Journal Pre-proof Salmon calcitonin distributes into the arcuate nucleus in mice to a subset of NPY neurons Hannah Louise Zakariassen, Linu Mary John, Jens Lykkesfeldt, Kirsten Raun, Tine Glendorf, Lauge Schaffer, Sofia Lundh, Anna Secher, Thomas Alexander Lutz, Christelle Le Foll PII:
S0028-3908(20)30053-8
DOI:
https://doi.org/10.1016/j.neuropharm.2020.107987
Reference:
NP 107987
To appear in:
Neuropharmacology
Received Date: 8 July 2019 Revised Date:
28 January 2020
Accepted Date: 2 February 2020
Please cite this article as: Zakariassen, H.L., John, L.M., Lykkesfeldt, J., Raun, K., Glendorf, T., Schaffer, L., Lundh, S., Secher, A., Lutz, T.A., Le Foll, C., Salmon calcitonin distributes into the arcuate nucleus in mice to a subset of NPY neurons, Neuropharmacology (2020), doi: https://doi.org/10.1016/ j.neuropharm.2020.107987. This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain. © 2020 Published by Elsevier Ltd.
1
Salmon calcitonin distributes into the arcuate nucleus in mice to a subset of NPY neurons
2 3 4 5
Hannah Louise Zakariassen1,2, Linu Mary John2, Jens Lykkesfeldt1, Kirsten Raun2, Tine
6
Glendorf4, †Lauge Schaffer5, Sofia Lundh3, Anna Secher4, Thomas Alexander Lutz6 and
7
Christelle Le Foll6,*
8 9
1
Section of Experimental Animal Models, Department of Veterinary and Animal Science, Faculty of
10
Health and Medical Sciences, University of Copenhagen, DK-1871 Frederiksberg C, Denmark.
11
2
12
Chemistry, Novo Nordisk A/S, 2760 Måløv, Denmark.
13
University of Zurich, CH-8057 Zurich, Switzerland
Obesity Pharmacology,
3
Pathology and Imaging, and
4
Diabetes Pharmacology 2, 6
5
Research
Institute of Veterinary Physiology,
14 15
*Corresponding authors:
[email protected]
16
†
Deceased on 27th August 2019
17 18
Number of figures: 6
19
Number of tables: 1
20
Number of videos (extended data): 3
21
Supplementary figures: 2
1
22
ABSTRACT
23
The amylin receptor (AMY) and calcitonin receptor (CTR) agonists induce acute suppression of food
24
intake in rodents by binding to receptors in the area postrema (AP) and potentially by targeting
25
arcuate (ARC) neurons directly. Salmon calcitonin (sCT) induces more potent, longer lasting
26
anorectic effects compared to amylin. We thus aimed to investigate whether AMY /CTR agonists
27
target key neuronal populations in the ARC, and whether differing brain distribution patterns could
28
mediate the observed differences in efficacy with sCT and amylin treatment. Brains were examined
29
by whole brain 3D imaging and confocal microscopy following subcutaneous administration of
30
fluorescently labelled peptides to male and female mice. We found that sCT, but not amylin,
31
internalizes into a subset of ARC NPY neurons, along with an unknown subset of ARC, AP and
32
dorsal vagal motor nucleus cells. ARC POMC neurons were not targeted. Furthermore, amylin and
33
sCT displayed similar distribution patterns when binding to receptors in the AP, the organum
34
vasculosum of the lamina terminalis (OVLT) and the ARC. Amylin was distributed within the
35
median eminence with only specs of sCT being present in this region, however amylin was only
36
detectable 10 minutes after injection while sCT displayed a residence time of up to 2 hours post
37
injection. We conclude that AMY /CTR agonists bind to receptors in a subset of ARC NPY neurons
38
and in circumventricular organs. Furthermore, the more sustained and greater anorectic efficacy of
39
sCT compared to rat amylin is not attributable to differences in brain distribution patterns but may
40
more likely be explained by greater potency at both CTR and AMY3.
41 42
KEYWORDS: amylin; agonist; whole-brain 3D imaging; area postrema; NPY
2
43 44
HIGHLIGHTS •
45
Whole brain 3D imaging showed that amylin and sCT present similar distribution patterns in the AP, the OVLT and the ARC.
46
•
sCT internalizes into a subset of ARC NPY neurons as well as in AP and DVM nucleus cells.
47
•
Peripherally injected AMY /CTR agonists target specific neuronal populations to induce their
48
metabolic effects.
3
49
1. INTRODUCTION
50 51
The pancreatic hormone amylin is secreted postprandially and induces satiation (Lutz et al., 1995) by
52
binding to amylin receptors (AMY), the constituents of which are distributed throughout the brain
53
(Becskei et al., 2004; Hilton et al., 1995; Nakamoto et al., 2000; Oliver et al., 2001). AMY are
54
composed of two components, the calcitonin receptor (CTR) and one of three receptor activity
55
modifying proteins (RAMP1-3) (Poyner et al., 2002). While amylin binds selectively to AMY,
56
salmon calcitonin binds to both CTR and AMY (Christopoulos et al., 1999; Tilakaratne et al., 2000).
57
Access of circulating factors to the brain are prevented by the blood brain barrier (BBB) except for
58
specific BBB free regions of the brain, namely the circumventricular organs (CVO, e.g., median
59
eminence (ME), subfornical organ (SFO)) that are infiltrated by highly permeable vasculature
60
(Abbott et al., 2010). Additionally, the ARC has been shown to be permeable to circulating factors
61
such as ghrelin by diffusion through the fenestrated capillaries of the ME that branch into the
62
ventromedial region of the arcuate hypothalamus to bind to cognate receptors in NPY/AgRP
63
(neuropeptide Y/ agouti-related protein) and POMC (pro-opiomelanocortin) expressing neurons
64
(Schaeffer et al., 2013). AMY /CTR agonists induce acute anorectic effects by activating neurons in
65
the area postrema (AP), a CVO in the hindbrain. AMY in the AP seem to be necessary for amylin’s
66
satiating effect (Braegger et al., 2014; Lutz et al., 2001; Riediger et al., 2004). From the AP, neuronal
67
projections send signals to other areas in the brain such as the nucleus of solitary tract (NTS), lateral
68
parabrachial nucleus (LPBN) and the lateral hypothalamic area (LHA) (Potes et al., 2010). In the
69
hypothalamus, amylin has been shown to potentiate leptin signaling (Dunn-Meynell et al., 2016;
70
Turek et al., 2010) and regulate several neuropeptide systems involved in energy balance regulation
71
(Barth et al., 2003; Roth et al., 2006).
72
POMC and NPY/AgRP co-expressing neurons in the arcuate nucleus (ARC) exert homeostatic
73
control of feeding behavior by inducing opposing effects of anorexigenic or orexigenic activity, 4
74
respectively (Loh et al., 2015; Yeo and Heisler, 2012). Recently, we found that amylin induces
75
pERK signaling in POMC but not NPY neurons independently of the AP (Lutz et al., 2018), but
76
others have shown that POMC neurons only express low levels of CTRs while NPY/AgRP neurons
77
express high levels (Campbell et al., 2017; Lam et al., 2017; Pan et al., 2018). Hence, these findings
78
indicate that AMY /CTR agonists target neuronal populations in the hypothalamus, but whether
79
NPY/AgRP and/or POMC neurons are targeted directly remains unclear. Treatment with both amylin
80
or calcitonin peptides decreases body weight in rodents (Andreassen et al., 2014; Feigh et al., 2011;
81
Roth et al., 2007; Trevaskis et al., 2010). Both rat amylin and salmon calcitonin have short half-lives
82
of approximately 10 and 30 minutes, respectively (Chaturvedula et al., 2005; Young et al., 1996), but
83
only salmon calcitonin induces a robust acute anorexic response past the reported half-life in plasma
84
(Reidelberger et al., 2001; Reidelberger et al., 2002). This has been suggested to be due to
85
irreversible binding of salmon calcitonin to the CTR (Houssami et al., 1994) and observed
86
differences in potency on cognate receptors. Nevertheless, differences in activation and regulation of
87
CNS signaling pathways have been reported (Braegger et al., 2014; Whiting et al., 2017), although
88
such differences have not been systematically studied.
89
Based on these earlier findings, we aimed to investigate whether fluorescently labelled rat amylin
90
and salmon calcitonin bind directly to POMC and/or NPY/AgRP neurons in the ARC, and if the two
91
peptides have similar distribution patterns within the brain to evaluate whether this could account for
92
part of the differences in acute anorectic effect. We hypothesized that both peptides bind to ARC
93
NPY/AgRP and POMC neurons along with cells in the AP, and that the peptides would show
94
differing distribution patterns. To test this hypothesis, wild type and transgenic mice models NPY-
95
GFP and POMC-cre:ERT2: td-tomato were used.
5
96
2. RESEARCH DESIGN AND METHODS
97 98 99
2.1. Peptides Native peptides
100
Novo Nordisk A/S (Måløv, Denmark) synthesized the native salmon calcitonin in an acetate buffer.
101
Native rat amylin (H9475 Bachem AG, Bubendorf, Switzerland) was diluted in saline at a stock
102
concentration of 0.25 mmol/L and further diluted at 10 nmol/ml in acetate buffer (pH= 4.0; 5 mM
103
acetate; 240 mM propylene glycol, 0.007% tween 20).
104
Synthesis of fluorescently labeled ligands
105
Novo Nordisk A/S (Måløv, Denmark) also synthesized the fluorescently labelled peptides.
106
Fluorescently labelled salmon calcitonin and rat amylin was synthesized by conjugating Alexa fluor
107
750-NHS ester (Invitrogen by Thermo Fisher Scientific, Waltham, MA, USA) to salmon calcitonin
108
(sCT750), Cyanin 7-NHS ester (Amersham by GE Healthcare Life Sciences, Chicago, IL, USA) to rat
109
amylin (rAMYCy7) and Cyanin 5-NHS ester (Amersham by GE Healthcare Life Sciences) to salmon
110
calcitonin (sCTCy5) and rat amylin (rAMYCy5).
111
The fluorescently labelled peptides, described in these experiments, were purified from excess
112
labelling agent by gel filtration on PD10 columns with isocratic elution of 20 mM NH4HCO3. The
113
fractions containing the pure peptide were freeze dried and used in the experiments. The labelled and
114
unlabeled peptide were validated by LC-MS and confirmed the presence of only one fluorophore per
115
peptide. sCTCy5 and sCT750 were confirmed to be single compounds, as the fluorophore sits on the N-
116
terminal amino group while rAMYCy5and rAMYCy7 may be a mixture of regioisomers modified at the
117
N-terminal or the epsilon-amino group of the lysine. All fluorescently labelled peptides were diluted
118
in acetate buffer. Control groups were dosed with saline or acetate buffer.
119
6
120
2.2. Animal husbandry and diet
121
Mice were group housed in temperature controlled (21 ± 2°C) room on a 12:12 h light:dark schedule
122
with ad libitum access to standard chow (no. 3436; Provimi Kliba, Kaiseraugst, Switzerland) and
123
water before and during experiments, unless otherwise stated. Animals were housed in an enriched
124
environment and handled before procedures. The experiments were carried out in accordance with
125
the EU Directive 2010/63/EU of the 22 September 2010 on the protection of animals used for
126
scientific purposes and approved by the Veterinary Office of the Canton Zurich (no. 102/2018) or the
127
Animal Experimentation Inspectorate, Ministry of Environment and Food, Denmark (no. 2012-15-
128
2934-0000069 and no. 2014-15-0201-00388). Studies investigating the efficacy of sCT750 and
129
localization of CTR and POMC in the mouse hypothalamus were performed at Novo Nordisk A/S
130
animal facilities in Måløv, Denmark, while all other studies were performed in the animal facilities at
131
the Institute for Veterinary Physiology, University of Zurich Switzerland.
132 133
2.3. Efficacy of fluorophore conjugated compounds on acute food intake suppression
134
Adult C57BL/6J male mice were purchased from Taconic (Lille Skensved, Denmark), fed a low-fat
135
diet (D12450B, Research Diets Inc., New Brunswick, NJ, USA) and acclimated to BioDAQ cages
136
for 2 weeks before study initiation (single housed with transparent divider with holes in a
137
temperature-controlled room (23 ± 2°C) on a 12-h light/12-h dark light cycle). Before study initiation
138
mice were randomized into three groups (n = 6 – 8), fasted for 4 hours before dark and injected
139
subcutaneously (SC) with vehicle, salmon calcitonin 100 nmol/kg or sCT750 100 nmol/kg 20 minutes
140
before dark initiation. At dark onset, food was returned, and food intake measured continuously by
141
the BioDAQ system (Research Diets Inc.) for 24 hours. After 24 hours, animals were sacrificed by
142
CO2 inhalation.
143
Adult male C57Bl6JRj mice (Elevage Janvier, France) fed a standard chow diet (no. 3436; Provimi
144
Kliba) were acclimated to BioDAQ cages for 1.5 weeks (temperature-controlled room (21 ± 2°C) on 7
145
a 12-h light/12-h dark light cycle). The mice were used in three separate experiments to validate the
146
in vivo efficacy of rAMYCy5, rAMYCy7 and sCTCy5: experiment 1) vehicle, rat amylin 100 nmol/kg or
147
rAMYCy5 100 nmol/kg; experiment 2) vehicle, rat amylin 300 nmol/kg or rAMYCy7 300 nmol/kg;
148
experiment 3) vehicle, salmon calcitonin 100 nmol/kg or sCTCy5 100 nmol/kg. Animals were
149
weighed before each experiment, randomized to new treatment groups (n = 8/group) and fasted for
150
12 hours before SC injection of peptides right before dark onset. At dark onset, food was returned,
151
and food intake measured every 15 min for 24 hours. A wash out period of 4 – 6 days was set
152
between each experiment. Due to unexpected errors in food intake measurements, one mouse was
153
excluded from the rat amylin group in experiment 1, one mouse was excluded from the rAMYCy7
154
group in experiment 2 and two mice were excluded from the vehicle group in experiment 3 before
155
statistical analysis.
156 157
2.4. In vitro validation of the fluorophore conjugated compounds
158
A BHK cell line was stably transfected with the human calcitonin receptor and a cAMP responsive
159
element (CRE) luciferase reporter gene. The cell line was further transfected with receptor modifying
160
protein 3 (RAMP3) thus generating the human amylin 3 receptor. When performing the assays,
161
frozen aliquots of the above mentioned BHK cell lines were thawed, washed in seeding medium
162
(DMEM with phenol red (no. 31966-021; Gibco ThermoFisher Scientific), 10% (v/v) FBS (no.
163
16140-071; Gibco) and 1% (v/v) Penicillin-Streptomycin (no. 15140-122; Gibco) and resuspended in
164
seeding medium. Cells were plated into 384-well plates at 4000 cells/well in a volume of 40 µL and
165
incubated O/N at 37°C and 5% CO2. On the following day, reference and test compounds were
166
prediluted to appropriate concentrations (approximately 10-100 nM) in assay buffer (DMEM w/o
167
phenol red (no. 11880-028; Gibco), 10 mM HEPES (no. 15630-056; Gibco), 1X GlutaMAX™ (no.
168
35020-038; Gibco), 0.1% (w/v) ovalbumin (no. A5503; Sigma Aldrich) and 1% (w/v) HSA (no.
169
A1887; Sigma Aldrich)) and transferred to 96-well plates where 7-fold serial dilutions of each 8
170
compound were performed in duplicates using a Biomek i7 liquid handler. Cells were washed three
171
times in assay medium without HSA and 30 µL of each dilution of reference and test compounds
172
were transferred to the 384-well assay plates. The plates were incubated for 3h at 37°C and 5% CO2
173
and 30 µL of steadyliteplus reagent (no.6066759; PerkinElmer) added. While protected from light,
174
plates were incubated with shaking for 5 minutes at 300 rpm and incubated for an additional 30
175
minutes at room temperature before measuring luminescence on a Synergy 2 (BioTek) plate reader.
176
Data were imported into GraphPad Prism (version 8.0.2 for Windows, GraphPad Software, San
177
Diego, California, USA) and EC50-values determined using non-linear regression (log(agonist) vs.
178
response (four parameters) with a Hill slope of 1.5 and a shared bottom for all curves). rAMYCy7
179
could not be assessed in vitro due to its low purity.
180 181
2.5. Salmon calcitonin and rat amylin binding to arcuate and hindbrain single cells
182
NPY-hrGFP [B6.FVB-Tg(Npy-hrGFP)1Lowl/J, no. 006417; The Jackson Laboratory] and POMC-
183
Cre:ERT2 (Gift from Pr. Joel Elmquist, UT Southwestern, USA) (Berglund et al., 2013) were bred in
184
the animal facilities at the Institute for Veterinary Physiology. NPY-hrGFP allows for identification
185
of NPY neurons without additional immunohistochemistry (IHC) staining due to endogenous GFP
186
fluorescence driven by the NPY promoter. Similarly, the POMC-Cre:ERT2 mice were crossed with
187
tdTomato reporter mice (B6.Cg-Gt(ROSA)26Sortm14(CAG-tdTomato)Hze/J, no. 007914; The
188
Jackson Laboratory) to allow for identification of POMC neurons expressing tdTomato induced by
189
tamoxifen injection. POMC-Cre:ERT2::tdTomato mice were treated daily for 5 days SC with
190
tamoxifen diluted in corn oil 100 mg/kg (T5648, Sigma Aldrich, St. Louis, MO, USA) to induce the
191
Cre recombination in POMC neurons. Mice were randomized into groups (n = 2 – 4/group/strain)
192
before study initiation and injected with either sCTCy5 or rAMYCy5 in the middle of the light period.
193
Both male and female were included in the study (5 males and 2 females from each strain). The
194
selection of dosing regimens was based on pilot studies performed with the fluorescently labelled 9
195
peptides. sCTCy5 100 nmol/kg was injected SC twice with 2 hours in between injections, before
196
termination 2 hours after the last injection. rAMYCy5 100 nmol/kg was injected SC twice with 30
197
minutes in between injections, before termination 10 minutes after the last injection. At termination,
198
mice were perfused with 0.1 mol/L phosphate buffer (PB) followed by 4% paraformaldehyde (PFA)
199
in 0.1 mol/l phosphate buffer (pH 7.2). Brains were post-fixed in 4 % PFA overnight and
200
cryoprotected in 20 % sucrose PB overnight and frozen in hexane before storage at –80°C.
201 202
2.6. Localization of calcitonin receptors on POMC and NPY neurons in the mouse ARC
203
Adult C57BL/6J male mice were purchased from Charles River Laboratories (Lyon, France), housed
204
at temperature-controlled room (23 ± 2°C) on a 12-h light/12-h dark light cycle and fed a low-fat diet
205
(D12450K, Research Diets Inc.). Animals were anaesthetized by isoflurane and were euthanized by
206
cardiac perfusion with a peristaltic pump (NaCl for 2 – 3 minutes and then 10% neutral buffered
207
formalin (NBF) for 10 minutes) (n = 4). The brains were removed and post-fixed in 10% NBF
208
overnight at room temperature.
209 210
2.7. Salmon calcitonin and rat amylin distribution to the whole brain
211
Adult male and female wild type mice (background 129S2/Sv issued from in-house colony) were
212
randomized into groups (n = 3 – 5/group) before study initiation, and injected with saline, sCT750 or
213
rAMYCy7 in the middle of the light period. Dosing regimen was based on findings from the above-
214
described study. sCT750 100 nmol/kg was injected SC twice with 2 hours between injections, before
215
termination at 30 minutes or 2 hours after the last injection. rAMYCy7 300 nmol/kg was injected SC
216
twice with 5 minutes between injections before termination 10 minutes after the last injection. At
217
termination, mice were perfused as described above, brains post-fixed in 4 % PFA for ~1 week,
218
transferred to PBS and stored at 5 °C.
219 10
220
2.8. Analysis of brains from the ARC and hindbrain cell binding study
221
2.8.1.
Brain sectioning
222
sCTCy5 and rAMYCy5 brains were cryosectioned into 20 µm thick coronal sections onto Superfrost
223
Plus slides (Life Technologies Europe, Zug, Switzerland), isolating the ARC and the AP/NTS/Dorsal
224
motor nucleus of the vagus (DMV) (bregma -1.23 mm to -2.53 mm and bregma -7.31 to -7.91 mm in
225
“Paxinos and Franklin's the Mouse Brain in Stereotaxic Coordinates” (Paxinos, 2013), respectively).
226
2.8.2. Immunohistochemistry (IHC)
227
DAPI IHC: All sections were stained with the nuclear dye DAPI (diamidino-2-phenylindole,
228
Thermo Fischer Scientific, Waltham, MA, USA). DAPI was diluted in PBS to a concentration of 25
229
mg/ml, and sections stained. Sections where then rinsed in PBS and coverslipped using
230
VECTASHIELD HardSet mounting medium (Vectorlabs, Servion, Switzerland).
231
POMC IHC: Comparison between POMC IHC and td-Tomato endogenous fluorescence was
232
assessed on slide from POMC-Cre:ERT2:td-tomato mice dosed with sCTCy5. Brain sections were
233
washed with 0.02M Potassium PBS (KPBS) for 30 min and blocked for overnight at 4°C (4% NDS,
234
1% bovine serum albumin (BSA), 0.4% Triton X-100 in KPBS). Sections were then incubated for 48
235
h at 4°C in blocking solution containing rabbit anti-POMC antibody (1:1.000; H-029-30, Phoenix
236
Europe, Karlsruhe, Germany). Following rinses, sections were placed in Alexa-Fluor 488 donkey
237
anti-rabbit for 2 hours (1:200; Jackson ImmunoResearch, Luzern, Switzerland), counterstained with
238
DAPI (Life Technologies Europe) and coverslipped using Vectashield Hardset mounting medium
239
(Vectorlabs, Servion, Switzerland) (Lutz et al., 2018).
240
2.8.3. Image analysis and quantitative analysis of fluorescently labelled cells in the ARC
241
Three sections from the ARC and from the AP/NTS/DMV areas of each animal were acquired using
242
Zeiss SP8 confocal system equipped with a 20X/0.75 objective performing sequential scans (DAPI:
243
HyD1 405, laser 7% with 100% gain; hrGFP: HyD3 552, laser 2% with 100% gain; tdTomato: HyD3
244
552, laser 2% with 20% gain; Cyanin 5: HyD3 638, laser 67% with 100% gain; Alexa Fluor 488: 11
245
HyD3 552, laser 5% with 15% gain; all scans: zoom 1, pinhole 1, Z-stack 20 µm, and step of 0.5
246
µm). Images from selected sections were furthermore acquired with an oil-immersed 63X objective
247
performing sequential tile scans (same settings as above).
248
Co-localization of Cyanin 5 with hrGFP/TdTomato: Image analysis was performed using Imaris
249
9.2.1 software (Bitplane AG, Zürich, Switzerland). The average count/ARC section was calculated
250
from three ARC sections for each animal. Quantification and co-localization of hrGFP/tdTomato
251
with Cyanin5 was performed using the spots function and spot co-localization function. hrGFP+,
252
tdTomato+ and Cyanin 5+ cells were defined as signal positive spots co-localizing or lying right
253
adjacent to a DAPI+ spot (spot size hrGFP/tdTomato/Cyanin 5: 12 µm; spot size DAPI: 8 µm; co-
254
localization threshold: 10 µm). Co-localization of hrGFP+/tdTomato+ cells with Cyanin 5+ cells was
255
defined as ≥40 % overlap of spots (co-localization threshold: 5 µm). Results are reported as average
256
count/section (means) and % co-localization (means).
257
Comparison between POMC IHC and td-tomato: Labelled cells were imaged using an L2 Imager
258
upright microscope (Zeiss, Germany). Images of single and double labelled cells were counted using
259
ImageJ (NIH, Bethesda, MD, USA) to allow for quantification. Three consecutive sections as
260
verified by the DAPI counterstain were used for quantification. To ensure similar imaging conditions
261
for all images, the same microscope set-up and acquisition settings were used to acquire all images
262
within the same experiment. Results are reported as average count/section (means) and % co-
263
localization (means). Co-localization of POMC IHC and sCTCy5 could not be performed as no
264
Cyanin 5 signal was evident after IHC processing.
265 266 267
2.9. Analysis of brains for the localization of calcitonin receptors, NPY and POMC in the ARC
268
Brains were paraffin embedded and the ARC (bregma -1.23 mm to -2.53 mm in “Paxinos and
269
Franklin's the Mouse Brain in Stereotaxic Coordinates” (Paxinos, 2013), respectively) cryosectioned 12
270
into 4.5 µm thick coronal sections. Duplex TSA+-based fluorescent in situ hybridization (ISH) for
271
CTR and POMC or NPY was performed on a Bond RX Fully Automated Research Stainer (Leica
272
Biosystems, Wetzlar, Germany). After bake and dewax (30 minutes at 60ºC) and ISH target retrieval
273
(15 minutes of ACD enzyme protease III and 15 minutes of ER2 protease treatment at 95ºC
274
(Advanced Cell Diagnostics, Newark, CA, USA)), the mRNA signal of interest was detected using
275
the RNAscope® LS Multiplex Fluorescent Reagent Kit (Cat No. 322800, Advanced Cell
276
Diagnostics). Specific probes that target mouse CTR, POMC or NPY (RNAscope® 2.5 LS Probe-
277
Mm-Calcr, Cat No. 494078; RNAscope® 2.5 LS Probe-Mm-Pomc-C2, Cat No. 314088-C2;
278
RNAscope® 2.5 LS Probe- Mm-Npy-C2, Cat No. 313328-C2 , Advanced Cell Diagnostics) were
279
used in combination with TSA+ Cy3 or Cy5 amplification systems, according to the manufacturer’s
280
instructions (Advanced Cell Diagnostics; PerkinElmer, Waltham, WA, USA).The slides were
281
counterstained with DAPI (Advanced Cell Diagnostics) and mounted in Fluorescent mounting
282
medium (S3023, Dako, Glostrup, Denmark) before being scanned in an Olympus VS120 slide
283
scanner using a 20x (NA 0.75, 0.33 µm/pixel) objective. 2 – 3 ARC sections were evaluated from
284
each mouse.
285 286
2.10. Analysis of brains from the whole brain distribution study
287
2.10.1. Tissue clearing and whole brain light sheet fluorescence microscopy (LSFM)
288
The sCT750 and rAMYCy7 brains were cleared using a modification of the tetrahydrofuran (THF)
289
clearing protocol (Ertürk et al., 2012). Briefly, brain tissue was dehydrated in THF diluted in dH2O
290
(w/v) 30/50/70/80/96%/2 x 100 % 6 – 12 hours for each step. The brains were subsequently cleared
291
in diBenzylether (DBE) for 6 hours in room temperature. Brains were imaged by LSFM. Image
292
stacks (16-bit tif) were acquired using the UltraMicroscope I or II LSFM system (LaVision Biotec,
293
Bielefeld, Germany) equipped with an Andor Neo 5.5 scmos camera (Andor Technology Ltd.,
294
Belfast, UK) and a SuperK Extreme EXR-15 laser (NKT Photonics, Birkerød, Denmark), with the 13
295
ImSpectorPro software (Lavision Biotec). Step size was set to 10 µm with a 0.8 X magnification.
296
Data acquisition was performed using a 545/25 nm excitation filter and 605/70 nm emission filter for
297
imaging auto-fluorescence and a 710/75 nm excitation filter and a 775/40 nm emission filter for
298
imaging specific signals (sCT750 and rAMYCy7). For quantification of signal intensity, images were
299
captured from all brains by the UltraMicroscope 1, and high-resolution images were captured from
300
selected brains with the UltraMicroscope II.
301
2.10.2. Quantification of brain distribution
302
Image analysis was performed utilizing the Imaris 7.6.5 software (Bitplane AG). Before
303
quantification of brain distribution, spectral unmixing was performed as described earlier (Salinas et
304
al., 2018) to minimize the contribution of tissue auto-fluorescence to the specific sCT750 and
305
rAMYCy7 distribution signals. In brief, the estimated auto-fluorescence contribution in the specific
306
channel was calculated and removed based on ratios of voxel intensities between selected voxels in
307
the unspecific channel, and corresponding voxel in the specific channel. A ratio was computed for 40
308
sets of voxels selected from the histogram of the unspecific channel. To determine regions which
309
displayed consistent specific signal, each brain was mapped to an integrated brain atlas and average
310
3D signals combining individual brain samples constructed for each group. Quantification of specific
311
(sCT750 and rAMYCy7) signal intensity in the ARC/ME and AP was performed by manual
312
segmentation of regions with the contour surfaces function in Imaris 7.6.5 (Bitplane AG). The total
313
signal intensity within each region of interest from the specific unmix channel was utilized for
314
analysis. The results are reported as fold changes (means ± SEM).
315 316
2.11. Statistical analysis
317
Differences in cumulative food intake over time in the validation of fluorophore conjugated peptides
318
study were tested with repeated measures ANOVA with an autocorrelation matrix, followed by a
319
Tukey’s post hoc test using SAS enterprise Guide 7.1 software (SAS Institute Inc., Cary, NC, USA). 14
320
Differences between groups in specific signal intensity was tested using a Kruskal Wallis analysis
321
followed by a Dunn’s multiple comparisons test or Mann Whitney test using GraphPad Prism 7
322
software (GraphPad Software, La Jolla, CA, USA). A p-value less than 0.05 was considered
323
statistically significant.
15
324
3. RESULTS
325 326
3.1. In vitro potency of fluorescently labelled peptides
327
To ensure that the fluorophore conjugation did not affect the receptor activation properties of rat
328
amylin and salmon calcitonin, in vitro potency assays were undertaken.
329
In cells transfected with the human amylin receptor 3, sCT750 and sCTCy5 displayed EC50-values in a
330
range similar to that of native sCT (Table 1, Suppl. Table 1, Suppl. Fig. 1) whereas rAMYCy5
331
presented a decreased potency compared to pramlintide even though it did not reach significance
332
(Suppl. Table 1). In cells transfected with the human calcitonin receptor, sCT750 and sCTCy5 also
333
displayed EC50-values comparable to that of native sCT, whereas rAMYCy5 and pramlintide
334
expectedly displayed much lower potencies since this cell type did not express RAMP3 (Table 1,
335
Suppl. Table 1).
336 337
3.2. Fluorescently labelled peptides retain anorectic efficacy
338
To ensure that fluorophore conjugation did not eliminate the anorectic properties of rat amylin and
339
salmon calcitonin, acute food intake measurements were compared between fluorescently labelled
340
and native compounds. sCTCy5 and rAMYCy5 had similar effects on suppressing food intake as the
341
native peptides, demonstrating intact in vivo efficacy (Fig 1A and B). Likewise, the efficacy of
342
sCT750 and rAMYCy7 to suppress food intake was on par with that of the native compounds, although
343
the effect of sCT750 was attenuated compared to native salmon calcitonin towards the end of the dark
344
phase (Fig 1C – D). Hence, the acute in vivo efficacy of the compounds was maintained, or slightly
345
attenuated, after fluorescent label conjugation.
346
16
347
3.3. Salmon calcitonin is internalized into a subset of ARC NPY, AP and DMV cells
348
Evaluation of ARC sections from hrGFP-NPY mice treated with sCTCy5 and terminated 2 hours after
349
the last injection revealed that an average of approximately 16 cells/section were sCTCy5 positive in
350
the sCTCy5 group. Furthermore, the sCTCy5 signal was found to be internalized into ARC cells. When
351
evaluated for co-localization with NPY, co-labelling for both sCTCy5 and NPY was observed in 7.5%
352
of all NPY+ neurons and 47 % of all sCTCy5 cells (Fig 2A, B, D and F). Similarly, on average about
353
14 cells/section from the ARC of POMC-Cre:ERT2::tdTomato mice were sCTCy5 positive 2 hours
354
after the last injection, but no co-localization with POMC positive neurons were identified (Fig 2C,
355
G and H). No rAMYCy5 positive cells were identified in either the hrGFP-NPY or POMC-
356
Cre:ERT2::td-tomato mice treated with rAMYCy5 and terminated 10 min post last injection (Fig 2A,
357
B, C and E).
358
Since a POMC-Cre:ERT2::td-tomato mouse model was used, a comparison between POMC td-
359
tomato expression and POMC IHC was performed. The comparison between endogenous POMC in
360
POMC-Cre:ERT2::td-tomato mouse and POMC IHC revealed that the number of POMC neurons was
361
similar between the two stainings, however, the percentage of co-localization of POMC IHC and
362
POMC::td-tomato was around 65-70% suggesting discrepancies between the two POMC labelling
363
techniques similar to what has been shown previously (Padilla et al., 2012; Rau et al., 2018) (Fig 3A
364
– C). No Cy5 fluorescent signal was preserved after POMC IHC staining procedure, whereby co-
365
localization between sCTCy5 and POMC IHC could not be performed. Earlier single cell sequencing
366
investigations suggest that POMC neurons only express low levels of the CTRs in the ARC and ME
367
unlike NPY neurons which display high CTR expression levels (Campbell et al., 2017; Lam et al.,
368
2017; Pan et al., 2018). We therefore investigated whether expression of the CTR co-localized with
369
POMC or NPY expressing neurons in sections from wild type mice. In line with earlier observations,
370
only few 25% of POMC neurons displayed CTR expression, while almost all NPY neurons had
371
abundant CTR expression (Fig 4A – C). 17
372
AP/NTS/DMV sections from hrGFP-NPY and POMC-Cre:ERT2::td-tomato mice treated with sCTCy5
373
or rAMYCy5 were also evaluated for binding to single cells. sCTCy5 positive cells were identified in
374
all AP sections from both mouse strains 2 hours post last injection, with the sCTCy5 being
375
internalized into cells (Fig 5A). No sCTCy5 signal was found in the NTS of any mice, but intra-
376
cellular sCTCy5 was identified in the DMV of most of sections from both mouse strains albeit with
377
lower signal intensity compared to the AP (Fig 5B). No rAMYCy5 positive cells were found in any
378
sections from either mouse strains 10 min after last injection (Fig 5C). This absence of rAMYCy5
379
signal may be explained by the lower in vitro potency compared to pramlintide or sCTCy5 (Table 1).
380 381 382
3.4. Whole brain 3D imaging show that salmon calcitonin and rat amylin distribute into the ARC, AP and OVLT of mouse brain
383
sCT750 was injected into wild type mice twice and brains collected 30 min and 2 hours post injection
384
after which the whole brain was scanned with LSFM. When evaluating the brains, a sCT750 signal
385
was consistently found in the ARC/ME, AP and OVLT both 30 minutes and 2 hours post injection
386
(Fig 6A – B). Supplementary video 6-1 is of a 3D reconstructed whole brain of a mouse
387
summarizing how sCT750 distributes in the brain. No signal was observed in the NTS or the DMV of
388
the hindbrain. sCT750 signal specks were also observed throughout the brain tissue indicative of
389
sCT750 bound to blood vessels. When the total sCT750 signal was quantified in the ARC/ME and AP
390
nuclei, a significantly higher signal was found 30 minutes post injection vs the control group (p<0.05
391
in ARC/ME, p<0.01 in AP) with the signal not significantly increased 2 hours post injection for both
392
regions (Fig 6C – F). sCT750 distribution did not change from 30 minutes to 2 hours post injection.
393
Notably, in the hypothalamus, the majority of the sCT750 signal was found in the medio-basal ARC
394
with only specs present in the ME, but the regions could not be separated in signal intensity analysis
395
(Fig 6D). Supplementary video 6-2 summarizes sCT750 distribution in ARC coronal sections.
18
396
As no signal was observed in the ARC or AP after rAMYCy5 treatment, a tripled dose of rAMYCy7
397
and shortened time interval between injections (5 rather than 30 min) of rAMYCy7 was used for the
398
whole brain distribution study. Ten minutes after the final injection of rAMYCy7 in WT mice, the
399
rAMYCy7 signal was observed in the ARC, ME, AP and OVLT of the brains. Again, rAMYCy7 signal
400
specs were observed in the brain tissue and in the choroid plexus of the brains indicating presence of
401
fluorescently labelled peptide in the vasculature. Quantification of total signal intensity in the
402
ARC/ME and the AP revealed a significantly higher signal in both regions relative to controls 10 min
403
post final injection (p<0.05 in both regions) (Fig 6G – J). Unlike the sCT750, a distinct rAMYCy7
404
signal was observed in the ME along with signal in the medio-basal ARC (Fig 6H). Supplementary
405
video 6-3 summarizes rAMYCy7 distribution in ARC coronal sections. Of note, capture of high-
406
resolution images of rAMYCy7 was not possible, as Cy7 is highly sensitive to light-exposure induced
407
degradation – as such the signal was abolished after the initial scan of the brains.
408
19
409
4. DISCUSSION
410
Our studies demonstrate that salmon calcitonin binds and internalizes into a subset of ARC NPY
411
neurons and unidentified cells in the ARC, AP and DMV of mice. A signal indicating internalization
412
into POMC neurons was not evident in POMC-Cre:ERT2::td-tomato mice. Furthermore, when
413
examining distribution into whole brains, salmon calcitonin distributed into the AP, OVLT and ARC
414
of mice 30 minutes and 2 hours after injection. While rat amylin was not found to bind to single
415
neurons in the ARC or hindbrain at equivalent doses as salmon calcitonin under current study
416
conditions, rat amylin showed a similar distribution pattern to salmon calcitonin in the whole brain
417
visualized by LSFM.
418
Salmon calcitonin has been found to internalize into cells expressing CTR in vitro (Houssami et al.,
419
1994), but our observation is the first to establish that salmon calcitonin also displays this property in
420
vivo directly targeting ARC NPY/AgRP neurons. Earlier studies have found that salmon calcitonin
421
activates NPY/AgRP neurons (Pan et al., 2018) and that amylin acutely suppresses the activity of
422
this neuronal population in vivo (Su et al., 2017). We have previously shown that pERK signaling in
423
ARC NPY neurons is unaltered after acute amylin treatment (Lutz et al., 2018) which indicates that
424
salmon calcitonin activates and/or suppresses other signaling pathways to elicit effects within this
425
neuronal population. The ARC NPY/AgRP neurons co-expressing leptin receptors and CTRs were
426
recently reported to be important for leptin-mediated metabolic effects since the deletion of leptin
427
receptors from these neurons results in hyperphagia and weight gain (Pan et al., 2018). Thus, the
428
NPY/AgRP neurons targeted by salmon calcitonin in our findings may include a subpopulation of
429
leptin receptor/CTR co-expressing NPY/AgRP neurons that potentially play a role in in leptin/amylin
430
synergism. Nonetheless, an inhibitory effect has not been identified on ARC NPY or AgRP gene
431
expression levels after acute or sub-chronic AMY /CTR agonist treatment in rats (Barth et al., 2003;
432
Le Foll et al., 2015; Roth et al., 2006) suggesting that AMY /CTR mediated satiety may not be a
433
result of direct effects on NPY or AgRP signaling in this neuronal population. However, it has been 20
434
reported that LPBN calcitonin gene related peptide (CGRP) neurons are activated by amylin, and
435
receive tonic GABAergic (γ-aminobutyric acid) inhibitory input from NPY/AgRP neurons in the
436
ARC (Carter et al., 2013; Wu et al., 2009). This suggests that the action of salmon calcitonin on
437
NPY/AgRP neurons with high CTR expression levels may be to regulate GABA levels, a
438
neurotransmitter that is co-expressed with NPY and AgRP in these neurons (Campbell et al., 2017).
439
Thus, AMY /CTR activation by salmon calcitonin on NPY/AgRP neurons may result in direct
440
inhibition of GABAergic input from ARC NPY/AgRP neurons to the LPBN or other neuronal
441
targets. Further studies are needed to ascertain the downstream targets of salmon calcitonin effects
442
on NPY/AgRP neurons.
443
We did not observe any sCTCy5 binding or internalization into ARC POMC neurons of POMC-
444
Cre:ERT2::tdTomato mice, suggesting that they may not be directly targeted by AMY /CTR agonists.
445
In support of this finding, Pan et al. found that salmon calcitonin only activates a low number of
446
ARC POMC neurons (Pan et al., 2018), and in line with earlier studies, we found that few POMC
447
neurons in the ARC displayed CTR expression. This suggests that earlier observed action of amylin
448
inducing intracellular pERK in POMC neurons and increasing ARC POMC expression (Lutz et al.,
449
2018; Roth et al., 2006) may be mediated via action upon other cell populations. AMY /CTR
450
agonists might indirectly activate IL-6 receptor expressing POMC neurons by inducing IL-6
451
production from hypothalamic microglia (Le Foll et al., 2015; Ropelle et al., 2010) or by suppressing
452
inhibitory NPY and GABAergic projections from NPY/AgRP neurons onto POMC neurons (Cowley
453
et al., 2001). However, as only 65-70% of POMC neurons were double labelled for IHC and
454
tdTomato it cannot be rejected that salmon calcitonin might target a subset of POMC neurons we did
455
not identify. Further, due to the labile nature of the sCTCy5 signal, co-localization with POMC-IHC
456
could not be performed. Considering that sCTCy5 was also found to be internalized into a subset of
457
unknown cells in the ARC, we performed Iba1 and GFAP IHC on ARC sections from mice treated
458
with sCTCy5 to test if microglia or astrocytes might be targeted. Unfortunately, the Cyanin 5 21
459
fluorescent signal was not visible after IHC processing and the unknown target cell population was
460
not identified. Furthermore, targeting of neurons producing other neuropeptides cannot be excluded.
461
Despite similar reported half-lives, we only managed to visualize rat amylin within the brain by
462
using a threefold higher dose compared to salmon calcitonin, combined with shorter dosing interval
463
and time to tissue sampling. Salmon calcitonin has been found to bind irreversibly to the CTR (Dal
464
Maso et al., 2018; Gingell et al., 2019; Hilton et al., 2000; Houssami et al., 1994), but this has not
465
been shown for amylin. Hence, rapid dissociation from AMY might diminish internalization
466
potential for amylin, whereby amylin is rapidly broken down and cleared from the extracellular
467
milieu. Rat amylin and salmon calcitonin on the other hand displayed similar distribution patterns
468
within the brain, suggesting that differential distribution does not underlie the observed
469
pharmacodynamic differences on acute food intake suppression. Further, it is likely that amylin
470
targets the same ARC neuronal populations as salmon calcitonin without being internalized. Thus,
471
salmon calcitonin prolonged effect is most likely due to longer residence time at the receptor
472
inducing long-lasting intracellular signaling which has been observed in vitro (Lamp et al., 1981).
473
One caveat of this study was that labelled amylin was not injected in NPY-GFP and POMC-
474
Cre:ERT2::tdTomato mice using the same paradigm as for the whole brain studies. Furthermore, the
475
in vitro potency assay highlighted the fact that rAMYCy5 displayed lower potency than pramlintide
476
and sCTCy5 at the CTR and AMY. These discrepancies in study design and receptor kinetics could
477
underlie the lack of amylin signal in NPY and POMC neurons. Thus, amylin internalization into
478
NPY and POMC neurons cannot be completely rejected based on our findings.
479
Both fluorescently labelled salmon calcitonin and rat amylin distributed into the AP, ARC and
480
OVLT. Alternatively, both rat amylin and salmon calcitonin might gain access to the ARC from the
481
ME via fenestrated capillaries near the ME-ARC border (Rodríguez et al., 2010). Salmon calcitonin
482
was not observed within the ME, while rat amylin was detected at the shorter 10-minute sampling
483
interval. Further, even though we did observe CTR expression in the ME of mice we did not observe 22
484
sCTCy5 internalized into ME cells, and it is unclear why the differential distribution at the ME
485
between sCT and amylin occurs and whether sampling at the shorter interval of 10 minutes would
486
have enabled detection of the salmon calcitonin signal.
487
While our studies confirm that AMY /CTR agonists bind to single cells in the AP, we also found
488
distribution into the OVLT. The OVLT is reported to regulate fluid balance and tonicity (Kaur and
489
Ling, 2017; McKinley et al., 2019), primarily by circulating hormones such as vasopressin and
490
angiotensin II. Several hormones that affect energy balance, have also been reported to influence
491
fluid intake (e.g. GLP-1, PYY CCK and amylin) (Zimmerman et al., 2017). Angiotensin II and
492
amylin has been found to increase water intake by activating the same neuronal population as
493
angiotensin II in the SFO (Riediger et al., 1999). It is thus plausible that AMY /CTRs display a
494
similar action in the OVLT.
495
Salmon calcitonin was also consistently found to be internalized into single DMV cells. However,
496
Sexton et al. reported low to undetectable radioactive amylin binding to the DMV of rat brain
497
sections and no other reports of CTRs in the DMV can be found (Sexton et al., 1994). Further, in
498
vitro studies suggest that calcitonin internalization is CTR mediated (Gingell et al., 2019). In view of
499
this and the lack of sCT750 signal from the DMV in the whole brain study, the significance of this
500
finding is uncertain. High doses of fluorescently labelled salmon calcitonin were utilized in this
501
study, whereby nonspecific binding to the DMV cannot be rejected. However, AMY /CTR agonist
502
have been found to reduce gastric emptying (Andreassen et al., 2014; Reidelberger et al., 2001;
503
Reidelberger et al., 2002), and the mechanism is thought to be centrally mediated being in part
504
dependent on both an intact AP and vagal nerve efferent signaling (Edwards et al., 1998; Wickbom
505
et al., 2008; Young, 2005). Thus, salmon calcitonin might in part modulate gastric motility by
506
binding directly to cells in the DMV, however, functional evidence to support this connection is
507
needed.
23
508
In conclusion, we find that salmon calcitonin distributes to the ARC, targeting and internalizing into
509
NPY/AgRP neurons but not POMC neurons. Further, rat amylin and salmon calcitonin display
510
similar distribution patterns within the mouse brain distributing to the ARC, AP and OVLT,
511
indicating that the underlying difference in their acute anorectic efficacy is due to salmon
512
calcitonin’s ability to be internalized into target cells rather than differential brain distribution.
513 514
5. CONCLUSION
515 516
Analogues of satiety-inducing hormones such as amylin/calcitonin receptor agonists that can act on
517
cognate receptors in the brain to induce beneficial effects on glucose homeostasis and body weight
518
loss are being developed for the treatment of metabolic diseases such as obesity and type 2 diabetes.
519
Earlier studies indicated that AMY /CTR agonists interact with regions of the brain that regulate
520
satiety and homeostatic energy balance, however, it remains unclear to what extent these peptides
521
directly interact with target neuronal populations. Our findings serve to clarify and envision how
522
peripherally injected AMY /CTR agonists target specific neuronal populations to induce their
523
metabolic effects.
524
24
525
ACKNOWLEDGEMENTS:
526
This work was supported by Novo Nordisk A/S, the Swiss National Science Foundation (SNF
527
31003A_175458) to TAL and by the Lifepharm Centre for In Vivo Pharmacology. We would like to
528
thank Wouter Hogendorf his help with the chemistry of the fluorescently labelled peptides along
529
with Lavinia Boccia, Bernd Coester, Salome Gamakharia and Stine Normann Hansen for assistance
530
with experimental work and analytical procedures.
531 532
Conflict of interest statement: KR, LMJ, SL, TG and AS are full-time employees of Novo Nordisk
533
and hold minor share portions as part of their employment. HLZ, JL, TL and CLF declare no
534
competing financial interests.
535
25
536
REFERENCES
537
Abbott, N. J., Patabendige, A. A., Dolman, D. E., Yusof, S. R., Begley, D. J., 2010. Structure and
538
function of the blood–brain barrier. Neurobiol. Dis. 37, 13-25.
539
Andreassen, K. V., Feigh, M., Hjuler, S. T., Gydesen, S., Henriksen, J. E., Beck-Nielsen, H.,
540
Christiansen, C., Karsdal, M. A., Henriksen, K., 2014. A novel oral dual amylin and calcitonin
541
receptor agonist (KBP-042) exerts antiobesity and antidiabetic effects in rats. Am. J. Physiol.
542
Endocrinol. Metab. 307, E24-E33.
543
Barth, S. W., Riediger, T., Lutz, T. A., Rechkemmer, G., 2003. Differential effects of amylin and
544
salmon calcitonin on neuropeptide gene expression in the lateral hypothalamic area and the
545
arcuate nucleus of the rat. Neurosci. Lett. 341, 131-134.
546 547
Becskei, C., Riediger, T., Zünd, D., Wookey, P., Lutz, T. A., 2004. Immunohistochemical mapping of calcitonin receptors in the adult rat brain. Brain Res. 1030, 221-233.
548
Berglund, E. D., Liu, C., Sohn, J.-W., Liu, T., Kim, M. H., Lee, C. E., Vianna, C. R., Williams, K.
549
W., Xu, Y., Elmquist, J. K., 2013. Serotonin 2C receptors in pro-opiomelanocortin neurons
550
regulate energy and glucose homeostasis. J. Clin. Invest. 123, 5061-5070.
551
Braegger, F. E., Asarian, L., Dahl, K., Lutz, T. A., Boyle, C. N., 2014. The role of the area postrema
552
in the anorectic effects of amylin and salmon calcitonin: behavioral and neuronal phenotyping.
553
Eur. J. Neurosci. 40, 3055-3066.
554
Campbell, J. N., Macosko, E. Z., Fenselau, H., Pers, T. H., Lyubetskaya, A., Tenen, D., Goldman,
555
M., Verstegen, A. M., Resch, J. M., McCarroll, S. A., 2017. A molecular census of arcuate
556
hypothalamus and median eminence cell types. Nat. Neurosci. 20, 484.
557 558
Carter, M. E., Soden, M. E., Zweifel, L. S., Palmiter, R. D., 2013. Genetic identification of a neural circuit that suppresses appetite. Nature 503, 111-+.
26
559
Chaturvedula, A., Joshi, D. P., Anderson, C., Morris, R. L., Sembrowich, W. L., Banga, A. K., 2005.
560
In vivo iontophoretic delivery and pharmacokinetics of salmon calcitonin. Int. J. Pharm. 297,
561
190-196.
562
Christopoulos, G., Perry, K. J., Morfis, M., Tilakaratne, N., Gao, Y. Y., Fraser, N. J., Main, M. J.,
563
Foord, S. M., Sexton, P. M., 1999. Multiple Amylin Receptors Arise from Receptor Activity-
564
Modifying Protein Interaction with the Calcitonin Receptor Gene Product. Mol. Pharmacol. 56,
565
235-242.
566
Cowley, M. A., Smart, J. L., Rubinstein, M., Cerdán, M. G., Diano, S., Horvath, T. L., Cone, R. D.,
567
Low, M. J., 2001. Leptin activates anorexigenic POMC neurons through a neural network in
568
the arcuate nucleus. Nature 411, 480-484.
569
Dal Maso, E., Just, R., Hick, C., Christopoulos, A., Sexton, P. M., Wootten, D., Furness, S. G., 2018.
570
Characterization of signalling and regulation of common calcitonin receptor splice variants and
571
polymorphisms. Biochem. Pharmacol. 148, 111-129.
572
Dunn-Meynell, A. A., Le Foll, C., Johnson, M. D., Lutz, T. A., Hayes, M. R., Levin, B. E., 2016.
573
Endogenous VMH amylin signaling is required for full leptin signaling and protection from
574
diet-induced obesity. Am. J. Physiol. Regul. Integr. Comp. Physiol. 310, R355-R365.
575
Edwards, G., Gedulin, B., Jodka, C., Dilts, R., Miller, C., Young, A., 1998. Area postrema (AP)-
576
lesions block the regulation of gastric emptying by amylin. Gastroenterology 114, A748.
577
Ertürk, A., Becker, K., Jährling, N., Mauch, C. P., Hojer, C. D., Egen, J. G., Hellal, F., Bradke, F.,
578
Sheng, M., Dodt, H.-U., 2012. Three-dimensional imaging of solvent-cleared organs using
579
3DISCO. Nat. Protoc. 7, 1983.
580
Feigh, M., Henriksen, K., Andreassen, K. V., Hansen, C., Henriksen, J. E., Beck-Nielsen, H.,
581
Christiansen, C., Karsdal, M. A., 2011. A novel oral form of salmon calcitonin improves
582
glucose homeostasis and reduces body weight in diet-induced obese rats. Diabetes Obes.
583
Metab. 13, 911-920. 27
584 585
Gingell, J. J., Hendrikse, E. R., Hay, D. L., 2019. New Insights into the Regulation of CGRP-Family Receptors. Trends Pharmacol. Sci. 40, 71-83.
586
Hilton, J. M., Chai, S. Y., Sexton, P. M., 1995. IN VITRO AUTORADIOGRAPHIC
587
LOCALIZATION OF THE CALCITONIN RECEPTOR ISOFORMS, C1a AND C1b, IN
588
RAT BRAIN. Neuroscience 69, 1223-1237.
589
Hilton, J. M., Dowton, M., Houssami, S., Sexton, P. M., 2000. Identification of key components in
590
the irreversibility of salmon calcitonin binding to calcitonin receptors. J. Endocrinol. 166, 213-
591
226.
592
Houssami, S., Findlay, D. M., Brady, C. L., Myers, D. E., Martin, T. J., Sexton, P. M., 1994.
593
Isoforms of the Rat Calcitonin Receptor: Consequences for Ligand Binding and Signal
594
Transduction. Endocrinology 135, 183-190.
595
Kaur, C., Ling, E.-A., 2017. The circumventricular organs. Histol. Histopathol. 32, 879-892.
596
Lam, B. Y., Cimino, I., Polex-Wolf, J., Kohnke, S. N., Rimmington, D., Iyemere, V., Heeley, N.,
597
Cossetti, C., Schulte, R., Saraiva, L. R., 2017. Heterogeneity of hypothalamic pro-
598
opiomelanocortin-expressing neurons revealed by single-cell RNA sequencing. Mol. Metab. 6,
599
383-392.
600
Lamp, S. J., Findlay, D. M., Moseley, J. M., Martin, T. J., 1981. Calcitonin Induction of a Persistent
601
Activated State of Adenylate Cyclase in Human Breast Cancer Cells (T 47D). J. Biol. Chem.
602
256, 2269-2274.
603
Le Foll, C., Johnson, M. D., Dunn-Meynell, A. A., Boyle, C. N., Lutz, T. A., Levin, B. E., 2015.
604
Amylin-Induced Central IL-6 Production Enhances Ventromedial Hypothalamic Leptin
605
Signaling. Diabetes 64, 1621-1631.
606 607
Loh, K., Herzog, H., Shi, Y. C., 2015. Regulation of energy homeostasis by the NPY system. Trends Endocrinol. Metab. 26, 125-135.
28
608
Lutz, T., Mollet, A., Rushing, P., Riediger, T., Scharrer, E., 2001. The anorectic effect of a chronic
609
peripheral infusion of amylin is abolished in area postrema/nucleus of the solitary tract
610
(AP/NTS) lesioned rats. Int. J. Obes. 25, 1005.
611
Lutz, T. A., Coester, B., Whiting, L., Dunn-Meynell, A. A., Boyle, C. N., Bouret, S. G., Levin, B. E.,
612
Le Foll, C., 2018. Amylin Selectively Signals Onto POMC Neurons in the Arcuate Nucleus of
613
the Hypothalamus. Diabetes 67, 805-817.
614 615
Lutz, T. A., Geary, N., Szabady, M. M., Delprete, E., Scharrer, E., 1995. Amylin Decreases Meal Size in Rats. Physiol. Behav. 58, 1197-1202.
616
McKinley, M. J., Denton, D. A., Ryan, P. J., Yao, S. T., Stefanidis, A., Oldfield, B. J., 2019. From
617
sensory circumventricular organs to cerebral cortex: Neural pathways controlling thirst and
618
hunger. J. Neuroendocrinol., e12689.
619
Nakamoto, H., Soeda, Y., Takami, S., Minami, M., Satoh, M., 2000. Localization of calcitonin
620
receptor mRNA in the mouse brain: coexistence with serotonin transporter mRNA. Brain Res.
621
Mol. Brain Res. 76, 93-102.
622
Oliver, K. R., Kane, S. A., Salvatore, C. A., Mallee, J. J., Kinsey, A. M., Koblan, K. S., Keyvan-
623
Fouladi, N., Heavens, R. P., Wainwright, A., Jacobson, M., Dickerson, I. M., Hill, R. G., 2001.
624
Cloning, characterization and distribution of receptor activity central nervous system
625
modifying proteins in the rat. Eur. J. Neurosci. 14, 618-628.
626
Padilla, S. L., Reef, D., Zeltser, L. M., 2012. Defining POMC neurons using transgenic reagents:
627
impact of transient Pomc expression in diverse immature neuronal populations. Endocrinology
628
153, 1219-1231.
629
Pan, W., Adams, J. M., Allison, M. B., Patterson, C., Flak, J. N., Jones, J., Strohbehn, G., Trevaskis,
630
J., Rhodes, C. J., Olson, D. P., 2018. Essential Role for Hypothalamic Calcitonin Receptor‒
631
Expressing Neurons in the Control of Food Intake by Leptin. Endocrinology 159, 1860-1872.
29
632 633 634 635
Paxinos, G., 2013. Paxinos and Franklin's the mouse brain in stereotaxic coordinates. In: Franklin, K. B. J., (Ed). Elsevier/Academic Press, Amsterdam. Potes, C. S., Lutz, T. A., Riediger, T., 2010. Identification of central projections from amylinactivated neurons to the lateral hypothalamus. Brain Res. 1334, 31-44.
636
Poyner, D. R., Sexton, P. M., Marshall, I., Smith, D. M., Quirion, R., Born, W., Muff, R., Fischer, J.
637
A., Foord, S. M., 2002. International Union of Pharmacology. XXXII. The mammalian
638
calcitonin gene-related peptides, adrenomedullin, amylin, and calcitonin receptors. Pharmacol.
639
Rev. 54, 233-246.
640
Rau, A. R., Hughes, A. R., Hentges, S. T., 2018. Various transgenic mouse lines to study
641
proopiomelanocortin cells in the brain stem label disparate populations of GABAergic and
642
glutamatergic neurons. Am. J. Physiol. Regul. Integr. Comp. Physiol. 315, R144-R152.
643
Reidelberger, R. D., Arnelo, U., Granqvist, L., Permert, J., 2001. Comparative effects of amylin and
644
cholecystokinin on food intake and gastric emptying in rats. Am. J. Physiol. Regul. Integr.
645
Comp. Physiol. 280, R605-R611.
646
Reidelberger, R. D., Kelsey, L., Heimann, D., 2002. Effects of amylin-related peptides on food
647
intake, meal patterns, and gastric emptying in rats. Am. J. Physiol. Regul. Integr. Comp.
648
Physiol. 282, R1395-R1404.
649 650
Riediger, T., Rauch, M., Schmid, H. A., 1999. Actions of amylin on subfornical organ neurons and on drinking behavior in rats. Am. J. Physiol. Regul. Integr. Comp. Physiol. 276, R514-R521.
651
Riediger, T., Zuend, D., Becskei, C., Lutz, T. A., 2004. The anorectic hormone amylin contributes to
652
feeding-related changes of neuronal activity in key structures of the gut-brain axis. Am. J.
653
Physiol. Regul. Integr. Comp. Physiol. 286, R114-R122.
654
Rodríguez, E. M., Blázquez, J. L., Guerra, M., 2010. The design of barriers in the hypothalamus
655
allows the median eminence and the arcuate nucleus to enjoy private milieus: the former opens
656
to the portal blood and the latter to the cerebrospinal fluid. Peptides 31, 757-776. 30
657
Ropelle, E. R., Flores, M. B., Cintra, D. E., Rocha, G. Z., Pauli, J. R., Morari, J., de Souza, C. T.,
658
Moraes, J. C., Prada, P. O., Guadagnini, D., 2010. IL-6 and IL-10 anti-inflammatory activity
659
links exercise to hypothalamic insulin and leptin sensitivity through IKKβ and ER stress
660
inhibition. PLoS Biol. 8, e1000465.
661
Roth, J. D., Coffey, T., Jodka, C. M., Maier, H., Athanacio, J. R., Mack, C. M., Weyer, C., Parkes,
662
D. G., 2007. Combination therapy with amylin and peptide YY 3-36 in obese rodents:
663
Anorexigenic synergy and weight loss additivity. Endocrinology 148, 6054-6061.
664
Roth, J. D., Hughes, H., Kendall, E., Baron, A. D., Anderson, C. M., 2006. Antiobesity effects of the
665
beta-cell hormone amylin in diet-induced obese rats: Effects on food intake, body weight,
666
composition, energy expenditure, and gene expression. Endocrinology 147, 5855-5864.
667
Salinas, C. B. G., Lu, T. T.-H., Gabery, S., Marstal, K., Alanentalo, T., Mercer, A. J., Cornea, A.,
668
Conradsen, K., Hecksher-Sørensen, J., Dahl, A. B., 2018. Integrated brain atlas for unbiased
669
mapping of nervous system effects following liraglutide treatment. Sci. Rep. 8, 10310.
670
Schaeffer, M., Langlet, F., Lafont, C., Molino, F., Hodson, D. J., Roux, T., Lamarque, L., Verdié, P.,
671
Bourrier, E., Dehouck, B., 2013. Rapid sensing of circulating ghrelin by hypothalamic
672
appetite-modifying neurons. Proc. Natl. Acad. Sci. U. S. A. 110, 1512-1517.
673 674 675 676 677
Sexton, P., Paxinos, G., Kenney, M., Wookey, P., Beaumont, K., 1994. In vitro autoradiographic localization of amylin binding sites in rat brain. Neuroscience 62, 553-567. Su, Z., Alhadeff, A. L., Betley, J. N., 2017. Nutritive, post-ingestive signals are the primary regulators of AgRP neuron activity. Cell Rep. 21, 2724-2736. Tilakaratne, N., Christopoulos, G., Zumpe, E. T., Foord, S. M., Sexton, P. M., 2000. Amylin receptor
678
phenotypes
derived
from
human
calcitonin
receptor/RAMP
coexpression
exhibit
679
pharmacological differences dependent on receptor isoform and host cell environment. J.
680
Pharmacol. Exp. Ther. 294, 61-72.
31
681
Trevaskis, J. L., Turek, V. F., Griffin, P. S., Wittmer, C., Parkes, D. G., Roth, J. D., 2010. Multi-
682
hormonal weight loss combinations in diet-induced obese rats: Therapeutic potential of
683
cholecystokinin? Physiol. Behav. 100, 187-195.
684
Turek, V. F., Trevaskis, J. L., Levin, B. E., Dunn-Meynell, A. A., Irani, B., Gu, G. B., Wittmer, C.,
685
Griffin, P. S., Vu, C., Parkes, D. G., Roth, J. D., 2010. Mechanisms of Amylin/Leptin Synergy
686
in Rodent Models. Endocrinology 151, 143-152.
687
Whiting, L., McCutcheon, J. E., Boyle, C. N., Roitman, M. F., Lutz, T. A., 2017. The area postrema
688
(AP) and the parabrachial nucleus (PBN) are important sites for salmon calcitonin (sCT) to
689
decrease evoked phasic dopamine release in the nucleus accumbens (NAc). Physiol. Behav.
690
176, 9-16.
691
Wickbom, J., Herrington, M. K., Permert, J., Jansson, A., Arnelo, U., 2008. Gastric emptying in
692
response to IAPP and CCK in rats with subdiaphragmatic afferent vagotomy. Regul. Pept. 148,
693
21-25.
694 695 696 697
Wu, Q., Boyle, M. P., Palmiter, R. D., 2009. Loss of GABAergic signaling by AgRP neurons to the parabrachial nucleus leads to starvation. Cell 137, 1225-1234. Yeo, G. S. H., Heisler, L. K., 2012. Unraveling the brain regulation of appetite: lessons from genetics. Nat. Neurosci. 15, 1343-1349.
698
Young, A., 2005. Inhibition of gastric emptying. Adv. Pharmacol. 52, 99-121.
699
Young, A. A., Vine, W., Gedulin, B. R., Pittner, R., Janes, S., Gaeta, L. S. L., Percy, A., Moore, C.
700
X., Koda, J. E., Rink, T. J., Beaumont, K., 1996. Preclinical pharmacology of Pramlintide in
701
the Rat: Comparisons With Human and Rat Amylin. Drug. Dev. Res. 37, 231-248.
702 703
Zimmerman, C. A., Leib, D. E., Knight, Z. A., 2017. Neural circuits underlying thirst and fluid homeostasis. Nat. Rev. Neurosci. 18, 459.
704 32
705
FIGURE LEGENDS
706 707
Figure 1. In vivo efficacy of fluorescently labelled peptides in mice. Cumulative food intake
708
(mean ± SEM) (g) after a single subcutaneous injection in C57Bl6 mice of A) vehicle, rAMY (100
709
nmol/kg) or rAMYCy5 (100 nmol/kg); B) vehicle, sCT (100 nmol/kg) or sCTCy5 (100 nmol/kg); C)
710
vehicle, rAMY (300 nmol/kg) or rAMYCy7 (300 nmol/kg); D) vehicle, sCT (100 nmol/kg) or sCT750
711
(100 nmol/kg). Cumulative food intake plotted every 1 hour 0 – 24 hours after injection. Shading =
712
dark period. N=6-8 per group. *p< 0.05: native peptide vs. vehicle; #p< 0.05: fluorescently labelled
713
peptide vs. vehicle. sCT = salmon calcitonin. rAMY = rat amylin.
714 715
Figure 2. Salmon calcitonin internalizes in a subset of NPY neurons but not POMC neurons in
716
reporter mice. hrGFP-NPY and POMC-Cre:ERT2::tdTomato mice were used to investigate whether
717
fluorescently labelled salmon calcitonin (sCTCy5) and rat amylin (rAMYCy5) distribute into the ARC
718
and binds to NPY and/or POMC neurons. A-D) Results from hrGFP-NPY mice injected with sCTCy5
719
or rAMYCy5 (n = 2 – 3/group) depicting average count/ARC section (mean) of NPY+, Cy5+ and
720
double-labelled cells (A), and the percentage of NPY+ and Cy5+ cells (mean) which were double
721
labelled in ARC sections (C). An orthogonal projection of a representative 20X image of the ARC
722
from hrGFP-NPY mice injected with rAMYCy5 showing NPY neurons (green), but no Cy5 cells (red)
723
(B). An orthogonal projection of a representative 20X image of Cy5 (red), NPY (green) and double
724
labelled (yellow, white arrows) cells in the ARC after sCTCy5 injection, where Cy5 internalization
725
into NPY neurons is visualized in the X, Y and Z planes (lines cross) (left). High resolution 63X
726
images of the marked section are depicted with (bottom) or without (top) the NPY channel showing
727
co-localization of Cy5 and NPY in the same neurons (right) (D). E-F) Results from POMC-
728
Cre:ERT2::tdTomato mice injected with sCTCy5 or rAMYCy5 (n = 3 – 4/group) depicting average
729
count/ARC section (mean) of POMC+, Cy5+ and double-labelled cells (E). An orthogonal projection 33
730
of a representative 20X picture of Cy5 (red, a selection marked with arrowheads) and POMC (green)
731
cells in the ARC after sCTCy5 injection, showing that Cy5 does not internalize into POMC neurons
732
(left). High resolution 63X image of the marked section where accumulation of Cy5 is not evident in
733
POMC expressing neurons (right) (F). 3V = 3rd ventricle. ARC = Arcuate nucleus. ME = Median
734
eminence.
735 736
Figure 3. Comparison between POMC positive cells in POMC-Cre:ERT2::tdTomato mice and
737
POMC IHC. ARC sections from POMC-Cre:ERT2::tdTomato mice expressing tdTomato in POMC
738
neurons were stained for POMC with IHC, and POMC expression compared (n = 4). The
739
identification number of the mouse is indicated next to the symbol to allow for direct comparison. A)
740
Average counts / ARC section (mean) of POMC+ after IHC staining or with endogenous tdTomato
741
expression. B) The percentage of POMC neurons which were double labelled for IHC+ and
742
tdTomato+ expression. C) Representative 20X image of DAPI, POMC-IHC and POMC tdTomato
743
expression in the ARC. 3V = 3rd ventricle. ARC = arcuate nucleus. IHC = immunohistochemistry.
744 745
Figure 4. CTR is more abundantly expressed in ARC NPY neurons than in POMC neurons.
746
ARC sections from wild type mice investigated for CTR expression in NPY and POMC expressing
747
neurons with ISH (n = 4). A) Depicted is a representative 20X image of an ARC/ME section with a
748
close-up of the marked section showing that few POMC expressing neurons (red) also express the
749
CTR (yellow) (white arrows). B) Depicted is a representative 20X image of an ARC/ME section
750
with a close-up of the marked section showing that almost all NPY expressing neurons (red) express
751
abundant levels of the CTR (yellow). C) Quantification of CTR-NPY and CTR-POMC neurons in
752
the ARC. 3V = 3rd ventricle. ARC = Arcuate nucleus. CTR = calcitonin receptor.
753
Immunohistochemistry. ISH = In situ hybridization.
IHC =
754 34
755
Figure 5. Salmon calcitonin internalizes into AP and DMV cells of mice. hrGFP-NPY and
756
POMC-Cre:ERT2::tdTomato mice were used to investigate if fluorescently labelled salmon calcitonin
757
(sCTCy5) and rat amylin (rAMYCy5) binds to cells in hindbrain nuclei (n = 5 – 7/group, both mouse
758
strains combined). A-B) An orthogonal projection of a representative 20X image from mice treated
759
with sCTCy5 of the AP (A) and DVM (B) showing that Cy5 (red) is internalized into cells in the X, Y
760
and Z planes (lines cross and marked with arrowheads) in both regions. High resolution 63x image of
761
marked section (insets) shows Cy5 accumulation in the cytoplasm of cells. C) An orthogonal
762
projection of a representative 20X image of the AP from mice treated with rAMYCy5, showing no
763
Cy5 signal (red) in the AP. AP = area postrema; DVM = Dorsal motor nucleus of the vagus; NTS =
764
Nucleus of solitary tract.
765 766
Figure 6. Distribution of fluorescently labelled salmon calcitonin and rat amylin in the mouse
767
brain. Wild type mice were injected with fluorescently labelled salmon calcitonin (sCT750) or rat
768
amylin (rAMYCy7) to investigate how the peptides distributes into the brain of mice (auto-
769
fluorescence from tissue = green, specific signal = white). A – B) Representative images of the
770
reconstructed 3D whole brain 30 min after last injection of sCT750 in the horizontal (A) and sagittal
771
(B) plane. C – F) Quantification (mean fold change ± SEM) of specific sCT750 signal in the
772
ARC/ME and AP of the brain 30 minutes and 2 hours post last injection of sCT750 in mice (C), and
773
representative images of the ARC/ME (D), AP (E) and OVLT (F) from mice terminated 30 min after
774
injection of sCT750. G – J) Quantification (mean fold change ± SEM) of specific rAMYCy7 signal in
775
the ARC/ME and AP of the brain 10 minutes post last injection of rAMYCy7 in mice (G), and
776
representative images of the ARC/ME (H), AP (I) and OVLT (J) from mice injected with rAMYCy7.
777
A-B) Scale bars = 1000 µm. D-F, H-J) scale bars = 100 µm. N=5 per group. *p< 0.05 and **p< 0.01
778
vs. control group. ARC = arcuate nucleus; AP = area postrema; ME = median eminence; OVLT =
779
vascular organ of the lamina terminalis. 35
780 781
Supplementary video 6-1.
782
A representative reconstructed 3D whole brain summarizing sCT750 distribution in the entire brain 2
783
hours post injection (autofluorescence from tissue = green, specific signal = white).
784 785
Supplementary video 6-2.
786
A representative reconstruction of ARC coronal sections summarizing sCT750 distribution in the
787
ARC 30 min post injection (autofluorescence from tissue = green, specific signal = white).
788 789
Supplementary video 6-3.
790
A representative reconstruction of ARC coronal sections summarizing rAMYCy7 distribution in the
791
ARC 10 min post injection (autofluorescence from tissue = green, specific signal = white).
36
792
Table 1: In vitro potency data (EC50-values) for pramlintide, sCT, sCT750, rAMYCy5 and sCTCy5
793
measured in BHK cell lines stably expressing the human calcitonin receptor (CTR) or amylin
794
receptor 3 (AMY3) and the CRE-luciferase reporter gene. Data are shown as means with 95%
795
confidence intervals (CI) based on three independent experiments. See statistical analysis in
796
supplementary table 1. Compound
AMY3
CTR
Pramlintide
EC50 (95% CI) [pM] 2.4 (1.3 to 4.3) 45.9 (21.2 to 99.4)
sCT sCT750 sCTCy5 rAMYCy5
0.9 (0.4 to 1.8) 1.9 (0.6 to 5.6) 0.8 (0.5 to 1.4) 9.8 (7.5 to 12.8)
1.7 (0.4 to 6.3) 2.6 (1.1 to 6.3) 1.6 (0.5 to 4.9) 167.7 (134.6 to 208.9)
797
37
B) Cumulative food intake (g)
Cumulative food intake (g)
A) 5 4 3
#
*, #
2
#
Vehicle rAMY 100 nmol/kg rAMYCy5 100 nmol/kg
#
1 0
*, #
4 3 2
Vehicle sCT 100 nmol/kg sCTCy5 100 nmol/kg
1 0
5
10 15 Time (h)
20
C)
0
5
10 15 Time (h)
20
D) *, #
5 4 #
3 2 Vehicle rAMY 300 nmol/kg rAMYCy7 300 nmol/kg
1 0
Cumulative food intake (g)
0
Cumulative food intake (g)
5
Vehicle sCT 100 nmol/kg sCT750 100 nmol/kg
5 4
*, # 3 2 1 0
0
5
10 15 Time (h)
20
0
5
10 15 Time (h)
20
120
rAMYCy5 sCTCy5
100 80 60
3V
40 20 0
NPY+
Cy5+
Cy5+ + NPY+
ME DAPI NPY rAMYCy5
ARC Cy5+ NPY+ co-loc. (%)
80 60 40
3V
20 0
Cy5+ NPY+ of NPY+
Cy5+ NPY+ of Cy5+
ME DAPI NPY sCTCy5 ARC
Average count / section
Average count / section
ARC
60 50 40 30 20
3V
10 0
POMC+
Cy5+
Cy5+ +POMC+
DAPI POMCsCTCy5
ME
B) 80
100
4 1
60
14 3
2
40
3
2
20 0
POMC IHC+ tdTomato+ co-loc. (%)
Average count / section
A)
IHC+ tdTomato+
C)
80 60
1 3 4
1 4 2 3
2
40 20 0 IHC+ tdTomato+ IHC+ tdTomato+ of IHC+ of tdTomato+
"5'
#%&'8 *+,-./*-
!"#$
#%&'($)'
"5' :;
6789.
#%&'(*+,-./*-
%0123/4
ARC
A)
ARC
DAPI POMC CTR
3V 3V
ME
B) DAPI NPY CTR
ARC
3V
Average count / section
ME
150
100
50
0
POMC+
NPY+
CTR+ CTR+ %CTR+ %CTR+ +POMC+ +NPY+ +POMC+ +NPY+
A)
B)
C)
AP AP
NTS DMV
DAPI sCTCy5
DAPI sCTCy5
DAPI rAMYCy5
A)
B)
OVLT
AP
ARC
AP OVLT
Fold change (sCT750 / Control)
C)
D)
sCT750 30 min sCT750 2 hours
35 30
E)
F)
**
25 20 15 10
ARC
*
5 ARC/ME 80 60
AP
ME
AP
G) Fold change (rAMYCy7 / Control)
ARC
H)
I)
OVLT
J)
rAMYCy7 10 min
*
*
40
ARC
20
AP 0 ARC/ME
AP
ME
OVLT
AUTHOR CONTRIBUTIONS:
Conceptualization, H.L.Z., L.M.J., T.A.L. and C.L.F.; Methodology, H.L.Z. and C.L.F.; Investigation, H.L.Z., T.G., S.L. and C.L.F.; Writing – Original Draft, H.L.Z and C.L.F. ; Writing – Review & Editing, H.L.Z., L.M.J., J.L., T.G., A.S., T.A.L. and C.L.F. Resources, L.M.J., K.R. and T.A.L.; Funding Acquisition, K.R., J.L. and T.A.L. C.L.F. is the guarantor of this work and, as such, had full access to all the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis.