Download embj201488977-sup-0010-Suppl

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Brain wikipedia , lookup

Types of artificial neural networks wikipedia , lookup

Adult neurogenesis wikipedia , lookup

Neurotransmitter wikipedia , lookup

Convolutional neural network wikipedia , lookup

Electrophysiology wikipedia , lookup

Endocannabinoid system wikipedia , lookup

Apical dendrite wikipedia , lookup

Activity-dependent plasticity wikipedia , lookup

Single-unit recording wikipedia , lookup

Connectome wikipedia , lookup

Synaptogenesis wikipedia , lookup

Artificial general intelligence wikipedia , lookup

Axon wikipedia , lookup

Biochemistry of Alzheimer's disease wikipedia , lookup

Axon guidance wikipedia , lookup

Metastability in the brain wikipedia , lookup

Caridoid escape reaction wikipedia , lookup

Molecular neuroscience wikipedia , lookup

Neural oscillation wikipedia , lookup

Chemical synapse wikipedia , lookup

Multielectrode array wikipedia , lookup

Stimulus (physiology) wikipedia , lookup

Mirror neuron wikipedia , lookup

Neural coding wikipedia , lookup

Development of the nervous system wikipedia , lookup

Central pattern generator wikipedia , lookup

Nervous system network models wikipedia , lookup

Clinical neurochemistry wikipedia , lookup

Premovement neuronal activity wikipedia , lookup

Neuroanatomy wikipedia , lookup

Pre-Bötzinger complex wikipedia , lookup

Synaptic gating wikipedia , lookup

Neuropsychopharmacology wikipedia , lookup

Feature detection (nervous system) wikipedia , lookup

Optogenetics wikipedia , lookup

Circumventricular organs wikipedia , lookup

Channelrhodopsin wikipedia , lookup

Transcript
Romanov, Alpar et al.
Date of submission: 06/05/2017
EMBOJ-2014-88977; revised submission
Supplementary Information to the manuscript:
A secretagogin locus of the mammalian hypothalamus controls
stress hormone release
Roman Romanov*, Alan Alpar*,#, Ming-Dong Zhang, Amit Zeisel, André
Calas, Marc Landry, Matthew Fuszard, Sally L. Shirran, Robert Schnell,
Arpad Dobolyi, Mark Olah, Lauren Spence, Henrik Marten, Miklos
Palkovits, Matthias Uhlen, Harald H. Sitte, Catherine M. Botting, Ludwig
Wagner, Sten Linnarsson, Tomas Hökfelt++ & Tibor Harkany++,#
*R.R. & A.A. contributed equally to this study.
++T.Hö.
& T.Ha. share senior authorship.
#Address
correspondence to:
Tibor Harkany (at the Karolinska
Institutet); Telephone: +46 8 524 87656; Fax: +46 8
341 960; e-mail: [email protected] or Alan Alpar (at
the
Semmelweis
University);
e-mail:
[email protected]
This file contains:
Supplementary Figures 1 - 6
Supplementary Tables 1 - 3
Legends to Supplementary Figures & Tables
Supplementary References
1
Romanov, Alpar et al.
Date of submission: 06/05/2017
EMBOJ-2014-88977; revised submission
Legends to Supplementary Tables & Figures
Suppl. Table 1
Electrophysiological parameters used to sub-classify PVN neurons in
ex vivo brain slice preparations.
Data are from: type Ia: n = 17; type Ib: n = 5; type IIa: n = 10; type IIb:
n = 7; type IIIa: n = 18; type IIIb: n = 16 neurons, and were expressed
as means ± s.e.m. Parameters were chosen according to published
protocols(Miyoshi et al, 2010). Sub-clustering was based on the posthoc identification of biocytin-filled neurons using AVP/oxytocin
(combined in triple-labeling experiments and used as “magnocellular
markers”) and secretagogin (scgn) as histochemical markers (see
also Fig. 3A,B).
Suppl. Table 2
List of primary antibodies used for multiple immunofluorescence
labeling.
All antibodies were affinity purified, and extensively tested in vitro
and in vivo, including relevant genetic controls (Benoit et al, 1980;
Mulder et al, 2009; Hartig et al, 2009; Westberg et al, 2009;
Dabrowska et al, 2011; Stanic et al, 2011; Attems et al, 2012; Shi et
al, 2012; Renner et al, 2012; Keimpema et al, 2013).
Suppl. Table 3
Proteins identified by differential interactome analysis of Ca 2+-bound
secretagogin.
A list of 97 proteins that were found when immunoprecipitation (IP) of
secretagogin was performed in the presence of 10 µM Ca2+. Criteria
for positive protein identification are shown at the top. Proteins were
classified by color coding to correspond to the cumulative data
shown in Fig. 5C1. Non-targeted IgG in the presence of 10 µM Ca2+
was used as negative control. Proteins (n = 15 in total) marked under
“anti-secretagogin IP, Ca2+-free” might have some ability to
constitutively interact with secretagogin. Note that the number of
peptides fragments is low, likely reflecting the sparsity of proteinprotein interactions under Ca2+-free conditions.
Suppl. Figure 1
Electrophysiological classification of PVN neurons.
2
Romanov, Alpar et al.
Date of submission: 06/05/2017
EMBOJ-2014-88977; revised submission
Criteria were chosen as per (Luther et al, 2000; Stern, 2001; Luther
et al, 2002; Wamsteeker Cusulin et al, 2013). Example traces are
known for each neuronal subgroup. Statistically-analyzed data are
presented in Supplementary Table 1.
Suppl. Figure 2
Secretagogin-expressing
PVN
neurons
exhibit
parvocellular
identities, including dendrite morphology.
(A) Variations of mRNA transcript levels in cells dissociated from the
mouse PVN. The x-axis is scaled to show only a fraction of nonexpressing neurons (at 0 levels) for visual clarity. Red circles
correspond to secretagogin+ neurons. Horizontal dashed lines show
the cut-off value used to scale positive neurons for each mRNA
transcript.
(B-B2)
Morphometric
secretagogin+
reconstruction
(B1) and
of
vasopressin+
vasopressin+/secretagogin+
(B),
(B2) neurons
isolated from mouse hypothalamus at postnatal days (P)1-2. Scale
bar = 30 µm.
(C) Morphological parameters of cultured hypothalamic neurons
identified post-hoc by multiple immunofluorescence cytochemistry.
Secretagogin+ neurons had smaller somata, with a higher number of
processes that were longer and arborized more extensively than
those of vasopressin+ neurons (11.61 ± 0.72 vs. 9.04 ± 0,21, p <
0.05). Vasopressin+/secretagogin+ neurons were morphologically
similar to vasopressin+/secretagogin- neurons (12.31 ± 0.61 vs. 11.61
± 0.72).
(D)
Representative
intracellular
Ca2+
responses
(ratiometric
measurements by using Fura-2AM) from n ≥ 3 neurons/group
exposed to depolarizing conditions (55 mM KCl).
Suppl. Figure 3
The effect of secretagogin on pharmacologically-induced Ca2+
responses in PVN neurons.
(A) Representative image of cultured hypothalamic neurons loaded
with Fura-2AM (left panel) and post-hoc histochemistry for AVP and
secretagogin (scgn). Scale bar = 60 µm.
3
Romanov, Alpar et al.
Date of submission: 06/05/2017
EMBOJ-2014-88977; revised submission
(B) Representative Ca2+ traces from cytochemically-identified PVN
neurons after ratiometric Fura-2AM imaging demonstrate the
existence of different response patterns to pharmacological stimuli.
Drug concentrations were as follows: 55 mM KCl, 50 µM glutamate,
50 µM NMDA + 100 µM glycine, 50 µM kainate, 50 µM carbachol, 10
µM ATP, 100 µM GABA and 50 µM bicuculline (Yamashita et al,
1987; Collden et al, 2010; Haam et al, 2012). Color-coded traces
correspond to responses from single cells.
(C) Cumulative (mean) responses per group upon depolarization by
55 mM KCl.
(D) Statistical analysis of Ca2+ responses, expressed as % change.
AVP+ (in blue; total n = 24), secretagogin+ (in red; total n = 69) and
other (unspecified, n = 14) neurons were analyzed; n = 14 - 69
neurons/group. ***p < 0.001; **p < 0.01; *p < 0.05; Student’s t-test.
Suppl. Figure 4
Effect of secretagogin overexpression on Ca2+ signaling in SH-SY5Y
human neuroblastoma cells.
(A) Overexpression of secretagogin in SH-SY5Y cells was used to
estimate the Ca2+-binding protein’s buffer capacity (B) in response to
excitatory stimuli as indicated.
(C) Immunofluorescence signal intensity (in green; A) was scaled
and correlated with Ca2+ responsiveness upon secretagogin
overexpression. Individual data points were presented. Dashed lines
were color-coded to identify specific stimuli and denote the lack of
correlation between Ca2+ peaks vs. secretagogin levels. Scale bar =
40 µm.
Suppl. Figure 5
In vivo siRNA-mediated secretagogin silencing does not affect
prominent hormone and neuropeptide systems in the mouse
paraventricular nucleus.
(A)
Arginine-vasopressin
(AVP)-immunoreactive
PVN
neurons
showed unchanged distribution in adult mice that had been injected
with secretagogin-specific siRNA in the paraventricular nucleus
(“scgn siRNA”) relative to control animals (“non-target siRNA”).
4
Romanov, Alpar et al.
Date of submission: 06/05/2017
EMBOJ-2014-88977; revised submission
(B,C) Likewise, the distribution of both neuropeptide Y-containing
processes (B) and oxytocin-containing neurons (C) in the PVN was
found unperturbed in animals exposed to secretagogin-specific
siRNA.
(D) In the arcuate nucleus, which borders the ventral-most wall of the
3rd ventricle and therefore can be accessible to siRNA molecules,
somatostatin immunoreactivity was unaffected.
Abbreviations: 3V, 3rd ventricle; PVN, paraventricular nucleus of the
hypothalamus. Scale bars = 100 µm (A-D).
Suppl. Figure 6
Summary
cartoon
for
secretagogin
action
and
interactome
engagement in CRH neurons.
(A) Secretagogin can affect CRH release either indirectly, by
affecting the function of key proteins involved in the vesicle formation
and cargo along the axons to the median eminence (“vesicle
logistics”), or more directly, by Ca2+-dependent modulation of the
exocytosis machinery in situ in nerve terminals (“release site”). This
duality of action suggests a significant role for secretagogin in
defining the dynamics of Ca2+-dependent neuropeptide release.
(B) Proteome profiling-based secretagogin interactions likely to
control neuropeptide release. Select protein-protein interactions are
indicated.
Note
that
secretagogin’s
localization
along
the
plasmalemma suggests its ability to also modulate intra-membrane
adaptor proteins. So far, our analysis leaves ambiguity as to the
molecular identity of the exact identity of the Rab3 family member
modulated by secretagogin, henceforth “Rab3” labeling is used.
5
Romanov, Alpar et al.
Date of submission: 06/05/2017
EMBOJ-2014-88977; revised submission
SUPPORTING REFERENCES
Attems J, Alpar A, Spence L, McParland S, Heikenwalder M, Uhlen M, Tanila H, Hokfelt TG, and
Harkany T (2012) Clusters of secretagogin-expressing neurons in the aged human olfactory tract lack
terminal differentiation. Proc Natl Acad Sci U S A 109: 6259-6264
Benoit R, Bohlen P, Brazeau P, Ling N, and Guillemin R (1980) Isolation and characterization of rat
pancreatic somatostatin. Endocrinology 107: 2127-2129
Collden G, Mangano C, and Meister B (2010) P2X2 purinoreceptor protein in hypothalamic neurons
associated with the regulation of food intake. Neuroscience 171: 62-78
Dabrowska J, Hazra R, Ahern TH, Guo JD, McDonald AJ, Mascagni F, Muller JF, Young LJ, and
Rainnie DG (2011) Neuroanatomical evidence for reciprocal regulation of the corticotrophin-releasing
factor and oxytocin systems in the hypothalamus and the bed nucleus of the stria terminalis of the rat:
Implications for balancing stress and affect. Psychoneuroendocrinology 36: 1312-1326
Haam J, Popescu IR, Morton LA, Halmos KC, Teruyama R, Ueta Y, and Tasker JG (2012) GABA is
excitatory in adult vasopressinergic neuroendocrine cells. J Neurosci 32: 572-582
Hartig W, Reichenbach A, Voigt C, Boltze J, Bulavina L, Schuhmann MU, Seeger J, Schusser GF,
Freytag C, and Grosche J (2009) Triple fluorescence labelling of neuronal, glial and vascular markers
revealing pathological alterations in various animal models. J Chem Neuroanat 37: 128-138
Keimpema E, Tortoriello G, Alpar A, Capsoni S, Arisi I, Calvigioni D, Hu SS, Cattaneo A, Doherty P,
Mackie K, and Harkany T (2013) Nerve growth factor scales endocannabinoid signaling by regulating
monoacylglycerol lipase turnover in developing cholinergic neurons. Proc Natl Acad Sci U S A 110:
1935-1940
Luther JA, Daftary SS, Boudaba C, Gould GC, Halmos KC, and Tasker JG (2002) Neurosecretory and
non-neurosecretory parvocellular neurones of the hypothalamic paraventricular nucleus express
distinct electrophysiological properties. J Neuroendocrinol 14: 929-932
Luther JA and Tasker JG (2000) Voltage-gated currents distinguish parvocellular from magnocellular
neurones in the rat hypothalamic paraventricular nucleus. J Physiol 523 Pt 1: 193-209
Miyoshi G, Hjerling-Leffler J, Karayannis T, Sousa VH, Butt SJ, Battiste J, Johnson JE, Machold RP,
and Fishell G (2010) Genetic fate mapping reveals that the caudal ganglionic eminence produces a
large and diverse population of superficial cortical interneurons. J Neurosci 30: 1582-1594
Mulder J, Zilberter M, Spence L, Tortoriello G, Uhlen M, Yanagawa Y, Aujard F, Hokfelt T, and
Harkany T (2009) Secretagogin is a Ca2+-binding protein specifying subpopulations of telencephalic
neurons. Proc Natl Acad Sci U S A 106: 22492-22497
Renner E, Puskas N, Dobolyi A, and Palkovits M (2012) Glucagon-like peptide-1 of brainstem origin
activates dorsomedial hypothalamic neurons in satiated rats. Peptides 35: 14-22
Shi TJ, Xiang Q, Zhang MD, Tortoriello G, Hammarberg H, Mulder J, Fried K, Wagner L, Josephson
A, Uhlen M, Harkany T, and Hokfelt T (2012) Secretagogin is expressed in sensory CGRP neurons
and in spinal cord of mouse and complements other calcium-binding proteins, with a note on rat and
human. Mol Pain 8: 80
Stanic D, Mulder J, Watanabe M, and Hokfelt T (2011) Characterization of NPY Y2 receptor protein
expression in the mouse brain. II. Coexistence with NPY, the Y1 receptor, and other neurotransmitterrelated molecules. J Comp Neurol 519: 1219-1257
Stern JE (2001) Electrophysiological and morphological properties of pre-autonomic neurones in the
rat hypothalamic paraventricular nucleus. J Physiol 537: 161-177
6
Romanov, Alpar et al.
Date of submission: 06/05/2017
EMBOJ-2014-88977; revised submission
Wamsteeker Cusulin JI, Fuzesi T, Watts AG, and Bains JS (2013) Characterization of corticotropinreleasing hormone neurons in the paraventricular nucleus of the hypothalamus of Crh-IRES-Cre
mutant mice. PLoS One 8: e64943
Westberg L, Sawa E, Wang AY, Gunaydin LA, Ribeiro AC, and Pfaff DW (2009) Colocalization of
connexin 36 and corticotropin-releasing hormone in the mouse brain. BMC Neurosci 10: 41
Yamashita H, Kannan H, Kasai M, and Osaka T (1987) Decrease in blood pressure by stimulation of
the rat hypothalamic paraventricular nucleus with L-glutamate or weak current. J Auton Nerv Syst 19:
229-234
7