Koda M, Ando F, Niino N, Ohta S, Shimokata H. 2003. Association of polymorphisms in the estrogen receptor a gene with body fat distribution. Int J Obes Relat Metab Disord 27:1020–1027 [DOI] [PubMed] [Google Scholar]
  • 87. Samaras K, Hayward CS, Sullivan D, Kelly RP, Campbell LV. 1999. Effects of postmenopausal hormone replacement therapy on central abdominal fat, glycemic control, lipid metabolism, and vascular factors in type 2 diabetes: a prospective study. Diabetes Care 22:1401–1407 [DOI] [PubMed] [Google Scholar]
  • 88. Krotkiewski M, Björntorp P, Sjöström L, Smith U. 1983. Impact of obesity on metabolism in men and women. Importance of regional adipose tissue distribution. J Clin Invest 72:1150–1162 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Lapidus L, Bengtsson C, Larsson B, Pennert K, Rybo E, Sjöström L. 1984. Distribution of adipose tissue and risk of cardiovascular disease and death: a 12-yr follow up of participants in the population study of women in Gothenburg, Sweden. Br Med J 289:1257–1261 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Dacks PA, Krajewski SJ, Rance NE. 2011. Activation of neurokinin 3 receptors in the median preoptic nucleus decreases core temperature in the rat. Endocrinology 152:4894–4905 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Endocrinology are provided here courtesy of The Endocrine Society

RESOURCES