You are on page 1of 31

Article

Fasting-Refeeding Impacts Immune Cell Dynamics


and Mucosal Immune Responses
Graphical Abstract Authors
Motoyoshi Nagai, Ryotaro Noguchi,
Daisuke Takahashi, ..., Keiyo Takubo,
Taeko Dohi, Koji Hase

Correspondence
hase-kj@pha.keio.ac.jp

In Brief
Temporary fasting drastically reduces the
levels of B cells in Peyer’s patches, with
germinal center B cells undergoing
apoptosis and naive cells migrating to the
bone marrow and only egressing upon
refeeding.

Highlights
d Fasting drastically reduces lymphocyte levels in Payer’s
patches

d Naive B cells migrate to bone marrow during fasting and then


back upon refeeding

d Nutritional signals are essential to maintain CXCL13


expression by stromal cells

d Fasting causes GC B cell death and attenuates antigen-


specific IgA response

Nagai et al., 2019, Cell 178, 1072–1087


August 22, 2019 ª 2019 Elsevier Inc.
https://doi.org/10.1016/j.cell.2019.07.047
Article

Fasting-Refeeding Impacts Immune Cell Dynamics


and Mucosal Immune Responses
Motoyoshi Nagai,1,2 Ryotaro Noguchi,1,2 Daisuke Takahashi,1 Takayuki Morikawa,3 Kouhei Koshida,1 Seiga Komiyama,1
Narumi Ishihara,1 Takahiro Yamada,1 Yuki I. Kawamura,2 Kisara Muroi,1 Kouya Hattori,1 Nobuhide Kobayashi,1
Yumiko Fujimura,1 Masato Hirota,1 Ryohtaroh Matsumoto,1 Ryo Aoki,4,5 Miwa Tamura-Nakano,6 Machiko Sugiyama,2,7
Tomoya Katakai,8 Shintaro Sato,9,10 Keiyo Takubo,3 Taeko Dohi,1,2 and Koji Hase1,10,11,*
1Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Science, Keio University, Tokyo 105-8512, Japan
2Department of Gastroenterology, Research Center for Hepatitis and Immunology, Research Institute, National Center for Global Health and
Medicine, Chiba 272-8516, Japan
3Department of Stem Cell Biology, Research Institute, National Center for Global Health and Medicine, Tokyo 162-8655, Japan
4Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
5Institute of Health Sciences, Ezaki Glico Co., Ltd., Osaka 555-8502, Japan
6Communal Laboratory, Research Institute, National Center for Global Health and Medicine, Tokyo 162-8655, Japan
7Laboratory for Immunobiology, Graduate School of Medical Life Science, Yokohama City University, Kanagawa 230-045, Japan
8Department of Immunology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan
9Mucosal Vaccine Project, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka

University, Osaka 565-0871, Japan


10International Research and Development Center for Mucosal Vaccines, the Institute of Medical Science, the University of Tokyo (IMSUT),

Tokyo 108-8639, Japan


11Lead Contact

*Correspondence: hase-kj@pha.keio.ac.jp
https://doi.org/10.1016/j.cell.2019.07.047

SUMMARY SanGiovanni, 1997). Furthermore, childhood malnutrition is a


predisposing factor for environmental enteropathy characterized
Nutritional status potentially influences immune re- by intestinal dysfunction, increased intestinal permeability, and
sponses; however, how nutritional signals regulate microbial dysbiosis (Brown et al., 2015; Humphrey, 2009). In
cellular dynamics and functionality remains obscure. industrialized countries, on the other hand, excessive food intake
Herein, we report that temporary fasting drastically accompanied by a lack of exercise has augmented the incidence
reduces the number of lymphocytes by 50% in of obesity (World Health Organization, 2016), which is a signifi-
cant risk factor for cardiovascular disease, metabolic syn-
Peyer’s patches (PPs), the inductive site of the gut
dromes, and cancer (Basen-Engquist and Chang, 2011; Grundy,
immune response. Subsequent refeeding seemingly
2004; Poirier et al., 2006). Low-grade inflammation due to
restored the number of lymphocytes, but whose obesity is significantly implicated in the development of these
cellular composition was conspicuously altered. A diseases (Visscher and Seidell, 2001). These observations
large portion of germinal center and IgA+ B cells indicate that nutritional status has a significant impact on the
were lost via apoptosis during fasting. Meanwhile, immune system.
naive B cells migrated from PPs to the bone marrow The gastrointestinal mucosa is directly exposed to exogenous
during fasting and then back to PPs during refeeding food ingredients and thus inevitably faces drastic changes in the
when stromal cells sensed nutritional signals and nutritional status of the lumen during food uptake and fasting.
upregulated CXCL13 expression to recruit naive We previously demonstrated that intestinal tissue is highly sus-
B cells. Furthermore, temporal fasting before oral ceptible to deprivation of luminal nutrients, as temporal fasting
arrested epithelial cell proliferation while refeeding induced hy-
immunization with ovalbumin abolished the induc-
perproliferation in the intestinal epithelium (Okada et al., 2013).
tion of antigen-specific IgA, failed to induce oral Given that epithelial cell turnover constitutes a robust first-line
tolerance, and eventually exacerbated food anti- barrier to external antigens, mucosal barrier function may be
gen-induced diarrhea. Thus, nutritional signals are more vulnerable during fasting than during feeding. Considering
critical in maintaining gut immune homeostasis. that fasting relieves the burden of food-borne antigens and
microorganisms on the gut mucosa, it is thus reasonable to
INTRODUCTION decelerate epithelial cell turnover temporarily to minimize energy
expenditure under nutrient deprivation.
Inappropriate calorie intake is a global health problem. In devel- The gut mucosal barrier consists of not only intestinal epithe-
oping countries, the nutritional deficiency often compromises lium but also an underlying immune system that establishes the
vaccination efficacy and increases the risk of infectious dis- second-line barrier. The gut mucosal immune response is charac-
eases (Kaufman et al., 2011; Savy et al., 2009; Scrimshaw and terized by the production of dimeric or polymeric immunoglobulin

1072 Cell 178, 1072–1087, August 22, 2019 ª 2019 Elsevier Inc.
A

E
D

Figure 1. Characterization of PPs during Fasting and Refeeding


(A) Serial sections of PP were stained with H&E. PPs were obtained from mice fed ad libitum (left), fasted for 36 h (middle), or refed with CE2 for 48 h (right). Scale
bar, 200 mm.
(B) Immunostaining of PPs from mice fed ad libitum (left), mice fasted for 36 h (middle), and mice refed with CE2 for 48 h (right). Scale bar, 200 mm.

(legend continued on next page)

Cell 178, 1072–1087, August 22, 2019 1073


A (IgA) to the mucosal surface (Lycke and Bemark, 2017). imal studies on calorie restriction and intermittent fasting have
Secretory IgA (S-IgA) plays vital roles in host defense against employed older mice, which may not adequately reflect biolog-
pathogens, inhibition of microbial metabolite penetration, and ical responses during childhood and children susceptible to
regulation of the gut microbial community (Mantis et al., 2011; nutritional deficiency.
Uchimura et al., 2018; Wei et al., 2011). To efficiently induce In the present study, we analyzed the influence of fasting-
S-IgA response, luminal antigens are actively taken to gut-associ- refeeding on cellular dynamics and functionality of the gut im-
ated lymphoid tissue, such as Peyer’s patches (PPs), that serve mune system mainly in juvenile mice. We observed that PP lym-
as an inductive site of mucosal immunity. In PPs, germinal center phocytes, particularly naive B cells, exhibit dynamic movement to
(GC) reactions, namely, class switch recombination to IgA as well the bone marrow (BM) during temporal fasting and then swiftly
as affinity maturation, occur continuously with the aid of follicular migrate back to PP in response to food intake. A similar oscillation
helper T (Tfh) cells. IgA class-switched B cells subsequently of naive B cells based on the circadian rhythm was observed to a
egress PPs and then home to the intestinal lamina propria via lesser extent in ad libitum-fed mice. Stromal CXCL13 expression
mesenteric lymph nodes (MLNs), the thoracic duct, and blood for recruiting and retaining B cell subsets was regulated by meta-
circulation, during which IgA+ B cells terminally differentiate into bolic status depending on aerobic glycolysis. The frequency of
IgA-producing plasma cells. memory-like B cell subsets markedly decreased after fasting-
Multiple lines of research have uncovered a link between im- refeeding treatment, leading to attenuation of antigen-specific
mune cell function and metabolic status (Kau et al., 2011; Man IgA production and oral tolerance to a food antigen, which even-
and Kallies, 2015). For example, upon T cell receptor (TCR) stim- tually increased susceptibility to antigen-induced diarrhea.
ulation, effector T (Teff) cells enhance the uptake and utilization
of glucose to promote aerobic glycolysis. Activated Teff cells RESULTS
also upregulate glutaminolysis. Such metabolic reprogramming
is essential for Teff cells to meet the energy demand of clonal Fasting Has a Profound Effect on the Morphology of PPs
expansion and effector functions, such as the production of To explore the impact of nutrient deficiency on the gut immune
inflammatory cytokines (Carr et al., 2010). Furthermore, IgA+ system, we maintained juvenile mice (around 6 weeks old) under
plasma cells in the intestine preferentially utilize glycolysis for en- fasting conditions for 36 h. The numbers of total cells, B cells, and
ergy metabolism, whereas naive B cells in PPs usually gain ATP T cells in PPs decreased by half after fasting compared with those
through aerobic metabolism in mitochondria (Kunisawa et al., of ad libitum-fed mice (Figure S1A). Such a drastic change was
2015). Stimulation with lipopolysaccharides (LPS) or B cell re- evident in PPs throughout the small intestine (Figure S1B). Immu-
ceptor (BCR) ligation upregulates glucose transporter 1 (Glut1) nofluorescent analysis demonstrated that the size of PPs was
expression in B cell activating factor (BAFF)-pretreated B cells, markedly reduced during fasting and was restored in response
which eventually undergo metabolic reprogramming to glycol- to refeeding. Despite this macroscopic alteration, the underlying
ysis (Caro-Maldonado et al., 2014). Because B cell-specific microstructure composed of B cell follicles and T cell regions was
Glut1 depletion leads to decreased B cell number and antibody not disturbed during fasting and refeeding (Figures 1A and 1B).
production, glycolytic rewiring is critical for B cell activation. To explore the cause of decrease in the number of lympho-
Upregulation of Glut1 expression in activated B cells is primarily cytes in PPs of fasted mice, we analyzed apoptotic cells and
mediated by activation of the phosphatidylinositol 3-kinase observed that fasting induced apoptosis in considerable number
(PI3K)-Akt pathway, which in turn enhances the mechanistic of B cell follicles, which were rescued by refeeding (Figure 1C).
target of rapamycin (mTOR) signaling. Excessive activation Apoptotic cells were mainly detected in the GC region as a
of the PI3K-Akt-mTOR pathway increases the frequencies of light-blue zone by hematoxylin counterstaining (Figure 1D).
Tfh cells and GC B cells in PPs, while the opposite is true, as Furthermore, transmission electron microscopy (TEM) showed
disruption of mTORC1 or mTORC2 diminishes GC reactions a higher frequency of apoptotic cells characterized by nuclear
and production of S-IgA (Zeng et al., 2016). Given that mTOR chromatin condensation and fragmentation (Figure 1E, upper
serves as a nutrient sensor, the whole-body nutritional status right) as well as phagocytes engulfing apoptotic bodies in the
in response to energy intake and starvation may considerably GC region during fasting (Figure 1E, lower right). Thus, a large
affect the immune response. Indeed, fasting or fasting-mimic portion of GC B cells, which include proliferating B cells and
diet exerts a protective effect on bacterial sepsis and colitis by IgA class-switched B cells, were eliminated in PPs by cell death
alleviating expression of proinflammatory cytokines (Okada followed by phagocytosis in fasted mice.
et al., 2017; Rangan et al., 2019), whereas glucose supplemen-
tation protects against influenza viral infection (Wang et al., Naive B Cells Circulate between PPs and BM in
2016). However, the impact of nutritional signals on mucosal bar- Response to Nutritional Status
riers, lymphocyte dynamics, and effector functions in the context Total cell number steeply declined with nutrient deprivation from
of fasting and feeding remains unknown. Furthermore, most an- 24 to 36 h and then gradually recovered by refeeding for an

(C and D) PPs were stained with TUNEL (C) or for cleaved caspase-3, counterstained with hematoxylin (D). PPs were obtained from mice fed ad libitum (left),
fasted for 36 h (middle), or refed with CE2 for 48 h (right). Scale bar, 200 mm (upper panel), 50 mm (lower panel). In quantification of TUNEL assay, PPs were
obtained from the ad libitum (n = 7), fasting (n = 6), or refeeding group (n = 8).
(E) Sections of PPs were observed by TEM. Scale bar, 10 mm (upper left and lower), 2.0 mm (upper right).
Data represent the means ± SEM. ANOVA followed by Tukey’s test (C). *p < 0.05; **p < 0.01.

1074 Cell 178, 1072–1087, August 22, 2019


A 2.5 2.0 4
Total cells (x107)

2.0
1.5 *

B cells (x107)
3

T cells (x106)
** **
1.5 ** ** *
** ** **
1.0 ** 2
1.0
0.5 1
0.5

0.0 0.0 0
0 24 48 72 96 0 24 48 72 96 0 24 48 72 96 (h)

B 1.5 2.5 2.5


Naive B cells (x107)

IgA+ B cells (x106)


GC B cells (x106)
2.0 2.0
CD95+ GL7+

** *
IgM+ IgD+

1.0
** 1.5 1.5
** **
** **
1.0 ** 1.0 **
0.5 ** **
0.5 0.5

0.0 0.0 0.0 (h)


0 24 48 72 96 0 24 48 72 96 0 24 48 72 96

C 5 ** 2.0 E
B220+ cells (x107)
Total cells (x107)

4
1.5 Fasting Refeeding
3
1.0 **
Total cell
2 B220+ cell
0.5 naive B cell 4
1

0.0
immature B cell
0
0 24 48 72 0 24 48 72 (h) CD4+ T cell
D CD8+ T cell
10
6 Granulocyte 3
Immature B cells (x106)

**
Naive B cells (x106)

**
8 Macrophage
GMP
IgM- IgD-
IgM+ IgD+

4 6 ** **
**
MEP
4
CMP
2
2 CLP 2
0 0
MPP(CD34Flt3)
0 24 48 72 0 24 48 72 ST-HSC(CD34Flt3)
LT-HSC(CD34Flt3)
1.5
MPP2(SLAM) 1
Ki67+ B cells (x107)

MPP3(SLAM)
1.0
MPP4(SLAM)
**
ST-HSC(SLAM)
0.5
** LT-HSC(SLAM)
0
0.0
0 24 48 72 (h)
F G
Naive B cells /PP # (x106)

1.6
ZT0 ZT8 ZT16
0.69 0.045 0.71 0.072 1.56 0.061 1.4
CD3ε

1.2
PP

1.0
n.s.
56.5 42.7 0.8
55.2 44.0 57.4 41.0 0 4 8 12 16 20 24
(ZT)
B220
B220+CD3ε- gated 2.0
Naive B cells (x106)

** **
0.94 6.06 1.18 8.88 1.81 14.0
1.5
IgD

BM

1.0

0.5

70.7 22.3 65.8 24.1 58.2 26.0


0.0
0 4 8 12 16 20 24
IgM (ZT)

Figure 2. Lymphocyte Dynamics in PPs and BM in Response to Fasting and Refeeding


(A–D) Numbers of the indicated cell subsets were measured in PPs (A and B) and the right tibia and femur BM (C and D) of mice fasted for 36 h (blue background)
and refed with CE2 (red background). 24, 36, and 60 h in (A) and (B), n = 12; the other datasets, n = 9.
(E) The mean numbers of the indicated cells from mice fasted for 36 h or refed for 48 h were normalized by the value for mice fed ad libitum (each group, n = 9).

(legend continued on next page)

Cell 178, 1072–1087, August 22, 2019 1075


additional 72 h reflecting the histological observations (Figures trafficking with knock-in mice carrying Kikume-Green Red (KikGR)
1A, 1B, and 2A). A similar tendency was observed for total B cells (Tomura et al., 2014). We selectively irradiated PPs with a 430-nm
(Figure 2A); however, cell behavior was different among the laser to induce photoconversion in PP lymphocytes before fasting
B cell subpopulations. IgM+IgD+ naive B cells were restored to (Figure 3D) and confirmed that PP naive B cells preferentially
pre-fasting (healthy) levels within 72 h after refeeding, whereas migrated to the BM, but not the spleen and MLN, during fasting
CD95+GL7+ GC and IgA+ B cells failed to recover (Figure 2B). (Figure 3E). Based on these observations, we considered that
Consequently, PPs were predominantly filled with naive B cells naive B cells might shuttle between the BM and PPs during the
after the mice experienced fasting and refeeding. Of note, total fasting and refeeding stages, respectively. Similar events were
CD4+ and CD8+ T cells also decreased after 36 h of fasting observed even under germ-free conditions (Figure S3), indicating
and slowly increased with refeeding (Figure S1C), but because that the gut microbiota is unlikely to contribute to the regulation
B cell subsets are deemed highly susceptible to food deprivation of lymphocyte dynamics in response to fasting and refeeding.
and intake, we focused on analyzing B cell dynamics. Notably, such B cell dynamics in response to food intake and
GC B cells were eliminated by apoptosis and would be newly deprivation was well conserved at all life stages (Figure S4).
induced in response to food intake, which explains the delayed
recovery during the refeeding period. On the other hand, the Circadian Oscillation in Lymphocyte Trafficking Was
rapid recovery of naive B cells during the refeeding stage raises Observed in Ad Libitum-Fed Mice
the possibility that this cell subset may migrate to effector sites The feeding behavior of mice is distinct between the daytime and
(i.e., intestinal lamina propria) and/or extraintestinal lymphoid tis- nighttime. Because mice are nocturnal, they do not feed during
sue during fasting and then gain re-entry to PPs during refeeding. the day. We observed slight body weight change of 25.0 ±
To examine this possibility, we carefully analyzed cell dynamics 0.10 g and 23.6 ± 0.10 g at zeitgeber time 0 (ZT0) and ZT12,
in multiple tissues during fasting and refeeding. Fasting did not respectively, in 6-week-old BALB/c male mice; body weight was
affect cell numbers of cecal patches (CPs), the MLNs, or the then restored by nighttime feeding. Therefore, we assumed that
small intestine lamina propria (SILP) (Figure S1A). The number PP lymphocytes may exhibit circadian oscillation in response to
of splenic B cells, which mainly consisted of naive B cells, signif- nutritional status. Indeed, our time course analysis demonstrated
icantly decreased during fasting and then recovered after 48 h of that the number of PP naive B cells slightly decreased until ZT12.
refeeding (Figure S1D). In sharp contrast, total BM cell number Conversely, in the BM, naive B cells increased with a peak value
increased during fasting and decreased after refeeding (Fig- around ZT12 and then gradually declined at ZT16–24 (Figures
ure 2C). In particular, the naive B cell number expanded more 2F and 2G). Given that feeding behavior is minimal at ZT0–12
than 4-fold during fasting and then steeply declined to basal and active at ZT12–24 under physiological conditions, lympho-
levels by refeeding (Figures 2D and 2E). These dynamics of cyte dynamics during daytime and nighttime were similar to those
BM naive B cells were complementary to that of PP and splenic seen in the fasting-refeeding model, albeit to a lesser extent (Fig-
B cells (Figures 2B and S1D). The accumulation of BM naive ures 2A–2E). We considered that food intake plays a central role in
B cells did not result from enhanced B cell generation because the circadian oscillations of naive B cells, and thus fasting disturbs
immature B (B220+IgMIgD) cells, as well as Ki67+ B cells, the oscillation by retaining this lymphocyte population in the BM.
decreased during fasting. Hematopoietic stem/progenitor cells
(HS/PCs) also decreased significantly during fasting and rapidly Nutritional Status Affects Chemokine Expression in PP
recovered in response to refeeding (Figures 2D, 2E, and S2A; and BM
Table S1). Moreover, the immunofluorescent analysis revealed We further explored the molecular basis controlling lymphocyte
that naive B cells were localized not only in the vascular region dynamics in response to nutritional status. Chemokine-chemo-
but also the BM cavity during fasting (Figures S2B and S2C). kine receptor interactions are critical in the regulation of immune
These data suggest that naive B cells most likely translocate to cell trafficking. Among them, the CXCL13-CXCR5 axis is essen-
the BM during nutrient deprivation. tial for the migration and retention of B cell subsets in lymphoid
To rigorously confirm the bias of lymphocyte trafficking under tissues including PPs (Ansel et al., 2000). We observed a signif-
fasting conditions, we adoptively transferred fluorescent dye- icant decrease in Cxcl13 expression in PPs during fasting, which
labeled PP cells from ad libitum-fed donors to either fasting or was recovered by refeeding (Figure 4A). Interestingly, the oppo-
ad libitum-fed recipients (Figure 3A). After 18 h, the transferred lym- site expression pattern was observed in the BM. Although the
phocytes preferentially migrated to the BM only when the recipient CCL20-CCR6 axis is also indispensable for the maturation of
mice were fasted (Figure 3B). These BM-migrating cells were B cell follicles in PPs (Varona et al., 2001), Ccl20 expression re-
mainly naive B cells (Figure 3C). We also assessed lymphocyte mained stable during fasting (Figure 4B). These observations

(F) Representative flow cytometry dot-plots of B220/CD3ε gated (upper panels) and IgM/IgD gated on B220+CD3ε B cells (lower panels) in the BM of mice fed ad
libitum.
(G) The circadian fluctuation in the number of naive B cells in PPs (upper) and the BM (lower). The mean number of PP cells was normalized by the number of PPs
(each time point, n = 8).
Data represent the means ± SEM. ANOVA followed by Dunnett’s test for comparison with mice fed ad libitum (A–D) or ZT0 (G). *p < 0.05; **p < 0.01. CMP,
common myeloid progenitor; GMP, granulocyte/macrophage progenitor; MEP, megakaryocyte/erythrocyte progenitor; MPP, multipotent progenitor; ST/LT-
HSC, short term/long term-hematopoietic stem cell.
See also Figures S1, S2, S3, and S4 and Table S1.

1076 Cell 178, 1072–1087, August 22, 2019


A B

Figure 3. Trafficking of Naive B Cells between PPs and the BM during Fasting
(A) The experimental protocols for adoptive cell transfer of CTV-labeled PP cells.
(B and C) Numbers of CTV+ total (B, n = 12–13) and indicated cell subsets (C, n = 7–8) in the ad libitum-fed and fasting groups.
(D) The experimental protocols for lymphocyte trafficking from PPs in KikGR mice.
(E) Numbers of KikGR-Red+ indicated cell subsets in the ad libitum-fed (n = 10) and fasting groups (n = 12).
Data represent the means ± SEM. Student’s t test. *p < 0.05; **p < 0.01; n.s., not statistically significant.

imply that differential CXCL13 expression in PPs and BM at least from PPs to the BM (Figure S5), indicating that the S1P-S1P1
partly account for the localization of naive B cells during fasting axis is likely not involved in regulating naive B cell dynamics in
and refeeding. response to nutritional status.
The sphingosine 1-phosphate (S1P)-S1P receptor type 1
(S1P1) axis promotes egress of lymphocytes from peripheral mTOR Signaling Partially Contributes to Lymphocyte
lymphoid tissue into circulatory fluids under physiological condi- Dynamics
tions (Matloubian et al., 2004). However, treatment with FTY720 To gain mechanistic insight into lymphocyte dynamics, we as-
failed to prevent the fasting-dependent naive B cell trafficking sessed the activation status of Akt-mTOR signaling, a sensor

Cell 178, 1072–1087, August 22, 2019 1077


A B

D E

Figure 4. Fasting Downregulates CXCL13 Expression in PPs Independent of mTOR Signaling


(A) Cxcl12 and Cxcl13 mRNA expression in PPs (upper) and the BM (lower). PPs and the BM were obtained from mice fed ad libitum, fasted for 36 h, or refed with
CE2 for 48 h (PP, each group, n = 24; BM, ad libitum, fasting, n = 14; refeeding, n = 8).
(B) Ccl20 mRNA expression in PPs. PPs were obtained from mice fed ad libitum or fasted for 36 h (each group, n = 9).
(C) Quantification of phosphorylated p70 S6 kinase (left) and Akt (right) in PPs (n = 18) and the BM (n = 6) using whole tissue lysates from ad libitum, fasting,
refeeding, and rapamycin-treated groups.
(legend continued on next page)

1078 Cell 178, 1072–1087, August 22, 2019


and integrator of external nutritional stimuli for regulating cellular 5I and 5J). These results indicate that metabolic reprogramming
metabolism and physiology, during fasting and refeeding. In the into Warburg-like aerobic glycolysis is a prerequisite for induc-
same experimental setting, we also treated the ad libitum-fed tion of CXCL13 in stromal cells.
group with rapamycin, which mainly inhibits mTORC1 as well
as mTORC2 to a lesser extent (Sarbassov et al., 2006), to deter- Repeated Fasting Attenuates Antigen-Specific IgA
mine mTOR signaling dependency. As anticipated, fasting sup- Response and Oral Tolerance Leading to Exacerbation
pressed phosphorylation of Akt-mTOR signaling molecules, of Food Antigen-Induced Diarrhea
such as p70 S6 kinase, Akt, IRS-1, and PTEN, in both PPs and Elimination of GC and IgA+ B cells from PPs during fasting
BM (Figures 4C and S6). Phosphorylation of these molecules increases the possibility of fasting-refeeding compromising im-
recovered to normal or even higher (e.g., p70 S6 kinase in PPs) mune responses against orally delivered antigens. To investigate
levels after 24-h refeeding compared with that of the control this possibility, mice were orally immunized with ovalbumin
ad libitum-fed group (Figure 4C). (OVA) and cholera toxin (CT) as an adjuvant once a week, four
Rapamycin treatment also decreased phosphorylation levels times. In the fasting group, immunization was performed after
of most Akt-mTOR signaling molecules in PPs and the BM, 48 h of refeeding that followed 36-h fasting (Figure 6A); this
except for Akt in PPs (Figures 4C and S6). Notably, rapamycin regimen did not affect final body weight (Figure 6B). Oral immu-
significantly reduced the number of all B cell subsets in PPs (Fig- nization with OVA/CT gradually increased fecal OVA-specific IgA
ure 4D). Consistent with previous studies (Limon and Fruman, until day 32 in the ad libitum-fed control group; however, the
2012; Zeng et al., 2016), rapamycin markedly decreased GC OVA-specific IgA response was markedly attenuated in the fast-
B cell number by less than 20% of vehicle control (Figure 4D). ing group (Figure 6C). Plasma OVA-specific IgA, IgM, and IgG
In the BM, rapamycin treatment decreased the levels of imma- were also significantly decreased in the fasting group (Figure 6D).
ture B cells, but not naive B cells (Figure 4D). Meanwhile, rapa- Thus, fasted mice failed to gain the booster effect of repeated
mycin treatment did not alter expression levels of chemokines, immunization. Considering that a subset of GC B cells differenti-
including Cxcl12 and Cxcl13, in PPs and the BM (Figure 4E). ates into memory B cells, the elimination of GC B cells from PPs
Thus, mTOR signaling may be critical for B cell survival but not by fasting may have resulted in this abnormality. Similarly, fasting
for chemokine expression. attenuated the generation of antigen-specific IgA in response to
oral infection with a recombinant Salmonella strain expressing a
CXCL13 Production by Stromal Cells Requires Warburg- tenuous toxoid fragment C (rSalmonella-ToxC) (Hase et al.,
like Aerobic Glycolysis 2009; VanCott et al., 1996) (Figure S7).
To further dissect the role of nutrient signals in the regulation of Because antigen-specific IgA and IgG are considered a pro-
chemokine expression, we focused on cellular metabolism. In tective factor against allergic symptoms (Aghamohammadi
juvenile mice, fasting drastically changed systemic nutritional et al., 2009; Strait et al., 2006; Yamaki et al., 2014), we explored
status with hypoglycemia accompanied by elevated plasma the effect of repeated fasting on an OVA-induced diarrhea model
b-hydroxybutyrate (BHB) levels (Figures 5A and 5B). We theo- (Figure 7A). We found that fasting promoted the development of
rized that such nutritional changes may affect chemokine diarrhea without significant increase of OVA-specific plasma IgE
expression in lymphoid tissue. To test this, we took advantage levels (Figures 7B and 7C), and repeated fasting diminished in-
of the lymph node-derived stromal cell line BLS12, which abun- duction of plasma OVA-specific IgG and fecal OVA-specific
dantly produces CXCL13 upon stimulation with tumor necrosis IgA on day 14 and 20, respectively (Figures 7D and 7E). Total
factor-a (TNF-a) and anti-lymphotoxin-b receptor (LTbR) antag- IgA in feces also significantly decreased in fasting group on
onistic antibodies (Katakai et al., 2008) (Figure 5C). We initially day 14 (Figure 7E). These results suggest that impaired produc-
investigated the intracellular metabolic status of BLS12 cells tion of antigen-specific IgG and IgA, as well as total IgA, may
by detecting Glut1 and MitoSOX expression, which represent have led to the exacerbation of diarrhea in the repeated fast-
functional markers for glycolysis and mitochondrial respiration, ing group.
respectively (Kunisada et al., 2017). We found that exposure to Furthermore, mice that were fasted before oral administration
TNF-a and anti-LTbR antibodies strongly skewed cellular meta- of OVA failed to induce oral tolerance to OVA, as evidenced by an
bolism toward glycolysis (Figure 5D). Correspondingly, glucose increase in auricular swelling due to delayed-type hypersensitiv-
deprivation in activated BLS12 cells prominently decreased ity after subcutaneous challenge (Figures 7F and 7G). In line with
Cxcl13 expressions (Figure 5E). Further, inhibition of glycolysis this, suppression of systemic IgG response to the antigen injec-
by 2-deoxy-D-glucose (2DG) markedly downregulated Cxcl13 tion by oral administration of OVA was not observed in the fasting
in activated BLS12 cells (Figure 5F) as well as in PP of mice mice, indicative of impaired systemic unresponsiveness (Fig-
fed ad libitum in association with decreases in B cell subsets ure 7H). Altogether, our findings indicate that exacerbation of
(Figures 5G and 5H). On the other hand, neither BHB nor rapa- diarrhea most likely resulted from disturbance in antigen-specific
mycin influenced chemokine expression in BLS12 cells (Figures mucosal immune responses as well as tolerogenic responses,

(D and E) Numbers of indicated cell subsets (D) and Cxcl12 and Cxcl13 mRNA expression (E) in PP and the BM of mice treated with vehicle (PBS) or rapamycin
(each group, n = 8).
Data represent the means ± SEM. ANOVA followed by Dunnett’s test (A) or Tukey’s test (C). Mann-Whitney U test (B and E). Student’s t test (D). *p < 0.05;
**p < 0.01; n.s., not statistically significant.
See also Figures S5 and S6.

Cell 178, 1072–1087, August 22, 2019 1079


A B

E F G

H I J

(legend on next page)

1080 Cell 178, 1072–1087, August 22, 2019


A B

C D

Figure 6. Repeated Fasting Suppresses Orally Induced Antigen-Specific Immune Responses


(A) Diagram illustrating the protocol for oral immunization with OVA and CT. Mice were fasted for 36 h and refed 48 h before immunization in the fasting-re-
fed group.
(B) Body weight of mice eating ad libitum or fasting-refed.
(C and D) OVA-specific IgA titers in feces (C) and OVA-specific IgA, IgM, and IgG titers in plasma (D) on day 32 from mice eating ad libitum or fasting-refed (each
group, n = 10).
Data represent the means ± SEM. Mann-Whitney U test (C and D). *p < 0.05; **p < 0.01; n.s., not statistically significant.
See also Figure S7.

underscoring the essential role of nutrient signals in the mainte- Under physiological conditions, a variety of food antigens
nance of gut immune homeostasis by securing PP cellularity. accompanied by opportunistic pathogens are continuously
delivered to the intestinal mucosa. The intestinal mucosa in adult
DISCUSSION humans possesses a total surface area of 200 m2. PPs conduct
immunosurveillance on the mucosal surface to eliminate
Our findings demonstrated the dynamic behavior of lympho- potentially hostile agents. A preconceived notion is that the gut
cytes during fasting and refeeding. Fasting decreased the mucosal immune system constitutively induces immune re-
number of PP lymphocytes while refeeding selectively restored sponses as evidenced by active GC reactions (Cesta, 2006).
naive—but not GC and IgA class-switched—B cells. GC B cells However, our findings revealed that immunological activity in
underwent massive apoptosis due to downregulation of PPs is nearly shut down during fasting where approximately
mTORC1 signaling during fasting, whereas PP-derived naive half of lymphocytes egress PPs or undergo apoptotic cell death.
B cells migrated into the BM in a CXCL13-dependent B cells comprise a major population of PPs, where the B cell/T
manner—at least in part. CXCL13 expression by peripheral cell ratio is 5-fold higher than in peripheral lymph nodes (Abbas
stromal cells was mostly dependent on glycolysis. Moreover, et al., 2014). We found that B cell populations were highly
repeated fasting attenuated antigen-specific IgA response susceptible to food deprivation; however, the physiological
and oral tolerance, which eventually exacerbated antigen- significance of this phenomenon remains to be elucidated. Given
induced diarrhea. that both luminal antigens and nutrient supply are greatly

Figure 5. Lymph Node-Derived Stromal Cells Depend on Glycolysis to Produce CXCL13


(A and B) Plasma glucose (A) and plasma b-hydroxybutyrate (BHB) (B) concentrations (each group, n = 8).
(C and D) BLS12 cells were stimulated with indicated stimulants for 24 h. CXCL13 transcripts were detected by qPCR (C, n = 6). The mitochondrial-specific
production of ROS (%MitoSOX+) and surface GLUT1 expression were detected by flow cytometry (D, n = 3). Data are representative of two independent
experiments.
(E) Cxcl13 expression in BLS12 cells cultured in control or glucose deprivation medium with TNF-a and anti-LTbR antibodies for 24 h. CXCL13 transcripts were
detected by qPCR (n = 6).
(F, I, and J) Cxcl13 expression in BLS12 cells treated with 2DG (F), BHB (I), and rapamycin (J), in the presence of TNF-a and anti-LTbR antibodies for 24 h, were
detected by qPCR (n = 6).
(G and H) Cxcl13 expression (G) and numbers of indicated cell subsets (H) in PPs of mice treated with vehicle (PBS) or 2DG (each group, n = 8).
Data represent the means ± SEM. ANOVA followed by Tukey’s test (A and B) or Dunnett’s test (C, D, F, and J). Mann-Whitney U test (E, G, and I). Student’s t test
(H). *p < 0.05; **p < 0.01; n.s., not statistically significant.

Cell 178, 1072–1087, August 22, 2019 1081


A B

C D

F
250 μl PBS

100 μl CFA

20 μl PBS (right ear)

G H

Figure 7. Repeated Fasting Exacerbates Food Antigen-Induced Diarrhea


(A) The protocol for OVA-induced diarrhea model. In the fasting group, mice were fasted for 36 h and refed 24 or 48 h before immunization. Related to Figures 7B–7E.
(B) Fecal clinical scores. Mice were fed ad libitum or were fasting-refed (each group, n = 14).
(C and D) Total and OVA-specific IgE (C), and IgG (D) in plasma (each group, n = 8).
(E) Total and OVA-specific IgA titers in feces (each group, n = 14).
(F–H) The protocol to assess oral tolerance (F). Mice were fed ad libitum or fasted for 48 h prior to oral administration with 25 mg OVA (tolerance) or PBS (control).
Delayed-type hypersensitivity (G), and OVA-specific IgG titers in plasma (H) (each group, n = 8).
Data represent the means ± SEM. Mann-Whitney U test (B, D, and E: OVA-specific IgG and IgA). Student’s t test (C–E: total IgE, IgA, and IgG, and OVA-specific
IgE). ANOVA followed by Tukey’s test (G) or Dunnett’s test (H). *p < 0.05; **p < 0.01; n.s., not statistically significant.

1082 Cell 178, 1072–1087, August 22, 2019


reduced during the fasting period, diminution of the lymphocyte rapidly by refeeding and PPs were mainly replenished by naive
pool may minimize energy expenditure. Despite hypoplastic B cells. Accordingly, the production of antigen-specific IgA in
morphology under fasting conditions, the fundamental micro- feces after repeated oral immunization with OVA and CT signifi-
structures of PPs remained intact and showed a rapid restora- cantly decreased when mice were fasted before immunization.
tion in response to refeeding. Such plasticity based on dynamic These results, together with previous observations, indicate
B cell movement characterizes PPs as mucosa-associated that repeated fasting may eliminate antigen-specific memory
lymphoid tissue. From another point of view, it is possible that B cells due to downregulation of mTOR signaling, which eventu-
the hypoplastic status is the default of PPs devoid of nutritional ally attenuates the mucosal immune response.
signals. Daily food intake most likely confers immunological ac- The importance of CXCL13 for the development and mainte-
tivity to PPs by recruiting lymphocytes, mainly naive B cells. nance of lymph nodes, including PPs, has been well docu-
The recirculation of naive B cells between PPs and the BM also mented (Ansel and Cyster, 2001; Ansel et al., 2000; Okada
occurred as circadian oscillation under physiological conditions. et al., 2002). This chemokine is mainly expressed by marginal
In mouse lymph nodes, noradrenalin-dependent b2-adrenergic reticular cells (MRCs) in lymphoid tissue (Katakai et al., 2008).
stimuli at night upregulate CCR7 and CXCR4 on lymphocytes Recent single-cell transcriptome analysis demonstrated that
to suppress cell egress from lymph nodes (Suzuki et al., 2016). MRCs and follicular dendritic cells (FDCs) individually express
Accordingly, lymphocytes accumulate in the lymph node at night CXCL13; however, the fact that MRCs outnumber FDCs impli-
and circulate in the blood during the day due to decreased cates MRCs as a significant source of CXCL13 (Rodda et al.,
noradrenaline levels. It remains an open question whether this 2018). BLS12 cells, which we used in this study to dissect the
mechanism also contributes to the circadian oscillation of PP molecular basis of CXCL13 production, share many characteris-
naive B cells. However, considering that fasting abrogated recir- tics with MRCs. For instance, BLS12 cells express several
culation of naive B cells between PP and the BM, food intake adherent molecules, including VCAM-1, ICAM-1, MAdCAM-1,
should serve as a primary element in the regulation of PP naive and RANKL. These molecules are also highly upregulated in
B cell dynamics. Indeed, the naive B cell population rapidly the MRC-like network of mucosa-associated lymphoid tissues,
expanded in response to refeeding; this rapid recovery cannot such as PPs, nasopharynx-associated lymphoid tissues, and
be explained by enhanced B cell generation, given that there CPs (Katakai et al., 2008). Furthermore, activation of protein
are multiple steps of differentiation from HSCs into naive B cells, kinase C and nuclear factor kB (NF-kB) pathways upon stimula-
via pro-B, pre-B, immature B, and transitional B cell stages tion with TNF-a and anti-LTbR agonist antibodies mediates
(Allman and Pillai, 2008; Shapiro-Shelef and Calame, 2005). CXCL13 expression in BLS12 cells (Katakai et al., 2008; Suto
Because naive B cells accumulate in the BM during fasting, we et al., 2009). We found that activated BLS12 cells undergo meta-
considered that the BM may function as a reservoir of naive bolic reprogramming to aerobic glycolysis, which is critical for
B cells to rapidly release the cells to mucosa-associated the production of CXCL13. This metabolic shift from oxidative
lymphoid tissue in response to food intake. In support of this phosphorylation to aerobic glycolysis is well characterized in
view, histological analysis of the BM of fasted mice detected activated M1 macrophages and monocytes as well as Th1 and
an accumulation of naive B cells in the BM cavity, especially in Th17 cells (Kelly and O’Neill, 2015; Michalek et al., 2011; O’Neill
the vicinity of blood vessels. Such a perivascular niche is also and Hardie, 2013). Unexpectedly, activation of mTOR signaling
known as a site for HSC differentiation and proliferation was found dispensable for metabolic reprogramming and
(Oh and Nör, 2015), suggesting that this region can establish a CXCL13 production in MRCs, because rapamycin treatment
microenvironment rich in cell survival factors (e.g., BAFF and failed to suppress both in in vitro and in vivo settings. Although
growth factors) for naive B cells (Schweighoffer and Tybulewicz, rapamycin administration did not influence Akt phosphorylation
2018; Zhang et al., 2004). in PPs, fasting markedly decreased phosphorylation levels. Akt
The Akt-mTOR signaling pathway is known to converge senses nutrients and upregulates not only the mTOR pathway
various external signals, including growth factors, insulin, but also other signaling pathways. For instance, Akt signaling
glucose, and amino acids (Laplante and Sabatini, 2012; Saxton activates phosphofructokinase 2/fructose-2,6-bisphosphatase
and Sabatini, 2017). Induction of GC B cells largely depends 2 (PFKFB2), a key regulator of glycolysis (Novellasdemunt
on mTOR signaling (Ersching et al., 2017). Over-activation of et al., 2013; Sreedhar et al., 2017). Notably, inhibition of glycol-
PI3K-Akt-mTOR and inhibition of mTOR exert positive and nega- ysis by 2DG or glucose deprivation significantly downregulated
tive effects on GC B in PPs, respectively (Zeng et al., 2016). Cxcl13 expression. Therefore, we speculate that CXCL13
Furthermore, B cell-specific deletion of Rictor, the core subunit expression by MRCs in PPs may be mediated by Akt signaling
of mTORC2, affects the survival and proliferation of B cells in an mTORC1-independent manner, although further investiga-
(Lee et al., 2013). Meanwhile, conditional deletion of a core tion is required to test this hypothesis.
mTORC1 protein in activated B cells arrests GC B cell differenti- We also found that repeated fasting exacerbates food anti-
ation, leading to a decrease in antigen-specific memory B cells gen-induced diarrhea in association with defects in antigen-
and plasma cells (Raybuck et al., 2018). In agreement with these specific IgA response and oral tolerance. In the gastrointestinal
studies, we also observed that rapamycin treatment prominently tract, dimeric IgA produced in the lamina propria is transported
reduced GC B cells in PPs. Because fasting mitigated mTOR to the mucosal surface by the action of polymeric Ig receptors
activity in PPs, we consider that the massive cell death of GC in intestinal epithelial cells (Corthésy, 2013; Pabst, 2012). Secre-
B cells during fasting is attributed to the downregulation of tory IgA may prevent translocation of luminal antigens in the
mTORC1 signaling. Furthermore, GC B cells did not recover body, and antigen-specific monomeric IgA in the blood prevents

Cell 178, 1072–1087, August 22, 2019 1083


anaphylaxis by competitively inhibiting the association of aller- d KEY RESOURCES TABLE
gens with antigen-specific IgE on mast cells (Yamaki et al., d LEAD CONTACT AND MATERIALS AVAILABILITY
2014). Selective IgA deficiency (IGAD) is the most common pri- d EXPERIMENTAL MODEL AND SUBJECT DETAILS
mary antibody deficiency, where IGAD patients frequently B Mice
develop allergic disorders, including asthma, atopic dermatitis, B Cell culture
and food allergies (Aghamohammadi et al., 2009; Schaffer d METHOD DETAILS
et al., 1991). Moreover, allergen-specific IgG protects against B Preparation of lymphocytes and BM cells
anaphylaxis and food allergy by neutralizing allergens and B Flow cytometry
cross-linking to an inhibitory IgG receptor, FcgRIIB (Strait B Adoptive transfer of PP cells
et al., 2006; Wagenaar et al., 2018). We consider that the exac- B Photoconversion of PPs from KikGR mice
erbation of diarrhea in fasting mice may be caused by the atten- B Oral immunization with OVA
uated induction of antigen-specific IgA and IgG, together with B Detection of antibody responses by ELISA
insufficient oral tolerance. The mechanism underlying the abnor- B Salmonella infection
mality in oral tolerance remains unknown. Early works have B Induction of oral tolerance
shown that PPs are essential for inducing oral tolerance to B Immunofluorescence
OVA (Fujihashi et al., 2001), although MLNs can also induce B Histological analysis
oral tolerance in the absence of PPs (Spahn et al., 2002). Several B Transmission electron microscopy
mechanisms have been proposed to account for oral tolerance, B Reverse transcription and quantitative PCR
which includes clonal deletion or anergy of allergen-specific B Bio-plex detection of phosphorylated proteins
T cells and the induction of regulatory T (Treg) cells (Pabst and B Plasma parameters
Mowat, 2012). Another study supports the central role of d QUANTIFICATION AND STATISTICAL ANALYSIS
effector/memory-type Treg cells in the establishment of oral
tolerance (Siewert et al., 2008). CD103+ dendritic cells drive SUPPLEMENTAL INFORMATION
the differentiation of Treg cells by secreting all-trans retinoic
acid. Furthermore, dietary antigens are required to induce pe- Supplemental Information can be found online at https://doi.org/10.1016/j.
cell.2019.07.047.
ripheral induction of Treg cells in the small intestine (Kim et al.,
2016). Fasting may thus affect the frequency of antigen-specific
ACKNOWLEDGMENTS
Treg and/or CD103+ dendritic cells. Collectively, our findings
demonstrate that nutritional stimuli are fundamental for main- We thank Yuuki Obata, Yutaka Nakamura, Naomi Hoshina, Hiroaki Shiratori,
taining gut immune homeostasis by facilitating antigen-specific Yuma Kabumoto, Hiyori Tanabe, Seiji Minegishi, and Teruki Hagiwara for
IgA and oral tolerance. technical support as well as Michio Tomura, Heiichiro Udono and Yun-Gi
Multiple studies have defined the beneficial effects of fasting or Kim for their valuable discussion and technical consultation. This work was
supported by AMED-Crest (16gm1010004h0101 and 17gm1010004h0102;
calorie restriction regarding metabolic diseases in overnutrition/
18gm1010004h0103 to K. Hase), the Japan Society for the Promotion of Sci-
obesity models. In these studies, time-restricted feeding, calorie ence (17KT0055, 16H01369, 18H04680, 25293114, and 26116709 to K. Hase),
restriction, fasting mimicking diets, and short/long-term fasting Keio Gijuku Academic Development Funds (to K. Hase), the SECOM Science
optimize nutritional balance to prevent or ameliorate multiple dis- and Technology Foundation (to K. Hase), the Takeda Science Foundation (to
orders, such as metabolic disorders, cardiovascular disease, and K. Hase.), the Science Research Promotion Fund, the Promotion and Mutual
autoimmune disorders (Brandhorst et al., 2015; Hatori et al., 2012; Aid Corporation for Private Schools of Japan (to K. Hase), Daiichi Sankyo
Foundation of Life Science (to K. Hase), Terumo Foundation for Life Science
Okada et al., 2017; Wei et al., 2017). In sharp contrast, a time-
and Arts (to K. Hase), Nagase Science Technology Foundation (to K. Hase),
restricted feeding regimen in juvenile mice exacerbated metabolic
The Tokyo Biochemical Research Foundation (to K. Hase), the National Center
disorders (Hu et al., 2019). This is analogous to our finding that for Global Health and Medicine (26-110 and 30-1006 to Y.I.K), Yoshida Schol-
repeated fasting in juvenile mice promoted food antigen-induced arship Foundation (to M.N.), and Keio University Doctorate Student Grant-
diarrhea. These observations suggest that fasting and time- in-Aid Program (to M.N.).
restricted feeding during growth may cause detrimental effects
depending on the feeding regimens and the age of test animals. AUTHOR CONTRIBUTIONS
In conclusion, we found that food intake secures the integrity
M.N. and R.N. performed most of the experiments and data analysis. M.N.
and function of the gut mucosal immune system through nutri-
wrote the manuscript. D.T., K.K., S.K., N.I., T.Y., Y.I.K., M.H., R.M., and M.S.
tional signaling. Nutritional deprivation impairs mucosal immunity, helped with animal experiments. K. Hattori and R.A. performed GF mouse ex-
leading to immune barrier dysfunction and excessive allergic periments. D.T. and K.M. performed KikGR mouse experiments. N.K. and Y.F.
response. Our study uncovered a novel link between nutritional performed infection experiments. M.T.-N. performed electron microscopy
signals and immune cell dynamics and functionality. Furthermore, analysis. T.Y. created the graphical abstract. Y.I.K. provided experimental re-
these findings may promote research and treatment courses for sources and discussed data. T.K. provided the cell line. S.S. provided tetanus
toxin. T.M. and K.T. analyzed the bone marrow data. T.D. conceived this study.
enhancing vaccine efficacy via dietary intervention.
K. Hase supervised the study. T.D. and K. Hase interpreted the data and
revised the manuscript.
STAR+METHODS
DECLARATION OF INTERESTS
Detailed methods are provided in the online version of this paper
and include the following: The authors declare no competing interests.

1084 Cell 178, 1072–1087, August 22, 2019


Received: December 5, 2018 Humphrey, J.H. (2009). Child undernutrition, tropical enteropathy, toilets, and
Revised: April 30, 2019 handwashing. Lancet 374, 1032–1035.
Accepted: July 25, 2019 Katakai, T., Hara, T., Sugai, M., Gonda, H., and Shimizu, A. (2004). Lymph node
Published: August 22, 2019 fibroblastic reticular cells construct the stromal reticulum via contact with
lymphocytes. J. Exp. Med. 200, 783–795.
REFERENCES
Katakai, T., Suto, H., Sugai, M., Gonda, H., Togawa, A., Suematsu, S., Ebi-
suno, Y., Katagiri, K., Kinashi, T., and Shimizu, A. (2008). Organizer-like
Abbas, A.K., Lichtman, A.H., and Pillai, S. (2014). Cellular and molecular immu-
reticular stromal cell layer common to adult secondary lymphoid organs.
nology, Eighth Edition (Elsevier), pp. 299–323.
J. Immunol. 181, 6189–6200.
Aghamohammadi, A., Cheraghi, T., Gharagozlou, M., Movahedi, M., Rezaei,
Kau, A.L., Ahern, P.P., Griffin, N.W., Goodman, A.L., and Gordon, J.I. (2011).
N., Yeganeh, M., Parvaneh, N., Abolhassani, H., Pourpak, Z., and Moin, M.
Human nutrition, the gut microbiome and the immune system. Nature 474,
(2009). IgA deficiency: correlation between clinical and immunological pheno-
327–336.
types. J. Clin. Immunol. 29, 130–136.
Kaufman, D.R., De Calisto, J., Simmons, N.L., Cruz, A.N., Villablanca, E.J.,
Allman, D., and Pillai, S. (2008). Peripheral B cell subsets. Curr. Opin. Immunol.
Mora, J.R., and Barouch, D.H. (2011). Vitamin A deficiency impairs vaccine-
20, 149–157.
elicited gastrointestinal immunity. J. Immunol. 187, 1877–1883.
Ansel, K.M., and Cyster, J.G. (2001). Chemokines in lymphopoiesis and
Kawamoto, T. (2003). Use of a new adhesive film for the preparation of multi-
lymphoid organ development. Curr. Opin. Immunol. 13, 172–179.
purpose fresh-frozen sections from hard tissues, whole-animals, insects and
Ansel, K.M., Ngo, V.N., Hyman, P.L., Luther, S.A., Förster, R., Sedgwick, J.D., plants. Arch. Histol. Cytol. 66, 123–143.
Browning, J.L., Lipp, M., and Cyster, J.G. (2000). A chemokine-driven positive
Kelly, B., and O’Neill, L.A. (2015). Metabolic reprogramming in macrophages
feedback loop organizes lymphoid follicles. Nature 406, 309–314.
and dendritic cells in innate immunity. Cell Res. 25, 771–784.
Basen-Engquist, K., and Chang, M. (2011). Obesity and cancer risk: recent re-
Kim, K.S., Hong, S.-W., Han, D., Yi, J., Jung, J., Yang, B.-G., Lee, J.Y., Lee, M.,
view and evidence. Curr. Oncol. Rep. 13, 71–76.
and Surh, C.D. (2016). Dietary antigens limit mucosal immunity by inducing
Brandhorst, S., Choi, I.Y., Wei, M., Cheng, C.W., Sedrakyan, S., Navarrete, G., regulatory T cells in the small intestine. Science 351, 858–863.
Dubeau, L., Yap, L.P., Park, R., Vinciguerra, M., et al. (2015). A Periodic Diet
Kunisada, Y., Eikawa, S., Tomonobu, N., Domae, S., Uehara, T., Hori, S., Fur-
that Mimics Fasting Promotes Multi-System Regeneration, Enhanced Cogni-
usawa, Y., Hase, K., Sasaki, A., and Udono, H. (2017). Attenuation of
tive Performance, and Healthspan. Cell Metab. 22, 86–99.
CD4+CD25+ Regulatory T Cells in the Tumor Microenvironment by Metformin,
Brown, E.M., Wlodarska, M., Willing, B.P., Vonaesch, P., Han, J., Reynolds, a Type 2 Diabetes Drug. EBioMedicine 25, 154–164.
L.A., Arrieta, M.C., Uhrig, M., Scholz, R., Partida, O., et al. (2015). Diet and spe-
Kunisawa, J., Sugiura, Y., Wake, T., Nagatake, T., Suzuki, H., Nagasawa, R.,
cific microbial exposure trigger features of environmental enteropathy in a
Shikata, S., Honda, K., Hashimoto, E., Suzuki, Y., et al. (2015). Mode of Bioen-
novel murine model. Nat. Commun. 6, 7806.
ergetic Metabolism during B Cell Differentiation in the Intestine Determines the
Caro-Maldonado, A., Wang, R., Nichols, A.G., Kuraoka, M., Milasta, S., Sun, Distinct Requirement for Vitamin B1. Cell Rep. 13, 122–131.
L.D., Gavin, A.L., Abel, E.D., Kelsoe, G., Green, D.R., and Rathmell, J.C.
Laplante, M., and Sabatini, D.M. (2012). mTOR signaling in growth control and
(2014). Metabolic reprogramming is required for antibody production that is
disease. Cell 149, 274–293.
suppressed in anergic but exaggerated in chronically BAFF-exposed B cells.
J. Immunol. 192, 3626–3636. Lee, K., Heffington, L., Jellusova, J., Nam, K.T., Raybuck, A., Cho, S.H.,
Thomas, J.W., Rickert, R.C., and Boothby, M. (2013). Requirement for Rictor
Carr, E.L., Kelman, A., Wu, G.S., Gopaul, R., Senkevitch, E., Aghvanyan, A.,
in homeostasis and function of mature B lymphoid cells. Blood 122,
Turay, A.M., and Frauwirth, K.A. (2010). Glutamine uptake and metabolism
2369–2379.
are coordinately regulated by ERK/MAPK during T lymphocyte activation.
J. Immunol. 185, 1037–1044. Limon, J.J., and Fruman, D.A. (2012). Akt and mTOR in B Cell Activation and
Differentiation. Front. Immunol. 3, 228.
Cesta, M.F. (2006). Normal structure, function, and histology of mucosa-
associated lymphoid tissue. Toxicol. Pathol. 34, 599–608. Lycke, N.Y., and Bemark, M. (2017). The regulation of gut mucosal IgA B-cell
Corthésy, B. (2013). Multi-faceted functions of secretory IgA at mucosal sur- responses: recent developments. Mucosal Immunol. 10, 1361–1374.
faces. Front. Immunol. 4, 185. Man, K., and Kallies, A. (2015). Synchronizing transcriptional control of T cell
Ersching, J., Efeyan, A., Mesin, L., Jacobsen, J.T., Pasqual, G., Grabiner, B.C., metabolism and function. Nat. Rev. Immunol. 15, 574–584.
Dominguez-Sola, D., Sabatini, D.M., and Victora, G.D. (2017). Germinal Center Mantis, N.J., Rol, N., and Corthésy, B. (2011). Secretory IgA’s complex roles in
Selection and Affinity Maturation Require Dynamic Regulation of mTORC1 immunity and mucosal homeostasis in the gut. Mucosal Immunol. 4, 603–611.
Kinase. Immunity 46, 1045–1058. Matloubian, M., Lo, C.G., Cinamon, G., Lesneski, M.J., Xu, Y., Brinkmann, V.,
Fujihashi, K., Dohi, T., Rennert, P.D., Yamamoto, M., Koga, T., Kiyono, H., and Allende, M.L., Proia, R.L., and Cyster, J.G. (2004). Lymphocyte egress from
McGhee, J.R. (2001). Peyer’s patches are required for oral tolerance to pro- thymus and peripheral lymphoid organs is dependent on S1P receptor 1. Na-
teins. Proc. Natl. Acad. Sci. USA 98, 3310–3315. ture 427, 355–360.
Grundy, S.M. (2004). Obesity, metabolic syndrome, and cardiovascular dis- Michalek, R.D., Gerriets, V.A., Jacobs, S.R., Macintyre, A.N., MacIver, N.J.,
ease. J. Clin. Endocrinol. Metab. 89, 2595–2600. Mason, E.F., Sullivan, S.A., Nichols, A.G., and Rathmell, J.C. (2011). Cutting
Hase, K., Kawano, K., Nochi, T., Pontes, G.S., Fukuda, S., Ebisawa, M., Kado- edge: distinct glycolytic and lipid oxidative metabolic programs are essential
kura, K., Tobe, T., Fujimura, Y., Kawano, S., et al. (2009). Uptake through for effector and regulatory CD4+ T cell subsets. J. Immunol. 186, 3299–3303.
glycoprotein 2 of FimH(+) bacteria by M cells initiates mucosal immune Novellasdemunt, L., Tato, I., Navarro-Sabate, A., Ruiz-Meana, M., Méndez-
response. Nature 462, 226–230. Lucas, A., Perales, J.C., Garcia-Dorado, D., Ventura, F., Bartrons, R., and
Hatori, M., Vollmers, C., Zarrinpar, A., DiTacchio, L., Bushong, E.A., Gill, S., Rosa, J.L. (2013). Akt-dependent activation of the heart 6-phosphofructo-
Leblanc, M., Chaix, A., Joens, M., Fitzpatrick, J.A.J., et al. (2012). Time- 2-kinase/fructose-2,6-bisphosphatase (PFKFB2) isoenzyme by amino acids.
restricted feeding without reducing caloric intake prevents metabolic diseases J. Biol. Chem. 288, 10640–10651.
in mice fed a high-fat diet. Cell Metab. 15, 848–860. O’Neill, L.A.J., and Hardie, D.G. (2013). Metabolism of inflammation limited by
Hu, D., Mao, Y., Xu, G., Liao, W., Ren, J., Yang, H., Yang, J., Sun, L., Chen, H., AMPK and pseudo-starvation. Nature 493, 346–355.
Wang, W., et al. (2019). Time-restricted feeding causes irreversible metabolic Oh, M., and Nör, J.E. (2015). The Perivascular Niche and Self-Renewal of Stem
disorders and gut microbiota shift in pediatric mice. Pediatr. Res. 85, 518–526. Cells. Front. Physiol. 6, 367.

Cell 178, 1072–1087, August 22, 2019 1085


Okada, T., Ngo, V.N., Ekland, E.H., Förster, R., Lipp, M., Littman, D.R., and for the induction of high-dose oral tolerance in the absence of Peyer’s patches.
Cyster, J.G. (2002). Chemokine requirements for B cell entry to lymph nodes Eur. J. Immunol. 32, 1109–1113.
and Peyer’s patches. J. Exp. Med. 196, 65–75. Sreedhar, A., Petruska, P., Miriyala, S., Panchatcharam, M., and Zhao, Y.
Okada, T., Fukuda, S., Hase, K., Nishiumi, S., Izumi, Y., Yoshida, M., Hagiwara, (2017). UCP2 overexpression enhanced glycolysis via activation of PFKFB2
T., Kawashima, R., Yamazaki, M., Oshio, T., et al. (2013). Microbiota-derived during skin cell transformation. Oncotarget 8, 95504–95515.
lactate accelerates colon epithelial cell turnover in starvation-refed mice. Strait, R.T., Morris, S.C., and Finkelman, F.D. (2006). IgG-blocking antibodies
Nat. Commun. 4, 1654. inhibit IgE-mediated anaphylaxis in vivo through both antigen interception and
Okada, T., Otsubo, T., Hagiwara, T., Inazuka, F., Kobayashi, E., Fukuda, S., Fc gamma RIIb cross-linking. J. Clin. Invest. 116, 833–841.
Inoue, T., Higuchi, K., Kawamura, Y.I., and Dohi, T. (2017). Intermittent fasting Suto, H., Katakai, T., Sugai, M., Kinashi, T., and Shimizu, A. (2009). CXCL13
prompted recovery from dextran sulfate sodium-induced colitis in mice. production by an established lymph node stromal cell line via lymphotoxin-
J. Clin. Biochem. Nutr. 61, 100–107. beta receptor engagement involves the cooperation of multiple signaling path-
Pabst, O. (2012). New concepts in the generation and functions of IgA. Nat. ways. Int. Immunol. 21, 467–476.
Rev. Immunol. 12, 821–832. Suzuki, K., Hayano, Y., Nakai, A., Furuta, F., and Noda, M. (2016). Adrenergic
Pabst, O., and Mowat, A.M. (2012). Oral tolerance to food protein. Mucosal control of the adaptive immune response by diurnal lymphocyte recirculation
Immunol. 5, 232–239. through lymph nodes. J. Exp. Med. 213, 2567–2574.
Poirier, P., Giles, T.D., Bray, G.A., Hong, Y., Stern, J.S., Pi-Sunyer, F.X., Tomura, M., Hata, A., Matsuoka, S., Shand, F.H.W., Nakanishi, Y., Ikebuchi,
and Eckel, R.H.; American Heart Association; Obesity Committee of the R., Ueha, S., Tsutsui, H., Inaba, K., Matsushima, K., et al. (2014). Tracking
Council on Nutrition, Physical Activity, and Metabolism (2006). Obesity and quantification of dendritic cell migration and antigen trafficking between
and cardiovascular disease: pathophysiology, evaluation, and effect of the skin and lymph nodes. Sci. Rep. 4, 6030.
weight loss: an update of the 1997 American Heart Association Scientific Uchimura, Y., Fuhrer, T., Li, H., Lawson, M.A., Zimmermann, M., Yilmaz, B.,
Statement on Obesity and Heart Disease from the Obesity Committee of Zindel, J., Ronchi, F., Sorribas, M., Hapfelmeier, S., et al. (2018). Antibodies
the Council on Nutrition, Physical Activity, and Metabolism. Circulation Set Boundaries Limiting Microbial Metabolite Penetration and the Resultant
113, 898–918. Mammalian Host Response. Immunity 49, 545–559.e5.
Rangan, P., Choi, I., Wei, M., Navarrete, G., Guen, E., Brandhorst, S., VanCott, J.L., Staats, H.F., Pascual, D.W., Roberts, M., Chatfield, S.N., Yama-
Enyati, N., Pasia, G., Maesincee, D., Ocon, V., et al. (2019). Fasting- moto, M., Coste, M., Carter, P.B., Kiyono, H., and McGhee, J.R. (1996). Regu-
Mimicking Diet Modulates Microbiota and Promotes Intestinal Regenera- lation of mucosal and systemic antibody responses by T helper cell subsets,
tion to Reduce Inflammatory Bowel Disease Pathology. Cell Rep. 26, macrophages, and derived cytokines following oral immunization with live
2704–2719. recombinant Salmonella. J. Immunol. 156, 1504–1514.
Raybuck, A.L., Cho, S.H., Li, J., Rogers, M.C., Lee, K., Williams, C.L., Shlom- Varanasi, S.K., Donohoe, D., Jaggi, U., and Rouse, B.T. (2017). Manipulating
chik, M., Thomas, J.W., Chen, J., Williams, J.V., and Boothby, M.R. (2018). Glucose Metabolism during Different Stages of Viral Pathogenesis Can Have
B Cell-Intrinsic mTORC1 Promotes Germinal Center-Defining Transcription either Detrimental or Beneficial Effects. J. Immunol. 199, 1748–1761.
Factor Gene Expression, Somatic Hypermutation, and Memory B Cell Gener-
Varona, R., Villares, R., Carramolino, L., Goya, I., Zaballos, A., Gutiérrez, J.,
ation in Humoral Immunity. J. Immunol. 200, 2627–2639.
Torres, M., Martı́nez-A, C., and Márquez, G. (2001). CCR6-deficient mice
Rodda, L.B., Lu, E., Bennett, M.L., Sokol, C.L., Wang, X., Luther, S.A., Barres, have impaired leukocyte homeostasis and altered contact hypersensitivity
B.A., Luster, A.D., Ye, C.J., and Cyster, J.G. (2018). Single-Cell RNA and delayed-type hypersensitivity responses. J. Clin. Invest. 107, R37–R45.
Sequencing of Lymph Node Stromal Cells Reveals Niche-Associated Hetero-
Visscher, T.L., and Seidell, J.C. (2001). The public health impact of obesity.
geneity. Immunity 48, 1014–1028.
Annu. Rev. Public Health 22, 355–375.
Sarbassov, D.D., Ali, S.M., Sengupta, S., Sheen, J.H., Hsu, P.P., Bagley, A.F.,
Wagenaar, L., Bol-Schoenmakers, M., Giustarini, G., Vonk, M.M., van Esch,
Markhard, A.L., and Sabatini, D.M. (2006). Prolonged rapamycin treatment
B.C.A.M., Knippels, L.M.J., Garssen, J., Smit, J.J., and Pieters, R.H.H.
inhibits mTORC2 assembly and Akt/PKB. Mol. Cell 22, 159–168.
(2018). Dietary Supplementation with Nondigestible Oligosaccharides Re-
Savy, M., Edmond, K., Fine, P.E.M., Hall, A., Hennig, B.J., Moore, S.E., Mulhol-
duces Allergic Symptoms and Supports Low Dose Oral Immunotherapy in a
land, K., Schaible, U., and Prentice, A.M. (2009). Landscape analysis of inter-
Peanut Allergy Mouse Model. Mol. Nutr. Food Res. 62, e1800369.
actions between nutrition and vaccine responses in children. J. Nutr. 139,
Wang, A., Huen, S.C., Luan, H.H., Yu, S., Zhang, C., Gallezot, J.D.,
2154S–2218S.
Booth, C.J., and Medzhitov, R. (2016). Opposing Effects of Fasting Meta-
Saxton, R.A., and Sabatini, D.M. (2017). mTOR Signaling in Growth, Meta-
bolism on Tissue Tolerance in Bacterial and Viral Inflammation. Cell 166,
bolism, and Disease. Cell 168, 960–976.
1512–1525.
Schaffer, F.M., Monteiro, R.C., Volanakis, J.E., and Cooper, M.D. (1991). IgA
Wei, M., Shinkura, R., Doi, Y., Maruya, M., Fagarasan, S., and Honjo, T. (2011).
deficiency. Immunodefic. Rev. 3, 15–44.
Mice carrying a knock-in mutation of Aicda resulting in a defect in somatic
Schmidt, T.H., Bannard, O., Gray, E.E., and Cyster, J.G. (2013). CXCR4 hypermutation have impaired gut homeostasis and compromised mucosal
promotes B cell egress from Peyer’s patches. J. Exp. Med. 210, defense. Nat. Immunol. 12, 264–270.
1099–1107.
Wei, M., Brandhorst, S., Shelehchi, M., Mirzaei, H., Cheng, C.W., Budniak, J.,
Schweighoffer, E., and Tybulewicz, V.L. (2018). Signalling for B cell survival. Groshen, S., Mack, W.J., Guen, E., Di Biase, S., et al. (2017). Fasting-
Curr. Opin. Cell Biol. 51, 8–14. mimicking diet and markers/risk factors for aging, diabetes, cancer, and car-
Scrimshaw, N.S., and SanGiovanni, J.P. (1997). Synergism of nutrition, infec- diovascular disease. Sci. Transl. Med. 9, eaai8700.
tion, and immunity: an overview. Am. J. Clin. Nutr. 66, 464S–477S. World Health Organization (2016). Report of the Commission on Ending Child-
Shapiro-Shelef, M., and Calame, K. (2005). Regulation of plasma-cell develop- hood Obesity (World Health Organization), pp. 1–68.
ment. Nat. Rev. Immunol. 5, 230–242. Yamaki, K., Nakashima, T., Miyatake, K., Ishibashi, Y., Ito, A., Kuranishi, A.,
Siewert, C., Lauer, U., Cording, S., Bopp, T., Schmitt, E., Hamann, A., and Taguchi, A., Morioka, A., Yamamoto, M., and Yoshino, S. (2014). IgA attenu-
Huehn, J. (2008). Experience-driven development: effector/memory-like al- ates anaphylaxis and subsequent immune responses in mice: possible appli-
phaE+Foxp3+ regulatory T cells originate from both naive T cells and naturally cation of IgA to vaccines. Immunol. Res. 58, 106–117.
occurring naive-like regulatory T cells. J. Immunol. 180, 146–155. Yamamoto, M., Fujihashi, K., Kawabata, K., McGhee, J.R., and Kiyono, H.
Spahn, T.W., Weiner, H.L., Rennert, P.D., Lügering, N., Fontana, A., (1998). A mucosal intranet: intestinal epithelial cells down-regulate intraepithe-
Domschke, W., and Kucharzik, T. (2002). Mesenteric lymph nodes are critical lial, but not peripheral, T lymphocytes. J. Immunol. 160, 2188–2196.

1086 Cell 178, 1072–1087, August 22, 2019


Yamazaki, S., Ema, H., Karlsson, G., Yamaguchi, T., Miyoshi, H., Shioda, S., Kinase Signaling and Glucose Metabolism Drive Follicular Helper T Cell Differ-
Taketo, M.M., Karlsson, S., Iwama, A., and Nakauchi, H. (2011). Nonmyelinat- entiation. Immunity 45, 540–554.
ing Schwann cells maintain hematopoietic stem cell hibernation in the bone Zhang, M., Ko, K.H., Lam, Q.L.K., Lo, C.K.C., Xu, D.J.L., Shen, L.,
marrow niche. Cell 147, 1146–1158. Zheng, B., Srivastava, G., and Lu, L. (2004). Novel function of TNF cyto-
Zeng, H., Cohen, S., Guy, C., Shrestha, S., Neale, G., Brown, S.A., Cloer, C., kines in regulating bone marrow B cell survival. Cell. Mol. Immunol. 1,
Kishton, R.J., Gao, X., Youngblood, B., et al. (2016). mTORC1 and mTORC2 447–453.

Cell 178, 1072–1087, August 22, 2019 1087


STAR+METHODS

KEY RESOURCES TABLE

REAGENT or RESOURCE SOURCE IDENTIFIER


Antibodies
Anti-cleaved caspase-3 (Asp175) (polyclonal) Cell Signaling Technology Cat#9661; RRID: AB_2341188
Anti-rat IgG (H+L) TRITC (polyclonal) Southern Biotech Cat#3030-03; RRID: AB_619945
Anti-rat IgG (H+L) Alexa Fluor 633 (polyclonal) Thermo Fisher Scientific Cat#A21094; RRID: AB_2535749
Anti-mouse TER-119 PerCP-Cy5.5 (TER-119) TONBO biosciences Cat#65-5921; RRID: N/A
Anti-mouse Lymphotoxin beta R/TNFRSF3 (polyclonal) R and D systems Cat#AF1008; RRID: AB_354531
Anti-mouse Ly6A/E PE-Cy7 (Sca-1) Biolegend Cat#122514; RRID: AB_756199
Anti-mouse Ly-6G/Gr1 PerCP-Cy5.5 (RB6-8C5) eBioscience Cat#45-5931; RRID: AB_906247
Anti-mouse Ki-67 PE-Cy7 (SolA15) eBioscience Cat#25-5698; RRID: AB_11220070
Anti-mouse IgM PE (R6-60.2) BD PharMingen Cat#553409; RRID: AB_394845
Anti-mouse IgM PE dozzle 594 (RMM-1) Biolegend Cat#406529; RRID: AB_2566585
Anti-mouse IgM HRP (polyclonal) Southern Biotech Cat#1020-05; RRID: AB_619903
Anti-mouse IgM Alexa Fluor 488 (polyclonal) Thermo Fisher Scientific Cat#A21042; RRID: AB_2535711
Anti-mouse IgG HRP (polyclonal) Southern Biotech Cat#1030-05; RRID: AB_2619742
Anti-mouse IgD APC (11-26c.2a) Biolegend Cat#405713; RRID: AB_10645480
Anti-mouse IgA HRP (polyclonal) Southern Biotech Cat#1040-05; RRID: AB_2714213
Anti-mouse IgA FITC(C10-3) BD PharMingen Cat#559354; RRID: AB_397235
Anti-mouse GL7 Pacific Blue (GL7) Biolegend Cat#144614; RRID: AB_2563292
Anti-mouse Endomucin (V.7C7.1) abcam Cat#ab106100; RRID: AB_10859306
Anti-mouse CD95 FITC (Jo2) BD PharMingen Cat#554257; RRID: AB_395329
Anti-mouse CD8a PerCP-Cy5.5 (53-6.7) TONBO biosciences Cat#65-0081; RRID: AB_2621882
Anti-mouse CD48 FITC (HM48-1) Biolegend Cat#103404; RRID: AB_313019
Anti-mouse CD45R/B220 V450 (RA3-6B2) BD Horizon Cat#560472; RRID: AB_1645276
Anti-mouse CD45R/B220 PE (RA3-6B2) eBioscience Cat#12-0452; RRID: AB_465671
Anti-mouse CD45R/B220 FITC (RA3-6B2) BD PharMingen Cat#553088; RRID: AB_394618
Anti-mouse CD45R/B220 APC-eFluor 780 (RA3-6B2) eBioscience Cat#47-0452; RRID: AB_1518810
Anti-mouse CD45R/B220 APC (RA3-6B2) eBioscience Cat#17-0452; RRID: AB_469395
Anti-mouse CD45R/B220 (RA3-6B2) eBioscience Cat#14-0452; RRID: AB_467254
Anti-mouse CD45R/B220 PerCP-Cy5.5 (RA3-6B2) TONBO biosciences Cat#65-0452; RRID: AB_2621892
Anti-mouse CD45 BV510 (30-F11) Biolegend Cat#103137; RRID: AB_2561392
Anti-mouse CD44 PE-Cy7 (IM7) eBioscience Cat#25-0441; RRID: AB_469623
Anti-mouse CD4 PerCP-Cy5.5 (RM4-5) TONBO biosciences Cat#65-0042; RRID: AB_2621876
Anti-mouse CD4 APC-eFluor 780 (GK1.5) eBioscience Cat#47-0041; RRID: AB_11218896
Anti-mouse CD3ε V500 (500A2) BD Horizon Cat#560771; RRID: AB_1937314
Anti-mouse CD3ε BV605 (145-2C11) Biolegend Cat#100351; RRID: AB_2565842
Anti-mouse CD3ε APC (145-2C11) TONBO biosciences Cat#20-0031; RRID: AB_2621537
Anti-mouse CD3ε (145-2C11) BD PharMingen Cat#550275; RRID: AB_393572
Anti-mouse CD34 FITC (RAM34) eBioscience Cat#11-0341; RRID: AB_465020
Anti-mouse CD31 (MEC 13.3) BD PharMingen Cat#550274; RRID: AB_393571
Anti-mouse CD16/CD32 Alexa Fluor 700 (93) eBioscience Cat#56-0161; RRID: AB_493994
Anti-mouse CD16/CD32 (2.4G2) TONBO biosciences Cat#70-0161; RRID: AB_2621487
Anti-mouse CD150/SLAM PE (TC15-12F12.2) Biolegend Cat#115904; RRID: AB_313683
Anti-mouse CD135 APC (A2F10) Biolegend Cat#135310; RRID: AB_2107050
Anti-mouse CD127/IL-7Ra PE (SB/119) Biolegend Cat#121111; RRID: AB_493510
(Continued on next page)

e1 Cell 178, 1072–1087.e1–e6, August 22, 2019


Continued
REAGENT or RESOURCE SOURCE IDENTIFIER
Anti-mouse CD11b PerC-/Cy5.5 (M1/70) TONBO biosciences Cat#65-0112; RRID: AB_2621885
Anti-mouse CD117/c-kit APC-Cy7 (2B8) Biolegend Cat#105826; RRID: AB_1626278
Anti-cleaved caspase-3 (Asp175) (polyclonal) Cell Signaling technology Cat#9661; RRID: AB_2341188
Anti-mouse GULT1 Alexa Fluor 647 (EPR3915) abcam Cat#ab195020; RRID: AB_2783877
Anti-hamster IgG (H+L) FITC (polyclonal) Southern Biotech Cat#6210-02; RRID:N/A
Bacterial and Virus Strains
rSalmonella–ToxC (DaroA, DaroD) VanCott et al., 1996 N/A
Chemicals, Peptides, and Recombinant Proteins
Cholera toxin (CT) List Biological Laboratories Cat#100B
Tetanus toxoid (TT) BIKEN foundation N/A
Freund’s Adjuvant, Complete (CFA) Sigma Aldrich Cat#F5881
Imject Alum Adjuvant Thermo Fisher Scientific Cat#77161
Alubmin, Chicken Egg (Ovalbumin) Grade V (OVA) Sigma Aldrich Cat#A5503; CAS: 9006-59-1
Rapamycin LC Laboratories Cat#R-5000; CAS: 53123-88-9
Fingolimod (FTY720) Cayman Chemical Company Cat#10006292; CAS: 162359-56-0
2-Deoxy-D-glucose (2DG) abcam Cat#ab142242; CAS: 154-17-6
3-Hydroxybutyric acid/b-hydroxybutyrate (BHB) Sigma Aldrich Cat#166898; CAS: 300-85-6
Murine Tumor Necrosis Factor-a (TNF-a) Peprotech Cat#315-01A
7-Aminoactinomycin D (7-AAD) TONBO Cat#13-6993
Fixable Viability Dye eFluor 780 Thermo Fisher Scientific Cat#65-0865
LIVE/DEAD Fixable Blue Dead Cell Stain Kit, for UV excitation Thermo Fisher Scientific Cat#L23105
SYTOX Blue Dead Cell Stain, for flow cytometry Thermo Fisher Scientific Cat#S34857
MitoSOX Red Mitochondrial Superoxide Indicator (MitoSOX-PE) Thermo Fisher Scientific Cat#M36008
CellTrace Cell Proliferation Kits CellTrace Violet (CellTrace Violet) Thermo Fisher Scientific Cat#C34557
Quetol 812 Nissin EM Cat#340
Deoxyribonuclease I (DNase I) Sigma Aldrich Cat#DN25; CAS: 9003-98-9
Collagenase Wako Cat#032-22364; CAS: 9001-12-1
cOmplete, mini Protease Inhibitor Cocktail Roche Cat#04-693-124-001
TRIzol Reagent Thermo Fisher Scientific Cat#15596026
EagleTaq Universal Master Mix (ROX) Roche Cat#07-260-288-190
Power SYBR Green PCR Master Mix Applied Biosystems Cat#4367659
Critical Commercial Assays
Bio-Plex Pro Cell Signaling Akt Panel 8-Plex Assay Bio Rad Cat#LQ0-0006JK0K0RR
LEGEND MAX Mouse OVA Specific IgE ELISA Kit with Biolegend Cat#439807
Pre-coated Plates
ELISA MAX Mouse Mouse IgE Biolegend Cat#432403
Mouse IgA ELISA Quantitation Set Bethyl Laboratories Cat#E90-103
Mouse IgG ELISA Quantitation Set Bethyl Laboratories Cat#E90-131
DeadEnd Colorimetric TUNEL System Promega Cat#G7130
Target Retrieval Solution Dako Cat#S1699
Protein Block Serum-Free Dako Cat#X0909
ENVISION+ System-HRP labeled Polymer Anti-Rabbit Dako Cat#K4002
VECTOR ImmPACT DAB Peroxidase Substrate Vector Laboratories Cat#SK4105
Lamina Propria Dissociation Kit Miltenyi Biotec Cat#130-097-410
Foxp3 / Transcription Factor Staining Buffer Set eBioscience Cat#00-5523-00
Rneasy Mini Kit QIAGEN Cat#74106
iScript Advanced cDNA Synthesis Kit for RT-qPCR BIO RAD Cat#1725038
Spotchem II glucose Arkray Cat#77301
(Continued on next page)

Cell 178, 1072–1087.e1–e6, August 22, 2019 e2


Continued
REAGENT or RESOURCE SOURCE IDENTIFIER
Ketone-H kit Serotech Cat#A350
Experimental Models: Cell Lines
BLS12 cell Katakai et al., 2004 N/A
Experimental Models: Organisms/Strains
Mouse: B6.B6129-Gt(ROSA)26Sor < tm1(CAG-kikGR) RIKEN RBC Cat# RBRC04847,
Kgwa > (KikGR mice) RRID:IMSR_RBRC04847
Mouse: BALB/cA mice CLEA Japan N/A
Oligonucleotides
Cxcl13 (Mm00444533_m1) Thermo Fisher Scientific Cat# 4331182
Primer: Rpl32 Forward: GGCTTTTCGGTTCTTAGAGGA Exigen N/A
Primer: Rpl32 Reverse: TTCCTGGTCCACAATGTCAA-30 ) Exigen N/A
Primer: Cxcl12 Forward: TTTCAGATGCTTGACGTTGG Exigen N/A
Primer: Cxcl12 Reverse: GCGCTCTGCATCAGTGAC Exigen N/A
Primer: Cxcl13 Forward: CTCCAGGCCACGGTATTCTG Exigen N/A
Primer: Cxcl13 Reverse: GGAGCTTGGGGAGTTGAAGA Exigen N/A
Primer; Ccl20 Forward: GGCAGAAGCAAGCAACTACG Exigen N/A
Primer; Ccl20 Reverse: CTTTGGATCAGCGCACACAG Exigen N/A
Software and Algorithms
Prism 8 GraphPad Software RRID:SCR_002798
FlowJo Version 10 FlowJo, LCC RRID:SCR_008520
DIVA software Version 6.2 BD Biosciences RRID:SCR_001456
IMARIS Version 9.2.0 ZEISS RRID:SCR_007370
ImageJ Software Version 1.49 NIH RRID:SCR_003070
Other
Mouse diet: CE-2 CLEA Japan N/A
Sporchem Arkray Cat#EZSP-4433
Fiber coupled Blue LED Light source Prizmatix Cat#Silver-LED-430
Dial-thickness gauge Mitsutoyo Cat#MDC-25PX

LEAD CONTACT AND MATERIALS AVAILABILITY

Further information and requests for reagents may be directed to, and will be fulfilled by the Lead Contact, Koji Hase (hase-kj@pha.
keio.ac.jp)

EXPERIMENTAL MODEL AND SUBJECT DETAILS

Mice
Unless otherwise stated, four to five-week-old male BALB/c mice were purchased from CLEA Japan Inc. (Tokyo, Japan) and were
acclimated for one week under specific pathogen-free (SPF) conditions at the animal facilities of the National Center for Global Health
and Medicine, Faculty of Pharmacy, Keio University (Tokyo, Japan). Knock-in mice carrying Kikume-Green Red (KikGR) cDNA under
the CAG promoter were obtained from RIKEN RBC (Tokyo, Japan) and were maintained under SPF conditions at the animal facilities
of Faculty of Pharmacy, Keio University (Tokyo, Japan). Germ-free (GF) BALB/cA mice (CLEA Japan Inc.) were maintained in GF vinyl
isolators at an animal facility in the Faculty of Medicine, Keio University. SPF and GF mice were fed with CE-2 (CLEA Japan) and were
kept under a 12:12 h light-dark cycle. During the fasting period, mice were kept in plastic cages without bedding chips or bait, with a
stainless mesh floor to avoid coprophagia, and with ad libitum drinking water. Irrespective of the fasting and refeeding period length,
refeeding or tissue collection was set to begin at 8:00 a.m. for all experiments. To examine the effect of rapamycin, 2DG and FTY720
in vivo, rapamycin (5 mg/kg; LC laboratories, Woburn, MA) was administrated intraperitoneally daily for seven consecutive days
(Zeng et al., 2016), 2DG (250 mg/kg; Abcam, Cambridge, UK) was also administrated i.p. three times every 12 h (Varanasi et al.,
2017), while FTY720 (1 mg/kg; Cayman Chemical Company, Ann Arbor, MI) was orally administrated three times every 12 h during
fasting. All animal experiments were performed according to the Institutional Guidelines for the Care and Use of Laboratory Animals in
Research with approval by the local ethics committees at the National Center for Global Health and Medicine, and Keio University.

e3 Cell 178, 1072–1087.e1–e6, August 22, 2019


Cell culture
BLS12 cells were cultured in DMEM (Sigma-Aldrich) supplemented with 10% FBS, GlutaMAX (GIBCO; Thermo Fisher Scientific),
1 mM sodium pyruvate (Sigma-Aldrich), and antibiotics, as previously described (Katakai et al., 2004). Cells were stimulated with
10 ng/mL murine TNF-a (PeproTech, Rocky Hill, NJ) and/or goat anti-mouse LTbR agonist antibodies (R&D Systems, Minneapolis,
MN) for 24 h. In a separate experiment, BLS12 cells were cultured in glucose-deprived medium containing 10% FBS, GlutaMAX,
MEM essential amino acids (Thermo Fisher Scientific), 1 mM sodium pyruvate, inorganic salts (1.8 mM CaCl2, 0.8 mM MgSO4,
5.3 mM KCl, 44 mM NaHCO3, 110 mM NaCl, and 0.9 mM NaH2PO4-H2O), and antibiotics in the presence of TNF-a and anti-
LTbR antibodies for 24 h. Control medium was supplemented with 1 mg/ml glucose in the glucose-deprived medium. For pharma-
cological inhibition of mTORC1 or glycolysis, BSL12 cells were treated with medium containing various concentrations of rapamycin
(LC Laboratories) or 2-deoxy-D-glucose (2DG; Abcam, Cambridge, UK) for 24 h. For analyzing the effect of b-hydroxybutyrate
(BHB; Sigma-Aldrich), BLS12 cells were treated with medium containing 25 mM HEPES (GIBCO) with or without 4 mM BHB. For
flow cytometric analysis, BLS12 cells were detached using Trypsin-EDTA solution (Nacalai Tesque, Kyoto, Japan) and washed
with PBS. Mitochondrial reactive oxygen species (ROS) production was detected using MitoSOX-PE (Thermo Fisher Scientific)
according to the manufacturer’s instructions.

METHOD DETAILS

Preparation of lymphocytes and BM cells


Peyer’s patches (PPs) were cut from the intestine and washed twice with phosphate-buffered saline (PBS; pH 7.2). PPs were then
minced and stirred in 30 mL RPMI 1640 medium (pH 7.2; Sigma-Aldrich, St. Louis, MO) containing 2% fetal bovine serum (FBS;
Sigma-Aldrich), 12.5 mM HEPES, 100 U/mL penicillin, 100 U/mL streptomycin, 0.5 mg/mL collagenase (Wako Pure Chemical Cor-
poration, Osaka, Japan), and 0.5 mg/mL DNase I (Sigma-Aldrich) for 30 min at 37 C. After filtration through a 70-mm cell strainer, the
cells were resuspended in PBS with 2% FBS. MLNs and CPs were mechanically dispersed into a single-cell suspension. Lamina
propria cells of the small intestine were prepared using the Lamina Propria Dissociation Kit (Miltenyi Biotec, Bergisch Gladbach, Ger-
many) according to manufacturer’s instructions. BM cells from the right femur and tibia, and splenocytes were mechanically
dispersed into a single-cell suspension. Red blood cells were removed by RBC lysis reagent (150 mM NH4Cl, 1 mM EDTA, and
0.4 mM NaHCO3) and then cells were suspended in PBS with 2% FBS.

Flow cytometry
For surface and intracellular staining, non-specific binding was blocked with Fc receptor antibody (clone: 2.4G2; Tonbo Biosciences,
San Diego, CA) prior to staining with fluorochrome-conjugated antibodies. For intracellular staining, lymphocytes were fixed,
permeabilized, and stained with monoclonal antibodies using the Foxp3 staining set (eBioscience, San Diego, CA) according to man-
ufacturer’s instructions. 7-AAD (Tonbo Biosciences), Fixable Viability Dye eFluor 780 (Thermo Fisher Scientific, Waltham, MA), LIVE/
DEAD Fixable Blue Dead Cell Stain Kit (Thermo Fisher Scientific), or SYTOX Blue Dead Cell Stain (Thermo Fisher Scientific) was used
to discriminate dead cells. The stained samples were analyzed using LSR II, LSRFortessa, and Aria III flow cytometers with DIVA
software (all from BD Biosciences, Franklin Lakes, NJ) and FlowJo software version 10 (FlowJo LLC, Ashland, OR).

Adoptive transfer of PP cells


PP cells from ad libitum-fed mice were prepared as described above. The cells were labeled with CellTrace Violet (Thermo
Fisher Scientific) according to the manufacturer’s instructions and then suspended in PBS. The fluorescence-labeled cells
(1 3 107 cells/mouse) were intravenously injected into recipient ad libitum-fed or 18 h-fasted recipients. Both groups were maintained
under the same conditions after cell transfer. Eighteen hours later, transferred cells were detected in PPs, BM, and MLNs by flow
cytometry.

Photoconversion of PPs from KikGR mice


Photoconversion of the PPs of KikGR mice was performed as described previously (Schmidt et al., 2013; Tomura et al., 2014). Briefly,
KikGR mouse was anesthetized with isoflurane, shaved with an electric razor, and antiseptically prepared with 10% povidone-iodine.
The skin was incised anteriorly at the midline below the costal margin, and then abdominal wall was incised. Each PP was sequen-
tially drawn out from abdominal cavity, and the surgical site was covered by a piece of sterile aluminum foil with a 5 mm hole punched
in it to leave only the PP exposed. A Silver LED 430 with a high numerical aperture polymer optical fiber light guide and fiber collimator
(Prizmatix) was used as a 430-nm blue light source. Each PP was exposed for 2 minutes and immediately replaced into the peritoneal
cavity to avoid drying. The abdominal cavity and skin were closed with 4–0 nylon suture (Natsume Seisakusho). After photoconver-
sion surgery, the mice fasted for 36 h or fed ad libitum. Photoconverted cells in the BM, MLNs and the spleen were analyzed by a
FACSAria III flow cytometer (BD Biosciences).

Oral immunization with OVA


In the fasting mouse group, 36-h fasts began at 8:00 p.m. on day 0. At 8:00 a.m. on day 4, all mouse groups were given 200 mL of 7.5%
sodium bicarbonate in HBSS to neutralize gastric acid 1 h prior to oral immunization with 250 mL OVA (4 mg/mL; Sigma Aldrich) and

Cell 178, 1072–1087.e1–e6, August 22, 2019 e4


CT (40 mg/mL; List Biological Laboratories, Campbell, CA) in PBS. This immunization protocol with or without fasting was repeated
on day 7, 14, and 21. Fecal samples were collected on day 0, 18, 25, and 32 to measure OVA-specific IgA levels by enzyme-linked
immunosorbent assays (ELISA), while plasma samples were collected on day 32 to measure OVA-specific IgM, IgA, and IgG levels
by ELISA.

Detection of antibody responses by ELISA


To measure fecal IgA, fecal samples were homogenized in PBS (1 mL/100 mg feces) containing 1 3 Complete Mini Protease Inhibitor
Cocktail (Roche, Basel, Switzerland) and 0.02% sodium azide (Wako Pure Chemical Corporation), followed by centrifugation to
collect the supernatant as a fecal extract. Microlon ELISA plates (96-well; Greiner Bio-One, Kremsmünster, Austria) were coated
with 1 mg/mL OVA in PBS at 4 C overnight. After washing, the wells were blocked with 4-fold diluted Block Ace (DS Pharma Biomed-
ical, Saita, Japan) for 1 h at room temperature. After washing four times with PBS containing 0.05% Tween 20 (PBS-T), serially diluted
plasma and fecal extracts were added in duplicate (100 mL/well). Fecal extracts from day 0 or plasma from non-immunized mice were
included as a negative control. Horseradish peroxidase (HRP)-conjugated anti-mouse IgA, IgM, or IgG (Southern Biotech, Birming-
ham, AL) were used as antibodies. After incubation at room temperature for 1 h, the plates were extensively washed with PBS-T.
Specific antibody binding was visualized by adding 3,30 ,5,50 -tetramethylbenzidine as a substrate (Sigma-Aldrich) and then the
reaction was terminated by 1.2 M H2SO4. Endpoint titers were expressed as the reciprocal log2 of the last dilution, giving OD450
values higher than control samples (Yamamoto et al., 1998). For measurements of plasma total IgG and fecal total IgA, Mouse
IgG ELISA Quantitation Set (Bethyl Laboratories) and Mouse IgA ELISA Quantitation Set (Bethyl Laboratories) were used. For mea-
surements of plasma total and OVA-specific IgE, ELISA MAX Mouse IgE (Biolegend) and LEGEND Mc Mouse OVA Specific IgE ELISA
Kit with Pre-coated Plates (Biolegend) were used according to the manufacturer’s instructions.

Salmonella infection
rSalmonella–ToxC (DaroA, DaroD) and TT were kindly provided by the BIKEN Foundation (Osaka, Japan)(VanCott et al., 1996).
Ad libitum or fasting group mice were orally immunized with 5 3 107 CFU of rSalmonella-ToxC on day 4. In the fasting group,
36-h fasts began at 8:00 p.m. on day 0, 17, and 31. TT-specific IgA in feces was measured by ELISA. Flat-bottomed, 96-well
MaxiSorp Nunc-Immuno plates were coated overnight with 500 ng/well of TT. Plates were blocked with 2% BSA in PBS, and optically
diluted fecal extracts and sera were added into the plate wells. The Mouse IgA ELISA Quantitation Set (Bethyl Laboratories) was used
for antibody detection. To produce HRP signals were visualized by adding 3,30 ,5,50 -tetramethylbenzidine as a substrate (Sigma-
Aldrich) and then the reaction was terminated by 1.2 M H2SO4.
Food antigen-induced diarrhea model
To establish diarrhea, female mice were injected intraperitoneally with 100 mL OVA (1 mg/mL) and alum (1 mg/mL; Thermo Fisher
Scientific) in PBS on day 0 and 7. Starting on day 14, OVA (50 mg) was administered orally every three days (on day 14, 17, and
20). In the fasting group, 36-h fasts began at 8:00 p.m. on day 10 and 17. The severity of allergic reactions to OVA was evaluated
based on total and OVA-specific IgE in plasma and diarrhea occurrence, which was assessed by visually monitoring the mice for
up to 1 h after oral challenge. Fecal and plasma samples were collected on day 14 and 20 to measure total and OVA-specific IgA
and IgG, respectively.

Induction of oral tolerance


Mice in the fasting group were fasted for 36 h. After 36-h refeeding (9:00 a.m. on day 3), the mice were gavaged with 25 mg OVA in
200 mL PBS. Control mice received PBS only. On day 10, the mice were immunized subcutaneously with 100 mg OVA in 100 mL com-
plete Freund’s adjuvant (CFA; Sigma-Aldrich). Delayed-type hypersensitivity (DTH) was measured on day 17 as described previously
(Fujihashi et al., 2001). Briefly, 20 mL PBS containing 10 mg OVA was injected into the left ear pinna of the mice, while the right ear pinna
received PBS as a negative control. After 24 h, ear swelling was measured using a dial thickness gauge (Mitutoyo, Kanagawa, Japan).
The DTH response was expressed as the difference in ear thickness between the right and left ears. Plasma samples were collected
on day 24 to measure OVA-specific IgG levels by ELISA.

Immunofluorescence
For immunostaining, PPs were snap-frozen in liquid nitrogen and embedded in OCT compound (Sakura, Tokyo, Japan). Frozen sec-
tions (4-mm thick) were fixed in dry ice-cold acetone for 15 min and then completely dried at room temperature for 1 h. After blocking
with an anti-CD16/CD32 antibody (Tonbo Biosciences) in 10-fold diluted Block Ace (blocking buffer; DS Pharma Biomedical) for
30 min, the sections were incubated with primary antibodies (hamster monoclonal anti-mouse CD3ε; BD pharmagen, or rat mono-
clonal anti-mouse CD45R/B220; eBiosciences) in blocking buffer overnight at 4 C. Bound antibodies were detected with FITC-
labeled anti-hamster (Southern Biotech) or TRITC-labeled anti-rat antibodies (Southern Biotech) and counterstained with DAPI.
The sections were then examined with a confocal microscope (BX50; Olympus, Tokyo, Japan).
Frozen BM sections (10-mm thick) were prepared according to the Kawamoto method (Kawamoto, 2003; Yamazaki et al., 2011)
and were fixed in 4% paraformaldehyde. After blocking with Protein Block (Dako, Jena, Germany) for 1 h, the fixed sections were
washed with 0.3% (v/v) Triton X-100 in PBS and incubated with Alexa Fluor 488-conjugated anti-IgM antibodies (Thermo Fisher
Scientific), anti-CD31 and anti-endomucin antibody, for 16 h at 4 C. The stained sections were again washed with 0.3% (v/v) Triton

e5 Cell 178, 1072–1087.e1–e6, August 22, 2019


X-100 in PBS and further stained with DAPI and secondary antibodies (Alexa Fluor 633-conjugated anti-rat IgG; Thermo Fisher Sci-
entific) for 4 h at room temperature. All antibodies were diluted in Protein Block. Immunofluorescence data were obtained and
analyzed with a confocal laser scanning microscope (FV1000; Olympus). B cell number and the distance between naive B cells
and vessels were determined using Imaris (Zeiss, Oberkochen, Germany) and ImageJ Software (NIH).

Histological analysis
PPs were fixed in 4% paraformaldehyde and embedded in paraffin. Tissue sections (3.5-mm thick) were then stained after deparaffi-
nization. For histological examination, the sections were stained with hematoxylin (Dako) and eosin (Wako Pure Chemical Corpora-
tion). Antigen retrieval was performed by autoclaving the sections in Target Retrieval Solution (Dako). Then, the sections were treated
with 3% H2O2 (Wako Pure Chemical Corporation) in methanol to inactivate endogenous peroxidase. After blocking with Protein Block
Serum-Free (Dako) for 1 h, the sections were incubated with anti-cleaved Caspase-3 (0.5 mg/mL; Cell Signaling Technology, MA,
USA) for 16 h at 4 C. After washing with PBS, sections were incubated with ENVISION+ System-HRP labeled Polymer Anti-Rabbit
(Dako) for 30 min at room temperature. The ImmPACTTM DAB peroxidase substrate (Vector Laboratories) was used for diaminoben-
zidine staining, and hematoxylin was used for counterstaining. For TUNEL staining, the DeadEnd Colorimetric TUNEL System
(Promega, Madison, WI) were used according to the manufacturer’s instructions. All sections were examined via confocal micro-
scopy (BX50).

Transmission electron microscopy


PPs were pre-fixed in an aldehyde mixture (2% paraformaldehyde and 2% glutaraldehyde in 30 mM HEPES buffer containing
100 mM NaCl and 2 mM CaCl2; pH adjusted to 7.4) for 1 h at room temperature and post-fixed in an aldehyde-OsO4 mixture (1%
OsO4, 1.25% glutaraldehyde, 1% paraformaldehyde, and 0.32% K3[Fe(CN)6] in 30 mM HEPES buffer; pH 7.4) for 1 h at room tem-
perature. The fixed blocks were washed three times with Milli Q water (Milli Q Integral; Merck Millipore, Burlington, MA), dehydrated
using a graded ethanol series, and then embedded in Quetol 812 (Nisshin EM, Tokyo, Japan). The resin blocks were sectioned
(70-nm thick) using an ultramicrotome (EM UC7; Leica, Wetzlar, Germany), contrasted with uranyl acetate and lead citrate, and finally
examined with a transmission electron microscope (JEM-1400; JEOL, Tokyo, Japan).

Reverse transcription and quantitative PCR


Total RNA from the BM was extracted using TRIzol Reagent (Thermo Fisher Scientific) while total RNA from PPs and BLS12 cells was
isolated using the RNeasy Mini Kit (QIAGEN, Hilden, Germany) according to manufacturer’s instructions. RNA was reverse-
transcribed to obtain cDNA using the iScript Advanced cDNA Synthesis Kit (Bio-Rad Laboratories, Hercules, CA). RT-qPCR was
performed using the 7900HT Fast Real-Time PCR System (Applied Biosystems, Foster City, CA) or the CFX384 Real-Time System
(Bio-Rad Laboratories) with SYBR Green (Applied Biosystems) and TaqMan assay (Roche). The oligonucleotide primers for Rpl32,
Cxcl12, Cxcl13, and Ccl20 were purchased from Exigen (Tokyo, Japan). TaqMan assay probe for Cxcl13 was obtained from Applied
Biosystems.

Bio-plex detection of phosphorylated proteins


Tissues were homogenized in 500 mL lysis solution (1X cell lysis factor QG and 2 mM phenylmethylsulfonyl fluoride in cell lysis buffer),
vortexed, and placed on ice. The tissue homogenate was transferred to a microcentrifuge tube and frozen at 70 C. The samples
were then thawed, sonicated, and centrifuged at 15,000 3 g for 10 min, after which the supernatant was collected. Protein concen-
trations were determined using the Pierce BCA Protein Assay Kit (Thermo Fisher Scientific) according to manufacturer’s instructions.
Lysates were adjusted to 200 mg/mL to detect the following key phosphorylated proteins of Akt downstream signaling, Akt (Ser473),
BAD (Ser136), GSK-3a/b (Ser21/Ser9), IRS-1 (Ser636/Ser639), mTOR (Ser2448), PTEN (Ser280), p70 S6 kinase (Thr389), and S6 ribosomal
protein (Ser235/Ser236), in PPs and the BM, followed by quantitative analysis with the Bio-Plex Phosphoprotein Assay (Bio-Plex Pro
Cell Signaling Akt Panel; 8-plex #lq00006jk0k0rr; Bio-Rad Laboratories) and the Bio-Plex 3D System (Bio-Rad Laboratories)
according to manufacturer’s instructions.

Plasma parameters
Plasma glucose levels were measured using Spotchem EZ SP-4430 (Arkray, Kyoto, Japan) with an automated dry chemistry system
(Spotchem II; Arkray). Plasma BHB concentration was measured using the Serotec Ketone-H Kit (Serotec Co., Ltd., Sapporo, Japan)
according to the manufacturer’s instructions.

QUANTIFICATION AND STATISTICAL ANALYSIS

For statistical analyses of two or more groups, we used Student’s t test or ANOVA followed by Tukey’s test. When variances were not
homogeneous, the data were analyzed by the non-parametrical Mann-Whitney U test or the Dunnett’s test. Two-way ANOVA was
applied to the time-course analysis of TT-specific IgA production. Differences with P-values < 0.05 were considered statistically sig-
nificant. Statistical analyses were performed using GraphPad Prism 8 software (GraphPad Software, Inc., La Jolla, CA). The exper-
iments were not randomized, and the investigators were not blinded to allocation during experiments and outcome assessment.

Cell 178, 1072–1087.e1–e6, August 22, 2019 e6


Supplemental Figures

Figure S1. The Behavior of Lymphocytes in Multiple Organs during Fasting and Refeeding, Related to Figure 2
(A) Numbers of the indicated cell subsets in PPs, the CPs, the MLNs, and SILP of mice fed ad libitum (PPs, n = 9; CPs, MLNs, and SILP, n = 10) or mice fasted for
36 h (PPs, n = 12; CPs, MLNs, and SILP, n = 10).
(B) Numbers of PP cells from proximal (Prox), middle (Mid), or distal (Dis) small intestine (each group, n = 10).
(C) Numbers of the indicated cell subsets from PPs at each time point. Mice were fasted for 36 h (blue background) and refed with CE2 for 72 h (red background)
(0, 84, and 108 h, n = 9; 24, 36, and 60 h, n = 12).
(D) Numbers of the indicated cell subsets in the spleen of mice fed ad libitum, mice fasted for 36 h, and mice refed with CE2 for 48 h (each group, n = 8).
Data represent the means ± SEM. Student’s t test (A and B). ANOVA followed by Dunnett’s test (C) or Tukey’s test (D). *p < 0.05; **p < 0.01; n.s., not significant.
(legend on next page)
Figure S2. Flow Cytometry Gating Strategies and Localization of Naive B Cells in the BM, Related to Figure 2
(A) Flow cytometry gating strategies on lymphocytes, granulocytes, macrophages, hematopoietic stem cells, and multipotent progenitors in the BM.
(B) Sections of the BM, which were obtained from mice fed ad libitum, fasted for 36 h, or refed with CE2 for 48 h, were stained with Abs against IgM (green) and
DAPI (gray) shown on the left and Abs against CD31 (blue), Emcn (blue), and DAPI (gray) shown on the right. The solid arrow indicates IgM+ naive B cells in
intravascular space. Scale bar: 20 mm.
(C) The ratio of IgM+ naive B cells in DAPI+. Calculated from Figure S2B (each group, n = 8).
Data represent the means ± SEM (C) ANOVA followed by Tukey’s test. *p < 0.05, **p < 0.01.
Figure S3. Minimal Contribution of Gut-Microbiota to the Behavior of Lymphocytes during Fasting, Related to Figure 2
Numbers of the indicated cell subsets in PPs and the BM of mice fed ad libitum (n = 9) or fasted for 36 h (n = 8). Six-week-old mice were kept under germ-free
conditions. Data represent the means ± SEM. Student’s t test. *p < 0.05; **p < 0.01.
Figure S4. The Behavior of Lymphocytes during Fasting and Refeeding in Aged Mice, Related to Figure 2
(A and B) Numbers of the indicated cell subsets in PPs and the BM of mice fed ad libitum, fasted for 36 h, or refed with CE2 for 48 h. Sixteen-week-old (A) and
40–50-week-old (B) mice were kept under SPF conditions (each group, n = 8).
Data represent the means ± SEM. ANOVA followed by Tukey’s test. *p < 0.05; **p < 0.01.
Figure S5. Minimal Effects of FTY720 on B Cell Dynamics in PPs and the BM, Related to Figure 4
(A) Diagram illustrating the FTY720 treatment protocol. FTY720 or vehicle (PBS) were orally administered into ad libitum-fed or fasting mice.
(B and C) Body weight (B) and numbers of the indicated cell subsets in PPs and the BM (C) of mice fed ad libitum or fasted for 36 h. Mice were treated with vehicle
or FTY720 (each group, n = 8).
Data represent the means ± SEM. ANOVA followed by Tukey’s test (B). Student’s t test (C).
*p < 0.05; **p < 0.01; n.s., not statistically significant.
Figure S6. Quantification of Phosphorylated Akt-mTOR Signaling Proteins in PPs and the BM, Related to Figure 4
(A and B) Quantification of phosphorylated Akt, BAD, GSK-3a/b, IRS-1, PTEN, S6 ribosomal protein, mTOR, and p70 S6 kinase in PPs (A, n = 18) and the BM
(B, n = 6) using whole tissue lysates. The mean of mean fluorescent intensity (MFI) from mice fasted for 36 h, mice refed with CE2 for 24 h and 48 h, and mice
treated with rapamycin was normalized by that of mice fed ad libitum. Data are represented as a heatmap.
Figure S7. The Effect of Fasting on Salmonella-Induced Mucosal Responses, Related to Figure 6
(A) Diagram illustrating the protocol for oral immunization model using S. Typhimurium expressing the fragment C of the tetanus toxoid (Salmonella-ToxC).
Mice were fasted for 36 h on day 0, 17, and 31 in the fasting-refed group.
(B) Absorbance of tetanus toxoid (TT)-specific IgA in feces from mice eating ad libitum (n = 8) or fasting-refed (n = 7). Data represent the means ± SEM.
Two-way ANOVA.

You might also like