PreprintPDF Available

Cold Exposure Protects from Neuroinflammation Through Immunologic Reprogramming

Authors:

Abstract

Autoimmunity is energetically costly, but the impact of a metabolically active state on immunity and immune-mediated diseases is unclear. Ly6Chi monocytes are key effectors in CNS autoimmunity with elusive role in priming naive autoreactive T cells. Here we provide unbiased analysis of the immune changes in various compartments during cold exposure, and show that this energetically costly stimulus markedly ameliorates active experimental autoimmune encephalomyelitis (EAE). Cold exposure decreases MHCII on monocytes at steady-state and in various inflammatory mouse models, and suppresses T cell priming and pathogenicity through the modulation of monocytes. Genetic, or antibody-mediated monocyte depletion, or adoptive transfer of Th1- or Th17-polarized cells for EAE abolish the cold-induced effects on T cells or EAE, respectively. These findings provide a mechanistic link between environmental temperature and neuroinflammation, and suggest competition between cold-induced metabolic adaptations and autoimmunity as energetic trade-off beneficial for the immune-mediated diseases.
1
Cold Exposure Protects from Neuroinflammation Through
Immunologic Reprogramming
Martina Spiljar1, Karin Steinbach2, Dorothée Rigo1, Nicolas Suárez-Zamorano1, Ingrid Wagner2,
Noushin Hadadi1, Ilena Vincenti2, Nicolas Page2, Bogna Klimek2, Mary-Aude Rochat1, Mario
Kreutzfeldt2, Claire Chevalier1, Ozren Stojanović1, Matthias Mack3, Dilay Cansever4, Melanie
Greter4, Doron Merkler2*, Mirko Trajkovski1*
1 Department of Cell Physiology and Metabolism, Faculty of Medicine, Centre Médical Universitaire
(CMU), University of Geneva, Switzerland.
2 Department of Pathology and Immunology, Faculty of Medicine, Centre Médical Universitaire (CMU),
University of Geneva, Switzerland.
3 Department of Internal Medicine II - Nephrology, University Hospital Regensburg, Germany.
4 Institute of Experimental Immunology, University of Zurich, Switzerland.
*Correspondence and equal contribution
SUMMARY
Autoimmunity is energetically costly, but the impact of a metabolically active state on immunity
and immune-mediated diseases is unclear. Ly6Chi monocytes are key effectors in CNS
autoimmunity with elusive role in priming naïve autoreactive T cells. Here we provide unbiased
analysis of the immune changes in various compartments during cold exposure, and show that
this energetically costly stimulus markedly ameliorates active experimental autoimmune
encephalomyelitis (EAE). Cold exposure decreases MHCII on monocytes at steady-state and in
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
2
various inflammatory mouse models, and suppresses T cell priming and pathogenicity through
the modulation of monocytes. Genetic, or antibody-mediated monocyte depletion, or adoptive
transfer of Th1- or Th17-polarized cells for EAE abolish the cold-induced effects on T cells or
EAE, respectively. These findings provide a mechanistic link between environmental temperature
and neuroinflammation, and suggest competition between cold-induced metabolic adaptations
and autoimmunity as energetic trade-off beneficial for the immune-mediated diseases.
INTRODUCTION
Maintaining immunity requires substantial metabolic resources (Buck et al., 2017; Demas and
Nelson, 2012; Ganeshan et al., 2019; Hotamisligil, 2017; McDade, 2005; O'Neill et al., 2016) for
steady-state systemic immune surveillance and during inflammation, leading to the generation of
millions of immune cells daily. The life-history theory (Stearns, 1992) proposes that prioritization
of resources between biological programs would depend on the environment. In hostile
environments, resources are shifted away from growth and reproduction programs into
maintenance programs (Okin and Medzhitov, 2012; Stearns, 1992; Wang et al., 2019). In addition
to immunity, these maintenance traits include adaptations to energy scarcity, but also adaptations
to changes in the environmental temperature. Exposure to cold provokes a high cost–high gain
physiologic response aimed at reducing heat dissipation (e.g. by vasoconstriction) and increasing
heat production, mainly by activating the brown adipose tissue (BAT) and browning of the
subcutaneous adipose tissue (SAT). BAT catabolizes energy to enable generation of heat, a
function conferred by the uncoupling protein 1 (UCP1). The cold-induced BAT activation is
triggered by sympathetic innervation (Cannon and Nedergaard, 2004; Stojanovic et al., 2018).
Norepinephrine released from the nerve endings stimulates the beta 3 adrenergic receptor, and
agonists of this receptor are frequently used to mimic cold exposure and experimentally induce
thermogenesis.
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
3
In autoimmunity, the organism develops an energetically costly pro-inflammatory immune
response (Okin and Medzhitov, 2012). Such high energy demanding processes may compete
with each other, and prioritization of one task over the other may be a result of an energetic trade-
off. This concept can be of particular interest for autoimmunity, in which introducing an additional
energy-costly program may result in milder immune response and disease outcome. In contrast
to the well-described immune status during obesity (Grant and Dixit, 2015; Hotamisligil, 2017;
Kohlgruber et al., 2016; Li et al., 2020; Man et al., 2017), the repercussions of a metabolically
active phenotype on the autoimmunity, and the potential systemic effects on the immune-
mediated diseases are unknown.
Experimental Autoimmune Encephalomyelitis (EAE) is an animal model for multiple sclerosis
(MS), the most frequent autoimmune demyelinating disease of the central nervous system (CNS).
Similar to MS, EAE is exacerbated by high caloric diet and obesity (Hasan et al., 2016; Piccio et
al., 2008; Timmermans et al., 2014; Winer et al., 2009), notably characterized by a positive energy
balance. CD4+ T cells and monocytes are important cellular players in the pathogenesis of EAE.
Among CD4+ T cells, distinct encephalitogenic subsets are essentially involved in disease
precipitation and are characterized by certain cytokine signatures, including interleukin17 (IL-17),
Granulocyte-Macrophage Colony Stimulating Factor (GM-CSF) and Interferon gamma (IFNg).
Monocytes and monocyte-derived cells are crucial for the resulting tissue destruction of the
central nervous system (CNS), particularly during the effector phase of EAE (Ajami et al., 2011;
Fife et al., 2000; Izikson et al., 2000; King et al., 2009; Mildner et al., 2009; Serbina and Pamer,
2006). Thereby, these monocytes and monocyte-derived cells can express pro-inflammatory
INOS (m1-like) or anti-inflammatory ARG1 (m2-like) and in addition can exert antigen presenting
function via major histocompatibility complex class II (MHCII) upregulation. The role of monocytes
in priming of autoreactive T cells in CNS autoimmunity, and whether cold exposure could affect
this interaction remain unclear.
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
4
Our study provides an unbiased characterization of the immune system in different compartments
in healthy conditions and during CNS autoimmunity following exposure to lower environmental
temperature. We demonstrate that cold exposure renders monocytes less activated in the bone
marrow, the circulation and the CNS. The change in the monocyte phenotype affects priming of
pathogenic T cells during neuroinflammation, resulting in reduced T cell cytokine expression and
consequently attenuated EAE. Our data suggest competition between cold exposure and
autoimmunity leading to a constrained immune response, which could be of therapeutic
importance in neuroinflammation, and potentially other autoimmune diseases.
RESULTS
Cold Exposure Modulates Myeloid Cells in Bone Marrow at Steady State
To explore the impact of cold exposure on the immune system, we first focused on the bone
marrow where immune cells originate from. We performed RNA sequencing of the bone marrow
from mice exposed to 10°C for two weeks following an acclimatization period, and compared them
to mice kept at room temperature (Figures 1A and S1A). We shortlisted the significantly different
genes (p<0.05) for pathway and gene ontology analyses and considered pathways and terms
significantly deregulated or enriched when p<0.05. Among the 5088 enriched terms identified by
Metacore Gene Ontology Processes, “Myeloid Leukocyte Activation” was among the top five most
enriched terms (Figure 1B). Metacore Metabolic Network analysis identified 41 enriched networks
(Figure S1B), whereas Metacore Pathway Maps revealed 885 pathways that were changed.
Among the top 10 most significantly regulated ones (Figure S1C), four pathways were consistently
changed (>80% of genes in same direction) (Figures 1C and S1D). These were Macrophage and
DC shift, G-CSF-induced myeloid differentiation, IFN-alpha/beta signaling and Transcription
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
5
regulation-granulocyte development, which mainly contain genes important for myeloid cell
activation and differentiation.
We then examined the impact of cold exposure at cellular and protein level within the bone marrow
by flow cytometry. While cold exposure did not affect the number of total bone marrow cells
(Figure S1E), there was a decrease in Ly6Chi but not Ly6Cint or Ly6Clo monocytes (Figure 1D).
Ly6Chi monocytes showed phenotypical changes characterized by decreased MHCII expression
(Figure 1E), a marker for antigen presentation and activation. Within the total MHCII+ cells, the
population of MHCII+Ly6Chi monocytes was decreased, while the percentage of total MHCII+ cells
within the bone marrow remained unchanged (Figure 1E, gating strategy S1F). Differences in the
MHCII expression on protein level were consistent with the observations from RNA sequencing,
where MHCII genes were within the top downregulated pathways (Figure 1C).
To address whether cold exposure decreases the monocytes by affecting hematopoiesis, we
analyzed hematopoietic progenitors in the bone marrow similarly as previously described
(Hettinger et al., 2013; Luo et al., 2015; Merad et al., 2013). In three out of five experiments, cold
exposure decreased the absolute cell numbers (data not shown) and the percentage of monocyte-
dendritic cell progenitors (MDPs), common dendritic cell progenitors (CDP) and tendentially
common monocyte progenitors (cMoP), while the common lymphoid progenitors (CLP) and most
hematopoietic cells (Lin- Sca1+ c-KIT+ or LSK) remained unchanged (Figure 1F, gating strategy
S1F).
Beta 3 adrenergic receptor signaling is one of the main pathways activated by cold exposure
(Cannon and Nedergaard, 2004; Stojanovic et al., 2018). The beta 3 adrenoreceptor agonist
CL316,243 (CL316) decreased Ly6Chi monocytes, common dendritic cell progenitors and
common monocyte progenitors (Figure 1G), suggesting that beta 3 adrenoreceptor stimulation is
sufficient to partly mimic the effects of cold on the myeloid bone marrow cells and their
progenitors. Together, these results suggest that cold exposure decreases myeloid cell
progenitors and monocytes in the bone marrow, and downregulates monocyte MHCII expression.
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
6
Cold Exposure Impacts Monocytes in Blood at Steady State
We next examined the systemic relevance of the cold exposure-mediated changes observed in
the bone marrow by FACS sorting CD115+ blood monocytes (Figure S2A) and subsequent
profiling them via RNA sequencing (Figures S2B and 2A). Among the 58 Gene Ontology
Biological Processes that were enriched, the top 15 terms included leukocyte migration, T cell co-
stimulation and antigen presentation via MHCII (Figure 2B), all processes important for the
immune system in health and disease. Such pathways were similarly found within the top 15 of
653 total Metacore Pathway Maps (no p-value cutoff) (Figure S2C). Metacore Metabolic Networks
analysis revealed that 13 metabolic terms were enriched (Figure S2D). Among the variety of
general cell membrane restructuring elements (p<0.05, total 12), “MHC class II protein complex”
was among the most enriched Cellular Components in Gene Ontology analysis (Figure S2E). To
further profile the effect of cold exposure on circulatory immune cells and in particular myeloid
cells, we performed high parameter flow cytometry (BD FACSymphony), followed by an unbiased
analysis using FlowSOM algorithm (Van Gassen et al., 2015) and visualization via UMAP plots.
Overall, we neither observed any major shifts in myeloid cell (Figure 2C), nor in T cell populations
(data not shown) or numbers, suggesting that cold exposure induces several gene expression
changes on the monocytes without significantly altering the overall immune cell composition.
These observations were further supported by flow cytometry, which revealed that cold exposure
consistently decreased MHCII expression of Ly6Chi monocytes, without altering their overall
number (Figures 2D and 2E, gating strategy Figure S2F). Similarly, as in the bone marrow, beta
3 adrenoreceptor stimulation mimicked the effects of cold exposure and reduced the MHCII
expression of Ly6Chi monocytes (Figure 2F). These data indicate that cold exposure induces
functional changes on the monocytes in the blood by reducing their MHCII expression, without
provoking major changes in the main immune cell populations.
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
7
Cold Decreases Monocyte MHCII Expression in Inflammatory Mouse Models
To study the functional relevance of the observed cold-induced monocyte modulations, we used
different immunologic models. First, we amplified circulating monocytes and their MHCII
expression using B16-GM-CSF tumor cells (Dranoff et al., 1993; Menezes et al., 2016) that were
subcutaneously injected into cold exposed-, or room temperature-kept mice. GM-CSF initiates an
inflammatory phenotype in monocytes and depletion of monocyte GM-CSF signaling renders
mice resistant to EAE (Croxford et al., 2015). Cold exposure decreased MHCII expression of
monocytes without changing the monocyte percentage after implantation of the tumor cells
(Figures 3A). To confirm that this effect is monocyte-specific we also analyzed total CD11c+ cells,
the majority of which are dendritic cells that remained unchanged (Figure 3B). Thus, cold
exposure counteracted GM-CSF-induced increase of MHCII on Ly6Chi monocytes.
Next, we triggered a thioglycolate elicited peritoneal inflammation, in which monocytes are highly
attracted into the peritoneum. Cold exposure decreased MHCII expression of recruited Ly6Chi
monocytes/monocyte-derived cells within the peritoneum (Figure 3C), without affecting the
proportion of several other immune cell populations (Figure S3A). These data show that cold
exposure leads to a preferential decrease in the monocyte MHCII expression in various mouse
models irrespective of the inflammatory trigger.
Monocytes can acquire antigen presenting functions, including antigen uptake and migration to
the lymph nodes (Guilliams et al., 2018; Jakubzick et al., 2017). We therefore investigated if
antigen presenting functions of monocytes, as well as dendritic cells (Ganguly et al., 2013), might
be changed upon cold exposure. For this purpose, we performed a FITC painting assay, in which
FITC is applied epicutaneously of one flank of the mouse and allows measuring migratory immune
cell properties into the draining lymph node (dLN) (Allan et al., 2006). We monitored the FITC
uptake in the ipsi- and contralateral LN at two different time points, where 12h indicates possible
initial differences in lymphatic draining, while 18h is the peak and allows an estimate of the overall
changes (Platt et al., 2013). At none of the monitored time points have we observed changes in
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
8
cell count, FITC uptake or MHCII expression on dendritic cells, monocytes and various dendritic
cell subsets (Figure S3B and data not shown), thus excluding that cold exposure affects antigen
drainage to the LN. Altogether, these data show that exposure to lower environmental
temperature leads to a decrease in the monocyte MHCII expression in various inflammatory
mouse models without affecting the lymphatic drainage.
Cold Exposure Attenuates Experimental Autoimmune Encephalomyelitis (EAE)
Monocytes and monocyte derived cells are important pathogenic players in chronic inflammatory
diseases, such as MS. Similarly, monocyte trafficking to the CNS is critical for EAE progression
(Ajami et al., 2011; King et al., 2009) and Ccr2-deficient mice, which lack monocytes outside the
bone marrow (Serbina and Pamer, 2006), show mild (Mildner et al., 2009) or no EAE (Fife et al.,
2000; Izikson et al., 2000). We therefore explored whether cold exposure could affect the EAE
disease outcome. For this purpose, we exposed mice to 10 °C for 2 weeks with an additional
initial acclimatization period of 5 days at 18°C and 5 days at 14°C before immunization, and kept
them at 10°C during the whole EAE (Figure 4A). Strikingly, exposing animals to cold led to a
pronounced attenuation of the EAE disease severity compared to room temperature kept controls
(Figures 4B and S4A). The cold exposed mice also showed delayed onset of the disease, together
with a lower maximum and cumulative EAE score, disease incidence and body weight loss
(Figures 4C-4G) compared to the RT control mice. Histologic evaluation revealed a reduced area
of demyelination in the spinal cord (Figure 4H) and a lower number of lesions (Figure S4B) in
spinal cords from cold exposed mice at the peak of EAE. Mac3+, CD3+ and APP lesions from
LFB-Pas per white matter area showed a tendential decrease (Figures S4C-S4G) without
reaching statistical significance. As the extent of EAE amelioration slightly differed between
experiments, a pool is shown (Figures 4 B-G) of experiments performed under the same
conditions except the housing room, which had no consistent impact.
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
9
To gain insights into the level of protection by cold exposure on the EAE-induced gene expression
alterations in the spinal cord, we performed RNA sequencing on mice kept at room or cold
temperature both at steady state, or at EAE peak. Principle component and multidimensional
scaling analysis revealed that when the room temperature-kept animals reached peak of the
disease after EAE induction (Figure 4I), the immunized cold-exposed mice clustered together with
the healthy, steady-state animals (Figures 4J and S4H). Interestingly, all genes that were
increased in the room temperature mice with EAE compared to healthy controls were left
unchanged, or deregulated to a lower extent in the cold exposed mice under EAE (Figure 4K).
These data show that the EAE-provoked spinal cord gene expression alterations are either
reduced, or prevented by cold exposure. Gene expression comparison between spinal cords from
cold exposed mice vs. room temperature controls during EAE revealed 61 up- and 1579
downregulated genes of 15467 total genes (p<0.05, logFC>1) (Figure 4L). Among the top 984
regulated pathways identified by the Metacore Pathway Maps, “Macrophage and Dendritic cell
phenotype shift…”, was the most significantly changed one. This pathway includes MHCII genes
and other activation markers (Figures 4M and S4I), further indicating that the myeloid cell and
antigen presentation are the most prominent changes induced by cold also during EAE. The
pathway further reveals downregulation of CXCL10, a recently identified marker of a pathogenic
monocyte subset (Giladi et al., 2020), which we found similarly decreased in the bone marrow
(Figure 1C).
To investigate whether the length of the cold exposure before the EAE induction is important for
the EAE onset and disease progression, we pre-exposed mice to cold two days before
immunization, and compared them to the animals that were kept 2 weeks at 10°C as in the
standard protocol. Two days cold pre-exposure was sufficient to attenuate EAE, although to a
slightly lower extent than the 2 weeks pre-exposed ones (Figures 4N and 4O), and to reduce
monocyte MHCII (Figure 4P), suggesting that the length of the cold pre-treatment plays a modest
role in the disease outcome.
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
10
To address whether warm exposure has an opposing effect on EAE, we exposed mice to 34°C
for seven days before immunization, and kept them at warm during EAE (Figure 4Q). Warm
exposure led to decreased food intake and body weight (Figure S4M, Figures 4O and U). Although
more mice died under warm exposure, the survival rate was not significantly different, and neither
was the day of onset (Figures S4N and S4O). However, in contrast to the effects seen during
cold, the MHCII expression of Ly6Chi monocytes was increased in the warm-exposed mice,
coupled to deteriorated EAE, enhanced body weight loss and increased maximum disease score
(Figures 4R-T and S4J, K). Together, these results suggest that the temperature is a critical
determinant for EAE severity, and that cold exposure is a potent stimulus that attenuates
neuroinflammation.
Long-term cold exposure increases energy expenditure by enhancing the thermogenesis in the
brown and the beige fat, enabling homeothermy (Chevalier et al., 2015). However, when
additional biologic maintenance programs are active, as for instance in inflammation, we
hypothesized that these responses (maintenance of homeothermy and autoimmunity) may be in
competition for energy. We therefore analyzed a variety of parameters involved in thermogenesis
in cold the exposed groups of mice. There was no difference in the body temperature between
the RT-kept, or cold exposed healthy vs. EAE mice (Figure S5A). While under RT, EAE reduced
the blood glucose levels, 3 hour fasting glycemia during cold was unchanged between EAE and
healthy mice (Figure S5B). To further address the functional properties of the BAT and SAT
during cold, we studied the oxygen consumption rates, which were similar between the cold
exposed EAE and healthy mice in both SAT and BAT (Figures S5C, D). Consistent with the lack
of functional differences, the weights of the BAT and SAT depots of the EAE mice compared to
the healthy cold exposed controls were unchanged (Figures S5E). While the expression of Cidea
and Pparg in the BAT was reduced in the cold exposed EAE mice, the rest of the brown fat
markers were unaltered, and there was no difference in expression in any of the browning markers
in the inguinal SAT (Figures S5F, G). Moreover, we detected no changes in the lipid droplet size
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
11
distribution between cold exposed healthy or EAE mice, together suggesting no detectable EAE-
induced limitation of the BAT and SAT thermogenic response during cold. However, the slightly
milder brown adipose tissue marker activity under EAE, but without overall reduction in the
thermogenesis and oxygen consumption rates may suggest competition for energetic resources
between of the two energy costly programs. Intriguingly, the lack of functional limitation in the
thermogenic response to cold, coupled to decreased neuroinflammation indicates prioritization of
the homeothermic response over the autoimmunity, resulting in ameliorated EAE severity.
Cold exposure activates BAT thermogenesis primarily through UCP1, which uncouples the
mitochondrial respiratory chain activity from the ATP biosynthesis. However, during prolonged
acclimatization to cold, the Ucp1-KO mice are able to compensate for the lack of UCP1 through
be additional UCP1-independent thermogenic mechanisms that can satisfy the thermogenic
needs, but at a very high energetic cost (Ikeda et al., 2017). Surprisingly, Ucp1 deletion rendered
mice even more protected from EAE compared to their wildtype littermates (Figure S5I),
associated with increased BW of the Ucp1 -KO compared to their WT littermates during cold and
EAE, consistent with the overall improved health status. These data demonstrate that the UCP1-
dependent BAT activity is dispensable for these effects. They also show that cold exposure
ameliorates EAE, but without a cost on the overall homeothermic response.
Cold Exposure Decreases Monocyte and T Cell Pathogenicity during EAE
Monocytes play an essential role in EAE development, progression and remission (Ajami et al.,
2011; Fife et al., 2000; Izikson et al., 2000; King et al., 2009; Mildner et al., 2009; Serbina and
Pamer, 2006), and they reach the CNS in inflammatory conditions through the blood stream. To
examine whether cold exposure affects monocytes in the circulation during EAE, we performed
RNA sequencing on blood monocytes after FACS sorting (Figure S5A, S6A, B) at the EAE onset.
Fold change analysis (p<0.05, logFC>1) revealed 660 up- and 86 downregulated genes of 10794
total genes (Figures 5A) in cold exposed compared to room temperature mice. Metacore
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
12
Metabolic Network analysis (p<0.05) showed 23 enriched networks (Figure 5B). Among the 3366
enriched terms identified using Metacore GO, the top 10 terms were mainly metabolic changes
(Figure 5C). Interestingly, Gene Ontology Cellular Components identified “MHC class II protein
complex” among the top 5 of 10 total enriched terms (Figure 5B middle). Within the Reactome
pathway analysis, pathways important for antigen presentation were within the 4 identified hits
(Figure 5B bottom).
The changes in MHCII antigen presentation machinery in blood monocytes at EAE onset between
the cold exposed and the room temperature mice were confirmed by flow cytometry (Figure S6C).
While the percentage of Ly6Chi/int/lo monocytes remained the same (Figure 5D), cold exposure led
to decreased MHCII of Ly6Chi monocytes, coupled to a partial lowering (p=0.069) in the
percentage of MHCII+Ly6Chi M within the total MHCII+ cells but no change in the percentage of
total MHCII expression of CD45+ cells (Figure 5E). Blood and lymph node immune cells at
different days of EAE were further investigated using high dimensional flow cytometry and
unbiased computed analysis and detected no alterations thus excluding changes in the myeloid
cell populations (Figures S6D and S6E). These analyses demonstrate that cold exposure reduced
MHCII expression of circulating Ly6Chi monocytes at EAE onset without shifting myeloid cell
populations.
Circulating Ly6Chi monocytes contribute to EAE pathology by migration to the inflamed CNS
(Ajami et al., 2011; Fife et al., 2000; Izikson et al., 2000; King et al., 2009; Mildner et al., 2009;
Serbina and Pamer, 2006). We therefore investigated if the changes detected in the circulating
monocytes may be seen within the inflamed CNS at EAE onset. Cold exposure decreased the
percentage of Ly6Chi monocytes/monocyte derived cells and their MHCII expression within the
CNS (Figure 5F). Furthermore, their Arg1 expression was increased, indicating a predominant
anti-inflammatory M2-like phenotype (Figure 5F). Percentage and MHCII expression of microglia
were similarly decreased (Figure 5G), together suggesting that they are activated to a lower extent
at EAE onset under cold exposure.
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
13
Apart from monocytes and monocyte-derived cells, T cells are the main drivers of EAE
pathogenicity. Cold exposure led to a decrease of pathogenic T cell cytokine signature and a
reduction of GM-CSF/IL-17 and IFNg/IL-17 double producers in the CNS (Figure 5H). We
detected similar changes in the dLN (Figure 5I), whereas in the mesenteric lymph nodes only GM-
CSF was decreased (Figure S6F upper), and no changes were seen in the spleen (Figure S6F
bottom). In the dLN, MHCII expression of Ly6Chi monocytes was lower on day two (Figure 5J) but
not at onset of EAE, or at steady state (data not shown). There were no consistent differences in
dendritic cell populations or their activation status in the dLN at any time (Figure S6G).
Collectively, these data show that cold exposure attenuates Ly6Chi monocyte and T cell
pathogenicity during active EAE.
T Cell Priming by Monocytes is Critical for the Cold-Induced Attenuation of EAE
The described changes in two of the main pathogenic cell types mediating EAE provide
mechanistic support for the observed amelioration of EAE by cold exposure. We therefore
investigated what is the connection between monocytes and T cells during cold and EAE. Since
cold exposure reduced T cell cytokine expression early during EAE and the monocyte changes
were already detected under steady-state conditions, we raised the questions whether cold
exposure affects the priming of T cells and what is the role of monocytes at regulating this stage.
To investigate this, we used an adoptive transfer model for passive EAE induction, in which in
vitro Th1 polarized, transgenic, MOG-specific 2D2 T cells were injected into room temperature or
cold exposed mice (Figure 6A). Similar to the active EAE, cold exposure reduced MHCII
expression of Ly6Chi blood monocytes during the passive EAE onset after transfer of differentiated
2D2 cells, while monocyte percentages remained unchanged (Figures 6B-D and 6E). Priming T
cells in vitro completely abolished the cold exposure-induced amelioration of EAE (Figures 6F-K
upper). Similar results were obtained after adoptive transfer of Th17 differentiated cells (Figures
6F-6K bottom). These data suggest that the effects of cold exposure during the T cell priming
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
14
phase are critical for the attenuation of EAE, and that cold exposure mediated MHCII
downregulation may be responsible for these effects. These conclusions were supported with
data from cold exposed mice that were shifted to room temperature on the day of immunization.
Transferring cold exposed mice to room temperature prior to the T cell priming stage (Figure S7A)
abolished the cold-induced effects on the active EAE disease (Figure S7B) and MHC expression
on monocytes (Figure S7C). This is in agreement with the observation that the MHCII
downregulation in monocytes under cold exposure was not sufficient to ameliorate EAE following
adoptive transfer of pre-activated encephalitogenic T cells, thus making it unlikely that the
observed monocyte changes are downstream mediators of effector T cells.
To directly test whether monocytes may influence T cells during their priming phase in cold, we
used two complementary approaches to deplete peripheral monocytes: anti-CCR2 (MC-21)
antibody and Ccr2-KO mice, both of which restrict monocytes to the bone marrow (Serbina and
Pamer, 2006). While anti-CCR2 completely abolished peripheral blood monocytes 2 days before
EAE onset, Ccr2-KO mice showed significant, but incomplete loss of monocytes from the blood
(Figure 6L), likely because during inflammation there could be a compensatory, CCR2-
independent mechanisms for partial monocyte exit from the bone marrow. While cold exposure
decreased pathogenic T cell cytokine expression in the isotype control mice, both monocyte
depletion mouse models did not show any T cell cytokine expression in the dLN (Figure 6 M)
irrespective of the housing temperature. Together, these data show that monocytes play a critical
role in T cell priming with subsequent implications on the T cell encephalitogenic capacities.
DISCUSSION
Our study has several fundamental implications for understanding the immune system functioning
in relation to an energy-demanding metabolic stimulus, such as the change in the environmental
temperature. The work provides a systematic overview of the immune cell changes in bone
marrow and blood as a consequence of cold exposure during steady-state, and shows that lower
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
15
environmental temperature markedly ameliorates neuroinflammation. The cold-induced gene
expression alterations were accompanied by phenotypic and functional changes of monocytes.
Reduced MHC class II expression and related pathways in these cells was accompanied with
lowered antigen presenting capacity following various inflammatory stimuli. This cold-induced
monocyte modulation resulted in reduced priming of autoreactive T cells, leading to attenuated
autoimmune CNS disease. These data reveal a hitherto underestimated role of monocytes during
T cell priming in autoimmunity.
Circulating Ly6Chi monocytes can acquire antigen presenting functions and can exert pro- and
anti-inflammatory tasks (Jakubzick et al., 2017). They therefore play an important role during
infections but are also involved in autoimmune pathogenesis. The importance of monocytes and
monocyte-derived cells during the effector phase of EAE is well established (Fife et al., 2000;
Izikson et al., 2000; King et al., 2009; Serbina and Pamer, 2006), and in particular the relevance
of migration of Ccr2+ monocytes into the CNS (Fife et al., 2000). This is in line with our observation
that cold exposure reduces monocyte-derived cells during EAE in the CNS. However, our data
also point out that the noted ameliorated EAE disease under cold exposure was associated with
a reduced generation of encephalitogenic T cells, which was most prominent during the early
phase of the disease. This suggests a cold-induced effect on the priming of naïve autoreactive
CD4+ T cells, since adoptive transfer of in vitro pre-activated 2D2 T cells abolished the observed
protection on EAE. While dendritic cells are supposed to be the primary antigen presenting cells
involved in T cell priming (Ganguly et al., 2013; Steinman and Banchereau, 2007), we did not find
indications that cold-exposure impacted dendritic cell function per se. Instead, our data rather
suggest that altered monocytic function was implied in the reduced priming of autoreactive T cells
under cold. Although it is known that monocytes can present antigens to T cells, their role in the
context of CNS autoimmunity for T cell priming remains debated (Guilliams et al., 2018; Jakubzick
et al., 2017). One previous study (Ko et al., 2014) described decreased but not abrogated IL-17
expression in Ccr2-DTR mice at EAE onset, and did not report differences in IFNg or GM-CSF.
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
16
However, the Ccr2+ cell depletion was induced transiently and after immunization, and therefore
likely did not address the initial priming phase. Another study found a minor decrease in IFNg/GM-
CSF double producing CD4+ T cells, but no abrogation and no effect on IL-17 in Ccr2-KO mice
before EAE onset (Ronchi et al., 2016). This mild effect on T cell pathogenicity could likely be
explained by the experimental conditions, or by an incomplete monocyte depletion from the
circulation in the Ccr2-KO mice under inflammatory conditions. Indeed, the monocyte depletion
in our study was milder in the Ccr2-KO compared to the anti-CCR2 treated mice. We observed
complete abrogation of pathogenic T cell cytokines after acute depletion of monocytes using the
antibody mediated approach – effects also evident in our genetic, Ccr2-KO mice.
In addition to the decrease in monocyte-derived cells, cold exposure led to reduction of monocyte
MHCII expression in bone marrow, blood and CNS at steady state, or during the early phase of
EAE, which provides a mechanistic link to the observed reduction in T cell priming and
consequently ameliorated disease course. The fact that MHCII in monocytes remained reduced
even after adoptive transfer of pre-activated 2D2 cells that abrogated the protective effect of cold
on EAE, strengthens the conclusions that the cold-induced monocyte changes interfere upstream
of T cell effector cells. We excluded other potential mechanisms by which monocytes could affect
T cell priming - neither the uptake and transport of antigen by monocytes in FITC painted mice,
nor monocyte cytokine expression on transcriptional level at steady-state and EAE onset were
modified under cold. Previous studies have suggested a positive feedback loop between T cells
and monocytes, where T cells secrete GM-CSF to induce monocyte IL-1b expression (Croxford
et al., 2015), which in return amplifies expansion of GM-CSF+ Th17 cells (Mufazalov et al., 2017).
Our study adds an additional layer to this feedback loop, stressing that as an initial step, monocyte
MHCII is critical for T cell priming.
Cold exposure induces beta adrenergic stimulation of the BAT to activate lipolysis, glucose uptake
and mitochondrial biogenesis, and is extensively studied in context of obesity and metabolic
disorders (Jastroch, 2017). Cold-induced BAT activation and white fat browning result in a
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
17
thermogenic response that is energy demanding. As such, this metabolic response may require
an energetic trade-off that competes with other energy costly programs, as for instance immune
responses (Wang and Medzhitov, 2019). Several studies addressed the energy trade-off concept
in the context of immune defense during infections and found that energy conserving
hypometabolic states may favor the utilization of tissue tolerance as a defense against bacterial
pathogens (Ganeshan et al., 2019; Weis et al., 2017). Similar to cold, caloric restriction also
induces negative energy balance, reduces and modulates circulating monocytes (Jordan et al.,
2019), and lowers CNS monocytes and their pathogenicity (Cignarella et al., 2018; Piccio et al.,
2008). In contrast to caloric restriction that requires an intervention to reduce food intake, cold
exposure ameliorates EAE despite the excessive caloric uptake. Certainly, a competition between
thermogenesis and a destructive immune response would be favorable with regard to
autoimmune diseases, as we see in our study. It was speculated that at low temperatures,
defense of remaining thermogenesis over immunity may take place (Wang and Medzhitov, 2019).
Our study is in line with this scenario and shows that cold exposure dampens autoimmunity, thus
providing evidence and a conceptual advance in our understanding of the energetic trade-off at
the cost of autoimmunity. In support of these conclusions, cold exposure ameliorates EAE to a
higher extend in absence of UCP1 and following acclimatization and activation of the UCP1-
independent thermogenic mechanisms, which function at a very high energetic cost (Ikeda et al.,
2017; Reynes et al., 2019; Roesler and Kazak, 2020). The cold-induced immune changes seem
to originate already in the bone marrow and can be partially provoked by beta adrenergic agonism,
which may suggest contribution of the nervous system to the energy trade-off. With regard to this
concept, it is important to stress that while evidently protective against autoimmunity, cold
exposure increases susceptibility to certain viral infections (Foxman et al., 2015; Jaakkola et al.,
2014). Thus, our work could be relevant not only for CNS autoimmunity, but also other immune-
mediated or infectious diseases, which warrants further investigations.
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
18
Our study reveals that cold exposure orchestrates immunologic reprogramming that protects from
CNS autoimmunity, showing that monocytes and the monocyte MHCII expression are crucially
implicated during T cell priming in the course of an autoimmune response. These findings provide
critical insights into the pathogenic mechanisms of neuroinflammation, and could lead to future
preventive and therapeutic approaches for MS and other autoimmune diseases.
ACKNOWLEDGEMENTS
We thank the iGE3 Genomics Platform at the University of Geneva for mRNA sequencing and
mapping reads, the Flow Cytometry core facility at the CMU of the University of Geneva for help
with flow cytometry, Christelle Veyrat-Duberex, Florian Visentin, Melis Colakoglu, Giovanni di
Liberto, Salvatore Fabbiano and Karim Hammad for experimental assistance, Sergei Startchik for
the help on automatized lipid droplet quantification, and Stephanie Hughes and Pierre
Guermonprez for organizing and providing B16-GMCSF cells, respectively, Thomas Korn for the
Th17 EAE protocol and Claes Wollheim for discussions. This work was supported by the
CONFIRM grant of the Hôpitaux Universitaires de Genève (HUG) Foundation (no. RC2-09) and
the Swiss Multiple Sclerosis Society Grant to DM and MT; the European Research Council (ERC)
under the European Union’s Horizon 2020 research and innovation programme (ERC
Consolidator Grant agreement No. 815962) to MT; Swiss National Science Foundation (SNSF)
grant to DM (310030_173010); and SNSF Professorship grants (PP00P3_144886 and
PP00P3_172906) to MT.
AUTHOR CONTRIBUTIONS
M.S. set up and performed experiments, analyzed and interpreted data, and drafted figures and
manuscript. K.S. set up and performed experiments, and analyzed and interpreted data. N.H. and
M.S. performed RNA-seq analysis. D.R., N.S.Z., M.A.R., C.C., I.V., N.P., B.K. and O.S. helped
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
19
with experiments. I.W. assisted in histological processing. M.G. assisted in setup of and D.C.
performed and analyzed FACSymphony. M.K. developed and applied computer-assisted image
analysis algorithms. MM provided the MC-21 antibody. D.M. and M.T. conceptualized, designed
and supervised the project, interpreted data, and wrote the manuscript.
DECLARATION OF INTERESTS
The authors declare no competing interests.
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
20
FIGURE LEGENDS
Figure 1. Cold Exposure Impacts Monocytes in the Bone Marrow
(A) Volcano plot showing the up- and downregulated transcripts by RNA sequencing in the bone
marrow of cold exposed mice for 2 weeks at 10°C compared to room temperature counterparts
(n=6 mice per group).
(B) Metacore Gene Ontology Processes analysis displaying the top 5 enriched processes of mice
as in (A).
(C) Shown are 3 of 4 heatmaps that were regulated in the same direction (> 80% of genes in the
same direction) from the top 10 differentially regulated Metacore Pathway Maps of mice as in (A).
Full pathway names are “Macrophage and dendritic cell phenotype shift in cancer”, “Immune
response_IFN-aplha/beta signaling via JAK/STAT” and “G-CSF induced myeloid differentiation”.
(D) Flow cytometry analysis of bone marrow cells of mice as in (A). Percentage of Ly6C high (hi),
intermediate (int) and low (lo) monocytes of total, single CD45+ alive cells.
(E) Percentage of MHCII+ cells of Ly6Chi monocytes (left), MHCII+ Ly6Chi monocytes of total
MHCII+ cells (middle) and total MHCII+ cells of CD45+ cells of mice as in (A), as determined by
flow cytometry.
(F) Bone marrow immune cell progenitors of mice as in (A) were analyzed by flow cytometry and
percentage of total alive single CD45+ cells is shown for common dendritic cell progenitors (CDP),
cMoP (common monocyte progenitors), monocyte-dendritic cell progenitors (MDP), common
lymphoid progenitors (CLP) and Lin- Sca1+ c-KIT+ cells (LSK), as determined by flow cytometry.
(G) Immune cell progenitor flow cytometry analysis of mice that were i.p. injected with beta 3
adrenoreceptor agonist CL316,243 (CL316) or vector (PBS) daily during one week. Percentage
of total alive CD45+ cells is shown of CDP, cMoP, MDP, CLP, LSK and Ly6Chi monocytes.
(B-C) The cutoff on differentially regulated genes considered for the pathway analysis is p<0.05
and pathways are considered deregulated with p<0.05. Shown are –log(p-value).
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
21
(D-G) Each dot represents one animal. Shown is mean ±SD. Significance was calculated using
student’s t test: p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001. Pool of five experiments (D), two experiments
(E), one representative experiment of three out of five that were similar (F) and one representative
of two are shown.
Figure 2. Cold Exposure Modulates Monocytes in the Circulation
(A) Volcano plot showing the up- and downregulated transcripts by RNA sequencing of
monocytes that were MACS (anti-PE) and FACS (CD115-PE+) sorted from blood of 2 weeks cold
exposed (10°C) mice compared to room temperature mice.
(B) 15 most enriched Gene Ontology Biological Processes of monocytes as in (A). Genes were
considered differentially regulated with p<0.05. Shown are –log(p-value).
(C) Blood immune cells of mice as in (A) visualized using UMAP and clustered using FlowSOM
algorithm in R. Ly6Chi monocytes (green), Ly6Clo monocytes (yellow), neutrophils (brown),
eosinophils (orange) and others. Heatmap shows median relative expression of all panel markers.
(D) Flow cytometry analysis of blood cells from mice as in (A). Percentage of Ly6C high (hi),
intermediate (int) and low (lo) monocytes of total, single, CD45+ cells.
(E) Percentage of blood MHCII+ cells of Ly6Chi monocytes (left), MHCII+ Ly6Chi monocytes of total
MHCII+ cells (middle) and total MHCII+ cells of total, single CD45+ cells of mice as in (A).
(F) Blood cell analysis after 1 week of daily i.p. injected beta 3 adrenoreceptor agonist CL316,243
or vector (PBS). Percentage of MHCII+ cells of Ly6Chi monocytes (left), MHCII+ Ly6Chi monocytes
of total MHCII+ cells (middle) and total MHCII+ cells of CD45+ cells.
(D-F) Each dot represents one animal. Shown is mean ±SD. Significance was calculated using
student’s t test: p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001. Pool of three experiments (D, E).
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
22
Figure 3. Cold Exposure Decreases Monocyte MHCII in Inflammatory Mouse Models
(A) Blood Ly6Chi monocytes and their MHCII expression were analyzed by flow cytometry on
different days after B16-GMCSF s.c. tumor implantation in mice that were cold (10°C) or room
temperature exposed for 2 weeks before and during tumor growth.
(B) Blood CD11c+MHCII+ cells and MHCII mean fluorescence intensity (MFI) of MHCII+CD11c+
cells were determined by flow cytometry from mice as in (A).
(C) Flow cytometry analysis of peritoneal fluid cells 24h after i.p. injection with thioglycollate into
2 weeks cold exposed (10°C) or room temperature mice.
(A-C) Each dot represents one animal. Shown is mean ±SD. Significance was calculated using
multiple t-test with Holm-Sidak correction (A, B) or student’s t test: (C), p < 0.05, ∗∗p < 0.01.
Figure 4. Cold Exposure Attenuates Neuroinflammation
(A-B) Scheme showing the experimental setup for EAE and cold exposure (A). EAE was induced
by s.c. immunization with MOG35-55 peptide in complete Freund’s adjuvant (day 0) and pertussis
toxin (day 0 and 2). Mice were exposed to cold or room temperature (10°C) for two weeks before
and during EAE. Clinical symptoms of EAE were monitored according to a standardized scoring
system (B).
(C-G) Day of EAE onset, i.e. when first symptoms occur (C), maximum disease score, i.e. the
highest score an individual mouse reached during the experiment (D), cumulative disease score,
i.e. the sum of all scores each individual mouse reached during the experiment (E), the
percentage of disease free mice (F) and body weight (G) of mice as in (A).
(H) Quantification of demyelinated area expressed as percentage of white matter (WM) detected
by Luxol Fast Blue and Periodic acid–Schiff (LFB-PAS) of spinal cords from mice as in (A).
(I-M) Spinal cords of healthy and peak EAE mice that were kept at room temperature or 2 weeks
cold exposed were used for RNA sequencing. Scores of the respective mice at sacrifice (I),
principal component analysis (J), relativeness analysis of logFC of room temperature healthy vs
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
23
EAE spinal cords (red) and cold exposure healthy vs EAE spinal cords (blue) with p<0.05, FC>2
(K), volcano plot (gene Gpx3 was excluded for better visualization) (L) and genes of top1
deregulated pathway “Macrophage and DC phenotype shift in cancer” from Metacore Pathway
Maps with p<0.05 (M).
(N-O) Scheme showing the experimental setup. Mice were either exposed to cold temperature
for two weeks or two days before and continued during EAE and compared to room temperature
controls (N). EAE was induced as in (A) and EAE scores are shown (O).
(P) Percentage of MHCII expression on Ly6Chi blood monocytes was determined using flow
cytometry on day zero and day four after EAE induction.
(Q-R) Scheme showing the experimental setup (Q). Mice were exposed to warm temperature of
34°C for one week before and during EAE (as in (A)) and compared to room temperature controls.
Each curve represents one individual mouse (R).
(S) Percentage of MHCII expression of Ly6Chi blood monocytes from mice as in (Q) was
determined using flow cytometry on day 0 of EAE.
(T-U) Maximum disease score (T) or delta of body weight gain from day zero to day six of warm
exposure (U).
(B-S) Data (B-G) represents pool of 3 experiments (n = 8-12 mice per group per experiment).
Shown is mean ±SEM, two-way ANOVA (B; G, O, R); student’s t test (C-E, I, S-U); Mantel-Cox
(F); multiple t-test with Holm-Sidak correction (P), mean ±SD, p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001.
Figure 5. Cold Exposure Reduces Monocyte and T Cell Pathogenicity during EAE
(A) Volcano plot identifying the up- and downregulated transcripts after RNA sequencing of
monocytes that were MACS (anti-PE) and FACS (CD115-PE+) sorted from blood of mice at onset
of EAE (as in Figure 4A) and exposed to cold (10°C) or room temperature for 2 weeks before and
continued during EAE.
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
24
(B-C) Top deregulated and enriched Metacore Metabolic Networks (B top), Gene Ontology
cellular components (B middle), Reactome pathways (B bottom) and Gene Ontology Processes
(C). Genes were considered when p<0.05 and for Reactome pathways in addition FC>2. Data of
sequencing as in (A).
(D-E) Flow cytometry analysis of blood cells of mice as in (A) at EAE onset. Percentage of Ly6C
high (left), intermediate (middle) and low (right) monocytes of total, single, CD45+ alive cells (D).
Percentage of MHCII+ cells of Ly6Chi monocytes (left), MHCII+ Ly6C+ monocytes of total MHCII+
cells (middle) and total MHCII+ cells of CD45+ cells (E). Pool of 3 experiments.
(F-H) Flow cytometry analysis of CNS cells from mice as in (A) at EAE onset. Percentage of
Ly6Chi monocytes/monocyte-derived cells of total, single, CD45+ alive cells (F, left). Percentage
of MHCII, arginase 1 (Arg1) and iNOS expression of Ly6Chi monocytes/monocyte-derived cells
(F, right). Percentage of microglia of total, single, CD45+ alive cells and percentage of MHCII of
microglia (G). Percentage of corresponding cytokine expression as indicated in CD4+ T cells (H).
(I) Flow cytometry analysis of dLN cells from mice as in (A) at EAE onset. Percentage of cytokine
expression in CD4+ T cells. Observed in 2 out of 3 experiments.
(J) Flow cytometry analysis of dLN cells from mice as in (A) on day two after EAE induction.
Percentage of Ly6Chi monocytes of total, single, CD45+ alive cells (upper panel). Percentage of
MHCII on Ly6Chi monocytes (lower panel).
(D-J) Shown is mean ±SD, significance was calculated using student’s t test, * p<0.05, ** p<0.01,
***p<0.001. Shown is 1 representative example of 3 (F), a pool of 2 experiments (H) or one
representative experiment of two out of three that were similar (I).
Figure 6. Monocyte Regulation of the T Cell Priming is Critical for Cold-Induced EAE
Attenuation
(A) Scheme showing experimental setup for cold exposure and adoptive transfer EAE. T cells
from transgenic 2D2 mice or actively immunized wildtype donor mice were in vitro differentiated
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
25
towards Th1 or Th17 respectively and transferred into room temperature or cold exposed (2
weeks, 10°C) mice.
(B-E) Flow cytometry analysis of blood cells of mice as in (A) at EAE onset. Percentage of Ly6Chi
(B), intermediate (C) and low monocytes of total, single, CD45+ cells (D). Percentage of MHCII+
cells of Ly6Chi monocytes (E).
(F-K) EAE symptoms of mice as in (A) were scored (F). Onset day of EAE disease (G), maximum
score (H), cumulative score (I), percentage of disease free animals (J) and body weight (K). All
panels are shown following adoptive transfer of Th1 (upper) and Th17 (lower) cells.
(L-M) Isotype control antibody, CCR2 antibody injected or Ccr2-knockout (KO) mice were housed
at room temperature or cold (10°C) for 2 weeks before and during EAE and were s.c. immunized
with MOG35-55 peptide in complete Freund’s adjuvant. Two days before disease onset,
monocyte depletion efficiency was analyzed in the blood (L) and at onset T cell cytokine
expression in the draining lymph nodes (M) via flow cytometry. Data represents mean ± SD,
significance was calculated using multiple t-test with Holm-Sidak correction, *p<0.05, **p<0.01,
***p<0.001.
(B-E, F-K, L-M) Pool of 3 experiments (B-E, F-K upper panel), student’s t test with mean ± SD (B-
E, G-I), two-way ANOVA with mean ± SEM (F,K) or Mantel-Cox (J). Multiple t-tests with Holm-
Sidak correction with mean ±SD, p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001 (L-M).
Supplemental Figure Legends
Figure S1. (Related to Figure 1)
(A) Multidimensional scaling and principal component analysis of the bone marrow from mice
after 2 weeks of cold exposure at 10°C compared to room temperature mice.
(B) 10 most enriched Metacore Metabolic Networks of mice as in (A).
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
26
(C) Metacore Pathway Maps analysis identifying the 10 most differentially regulated pathways of
mice as in (A).
(D) 1 of 4 heatmaps that were consistently changed (> 80% of genes in the same direction) from
the top 10 deregulated Metacore Pathway Maps of mice as in (A).
(E) Absolute lymphocyte numbers within the bone marrow of mice as in (A) as determined by flow
cytometry. Shown is a pool of 5 experiments, mean ±SD.
(F) Bone marrow monocytes were gated as live, single, CD45+, Ly6G-, CD11b+, CD115+, Ly6C+
cells and MHCII+ population determined. Bone marrow progenitors were all gated as single, live,
CD45+, Lin- (NK1.1, CD3, CD19, Ter119, Ly6G) and CD11b-, Sca1+, CD117hi, CD115+, Ly6Chi
(cMoP), CD11b-, Sca1+, CD117hi, CD115+, Ly6C- (MDP), CD11b-, Sca1+, CD117-, CD115+,
CD135+ (MDP), CD117+, CD127+, Sca-1+ (CLP), Sca-1+, CD117+ (LSK).
(C-D) The cutoff on differentially regulated genes considered for the pathway analysis is p<0.05
and pathways are considered deregulated with p<0.05. Shown are –log(p-value).
Figure S2. (Related to Figure 2)
(A) Gating strategy for the FACS (CD115-PE+) sort after MACS (anti-PE) sort before RNA
sequencing from blood of 2 weeks cold exposed (10°C) compared to room temperature mice.
Sorted were DAPI-, single, CD115+ cells.
(B) Multidimensional scaling (left) and principal component analysis (right) after RNA sequencing
of monocytes as in (A).
(C-E) 10 most differentially regulated Metacore Pathway Maps without p-value cutoff on
considered genes (C), Metacore Metabolic Networks (D) and Gene Ontology Cellular
Components (E). Genes were considered as differentially regulated genes with p<0.05 and
pathways are considered deregulated or enriched with p<0.05. Shown are –log(p-value).
(F) For flow cytometry analysis, blood monocytes were gated as Ly6G-, CD11b+, CD115+,
Ly6Chi/int/lo and MHCII+ positive population was gated.
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
27
Figure S3. (Related to Figure 3)
(A) Peritoneal fluid cells were analyzed by flow cytometry 24h after i.p. injection with thioglycollate
into two weeks cold exposed (10°C) or room temperature mice. Shown is mean ±SD.
(B) Flow cytometry analysis of lymph node draining cells 12 and 18 hours after FITC skin painting
on one flank of 2 weeks cold exposed (10°C) or room temperature mice. Percentage of panDCs
(upper) and Ly6Chi monocytes (bottom), their FITC uptake and MHCII expression. Shown is mean
±SD, two-way ANOVA. Every dot represents one animal and the experiment is representative of
two.
Figure S4. (Related to Figure 4)
(A) Individual EAE (as in Figure 4A) curve of each room temperature and cold exposed mouse.
(B-G) Histology on sagittal spinal cord sections of cold exposed and room temperature peak EAE
mice. Number of lesions that were found in both LFB-PAS and APP staining were quantified in
APP staining (B). Immunohistochemistry using anti-mac3 (C) and anti-CD3 antibody (D). APP
staining was quantified within areas identified as demyelinated in LFB-Pas staining and presented
as percentage per white matter area (E). EAE disease score at sacrifice of mice in which histology
was performed (F). LFB-Pas stain shows demyelinated areas within the white matter in purple
(first panel), anti-mac3 stains mac3+ macrophages (second panel), anti-CD3 stains T cells in
brown (third panel) and amyloid precursor protein (APP) accumulates at axons with impaired
axonal transport, a surrogate for acute axonal damage (fourth panel). Arrows identify punctuated
APP-positive axonal spheroids. (G). Anti-mac3, anti-CD3 and anti-APP stainings were developed
with DAB and nuclei counterstained with hemalum.
(H, I) Multidimensional scaling analysis of spinal cord from mice as in Figure 4J (H). Top 35
Metacore Metabolic Network pathways of mice with EAE, genes with p<0.05 were included for
analysis (I).
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
28
(J-O) EAE disease curve of warm exposed (34°C, started one week before immunization) or room
temperature mice. Arrows indicate individual mice that died or had to be sacrificed, which received
a score of five on the day of death (J). Body weight during the course of EAE (K). Food intake of
two mice per cage within 24 h on day 6 of warm exposure (L) and body weight on day six of warm
exposure (M), before immunization. Survival of mice during EAE (N) and day of EAE onset (O).
(B-F, J-O) Shown is mean ±SEM, two-way ANOVA (J, K); Mantel-Cox (N), student’s t test (B-F,
L-M, O) with mean ±SD, p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001.
Figure S5. (Related to Figure 5)
(A) Body temperature shown is the mean temperature of the two eyes as determined via infrared
images of mice on day 7 of EAE and steady state. Mice were cold exposed for 2 weeks before
and during EAE or steady state. Control mice were at room temperature for the same duration.
(B) Glycemia of mice as in (A) on day 8 of EAE after 3 hours of fasting.
(C-D) Oxygen consumption rate of interscapular brown (BAT, C) and inguinal subcutaneous
adipose tissue (SAT, D) on day 8 of EAE or in steady state after 3 weeks of cold exposure (CE)
and 3 hours of fasting. Technical replicates were used for BAT.
(E) Adipose tissue weights of interscapular brown (BAT, left), inguinal subcutaneous adipose
tissue (SAT, middle) and perigonadal visceral adipose tissue (VAT, right) of mice as in (C).
(F-G) mRNA expression of browning markers relative to B2m and 36b4 in interscapular brown
(BAT, F) and inguinal subcutaneous adipose tissue (SAT, G) of mice as in (B) as determined by
qPCR.
(H) Quantification of lipid droplet size distribution in ingSAT. Slides were analyzed in technical
duplicates from different layers of the tissue and averaged.
(I) Symptoms of EAE (left) and body weight (right) in Ucp1-knockout (KO) and their respective
wildtype (WT) littermate controls housed at room temperature or cold for 1 week before and during
EAE. Shown is 1 representative (n=10-15 per group) of 2 experiments.
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
29
(A-G, I) Multiple t-test with Holm-Sidak correction (A-B, F-G), student t-test (C, D), mean ±SD,
two-way ANOVA with mean ±SEM (I), p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001.
Figure S6. (Related to Figure 6)
(A) Gating strategy for the FACS sort after MACS sorting before RNA sequencing (as in Figure
6A). Sorted were DAPI-, CD115+, single cells.
(B) Multidimensional scaling and principal component analysis of FACS sorted blood monocytes
at EAE onset as in Figure 6A.
(C) Gating strategy for MHCII expression on Ly6Chi blood monocytes of mice as in Figure 6A.
Cells were gated as single, Ly6G-, CD115+CD11b+, Ly6Chi, MHCII+.
(D-E) Blood (D) and lymph node (E) immune cells of mice as in Figure 6A on different days after
immunization were visualized using UMAP and clustered using the FlowSOM algorithm in R.
Heatmap shows median relative expression of all panel markers.
(F) Flow cytometry analysis of mesenteric LN (upper panel) or spleen cells (lower panel) of mice
as in Figure 6A at EAE onset. Percentage of cytokine expression in CD4+ T cells. Shown is mean
±SD, significance was calculated using student’s t test, * p<0.05.
(G) Flow cytometry analysis of lymph node dendritic cells and their MFI of MHCII on different days
after immunization.
Figure S7. (Related to Figure 6)
(A-B) Scheme showing experimental setup for cold exposure and EAE (A). Cold exposure either
started two weeks before and continued during EAE (blue, CE) or switched to room temperature
from the day of immunization (violet, CE-RT) and compared to room temperature mice (red, B).
EAE disease curve shown as mean ±SEM, two-way ANOVA, p < 0.05.
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
30
(C) Percentage of MHCII expression of Ly6Chi blood monocytes via flow cytometry on day zero,
day three and day seven after EAE induction. Multiple t-test with Holm-Sidak correction with mean
±SD, p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001.
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
31
REFERENCES
Ajami, B., Bennett, J.L., Krieger, C., McNagny, K.M., and Rossi, F.M. (2011). Infiltrating
monocytes trigger EAE progression, but do not contribute to the resident microglia pool. Nat
Neurosci 14, 1142-1149.
Allan, R.S., Waithman, J., Bedoui, S., Jones, C.M., Villadangos, J.A., Zhan, Y., Lew, A.M.,
Shortman, K., Heath, W.R., and Carbone, F.R. (2006). Migratory dendritic cells transfer antigen
to a lymph node-resident dendritic cell population for efficient CTL priming. Immunity 25, 153-
162.
Buck, M.D., Sowell, R.T., Kaech, S.M., and Pearce, E.L. (2017). Metabolic Instruction of
Immunity. Cell 169, 570-586.
Cannon, B., and Nedergaard, J. (2004). Brown adipose tissue: function and physiological
significance. Physiol Rev 84, 277-359.
Cignarella, F., Cantoni, C., Ghezzi, L., Salter, A., Dorsett, Y., Chen, L., Phillips, D., Weinstock,
G.M., Fontana, L., Cross, A.H., et al. (2018). Intermittent Fasting Confers Protection in CNS
Autoimmunity by Altering the Gut Microbiota. Cell Metab 27, 1222-1235 e1226.
Croxford, A.L., Lanzinger, M., Hartmann, F.J., Schreiner, B., Mair, F., Pelczar, P., Clausen,
B.E., Jung, S., Greter, M., and Becher, B. (2015). The Cytokine GM-CSF Drives the
Inflammatory Signature of CCR2+ Monocytes and Licenses Autoimmunity. Immunity 43, 502-
514.
Davies, L.C., Rosas, M., Jenkins, S.J., Liao, C.T., Scurr, M.J., Brombacher, F., Fraser, D.J.,
Allen, J.E., Jones, S.A., and Taylor, P.R. (2013). Distinct bone marrow-derived and tissue-
resident macrophage lineages proliferate at key stages during inflammation. Nat Commun 4,
1886.
Demas, G., and Nelson, R. (2012). Ecoimmunology (Oxford University Press).
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
32
Dobin, A., Davis, C.A., Schlesinger, F., Drenkow, J., Zaleski, C., Jha, S., Batut, P., Chaisson,
M., and Gingeras, T.R. (2013). STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29,
15-21.
Dranoff, G., Jaffee, E., Lazenby, A., Golumbek, P., Levitsky, H., Brose, K., Jackson, V.,
Hamada, H., Pardoll, D., and Mulligan, R.C. (1993). Vaccination with irradiated tumor cells
engineered to secrete murine granulocyte-macrophage colony-stimulating factor stimulates
potent, specific, and long-lasting anti-tumor immunity. Proc Natl Acad Sci U S A 90, 3539-3543.
Fife, B.T., Huffnagle, G.B., Kuziel, W.A., and Karpus, W.J. (2000). CC chemokine receptor 2 is
critical for induction of experimental autoimmune encephalomyelitis. J Exp Med 192, 899-905.
Foxman, E.F., Storer, J.A., Fitzgerald, M.E., Wasik, B.R., Hou, L., Zhao, H., Turner, P.E., Pyle,
A.M., and Iwasaki, A. (2015). Temperature-dependent innate defense against the common cold
virus limits viral replication at warm temperature in mouse airway cells. Proc Natl Acad Sci U S
A 112, 827-832.
Ganeshan, K., Nikkanen, J., Man, K., Leong, Y.A., Sogawa, Y., Maschek, J.A., Van Ry, T.,
Chagwedera, D.N., Cox, J.E., and Chawla, A. (2019). Energetic Trade-Offs and Hypometabolic
States Promote Disease Tolerance. Cell 177, 399-413 e312.
Ganguly, D., Haak, S., Sisirak, V., and Reizis, B. (2013). The role of dendritic cells in
autoimmunity. Nat Rev Immunol 13, 566-577.
Giladi, A., Wagner, L.K., Li, H., Dorr, D., Medaglia, C., Paul, F., Shemer, A., Jung, S., Yona, S.,
Mack, M., et al. (2020). Cxcl10(+) monocytes define a pathogenic subset in the central nervous
system during autoimmune neuroinflammation. Nat Immunol 21, 525-534.
Grant, R.W., and Dixit, V.D. (2015). Adipose tissue as an immunological organ. Obesity (Silver
Spring) 23, 512-518.
Guilliams, M., Mildner, A., and Yona, S. (2018). Developmental and Functional Heterogeneity of
Monocytes. Immunity 49, 595-613.
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
33
Hasan, M., Seo, J.E., Rahaman, K.A., Kang, M.J., Jung, B.H., and Kwon, O.S. (2016).
Increased levels of brain serotonin correlated with MMP-9 activity and IL-4 levels resulted in
severe experimental autoimmune encephalomyelitis (EAE) in obese mice. Neuroscience 319,
168-182.
Hettinger, J., Richards, D.M., Hansson, J., Barra, M.M., Joschko, A.C., Krijgsveld, J., and
Feuerer, M. (2013). Origin of monocytes and macrophages in a committed progenitor. Nat
Immunol 14, 821-830.
Hotamisligil, G.S. (2017). Foundations of Immunometabolism and Implications for Metabolic
Health and Disease. Immunity 47, 406-420.
Ikeda, K., Kang, Q., Yoneshiro, T., Camporez, J.P., Maki, H., Homma, M., Shinoda, K., Chen,
Y., Lu, X., Maretich, P., et al. (2017). UCP1-independent signaling involving SERCA2b-
mediated calcium cycling regulates beige fat thermogenesis and systemic glucose homeostasis.
Nat Med 23, 1454-1465.
Izikson, L., Klein, R.S., Charo, I.F., Weiner, H.L., and Luster, A.D. (2000). Resistance to
experimental autoimmune encephalomyelitis in mice lacking the CC chemokine receptor
(CCR)2. J Exp Med 192, 1075-1080.
Jaakkola, K., Saukkoriipi, A., Jokelainen, J., Juvonen, R., Kauppila, J., Vainio, O., Ziegler, T.,
Ronkko, E., Jaakkola, J.J., Ikaheimo, T.M., et al. (2014). Decline in temperature and humidity
increases the occurrence of influenza in cold climate. Environ Health 13, 22.
Jakubzick, C.V., Randolph, G.J., and Henson, P.M. (2017). Monocyte differentiation and
antigen-presenting functions. Nat Rev Immunol 17, 349-362.
Jastroch, M. (2017). Uncoupling protein 1 controls reactive oxygen species in brown adipose
tissue. Proc Natl Acad Sci U S A 114, 7744-7746.
Jordan, S., Tung, N., Casanova-Acebes, M., Chang, C., Cantoni, C., Zhang, D., Wirtz, T.H.,
Naik, S., Rose, S.A., Brocker, C.N., et al. (2019). Dietary Intake Regulates the Circulating
Inflammatory Monocyte Pool. Cell 178, 1102-1114 e1117.
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
34
King, I.L., Dickendesher, T.L., and Segal, B.M. (2009). Circulating Ly-6C+ myeloid precursors
migrate to the CNS and play a pathogenic role during autoimmune demyelinating disease.
Blood 113, 3190-3197.
Ko, H.J., Brady, J.L., Ryg-Cornejo, V., Hansen, D.S., Vremec, D., Shortman, K., Zhan, Y., and
Lew, A.M. (2014). GM-CSF-responsive monocyte-derived dendritic cells are pivotal in Th17
pathogenesis. J Immunol 192, 2202-2209.
Kohlgruber, A.C., LaMarche, N.M., and Lynch, L. (2016). Adipose tissue at the nexus of
systemic and cellular immunometabolism. Semin Immunol 28, 431-440.
Li, C., Spallanzani, R.G., and Mathis, D. (2020). Visceral adipose tissue Tregs and the cells that
nurture them. Immunol Rev 295, 114-125.
Luo, Y., Chen, G.L., Hannemann, N., Ipseiz, N., Kronke, G., Bauerle, T., Munos, L., Wirtz, S.,
Schett, G., and Bozec, A. (2015). Microbiota from Obese Mice Regulate Hematopoietic Stem
Cell Differentiation by Altering the Bone Niche. Cell Metab 22, 886-894.
Man, K., Kutyavin, V.I., and Chawla, A. (2017). Tissue Immunometabolism: Development,
Physiology, and Pathobiology. Cell Metab 25, 11-26.
McCarthy, D.J., Chen, Y., and Smyth, G.K. (2012). Differential expression analysis of multifactor
RNA-Seq experiments with respect to biological variation. Nucleic Acids Res 40, 4288-4297.
McDade, T.W. (2005). Life history, maintenance, and the early origins of immune function. Am J
Hum Biol 17, 81-94.
McInnes, L., Healy, J., and Melville, J. (2018). Umap: Uniform manifold approximation and
projection for dimension reduction. arXiv preprint arXiv:180203426.
Menezes, S., Melandri, D., Anselmi, G., Perchet, T., Loschko, J., Dubrot, J., Patel, R., Gautier,
E.L., Hugues, S., Longhi, M.P., et al. (2016). The Heterogeneity of Ly6C(hi) Monocytes Controls
Their Differentiation into iNOS(+) Macrophages or Monocyte-Derived Dendritic Cells. Immunity
45, 1205-1218.
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
35
Merad, M., Sathe, P., Helft, J., Miller, J., and Mortha, A. (2013). The dendritic cell lineage:
ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed
setting. Annu Rev Immunol 31, 563-604.
Mildner, A., Mack, M., Schmidt, H., Bruck, W., Djukic, M., Zabel, M.D., Hille, A., Priller, J., and
Prinz, M. (2009). CCR2+Ly-6Chi monocytes are crucial for the effector phase of autoimmunity
in the central nervous system. Brain 132, 2487-2500.
Mufazalov, I.A., Schelmbauer, C., Regen, T., Kuschmann, J., Wanke, F., Gabriel, L.A.,
Hauptmann, J., Muller, W., Pinteaux, E., Kurschus, F.C., et al. (2017). IL-1 signaling is critical
for expansion but not generation of autoreactive GM-CSF+ Th17 cells. EMBO J 36, 102-115.
O'Neill, L.A., Kishton, R.J., and Rathmell, J. (2016). A guide to immunometabolism for
immunologists. Nat Rev Immunol 16, 553-565.
Okin, D., and Medzhitov, R. (2012). Evolution of inflammatory diseases. Curr Biol 22, R733-740.
Piccio, L., Stark, J.L., and Cross, A.H. (2008). Chronic calorie restriction attenuates
experimental autoimmune encephalomyelitis. J Leukoc Biol 84, 940-948.
Platt, A.M., Rutkowski, J.M., Martel, C., Kuan, E.L., Ivanov, S., Swartz, M.A., and Randolph,
G.J. (2013). Normal dendritic cell mobilization to lymph nodes under conditions of severe
lymphatic hypoplasia. J Immunol 190, 4608-4620.
Reynes, B., van Schothorst, E.M., Keijer, J., Palou, A., and Oliver, P. (2019). Effects of cold
exposure revealed by global transcriptomic analysis in ferret peripheral blood mononuclear
cells. Sci Rep 9, 19985.
Roesler, A., and Kazak, L. (2020). UCP1-independent thermogenesis. Biochem J 477, 709-725.
Ronchi, F., Basso, C., Preite, S., Reboldi, A., Baumjohann, D., Perlini, L., Lanzavecchia, A., and
Sallusto, F. (2016). Experimental priming of encephalitogenic Th1/Th17 cells requires pertussis
toxin-driven IL-1beta production by myeloid cells. Nat Commun 7, 11541.
Serbina, N.V., and Pamer, E.G. (2006). Monocyte emigration from bone marrow during bacterial
infection requires signals mediated by chemokine receptor CCR2. Nat Immunol 7, 311-317.
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
36
Stearns, S.C. (1992). The evolution of life histories.
Steinman, R.M., and Banchereau, J. (2007). Taking dendritic cells into medicine. Nature 449,
419-426.
Stojanovic, O., Kieser, S., and Trajkovski, M. (2018). Common traits between the beige fat-
inducing stimuli. Curr Opin Cell Biol 55, 67-73.
Timmermans, S., Bogie, J.F., Vanmierlo, T., Lutjohann, D., Stinissen, P., Hellings, N., and
Hendriks, J.J. (2014). High fat diet exacerbates neuroinflammation in an animal model of
multiple sclerosis by activation of the Renin Angiotensin system. J Neuroimmune Pharmacol 9,
209-217.
Van Gassen, S., Callebaut, B., Van Helden, M.J., Lambrecht, B.N., Demeester, P., Dhaene, T.,
and Saeys, Y. (2015). FlowSOM: Using self-organizing maps for visualization and interpretation
of cytometry data. Cytometry A 87, 636-645.
Wang, A., Luan, H.H., and Medzhitov, R. (2019). An evolutionary perspective on
immunometabolism. Science 363.
Wang, A., and Medzhitov, R. (2019). Counting Calories: The Cost of Inflammation. Cell 177,
223-224.
Weis, S., Carlos, A.R., Moita, M.R., Singh, S., Blankenhaus, B., Cardoso, S., Larsen, R.,
Rebelo, S., Schauble, S., Del Barrio, L., et al. (2017). Metabolic Adaptation Establishes Disease
Tolerance to Sepsis. Cell 169, 1263-1275 e1214.
Winer, S., Paltser, G., Chan, Y., Tsui, H., Engleman, E., Winer, D., and Dosch, H.M. (2009).
Obesity predisposes to Th17 bias. Eur J Immunol 39, 2629-2635.
Yu, G., and He, Q.Y. (2016). ReactomePA: an R/Bioconductor package for reactome pathway
analysis and visualization. Mol Biosyst 12, 477-479.
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
37
METHODS
RESOURCES TABLE
REAGENT or RESOURCE
SOURCE
IDENTIFIER
Chemicals, Peptides, Recombinant Proteins
2-Mercaptoethanol
Biorad
1610710
Acetone
Sigma
179124
Brefeldin A
BioLegend
420601
CL316,243
Sigma
C5976
Collagenase A
Roche
11088882001
Collagenase D
Sigma
11088866001
DAB
DAKO
K5001
DAPI
ThermoFisher
D1306
Dibutyl phthalate,99%
Sigma
524980
DMEM
ThermoFisher
41965-039
DNAse
Roche
10104159001
FA Folic Acid solution
Sigma
F8758
Fluorescein isothiocyanate isomer I
Sigma
F7250
Hemalun
Merck
1092490.500
Heparin
Sigma
H3393
hTGF-beta
Peprotech
100-21C
IL-12
Peprotech
210-12
IL-18
MBL (Biozol)
B004-5
IL-2
ebioscience
34-8021-82
IL-23
Miltenyi
130-096-676
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
38
IL-6
Peprotech
216-16
IL-7
Peprotech
217-17
Ionomycin
Sigma
IO634
L-Arginine
Sigma
11009
L-Asparagine
Sigma
A0884
L-Glutamin
Bioconcept
5-10K00H
Lithium Carbonate
Merck
5671.0250
Luxol Fast Blue
BDH laboratories
340443P
Monensin
BioLegend
420701
NEAA 100x
Gibco
11140-035
Penicilin-Streptomycin solution
AMIMED
4-01F-00H
Percoll
GE Healthcare
17-0891-01
Periodoc Acid
Merck
1005240100
PMA
Sigma
P1585
Schiff's reagent
Merck
1.09033.0500
Sodium Pyruvate
Sigma
S8636
Thioglycollate medium
Sigma
T9032
VIT Vitamin mix solution
Sigma
M6985
Critical Commercial Assays
AccuCheck Counting Beads
Molecular Probes
PCB100
BD FACS Lysing Solution
BD
349202
CD4(l3t4) microbeads mouse
Miltenyi
130-117-043
cDNA Reverse Transcription Kit
Applied Biosystems
4368814
Fixation buffer
BioLegend
420801
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
39
FoxP3/Transcription Factor Staining Buffer
Set
eBioscience
00-5523-00
heat inactivated mycobacterium tuberculosis
H3/RA
BD Difco
231141
Incomplete Freund`s adjuvant
BD Difco
263910
Intracellular Staining Perm Wash Buffer
BioLegend
421002
MOG peptide 35-55
Peptides&elephants
N/A
Nextera kit
Illumina
N/A
Pertussis toxin
Sigma/ Calbiochem
P7208/ 516560
RBC Lysis buffer
BioLegend
420301
RNAlater stabilization solution
ThermoFisher
AM7020
RNeasy plus Micro Kit
QIAGEN
74034
RNeasy plus Mini Kit
QIAGEN
74134
PowerUp SYBR Green Master Mix
Applied Biosystems
A25741
SMART-Seq v4 ultra-low input kit
Clontech
N/A
TRIzol reagent
ThermoFisher
15596026
TruSeq stranded mRNA kit
Illumina
N/A
Versacomp beads
Beckman Coulter
B22804
Zombie NIR Fixable Viability Kit
BioLegend
423105
Antibodies
Anti-mouse HRP
DAKO
K4001
Anti-rabbit HRP
DAKO
K4003
Anti-rat HRP
Vectorlab
MP-7404-50
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
40
Armenian hamster anti-CD11c (clone N418)
BioLegend
117328, 117308,
117343, 17309,
117312
Armenian hamster anti-CD11c (clone N418)
eBioscience
103618
Armenian hamster anti-CD3 (clone 145-2C11)
ThermoFisher,
BioLegend
11-0031-82,
100328
Armenian Hamster anti-CD3 (clone 145-
2C11)
BioLegend
100314
Armenian hamster anti-CD49a (clone Ha31/8)
BD
564667
Mouse anti-APP A4 a.a. 66-81 (clone 22C11)
Sigma (formerly Millipore)
MAB348
Mouse anti-CD64 (clone X54-5/7.1)
BioLegend
139308, 139309
Mouse anti-CX3CR1 (clone SA011F11)
BioLegend
149013, 123135
Mouse anti-NK1.1 (clone PK136)
BioLegend
108727
Mouse anti-XCR1 (clone ZET)
BioLegend
562290
Rabbit anti-CD3e (polyclonal)
DAKO
A0452
Rat anti-B220 (clone RA3-6B2)
BioLegend
740262
Rat anti-CCR2 (MC-21)
Provided by Matthias
Mack
N/A
Rat anti-CD103 (clone M290)
BD
564322
Rat anti-CD107b (Mac3) (clone M3/84)
BioLegend
108502
Rat anti-CD115 (clone AFS98)
BioLegend
135505, 135515,
135523
Rat anti-CD117 (clone ACK2, 2B8)
BioLegend
135111, 105827
Rat anti-CD11b (clone M1/70)
BioLegend
101242, 101228,
101222, 101263
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
41
Rat anti-CD11b (clone M1/70)
BD
35011482
Rat anti-CD127 (clone A7R34)
BioLegend
135009
Rat anti-CD135 (clone A2F10)
BioLegend
135310
Rat anti-CD16/32 TruStain FcX (clone 93)
BioLegend
101320
Rat anti-CD19 (clone 6D5)
BioLegend
115534
Rat anti-CD4 (clone GK1.5)
BioLegend
100422
Rat anti-CD45 (clone 30-F11)
BioLegend
103114, 103108
Rat anti-CD8 (clone 53-6.7)
BD
563786
Rat anti-F4/80 (clone BM8)
BioLegend
123114
Rat anti-F4/80 (clone BM8)
BioLegend
107622
Rat anti-GM-CSF (clone MP1-22E9)
ThermoFisher
11733181
Rat anti-IFNg (clone XMG1.2)
BioLegend
508510, 505830
Rat anti-IFNg (XMG1.2)
Bio X Cell
BE0055
Rat anti-IL-17 (clone TC11-18H10.1)
BioLegend
506904
Rat anti-Ly6C (clone HK1.4)
BioLegend
128036, 128037
Rat anti-Ly6G (clone 1A8)
BioLegend
127622, 126615
, 565707
Rat anti-MHCII (clone M5/114.15.2)
BioLegend
107620, 107606,
107631, 107622
Rat anti-mouse CD49d (clone R1-2)
BioLegend
149027
Rat anti-NOS2 (clone CXNFT)
ThermoFisher
53-5920-82
Rat anti-PDCA-1 (clone 927)
BioLegend
127012
Rat anti-Sca1 (clone D7)
BioLegend
108116
Rat anti-Siglec-F (clone E50-2440)
BD
108749
Rat anti-Ter119 (clone Ter119)
BioLegend
116228
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
42
Rat IgG2b isotype
Bio X Cell
BE0090
Sheep anti-Arg1
R&D
IC5868P
Syrian Hamster anti-CD28 (clone 37.51)
BioLegend
102112
Other
Attune NxT
ThermoFisher
N/A
AutoMACS Pro
Miltenyi
N/A
AxioScan Z1
Zeiss
N/A
Bioanalyzer Agilent 2100
Agilent
N/A
BD FACSAria II
BD Biosciences
N/A
BD FACSymphony
BD Biosciences
N/A
BD LSRFortessa
BD Biosciences
N/A
TProfessional Basic Thermocycler
Biometra
N/A
Bio-Rad S3
Bio-Rad
N/A
FLIR E60 thermal camera
FLIR systems
N/A
HiSeq 4000
Illumina
N/A
Light Cycler 480-II
Roche
N/A
MoFlo Astrios
Beckman Coulter
N/A
NanoDrop
ThermoFisher
N/A
Pannoramic 250 FLASH II
3Dhistech
N/A
Seahorse XF24 Analyzer
Agilent
N/A
Tissue Lyser II
QIAGEN
N/A
Experimental Models: Organisms/ Strains
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
43
Mouse: 2D2
The Jackson Laboratory
006912
Mouse: C57BL/6
Charles River, Janvier
Mouse: Ccr2-KO
The Jackson Laboratory
004999
Mouse: Ucp1-KO
The Jackson Laboratory
003124
Experimental Models: Cell Lines
B16-GMCSF cells
Dranoff et al., 1993
N/A
Name
Forward Primer
Reverse Primer
M. musculus
Ucp1
5′-GGCCTCTACGACTCAGTCCA-3′
5′-TAAGCCGGCTGAGATCTTGT-3′
M. musculus
Cidea
5′-TGCTCTTCTGTATCGCCCAGT-3′
5′-GCCGTGTTAAGGAATCTGCTG-3′
M. musculus
Ppargc1a
5′-
AAGTGTGGAACTCTCTGGAACTG-
3′
5′-
GGGTTATCTTGGTTGGCTTTATG-3′
M. musculus
Ppara
5′-AGAGCCCCATCTGTCCTCTC-3′
5′-ACTGGTAGTCTGCAAAACCAAA-
3′
M. musculus
Pparg
5′-AGGCGAGGGCGATCTTGACAG-
3′
5′-AATTCGGATGGCCACCTCTTTG-
3′
M. musculus
Prdm16
5′-CAGCACGGTGAAGCCATT-3′
5′-GCGTGCATCCGCTTGTG-3′
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
44
M. musculus
36B4
5′-
GTACCCATTGATGATGGAGTGT-3′
5′-TGAAGTGCTCGACATCACAG-3′
M. musculus
B2m
5′-TTGTCTCACTGACCGGCCT-3′
5′-TATGTTCGGCTTCCCATTCTCC-
3′
Software and Algorithms
FastQC v.0.11.5
https://www.bioinformatics.babraham.a
c.uk/projects/fastqc/
N/A
FLIR Tools+ software
FLIR technologies
FlowJo 10.2
TreeStar
N/A
GraphPad 8
Prism
N/A
HTSeq v0.9.1
https://www-
huber.embl.de/users/anders/HTSeq/
N/A
MetaCore
Clarivate Analytics
N/A
Pannoramic Viewer software
3DHistech
N/A
Picard tools v.1.141
https://picard.sourceforge.net
N/A
QuPath v0.2.2
QuPath
N/A
R language and environment for statistical
computing and graphics
https://www.r-project.org
N/A
R/Bioconductor package EdgeR v. 3.4.2
(McCarthy et al., 2012)
N/A
R/Bioconductor package topGO
https://bioconductor.org/packages/rele
ase/bioc/vignettes/topGO/inst/doc/topG
O.pdf
N/A
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
45
EXPERIMENTAL MODEL AND SUBJECT DETAILS
Mice
Male 8 weeks old C57BL/6J mice were obtained from Charles River, or Janvier (France). Ucp1-
KO (The Jackson Laboratory), their littermate controls, Ccr2-KO (The Jackson Laboratory) and
2D2 (The Jackson Laboratory) mice were bred in house. Mice were housed in a specific pathogen
free (SPF) facility in 12 h day/night cycles, fed irradiated standard chow diet and given water from
autoclaved bottles. Cold exposure was applied in a light- and humidity-controlled climatic chamber
(TSE, Germany) under SPF conditions, at 10 °C for 2 weeks before immunization with an
additional initial acclimatization period of 5 days at 18°C and 5 days at 14°C, as well as during
EAE. Warm exposure of 34°C was applied for 7 days before immunization, and during EAE. All
climate chamber experiments were performed with 2 mice per cage without bedding and nesting
material for enrichment to ensure controlled temperature conditions. CL316,243 (Sigma) was i.p.
injected daily 7 days before immunization and until sacrifice. Body temperature was determined
with infrared camera FLIR E60 (FLIR, UK) taking the mean temperature of both eyes using the
FLIR Tools+ software. All animal experiments were performed at the University of Geneva with
authorization by the responsible Cantonal authority and in accordance with the Swiss law for
animal protection.
ReactomePA R-package
(Yu and He, 2016)
N/A
Rgraphviz
https://www.bioconductor.org/package
s/release/bioc/html/Rgraphviz.html
N/A
STAR aligner v.2.6.0c
(Dobin et al., 2013)
N/A
Seahorse Wave Desktop Software
Agilent
N/A
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
46
METHODS DETAILS
EAE induction and scoring
For active EAE immunization, complete Freund`s adjuvant was prepared by mixing 10mg/ml heat
inactivated mycobacterium tuberculosis H3/RA (Difco) with incomplete Freund`s adjuvant (Difco).
To create an emulsion, this was mixed 1:1 with MOG peptide 35-55 (4 mg/ml, peptides &
elephants). Mice were anesthetized and 50 ul of the emulsion was injected into the left and right
flank each (200 ug peptide/mouse), followed by i.v. injection of 300 or 400 ng pertussis toxin
(Sigma or Calbiochem). After two days, mice received additional 300 or 400 ng pertussis toxin
i.p.
For Th1 adoptive transfer EAE, splenocytes of 2D2 T cell receptor transgenic mice were cultured
for 2 days with MOG35-55 peptide (20 µg/ml), IL-2 (5 ng/ml, ebioscience) and IL-7 (5 ng/ml,
Peprotech), 4 days with IL-2 (5 ng/ml) and IL-7 (5 ng/ml) and 1 day with anti-CD3/28 (0.5 ng/ul,
Biolegend), IL-12 (20 ng/ml, Peprotech) and IL-18 (25 ng/ml, MBL (Biozol)) at 37°C. Per mouse,
1-2 million cells were transferred (i.p.) and animals were injected with 67 ng pertussis toxin on
day 0 (i.v.) and day 2 (i.p.) of EAE. Congenic animals (CD45.1 and CD45.2) were used to later
distinguish endogenous and transferred T cells.
For Th17 adoptive transfer EAE, wildtype donor mice were actively immunized as described
above and after 7 days their splenocytes lysed, cultured together with cells from dLNs for 3 days
with IL-6 (5 ng/ml, Peprotech), hTGF-beta (0.25 ng/ml, Peprotech), anti-IFNg (14 ug/ml, Bio X
Cell), IL-23 (6.5 ng/ml, Miltenyi) and MOG peptide (35 ug/ml) and sorted via positive selection of
CD4+ T cells (MACS, Miltenyi). Per recipient, 3 million CD4+ T cells were transferred by iv. injection
and animals were injected i.p. with pertussis toxin on day 0 and day 2 of EAE (300 ng).
Starting at the disease onset, approx. 8-10 days after induction of EAE, disease severity was
monitored using an established scale from 0 to 4 to describe clinical symptoms: 0, no detectable
signs of classical EAE; 0.5, paresis of distal limb tail; 1.0, tail paralysis; 1.5, hind-limb weakness;
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
47
2, hind-limb paresis; 2.5, partial hind-limb paralysis; 3, complete bilateral hind-limb paralysis; 3.5,
ascending paralysis to torso; 4, paralysis of forelimbs and hind limbs. Mice with scores >3.5 were
sacrificed. Dead animals were excluded or in warm exposure experiment given a score of 5 on
death day due to increased deaths and high scores. Animals that reach a score >1.5 were
provided with their chow diet, watered, inside the cage.
B16-GMCSF tumor and thioglycolate elicited peritoneal inflammation
GMCSF-expressing B16 melanoma tumor cells (Dranoff et al., 1993) were cultured and s.c.
engrafted as described (Menezes et al., 2016) into cold exposed and room temperature mice.
Blood was drawn on several days after tumor implantation in heparin (Sigma) to analyze
circulating monocyte expansion and phenotype using flow cytometry. For peritoneal inflammation,
cold-exposed and room temperature mice were i.p. injected with 1ml of 4% thioglycolate (Sigma),
as described before (Davies et al., 2013). Animals were sacrificed after 24h for analysis of
peritoneal monocytes and monocyte derived cells using flow cytometry.
FITC migration of DCs using skin painting of the flank
Mice were anesthetized with isoflourane, a small area of one flank (about 1x1cm) was shaved
and 20 ul of FITC solution was applied, consisting of 1% FITC (Sigma-Aldrich: Fluorescein
isothiocyanate isomer I) in Aceton/Dibutylphthalate (1:1, Sigma). After 12h and 18h, the 3 draining
lymph nodes (axillary, brachial and inguinal) of ipsi- and contralateral side were harvested and
cells harvested as described above.
Antibody treatment for CCR2+ monocyte depletion
Monocytes were depleted by daily i.p. injections of anti-CCR2 mAb (clone: MC-21; 40 μg), starting
one day before immunization on 8 consecutive days until sacrifice. Control mice received isotype
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
48
rat IgG2b (Bio X Cell) at the same dose. Depletion efficiency was tested 2 days before EAE onset
and sacrifice.
Histology
For histologic analyses of spinal cord, mice were kept until 1-2 days after peak, perfused with
PBS and subsequently 4% paraformaldehyde (PFA) and additionally immersion fixed in 4% PFA.
Sectioning, embedding and staining were performed as previously described (Steinbach et al.,
2016). Mice shown for histologic evaluation were control mice of an experiment and therefore
injected with diphtheria toxin (Sigma). Stainings were developed with DAB (DAKO) and nuclei
counterstained with hemalum (Merck). Sections were scanned using Pannoramic Digital Slide
Scanner 250 FLASH II (3DHISTECH). Quantifications were performed blinded to the
experimental condition. Anti-CD3 (polyclonal, DAKO) and anti-mac3 (M3/84, Biolegend) stainings
were automatically quantified using Definiens Developer software with a custom-made script.
Quantifications of Luxol Fast Blue (BDH laboratories)-Perdiodic Acid (Merck)-Schiff’s (Merck)
(LFB-Pas) lesion areas, anti-APP (22C11, Millipore) and lesion number were performed manually
using Pannoramic Viewer software (3DHISTECH). Number of lesions were counted in APP
staining and only lesions were taken into account that were found in LFB-Pas as well. APP was
quantified within lesions marked in LFB-Pas staining and reported per total white matter area. For
representative LFB-Pas images, contrast was equally adjusted in the compared conditions in
Adobe Photoshop.
For histologic analyses of adipose tissues, tissues were fixed in 4% paraformaldehyde (Sigma-
Aldrich), paraffin embedded, cut in sections (5 μm) and stained with hematoxylin-eosin (H&E).
Images were acquired using AxioScan Z1 (Zeiss) and the cell/lipid droplet size quantified
with Qupath software (V0.2.2) using machine-learning classifiers.
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
49
Immune cell isolation from tissues
Peripheral blood samples (10-30 ul) were obtained by facial vein puncture in 1 ul heparin (10U/ul,
Sigma). Blood erythrocytes were lysed and cells fixed using BD FACS Lysing Solution. For other
tissues that required red blood cell lysis, RBC Lysis buffer (Biolegend) was used. For the isolation
of immune cells from the CNS, mice were anesthetized and transcardially perfused with PBS.
Brain and spinal cord were minced, digested in DMEM (ThermoFisher) with Collagenase A
(1mg/ml, Sigma) and DNaseI (0.1 mg/ml, Roche) for 1h at 37°C and homogenized using 70-μm
cell strainers (BD). Leukocytes were separated using a discontinuous Percoll (GE healthcare)
gradient (30% / 70%, 2500 RPM, 30 min, 4°C). Bone marrow cells were flushed from 1 femur per
mouse. For analysis of DC and monocyte populations, axillary and brachial lymph nodes or spleen
were cut into small pieces and digested in DMEM with collagenase D (1mg/ml, Roche) and
DNaseI (10ug/ml, Roche) for 30-35 min at 37°C under constant pipetting. For studies of all other
immune cells, lymph nodes or spleens were mashed through a 70 um cell strainer. For
splenocytes, red blood cell lysis was performed (BioLegend).
Flow cytometry
Dead cells were excluded from the analysis using Zombie NIR or Aqua Fixable Viability kit
(BioLegend) for all tissues except blood. Surface staining was performed with directly labeled
antibodies in FACS buffer (DPBS, 2.5% FCS, 10mM EDTA). Compensation controls were set up
using Versacomp beads (Beckman Coulter). Antibodies were from BioLegend if not stated
otherwise. For staining of bone marrow progenitors and mature monocytes, the following
antibodies were used: Lin (CD3 (145-2C11), CD19 (6D5), NK1.1 (PK136), Ly6G (1A8), Ter-119),
CD127 (A7R34), CD117 (ACK2, 2B8), CD115 (AFS98), CD135 (A2F10), Ly6C (HK1.4), CD11b
(M1/70), Sca1 (D7), CD11c (N418), MHCII (M5/114.15.2). In figures, monocytes and monocyte-
derived cells were abbreviated as “M” for simplicity reasons. Both Ly6Chi monocytes and
monocyte derived cells were defined mainly by their Ly6C expression (in addition to and exclusion
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
50
of other markers), but irrespective of a co-expression of CD11c or pro/anti-inflammatory
cytokines. “M” therefore comprises both, undifferentiated Ly6Chi monocytes and monocytes that
already gained APC or macrophage like functions but still express Ly6C. For characterization of
blood and bone marrow monocytes, the following antibodies were used: CD45 (30-F11), Ly6G,
CD11b, CD115, Ly6C, MHCII. For characterization of T cells in various tissues, the following
antibodies were used: CD45, CD11b, Ly6G, CD4 (GK1.5), CD3, CD8 (53-6.7, BD), GM-CSF
(MP1-22E9, ThermoFisher), IL-17 (TC11-18H10.1), IFNg (XMG1.2). Unspecific binding to FC
receptors was blocked using anti-CD16/32 (BioLegend). Intracellular stainings were performed
according to manufacturer’s instructions: Fixation/Intracellular Staining Permeabilization Wash
Buffer (BioLegend) and Fixation buffer (BioLegend) were used for Arg1 (IC5868P, R&D), iNOS
(CXNFT), IL-17, IFNg and GM-CSF. To assess for intracellular cytokine production of T cells,
leukocytes were cultured for 4 h in the presence of PMA (end 50ng/ml, Sigma) and Ionomycin
(end 1ug/ml) (Sigma), monensin (1:1000, BioLegend), and brefeldin A (1:1000, BioLegend).
Absolute cell numbers were quantified using AccuCheck Counting Beads (Invitrogen). Flow
cytometric samples were acquired on an Attune NxT (ThermoFisher) or LSRFortessa (BD) flow
cytometer using compensation controls. FlowJo software (Treestar, Version 10) was used for
analysis. Further antibodies used in DC analysis or high dimensional analysis, that were not yet
mentioned before are CD49a (Ha31/8, BD), CD64 (X54-5/7.1), CX3CR1 (SA011F11), XCR1
(ZET), B220 (RA3-6B2), CD103 (M290, BD), F4/80 (BM8), CD49d (R1-2), PDCA-1 (927), Siglec-
F (E50-2440, BD). High-dimensional flow cytometry analysis was performed using BD
FACSymphony. Raw data was pre-processed using FlowJo followed by transformation, percentile
normalization in R, dimensionality reduction and visualization by Uniform Manifold Approximation
and Projection (UMAP) (McInnes et al., 2018). The FlowSOM algorithm was used for automated
clustering (Van Gassen et al., 2015) using UMAP with overlaid mean marker expression values
and a heatmap of median expression values (Hartmann et al., 2016; Mrdjen et al., 2018). Data
shown in Figure 2C is the same as in Figure 5E but was visualized together with data from other
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
51
time points to highlight shifts of populations over the time in Figure 5E.
Oxygen Consumption Rate (OCR). OCR was determined as before (Dunham-Snary et al.,
2014), in 3.2-8mg punches (2.5 mm) of adipose tissue. After washing in wash buffer (25mM
HEPES, 1% fatty acid-free BSA, 2.5 mM glucose, in salt solution: 4mM KCl, 115 mM NaCl, 1 mM
MgCl2, 1.5 mM KH2PO4, 2 mM MgSO4, 1.5 mM CaCl2 at pH 7.4), tissue punches were placed
in islet capture plates (model XF24, Seahorse Bioscience) and washed with wash and
subsequently with respiration buffer (0.1% fatty acid-free BSA, 10 mM glucose, 2 mM L-glutamine,
0.5 mM L-carnitine, 1 mM sodium pyruvate in salt solution at pH 7.4), in which they were further
equilibrated for 50 or 60 min at 37°C. Samples were analyzed using a Seahorse XF24 Analyzer
(Seahorse Bioscience) and Wave software with 3 minutes mix; 2 minutes rest; 3 minutes
measurements in basal condition.
RNA Extraction for RNA sequencing and qPCR
After preparation from mice, spinal cord and adipose tissues were stored at -80°C until further
processed. For RNA isolation, frozen tissues were mechanically homogenized in 1 ml Trizol
(Thermo Fisher Scientific) with 1 stainless steel bead (5 mm) per sample using the TissueLyserII
(Qiagen) by shaking for 50 s at 30 Hz. 200 μL chloroform was added to homogenized samples in
trizol, followed by shaking for 15 s and centrifugation (15 min, 12000 RCF, 4°C). The chloroform
phase was collected, shaken for 15 s with 500 μL isopropanol and centrifuged (15 min, 12000
RCF, 4°C). The pellet obtained was washed twice with 70% ethanol (10 min, 8000 RCF, 4°C) and
resuspended in 50 μL PCR-grade water. Bone marrows were flushed right after sectioning from
mouse, cells centrifuged, loaded onto shredder columns (Qiagen). Shredded bone marrow cells
or FACS sorted cells were frozen (-80°C) in RLT buffer with 1% beta-mercaptoethanol until RNA
extraction via RNA mini kit or micro kit (Qiagen), respectively. For Sequencing, RNA integrity
number (RIN) was determined (Bioanalyzer 2100, Agilent). For qPCR, RNA concentration and
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
52
purity were determined (NanoDrop, Thermo Fisher Scientific) and 1 μg RNA was used per sample
for retrotranscription using the High-Capacity cDNA Reverse Transcription Kit (Applied
Biosystems). Expression of mRNA was measured by qPCR on a LightCycler 480 II (Roche) with
PowerUp SYBR Green Master Mix (Applied Biosystems). Relative mRNA expression was
normalized to at least two house keeping genes and to room temperature control mice.
Blood monocyte purification for RNA sequencing
Blood was taken retro-orbital under terminal anesthesia in polypropylen facs tubes containing 7ul
heparin (10U/ul) and coated with 100 IU/mL heparin. Anti-CD115-PE was directly added and
MACS separation was performed using Anti-PE MicroBeads (Miltenyi) and AutoMACS Pro
Seperator (Miltenyi). Immediately after, monocytes were sorted as CD115+, DAPI-
(ThermoFisher) cells (FACSAriaII, BD, Bio-Rad S3 or MoFlo Astrios, Beckman Coulter).
RNAseq sequencing and analysis
mRNA sequencing was performed at the iGE3 Genomics Platform at the CMU of the University
of Geneva. Libraries for sequencing were prepared with the TruSeq stranded mRNA kit (Illumina)
for bone marrow and spinal cord. The SMART-Seq v4 ultra-low input kit (Clontech) combined with
the Nextera kit (Illumina) were used respectively for cDNA amplification and library preparation
for monocytes (steady state and EAE). Bone marrow, spinal cord, and monocytes (EAE) were
sequenced with read length SR50 and steady state monocytes with read length SR100 (Illumina
HiSeq 4000). The sequencing quality control was done with FastQC v.0.11.5
(http://www.bioinformatics.babraham.ac.uk/projects/fastqc/). The reads were mapped
with STAR aligner (Dobin et al., 2013) to the UCSC Mus musculus mm10 reference. The
transcriptome metrics were evaluated with the Picard tools v.1.141
(http://picard.sourceforge.net/). The table of counts with the number of reads mapping to each
gene feature of the Mus musculus mm10 reference was prepared with HTSeq v0.9.1 (htseq-
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
53
count, http://www-huber.embl.de/users/anders/HTSeq/). Raw counts were then further processed
and analyzed by R/Bioconductor package EdgeR v. 3.4.2 (McCarthy et al., 2012a), for differential
expression analysis. The counts were normalized according to the library size and filtered. Only
genes having log count per million reads (cpm)>0 were kept for further analyses. After
normalization of the counts, transcript abundances were compared in pairwise conditions in a
modified Fischer exact test (as implemented in edgeR). The differentially expressed genes p-
values were corrected for multiple testing error with a 5% FDR (false discovery rate). The
correction used is Benjamini-Hochberg (BH). Transcripts with log(cpm)>0 and p≤0.05 were
subsequently subjected to pathway analysis. For the relativeness and reactome analysis, genes
were called significantly differentially expressed between any given two conditions when p<0.05
and FC>2. Reactome pathways database was used with ReactomePA R-package (Yu and He,
2016), reporting the enrichment ratio (# DEGs/Total Genes in the dataset) and FDR-adjusted
Pvalue computed by Fisher exact test. For gene ontology analysis, R/Bioconductor package
topGO (https://bioconductor.org/packages/release/bioc/vignettes/topGO/inst/doc/topGO.pdf),
together with Rgraphviz was used. MetaCore (Clarivate Analytics) Pathway Maps, Gene Ontology
and Metabolic Networks were used.
Statistical analysis
To assess significant differences, GraphPad 8.0 (Prism) was used. For two groups, two-tailed
Student’s t test was used. For more than two groups, multiple t-tests were applied with Holm-
Sidak correction. Two-waxy ANOVA was used to compare two variables between groups. Mantel-
Cox was applied to test the incidence or survival distributions of two groups. P < 0.05 was
considered significant (*), p < 0.01 (**), p < 0.001 (***).
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
Figure 1
A
D
RT CE
0.0
0.1
0.2
0.3
% Ly6Cint M of alive
RT CE
0
2
4
6
% MHCII of Ly6Chi M
***
RT CE
0
1
2
3
4
%MHCII+Ly6Chi of MHCII+
**
RT CE
0
10
20
30
40
50
%MHCII+ of CD45+
RT CE
0.00
0.05
0.10
0.15
0.20
% CDP
*
RT CE
0.00
0.02
0.04
0.06
0.08
0.10
% MDP
*
RT CE
0.00
0.01
0.02
0.03
0.04
0.05
% CLP
RT CE
0.00
0.05
0.10
0.15
0.20
0.25
% LSK
E
F
B
C
G
RT CE
0.0
0.2
0.4
0.6
0.8
1.0
% Ly6Clo M of alive
RT CE
0
2
4
6
8
% Ly6Chi M of alive
*
RT CE
0.10
0.15
0.20
0.25
% cMoP
p=0.095
PBS CL316
0.00
0.05
0.10
0.15
% cMoP
*
PBS CL316
0.00
0.02
0.04
0.06
0.08
% CLP
PBS CL316
0.0
0.2
0.4
0.6
% LSK
PBS CL316
0.00
0.05
0.10
0.15
0.20
% CDP
*
PBS CL316
0
1
2
3
4
%Ly6Chi M of alive
*
0 50 100 150
Myeloid leucocyte activation
Export from cell
Secretion
Cell activation
Immune system process
-log(p-value)
GO Processes
PBS CL316
0.00
0.02
0.04
0.06
0.08
% MDP
p=0.1
RT1
RT2
RT3
RT4
RT5
RT6
CE1
CE2
CE3
CE4
CE5
CE6
B2m
CD80
Csf1
Csf1r
Csf2ra
Csf2rb
Cxcl10
Ern1
Gas6
H2-D1
H2-K1
H2-Q10
Hgf
Hif1a
Ifngr1
Il12a
Il12rb1
Il12rb2
Il13ra1
Il1b
Il1r1
Il4
Il4ra
Inhba
Irf4
Irf5
Itgam
Itgb2
Jag1
Kdm6b
Lilrb4
Mapk13
Mapk14
Mertk
Msr1
Notch1
Pik3r3
Postn
Ptger2
Ptpn6
Rbpj
Ripk1
Socs3
Stat1
Stat3
Stat6
Tgfbr1
Thbs1
Tlr2
Tlr4
Tlr7
Tnf
Tnfrsf1a
Tnfsf10
Xbp1
Macrophage and DC shift
-2
0
2
RT1
RT2
RT3
RT4
RT5
RT6
CE1
CE2
CE3
CE4
CE5
CE6
Adar
B2m
Bcl2
Casp-7
Cbl
Cxcl10
Cxcl9
Eif2ak1
Eif2ak2
Gbp3
Gbp4
H2-D1
H2-K1
H2-Q10
HIF1A
Ifit3
Irf1
Irf7
Irf9
Isg20
Mx1
Ncoa7
Oas1a
Oas1b
Oas1g
Socs3
Stat1
Stat2
Stat3
Stat4
Stat6
Tap1
Tnfsf10
Usp18
Xaf1
IFN-alpha/beta signaling
-2
0
2
RT1
RT2
RT3
RT4
RT5
RT6
CE1
CE2
CE3
CE4
CE5
CE6
Ccr2
Cebpa
Cebpe
Csf3r
Gfi1
Itgam
Itgb2
Lcn2
Ltf
Map2k1
Mapk3
Mpo
Myc
Prtn3
Ptpn11
Ptpn6
Rasgrp1
Rb1
Sfpi1
Socs3
Stat3
G-CSF-induced myeloid differentiation
-2
-1
0
1
2
8430408G22Rik
Apol11a
C130026I21Rik Cd209f
Col1a1
Ctsk
Oasg1
Cyr61
Hspa1a
Ifitm1
Marco
Upp1
0
5
10
Log2
fold change
-Log10 P
Hspa1b
Col1a1
Vsig4
Rspo2
Mmp13
Cxcr1
Irg1
Oas2
NS
p-value
2
p-value and log FC
-1 1
02
(which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.
The copyright holder for this preprintthis version posted August 30, 2020. ; https://doi.org/10.1101/2020.08.28.269563doi: bioRxiv preprint
Figure 2
A
RT CE
0
1
2
3
4
% Ly6Clo M of CD45+
0
5
10
15
20
% MHCII of Ly6Chi M
***
RT CE
RT CE
0.0
0.5
1.0
1.5
2.0
% Ly6Cint M of CD45+
RT CE
0
20
40
60
80
% MHCII+ of CD45+
B
D
E
RT CE
0
5
10
% Ly6Chi M of CD45+
PBS CL316
0
5
10
15
% MHCII+ of Ly6Chi M
*
PBS CL316
0.0
0.5
1.0
1.5
2.0
2.5
%MHCII+Ly6Chi of MHCII+
*
0 2 4 6 8
Positive regulation of macrophage chemotaxis
Ureteric bud development
T cell activation
Regulation of fever generation
Positive regulation of heat generation
Antigen processing, presentation of peptide or polysaccharide antigen via MHCII
T cell costimulation
Regulation of cytosolic Ca2+ in PLC-activating G protein-coupled signaling
Negative regulation of interleukin-6 secretion
Response to bacterium
Regulation of B cell proliferation
Neutrophil chemotaxis
Leukocyte migration involved in inflammatory response
Adaptive immune response
Positive regulation of cytosolic calcium ion concentration
-log(p-adjust.)
GO BP
PBS CL316
0
20
40
60
80
% MHCII+ of CD45+
RT CE
0
1
2