Download as pdf or txt
Download as pdf or txt
You are on page 1of 23

nature aging

Letter https://doi.org/10.1038/s43587-023-00473-3

Induction of mitochondrial recycling reverts


age-associated decline of the hematopoietic
and immune systems

Received: 8 September 2021 Mukul Girotra1,8, Yi-Hsuan Chiang 1,8, Melanie Charmoy2, Pierpaolo Ginefra 1
,
Helen Carrasco Hope 1, Charles Bataclan 3, Yi-Ru Yu 1, Frederica Schyrr 3
,
Accepted: 24 July 2023
Fabien Franco1, Hartmut Geiger4, Stephane Cherix5, Ping-Chih Ho1,
Published online: 31 August 2023 Olaia Naveiras3,6, Johan Auwerx 7, Werner Held2 & Nicola Vannini 1

Check for updates


Aging compromises hematopoietic and immune system functions, making
older adults especially susceptible to hematopoietic failure, infections and
tumor development, and thus representing an important medical target for
a broad range of diseases. During aging, hematopoietic stem cells (HSCs)
lose their blood reconstitution capability and commit preferentially toward
the myeloid lineage (myeloid bias)1,2. These processes are accompanied by
an aberrant accumulation of mitochondria in HSCs3. The administration of
the mitochondrial modulator urolithin A corrects mitochondrial function
in HSCs and completely restores the blood reconstitution capability of
‘old’ HSCs. Moreover, urolithin A-supplemented food restores lymphoid
compartments, boosts HSC function and improves the immune response
against viral infection in old mice. Altogether our results demonstrate
that boosting mitochondrial recycling reverts the aging phenotype in the
hematopoietic and immune systems.

Hematopoietic stem cells (HSCs) generate all the blood and immune concept that cellular metabolism operates as a potent regulator of the
cells throughout the entire life of an organism and they ensure the cor- HSC pool12–14, and more broadly, of stem cell function and fate11,15,16.
rect homeostasis between lymphoid and myeloid lineages. However, Defective autophagy and consequent accumulation of mitochondria
aging dramatically reduces the blood reconstitution capability of HSCs in HSCs has been associated with aging processes3, providing evidence
and skews their fate toward myeloid lineages to the detriment of lym- that cellular quality control processes are crucial for the maintenance
phoid cell production1,2. In fact, specific age-related dysfunctions of of a functional HSC pool both in homeostasis and in stress condi-
the immune system stem from the deteriorated HSC pool4,5. Thus, it has tions17–19. Even though HSCs are refractory to systemic rejuvenating
been proposed that reversing HSC aging would improve both hemato­ approaches20, nutritional interventions, such as prolonged fasting and
poietic and immune functions6. HSC aging is coupled with increased nicotinamide adenine dinucleotide (NAD) boosting strategies, have
DNA damage due to an impairment of DNA repair enzymes7,8 and epi- been demonstrated to partially restore HSC function in old mice21,22.
genetic changes9,10. Interestingly, aging has also been directly linked In particular, NAD precursors have been shown to exert their effect by
to metabolic alterations in HSCs11. These studies led to the emerging directly modifying HSC mitochondrial function18,22.

Department of Oncology, Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland. 2Department of Oncology, University of
1

Lausanne, Epalinges, Switzerland. 3Laboratory of Regenerative Hematopoiesis, Department of Biomedical Sciences, University of Lausanne and ISREC,
School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland. 4Institute of Molecular Medicine, Ulm University, Ulm,
Germany. 5Orthopedic and Traumatology Service, Lausanne University Hospital, Lausanne, Switzerland. 6Hematology Service, Department of Oncology,
Lausanne University Hospital, Lausanne, Switzerland. 7Laboratory of Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique
Fédérale de Lausanne, Lausanne, Switzerland. 8These authors contributed equally: Mukul Girotra, Yi-Hsuan Chiang. e-mail: nicola.vannini@unil.ch

Nature Aging | Volume 3 | September 2023 | 1057–1066 1057


Letter https://doi.org/10.1038/s43587-023-00473-3

a Primary transplant Secondary transplant

Spleen Spleen
Competitor 0 4 . . 24 analysis
Ex vivo culture 0 4 . . 24 analysis
± UA
BM
Recipient BM Secondary
Old mouse analysis
LKSCD150+CD48− analysis recipient
(>100 weeks)

b Primary transplant c
Total Lymphoid Myeloid BM Spleen
80 60
***
100 ******* 125 NS 125 NS
*** *** *** **
*** *** **** *** **

Donor chimerism
**
Donor chimerism

60 *** *** *** 80 *** 100 100


*
40
60 75 75 Young control
40 Old control
40 50 50
20 Old UA
20 Young control 25 25
20 Old control
Old UA 0 0
0 0 0
0 4 8 12 16 20 24 0 4 8 12 16 20 24 0 4 8 12 16 20 24
Weeks Weeks Weeks

d Secondary transplant e Spleen BM


Total Lymphoid Myeloid Lymphoid Myeloid LKS
100 80 0.06 100 125 NS 125 125 NS
0.07
* * ** *
** *
NS
0.05 NS NS
** *

Donor chimerism
Donor chimerism

80 * * * 0.06 60
80 NS 100 100
** *
100

60 60 75 75 75
40
40 40 50 50 50
20 Young control 25 25 25
20 20
Old control
0 0 0 Old UA 0 0 0
0 4 8 12 16 20 0 4 8 12 16 20 0 4 8 12 16 20
Weeks Weeks Weeks

Fig. 1 | UA in vitro treatment reverses age-associated defects in HSCs. P < 0.0001; old control versus old UA P = 0.0003). d, Blood donor chimerism of
a, HSCs isolated from old mouse (100 weeks) BM were cultured for 3 days in the secondary transplant at the indicated time points (total: 4 weeks P = 0.0181;
presence of 20 μM UA. After culture, 2,000 cells were transplanted in lethally 8 weeks P = 0.0352; 12 weeks P = 0.0460; 16 weeks P = 0.0584; 20 weeks P = 0.0504;
irradiated primary recipient mice together with BM derived from competitor lymphoid: 4 weeks P = 0.006; 8 weeks P = 0.0455; 12 weeks P = 0.0669; 16 weeks
mice. At the endpoint, BM from primary recipient mice was transplanted into P = 0.0611; 20 weeks P = 0.0732; myeloid: 4 weeks P = 0.0414; 8 weeks P = 0.0352;
lethally irradiated secondary recipient mice. b, Blood donor chimerism analyses 12 weeks P = 0.1019; 16 weeks P = 0.1133; 20 weeks P = 0.0872). e, Spleen analyses
of primary transplant at the indicated time points (total: 4 weeks P = 0.0001; of donor chimerism at the endpoint and analysis of donor-derived stem and MPP
8 weeks P = 0.0002; 12 weeks P = 0.0001; 16 weeks P = 0.0001; 24 weeks P = 0.0001; population LKS (spleen lymphoid: young control versus old control P = 0.0034;
lymphoid: 8 weeks P = 0.0023; 12 weeks P = 0.0009; 16 weeks P = 0.0006; old control versus old UA P = 0.0231; spleen myeloid: young control versus old
24 weeks P = 0.0006; myeloid: 4 weeks P < 0.0001; 8 weeks P = 0.0001; 12 weeks control P = 0.0058; old control versus old UA P = 0.0404; LKS: young control
P = 0.0011; 16 weeks P = 0.0007; 24 weeks P = 0.0007). c, BM and spleen donor versus old control P < 0.0055; old control versus old UA P = 0.0156). n = 10; values
chimerism at the endpoint (BM: young control versus old control P < 0.0001; are the mean ± s.e.m., two-tailed Student’s t-test; *P ≤ 0.05, **P ≤ 0.01, ***P ≤ 0.001,
old control versus old UA P = 0.0003; spleen: young control versus old control ****P ≤ 0.0001. NS, not significant.

In this context, urolithin A (UA), a natural compound belonging secondary recipient mice, and donor chimerism analysis was followed
to the ellagitannin family23, has been shown to improve metabolic over a period of 20 weeks (Fig. 1a). The UA effect was maintained dur-
fitness in humans and rodents, and prolong the life span in worms ing secondary transplantation, where UA-treated old HSCs sustained
by sustaining mitophagy and mitochondrial function24–27. Because an identical blood reconstitution capability of young HSCs (Fig. 1d), pro-
HSCs have defective autophagy and accumulate mitochondria during duced a comparable donor-derived chimerism in primary and secon­
aging3,28 (Extended Data Fig. 1), we tested the capacity of UA to recover dary lymphoid tissues (Fig. 1e and Extended Data Fig. 2b), restored
the functions of HSCs purified from mice older than 18 months (‘old multipotent stem and progenitor population (Lin−c-Kit+Sca1+ (LKS))
HSCs’). Purified old HSCs (Lin−Sca1+c-Kit+CD48−CD150+) cultured for (Fig. 1e) and pure HSC pool (LKS CD150+CD48−) in the BM (Extended
3 days in the presence of 20 μM UA were transplanted into lethally irradi­ Data Fig. 2d). Similarly, UA treatment improved the hematopoietic
ated recipient mice and blood chimerism was assessed over a period of stem and progenitor activities of CD34+CD38−CD45RA− human HSCs
24 weeks by exploiting the CD45.1 and CD45.2 double congenic allelic purified from old adults in a colony-forming assay (Extended Data
system (Fig. 1a). Comparison of the blood reconstitution capabilities Fig. 3a), as evidenced by improved colony-forming capacity both in
revealed that exposure to UA restored the reconstitution function of primary (Extended Data Fig. 3b) and long-term secondary colony-
old HSCs to levels seen in HSCs purified from young mice (8–12 weeks forming assay, as measured by colony-forming units (CFUs) (Extended
old; ‘young HSCs’) (Fig. 1b) without significantly affecting the lym- Data Fig. 3c). Interestingly, UA treatment enhances the reconstitution
phoid and myeloid proportion during blood recovery (Extended Data capability of young HSCs even though to a minor extent compared to
Fig. 2a). The functional restoration of old HSCs was further confirmed by old HSCs (Extended Data Fig. 2e). Taken together, these data highlight
an increased donor chimerism in primary and secondary lymphoid tis- that a short in vitro exposure (3 days) to UA restores the long-term
sues, bone marrow (BM) and spleen, respectively (Fig. 1c and Extended blood reconstitution capability of old HSCs.
Data Fig. 2b). To ascertain the long-term HSC gain of function, BM To study whether UA recovers hematopoietic and immune
from primary recipient mice was transplanted into lethally irradiated functions when delivered systemically in vivo, we fed mice with

Nature Aging | Volume 3 | September 2023 | 1057–1066 1058


Letter https://doi.org/10.1038/s43587-023-00473-3

a Control diet
Competitor
UA diet
Weeks 0 16 Spleen Weeks 0 4 .. 20
analysis
BM Recipient Secondary
analysis recipient
Old

b Cellularity HSCs MPP CLPs


60 0.8 0.8 0.3
*
No. of cells (106)
50
*
0.6 0.6

Percentage
Percentage

Percentage
40 0.2

30 0.4 0.4

20 0.1
0.2 0.2
10
0 0 0 0
BM

CMP GMP MEPs


0.8 1.6 1.5

0.6
*

Percentage
Percentage

1.4 1.0

0.4
Old control
1.2 0.5 Old UA
0.2

0 1.0 0

c
Cellularity CD3+ 1.5 CD4+ 0.5 CD8+
30
2.0
No. of cells (106)

No. of cells (106)


0.4
No. of cells (106)

No. of cells (106)

20 1.5 1.0
Spleen

0.3
1.0
0.2
10 0.5
0.5 0.1

0 0 0

d Total Lymphoid Myeloid


100 *** 100 125
** ** *** **** ** ** *******
** ** *** *** ***
Donor chimerism
Donor chimerism
Donor chimerism

80 80 100
**
60 60 75

40 40 50

20 Old control 20 Old control 25 Old control


Old UA Old UA Old UA
0 0 0
0 4 8 12 16 20 0 4 8 12 16 20 0 4 8 12 16 20
Weeks Weeks Weeks
e Control diet
Competitor
UA diet
Weeks 0 16
Weeks 0 4 .. 16

Recipient
200 HSCs
Old LKS CD150+CD48–

f g Proportion of lymphoid and myeloid cells


50 ** 0.059
* * 0.068 0.14
Donor chimerism (%)

0.068
Percentage of cells

40 100

30

20 50 Myeloid
Young control
Lymphoid
10 Old control
Old UA
0 0
0 4 8 12 16
Young
Old
Old + UA

Young
Old
Old + UA

Young
Old
Old + UA

Young
Old
Old + UA

Weeks

4 weeks 8 weeks 12 weeks 16 weeks

a UA-enriched diet (50 mg kg day−1) for a period of 4 months, char- which was monitored monthly, was not different between UA-treated
acterized hematopoietic stem and progenitor pools and analyzed and untreated groups (Extended Data Fig. 4). However, analyses of
immune compartments (Fig. 2a). The cellular composition of blood, BM hematopoietic stem and progenitor populations (Extended

Nature Aging | Volume 3 | September 2023 | 1057–1066 1059


Letter https://doi.org/10.1038/s43587-023-00473-3

Fig. 2 | UA oral supplementation rejuvenates the hematopoietic system of old 12 weeks P = 0.0010; 16 weeks P = 0.0002; 20 weeks P < 0.0001; myeloid: 4 weeks
mice. a, Old mice (81–84 weeks old) were fed with a control or UA-supplemented P = 0.0021; 8 weeks P = 0.0094; 12 weeks P = 0.0009; 16 weeks P = 0.0003;
diet (50 mg kg day−1) for 16 weeks. Mice were euthanized to analyze the spleen 20 weeks P = 0.0002). n = 5; values are the mean ± s.e.m.; two-sided Student’s
and BM. A part of the BM was transplanted into lethally irradiated primary t-test; **P ≤ 0.01, ***P ≤ 0.001, ****P ≤ 0.0001. e, Two hundred HSCs purified
and secondary recipient mice. b, Analysis of different stem and progenitor from young, old and UA-fed old mice were transplanted into lethally irradiated
populations of the BM using flow cytometry (HSCs: P = 0.0147; CLPs: P = 0.0492; recipient mice. f, Blood donor chimerism analyses of the primary transplant
P = 0.0298). n = 5; values are the mean ± s.e.m.; two-sided Student’s t-test; at the indicated time points (4 weeks P = 0.0023; 8 weeks P = 0.068; 12 weeks
*P ≤ 0.05. c, Analysis of different T cell compartments in the spleen using flow P = 0.0599; 16 weeks P = 0.0455). n = 5; values are the mean ± s.e.m.; one-tailed
cytometry (n = 5; values are the mean ± s.e.m.). d, Blood donor chimerism Student’s t-tests; *P ≤ 0.05, **P ≤ 0.01. g, Analyses of donor lymphoid versus
analyses of the primary transplant at the indicated time points (total: 4 weeks myeloid compartments of primary transplants at the indicated time points
P = 0.0011; 8 weeks P = 0.0017; 12 weeks P = 0.0008; 16 weeks P = 0.0001; (8 weeks P = 0.0259; 12 weeks P = 0.068; 16 weeks P = 0.14). n = 5; values are the
20 weeks P < 0.0001; lymphoid: 4 weeks P = 0.0059; 8 weeks P = 0.0018; mean ± s.e.m.; one-tailed Student’s t-tests; *P ≤ 0.05.

Data Fig. 5a) revealed an expansion of HSCs and common lymphoid virus (LCMV) Armstrong strain, which resulted in an acute resolved
progenitor (CLP) cells (Lin−CD127+Sca1lowc-Kitlow) and a reduction of infection mediated by a CD8+ T cell response. The specific immune
megakaryocyte erythroid progenitor (MEP) cells (Lin−c-Kit+Sca1− response against the virus was assessed at day 8 after infection (Fig. 3a,b).
CD16/32−CD34−) (Fig. 2b), while no changes were observed in common Aging strongly reduced the abundance of CD8+ T cells specific for the
myeloid progenitor (CMP) cells (Lin−c-Kit+Sca1−CD16/32−CD34+), gp33 and np396 viral epitopes (Fig. 3a,b)30,31. UA supplementation
granulocyte monocyte progenitor (GMP) cells (Lin−c-Kit+Sca1−CD16/32+ increased the presence of np396-specific CD8+ T cells (Fig. 3c). Impor-
CD34−) and multipotent progenitor (MPP) cells (Lin−Sca1+c-Kit+ tantly, UA treatment improved the effector differentiation of old CD8+
CD48−CD150−) (Fig. 2b). Indeed, during aging CLPs are reduced while T cells as indicated by an increase in KLRG1+ cells (Fig. 3d) and granzyme
megakaryocyte progenitors expand29; thus, UA supplementation B (GzmB) expression by gp33- and np396-specific cells and increased
reverted the aging-associated features of the hematopoietic system. interferon-γ (INFγ) expression by np396 cells (Fig. 3e,f) (Extended Data
Interestingly, CLP expansion correlated with a partial expansion of lym- Fig. 6a). Moreover, UA-treated, virus-specific CD8+ T cells expressed
phocytes in the spleen (Fig. 2c). To test the BM functionality of old mice reduced levels of the inhibitory programmed cell death protein 1 (PD-1)
supplemented with UA, we transplanted whole BM into lethally irradi- and lymphocyte-activation gene 3 (LAG-3) receptors (Extended Data
ated mice (Fig. 2a). Donor chimerism analyses revealed that BM derived Fig. 6b). Consistent with increased effector differentiation and func-
from UA-treated old mice had a higher reconstitution capacity com- tion, UA-supplemented old mice showed better virus control (Fig. 3g).
pared to the untreated group (Fig. 2d) and no significant changes in the Next, we addressed whether the effects of UA feeding on the CD8+
lymphoid and myeloid proportion during blood recovery (Extended T cell response were directly due to the recovery of HSC function. To
Data Fig. 5b). Importantly, the improved reconstitution capability this end, mice were transplanted with a fixed number of HSCs derived
of HSCs was maintained on secondary transplantation (Extended from UA-fed mice (Fig.3h). Sixteen weeks after transplantation, mice
Data Fig. 5c). These data indicate that UA dietary supplementation were infected with LCMV (Armstrong strain), and donor-derived CD8+
boosts hematopoietic stem and progenitor functions and restores T cells were monitored by exploiting the double congenic allelic system
the CLP population in old mice. Moreover, to assess the cell-intrinsic (Fig. 3h). Despite the presence of virus-specific CD8+ T cells (Extended
effect of UA feeding on HSC function, a fixed number of HSCs purified Data Fig. 7a), the proportion of KLRG1+ cells were not different
from UA-fed mice were transplanted in lethally irradiated mice and (Extended Data Fig. 7b), while virus-specific CD8+ T cells derived from
blood chimerism was monitored over time (Fig. 2e). HSCs purified UA-treated HSCs showed improved GzmB expression (Fig. 3i), which
from UA-supplemented old mice had better blood reconstitution was more evident in GzmB-producing IFNγ+ cells (Fig. 3j). These results
capability, with an efficiency comparable to HSCs purified from young indicate that HSCs contribute directly to the improved functionality of
mice (Fig. 2f) and restored lymphoid lineage production (Fig. 2g). Taken CD8+ T cells. Importantly, UA in vitro treatment of splenocytes derived
together, these results indicate that UA supplementation improves the from old mice (Extended Data Fig. 8a) drove a mild modulation of
cell-intrinsic function of old HSCs. mitochondrial membrane potential without affecting mitochondrial
To evaluate whether the boost in hematopoietic capacity mass (Extended Data Fig. 8b) and enhanced functionality, as shown by
improves immune function, old mice supplemented for 8 weeks with a increased cytokine production (Extended Data Fig. 8c) in both CD8+
UA-enriched diet were infected with lymphocytic choriomeningitis and CD4+ T cells. Taken together, these data suggest that the UA-driven

Fig. 3 | UA supplementation improves the CD8+ T cell response to viral P = 0.0003; old control versus old UA P = 0.0005; np396+: young control versus
infection in old mice. a, Old mice (84 weeks old) were fed with a control or a old control P < 0.0001; old control versus old UA P = 0.0002). n = 10; values are
UA-supplemented diet (50 mg kg day−1) for 8 weeks before infection with LCMV the mean ± s.e.m.; one-way ANOVA; ***P ≤ 0.001, ****P ≤ 0.0001. f, INFγ+CD8+ T
(Armstrong strain). Mice were euthanized 8 days later and spleen cells were cells (np396+: young control versus old control P = 0.0044; old control versus old
analyzed. b, Gating strategy for virus-specific CD8+ T cells and their immune UA P = 0.0259). n = 10; values are the mean ± s.e.m.; one-way ANOVA; *P ≤ 0.05,
profile. FSC-A, forward scatter-area. c, Quantification of CD8+ T cells specific **P ≤ 0.01. g, Quantification of viral titers in the spleen (young control versus old
for the gp33 (gp33+) or np307 (np396+) epitope (gp33+: young control versus control P = 0.0092; old control versus old UA P = 0.0151). n = 10; values are the
old control P = 0.049; young control versus old UA P = 0.0007; np396+: young mean ± s.e.m.; one-way ANOVA; *P ≤ 0.05, **P ≤ 0.01. h, Mice stably reconstituted
control versus old control P < 0.0001; young control versus old UA P = 0.0002; with 200 HSCs derived from old mice fed with a control or a UA-supplemented
old control versus old UA P = 0.0222). n = 10; values are the mean ± s.e.m.; diet (50 mg kg day−1) were infected with LCMV (Armstrong strain) and euthanized
one-way analysis of variance (ANOVA); **P ≤ 0.01, ***P ≤ 0.001, ****P ≤ 0.0001. 8 days after infection to analyze the spleen. i, GzmB expression level in donor-
d, Gated gp33+ and np396+ CD44+CD8+ T cells were analyzed for KLRG1 derived, virus-specific CD8+ T (gp33+: old control versus old UA P = 0.07; np396+:
expression (gp33+: young control versus old control P < 0.0001; young control old control versus old UA P = 0.0354). n = 10; values are the mean ± s.e.m.;
versus old UA P = 0.0002; old control versus old UA P = 0.0007; np396+: young one-way ANOVA; *P ≤ 0.05. j, INFγ+ donor-derived, virus-specific CD8+ T cells
control versus old control P < 0.0001; young control versus old UA P = 0.0002; (gp33+: old control versus old UA P = 0.0249; np396+: young control versus
old control versus old UA P = 0.0222). n = 10; values are the mean ± s.e.m.; old UA P = 0.0149; old control versus old UA P = 0.0030). n = 5; values are the
one-way ANOVA; **P ≤ 0.01, ***P ≤ 0.001, ****P ≤ 0.0001. e, Analysis of GzmB mean ± s.e.m.; one-way ANOVA; *P ≤ 0.05, **P ≤ 0.01. NS, not significant.
expression in virus-specific CD8+ T cells (gp33+: young control versus old control

Nature Aging | Volume 3 | September 2023 | 1057–1066 1060


Letter https://doi.org/10.1038/s43587-023-00473-3

amelioration of immune function in old mice is due both to a direct based on the shift in the GFP signal18,32,33. Indeed, UA treatment induced
modulation of HSC and T cell functions. mitochondrial clearance in HSCs, as indicated by the increased propor-
Το assess whether the UA effect was exerted through mitochon- tion of cells with a high mitophagy rate (Fig. 4a) and by the decreased
drial recycling, we first tested the capacity of UA to clear mitochon- GFP to mCherry ratio on the whole HSC population (Extended Data
dria in HSCs. HSCs purified from mito-QC mitophagy reporter mice Fig. 9a). These results were further corroborated based on the increased
were cultured for 3 days in the presence of 20 μM UA, and mitophagy expression of key genes involved in autophagy and mitophagy (Atg5,
induction was measured using flow cytometry. Mito-QC mice express Park2 and p62) (Fig. 4b).
a tandem mCherry-green fluorescent protein (GFP) targeted to the Importantly, induction of mitochondrial clearance did not change
mitochondrial outer membrane. When mitochondria are degraded cellular basal and maximal respiration (Fig. 4c–e) of c-Kit+ hematopoi-
in lysosomes, the acidic environment quenches GFP but not mCherry etic progenitor cells, but was accompanied by an improved spare res-
fluorescence; consequently mitochondrial clearance can be measured piratory capacity (% SRC) (Fig. 4f), indicating improved mitochondrial

a LCMV b
(Armstrong strain)

CD44+ Tet CD44+ Tet+ KLRG1+
Control diet
CD8+

FSC-A

KLRG1
CD44
UA diet Day 8
after infection
Weeks 0 8

CD44

4 Days CD8 Tetramer CD127


washout (virus-specific)
Old

c d KLRG1+ KLRG1+
+ + (gp33+) (np396+)
gp33 np396
** ***
*** 80 80 NS Young control
4
***
6
**** Young control ****
Number of cells (106)

*** ****
Percentage of cells

Old control Old control


3
Old UA
60 60 ** Old UA
4
*
2 NS 40 40

2
1 20 20

0 0 0 0

e GzmB GzmB f GzmB GzmB g Viral titer


(gp33+) (np396+) (gp33+ IFNγ+) (np396+ IFNγ+) NS
NS NS
NS ** ** * Young control
200 150 **** 250
NS
NS 250
NS
*
107
Old control
*** *** 10 6
200 200
Normalized MFI

150 *** 105


Old UA
PFU/spleen

100
150 150 104
100
100 100 103
50 102
50 50 50
101
0 0 0 0 0

LCMV
(Armstrong strain)
h
Control diet
Competitor
CD45.1 and CD45.2
UA diet
Day 8
Weeks 0 16 after infection
Weeks 0 4 .. 16

Recipient
200 HSCs CD45.2
Old LKS CD150+CD48–
CD45.1

i GzmB GzmB j GzmB GzmB


(gp33+) (np396+) (gp33+ IFNγ+) (np396+ IFNγ+)
NS NS NS
5,000 10,000 15,000
P = 0.07 6,000
* *
*
4,000 8,000 NS **
Young control 10,000
NS 4,000 6,000 Young control
3,000 NS NS
Old control
MFI

Old control
MFI

2,000 4,000
2,000
Old UA 5,000 Old UA
1,000 2,000

0 0 0
0

Nature Aging | Volume 3 | September 2023 | 1057–1066 1061


Letter https://doi.org/10.1038/s43587-023-00473-3

a b 2.0
Old Control **
8 Old UA ** **
Mitophagy Mitophagy ** 1.5

Percentage of cells

Fold expression
6.7 6 NS
2.6
*
1.0
4
mCherry

2 0.5
Control
GFP UA
0 0
Pik3c3 Becn1 Atg5 Park2 p62

c d e f g
70 30 60
P = 0.05
Old Control NS NS 250 10
60 UA **
OCR (pmol min−1)

SRC (% of baseline)
OCR (pmol min−1)

OCR (pmol min−1)


200 8
50
20 40

MFI (103)
40 150 6
30
100 4
20 10 20
Old Control
10 50 2 Old UA

0 0 0 0 0
0 20 40 60 80
Time (min)

h 8 i 10 j 8
*
4 **
* high **
CD150 8
6 3 6

MFI (103)

MFI (103)
MFI (103)

MFI ratio

6
CD150

** 4
4 2
4
low
Old Control CD150 Old Control Old Control
2 1 Old Control 2
Old UA 2 Old UA Old UA
Old UA

0 0 FCS 0 0

gh

w
lo
hi

50
50

D1
D1

C
C

k l m n o
* 3
80 250
Control 50
NS 150 *
70 UA
SRC (% of baseline)
OCR (pmol min−1)

200
OCR (pmol min−1)

OCR (pmol min−1)

60 40 NS
2
50
30
100
150 MFI (103)
40
30 20 100 1
50
20
10 50
10
0 0 0
0 0
0 20 40 60 80
Time (min)

Fig. 4 | UA induces mitochondrial recycling and restores mitochondrial methyl ester (TMRM)/MitoTracker Green FM (MTG) (right) using flow cytometry
fitness. a, Analysis of mitochondrial clearance in HSCs purified from of cultured HSCs in the presence of UA (P = 0.0324); values are expressed as mean
mitophagy reporter mice (mito-QC) on a 3-day UA treatment in vitro measured fluorescence intensity (MFI). n = 3; values are the mean ± s.e.m.; two-tailed paired
as percentage of cells in the mitophagy gate (left) (P = 0.0037). n = 5; values Student’s t-test; *P ≤ 0.05. i, Measurement of mitochondrial mass by MitoTracker
are the mean ± s.e.m.; two-tailed paired Student’s t-test; *P ≤ 0.05, **P ≤ 0.01. Green staining in CD150high (P = 0.0028) and CD150low (P = 0.0048) HSCs cultured
b, Gene expression analyses by quantitative PCR of key genes involved in the for 3 days in the presence of UA; data are expressed as the MFI. n = 3; values are
autophagy and mitophagy processes in HSCs cultured for 3 days in the presence the mean ± s.e.m.; two-way ANOVA; **P ≤ 0.01. j, Analysis of PGC1α expression in
of UA (whole progeny) (Becn1 P = 0.035; Atg5 P = 0.0049; Park2 P = 0.0040; p62 HSCs cultured for 3 days in the presence of UA using flow cytometry (P = 0.0032).
P = 0.0058). n = 3; values are the mean ± s.e.m.; two-tailed paired Student’s t-test; n = 4; values are the mean ± s.e.m.; Student’s t-test; **P ≤ 0.01. k–n, Measurement
*P ≤ 0.05, **P ≤ 0.01). c–f, Measurement of OCR (c), basal respiration (d), maximal of OCR (k), basal respiration (l), maximal respiration (m) and SRC (% baseline) (n)
respiration (e) and SRC (% baseline) (f) of 3-day cultured Lin−c-Kit+ hematopoietic of Lin−c-Kit+ hematopoietic stem and progenitor population derived from mice
stem and progenitor population in the presence of UA (P = 0.0526) (n = 3; values fed with UA for 4 months (P = 0.0424). n = 3; values are the mean ± s.e.m.; two-
are the mean ± s.e.m.; paired two-tailed Student’s t-test). g, Analysis of CD150 tailed Student’s t-test; *P ≤ 0.05. o, Analysis of the mitochondrial mass of HSCs
expression (P = 0.0036). n = 3; values are the mean ± s.e.m.; two-tailed paired derived from mice fed with UA for 4 months. n = 3; values are the mean ± s.e.m.;
Student’s t-test; **P ≤ 0.01. h, Mitochondrial mass (MitoTracker) (P = 0.0127) Student’s t-test; *P ≤ 0.05,). NS, not significant.
(left) and mitochondrial activity per mitochondrial mass tetramethylrhodamine,

performance. As HSCs accumulate mitochondria due to autophagy reduced CD150 expression and mitochondrial mass in purified old
defects (Extended Data Fig. 1) and have increased expression of CD150 HSCs (Fig. 4g,h). Interestingly, old CD150high HSCs, which have a marked
during aging (Extended Data Fig. 9b)3,28, we investigated the capacity of myeloid bias and aging phenotype28, have higher mitochondrial con-
UA to revert these aging phenotypes in old HSCs. UA in vitro treatment tent compared to CD150low HSCs. Treatment with UA decreased the

Nature Aging | Volume 3 | September 2023 | 1057–1066 1062


Letter https://doi.org/10.1038/s43587-023-00473-3

mitochondrial content in both CD150high and CD150low HSCs compara- BM extraction and cell sorting
bly (Fig. 4i). Importantly, UA treatment increased the expression of the Flow cytometry analysis was performed on freshly isolated BM from
master regulator of mitochondrial biogenesis PGC1α in old HSCs C57BL/6 mice (young: 8 weeks old; old more than 80 weeks old). BM was
(Fig. 4j). These data indicate that UA treatment reestablishes mito­ extracted from crushed femur, tibia and pelvis. The cell suspension was
chondrial function and fitness by promoting mitochondrial recycling, filtered through a 70-μm cell strainer and erythroid cells were eliminated
which affects both mitochondrial clearance and biogenesis. by incubation with red blood cell lysis buffer (eBioscences). Isolation
We further investigated the effect of dietary UA supplementation and stains were performed in ice-cold PBS 1 mM EDTA. Lineage-positive
on metabolic features of hematopoietic stem and progenitor popula- cells were then removed with a magnetic lineage depletion kit (BD Bio-
tions. As observed in in vitro treatment, UA dietary supplementation sciences). The cell suspension was then stained with specific antibodies
did not change cellular basal and maximal respiration (Fig. 4k–m) but for progenitor and stem cell compartments and sorted on a BD FACSAria
improved SRC (Fig. 4k,n) of hematopoietic progenitor cells. Congru- III into 1.5-ml Eppendorf tubes. The HSC compartment was identified
ently, these metabolic changes were accompanied by a reduction of and sorted with the following cell surface phenotype LKS CD150+CD48−.
mitochondrial mass in the HSC population (Fig. 4o). Overall, our data
indicate that induction of mitochondrial recycling in old HSCs allows Antibodies
the restoration of mitochondrial function both in vitro and in vivo. The following antibodies were used in this study: rat monoclonal anti-
Interestingly, HSCs derived from Mx1-PARK2−/− mice, bearing spe- bodies (mAbs) against streptavidin− Texas Red, c-Kit-PeCy7 (clone 2B8),
cific deletion of the mitophagy gene PARK2 in the hematopoietic line- Sca1-allophycocyanin (APC) (clone D7), CD150-PE (clone TC-15-12F12.2),
age, fed with UA and transplanted in lethally irradiated mice (Extended CD48-PB (clone HM48-1), CD45.2-PB (clone 104), CD45.1-FITC (clone A20),
Data Fig. 9c), did not exhibit better blood reconstitution capacity Gr1-APC (clone RB6-8C5), F4/80-APC (clone BM8), CD19-PE (clone 6D5),
(Extended Data Fig. 9d). Consistently, deletion of PARK2 exacerbated CD3-PE (clone 17A2) and CD16/CD32 (clone 2.4G2). The antibodies were
the aging phenotype in HSCs, leading to a further shrinkage of the purchased from BioLegend, eBioscience and BD Biosciences. A mixture
lymphoid lineage (Extended Data Fig. 9e). These data indicate that of biotinylated mAbs against CD3, CD11b, CD45R/B220, Ly-6G, Ly-6C
mitochondrial clearance is necessary to restore HSC function. In fact, and TER-119 was used as lineage marker (‘lineage cocktail’) and was pur-
the maintenance of metabolic fitness and function of HSCs requires a chased from BD Biosciences. Human-specific antibodies were: hCD56
fine balance between the regeneration of newly functional mitochon- (clone NCAM16.2), hCD16 (clone 3G8), hCD45 (clone HI30), hCD19 (clone
dria and the clearance of old defective ones. HIB19), hCD4 (clone RPA-T4), hCD3 (clone SK7), hCD14 (clone M5E2),
Interestingly, NAD booster supplementation can partially reju- hCD8b (clone SIDI8BEE), hCD34 (clone 8G12) and hCD38 (clone HB-7),
venate ‘old’ HSCs through the restoration of the metabolic features of and were either from eBioscience or BD Biosciences; 4,6-diamidino-
‘young’ HSCs22. As we have observed on UA treatment, ‘old’ HSCs reduce 2-phenylindole staining was used for LIVE/DEAD cell discrimination.
their mitochondrial mass on nicotinamide riboside supplementation, For the viral infection analyses, the following antibodies were
reinforcing the concept that mitochondria are key regulators of the used: CD44-PB (clone IM781), KLRG1-APC (clone 2F1), CD127-BV786
aging process in HSCs3,22,34. Differently, UA treatment did not require (clone A7R34), PD-1-PE-Cy7 (clone RMP1-30), LAG3-PercPCy5.5 (clone
continuous supplementation to restore ‘old’ HSC function and, most eBioC9B7W), GrzB-PB (clone GB11), IFNγ-PercPCy5.5 (clone XMG1.2),
importantly, we showed that the effect observed in HSCs translated TNFα-PE-Cy7 (clone MP6-XT22) and IL2-APC (clone JES6-5H4).
into amelioration of immune functions22. Our data evidence that UA For the splenocyte culture assays, the following antibodies were
supplementation might have a major impact on older adults by rein- used: CD3ε (clone 145-2C11), CD8α (clone 53.6.7), CD4 (clone RM4-5),
forcing both their hematopoietic and immune systems, therefore IFNγ (catalog no. 505810, BioLegend) and TNFα (catalog no. 11-7321-82,
reducing the risk of hematopoietic failure, boosting immune surveil- Thermo Fisher Scientific).
lance and improving their response to vaccination. Another category
of patients who could benefit from UA supplementation is patients Mouse HSC, progenitor and human CD34+ cell culture
undergoing chemotherapy, which induces immunosenescent features, Murine HSCs were sorted in 1.5-ml Eppendorf tubes. Cells were seeded
increases the susceptibility to infections and reduces the response to in 96-well round bottom plates and cultured in Stemline II (Sigma-
vaccination. Aldrich) supplemented with 100 ng ml−1 stem cell factor (SCF) (R&D)
The loss of cellular quality control processes during aging leads and 2 ng ml−1 Flt3 (R&D). UA (Sigma-Aldrich) was added at the indicated
to the accumulation of defective cellular machineries and to a rapid concentrations (dissolved in dimethylsulfoxide (DMSO)); an equal
deterioration of cellular functions35. Our results demonstrate that UA quantity of DMSO was added to the control wells. All cultures were
treatment is capable to revert the metabolic defects of old HSCs and maintained at 5% CO2 at 37 °C. Cryopreserved CD34+ cells isolated
rejuvenates hematopoietic and immune system functions, providing from patients with hip replacement orthopedic surgery were thawed
further evidence on the important interplay between cellular metabo- and cultured in StemSpan (STEMCELL Technologies) medium sup-
lism and aging-related cellular dysfunctions. plemented with human SCF (100 ng ml−1), human FLT3L (100 ng ml−1),
human thrombopoietin (50 ng ml−1) and human low-density lipoprotein
Methods (10 μg ml−1), and different concentrations of UA (dissolved in DMSO)
This study complies with all relevant ethical regulations. For human were added; an equal amount of DMSO was added to the control wells.
sample collection the protocol has been approved by Commission All cultures were maintained at 5% CO2 at 37 °C.
Cantonale d’Éthique de la Recherche sur l’Être Humain CER-VD
(authori­zation no. CER-VD 2017-00846), where human CD34+ cells Analysis of mitochondrial mass using flow cytometry
were collected from patients who were informed about the research Postculture cells were incubated at 37 °C for 45 min with 100 nM
project according to a written consent. For the animal study the pro- MitoTracker Green. Cells were then washed with fluorescence-acti-
tocols have been approved by the Service de la Consummation et des vated cell sorting (FACS) buffer and analyzed by flow cytometry on a
Affaires Vétérinaires (authorization nos. VD3684 and VD3572). BD LSR II flow cytometer.

Animals DNA extraction and mitochondrial DNA and nuclear DNA


Experiments were performed on C57BL/6 female mice throughout estimation
the study. Young mice were 8 weeks old while old mice were more than A progeny of 1,000 cells were collected at the end of the culture period
80 weeks old. and DNA was isolated using the DNeasy kit (QIAGEN) according to

Nature Aging | Volume 3 | September 2023 | 1057–1066 1063


Letter https://doi.org/10.1038/s43587-023-00473-3

the manufacturer’s instructions. Quantitative PCR was carried out catalog no. 88-8824, eBioscience) followed by intracellular staining in
to estimate the relative values for mitochondrial (COX1) and nuclear 1× permeabilization buffer.
(NDUFA2) DNA to estimate the mitochondrial to nuclear DNA ratio.
Metabolic assay
Murine BM transplantations The oxygen consumption rate (OCR) was measured using a 96-well
C57BL/6 Ly5.2 adult mice (8–12 weeks old) were lethally irradiated Seahorse Bioanalyzer XF96 according to the manufacturer’s instruction
with a total 8.5 Gy dose in an X-ray irradiator (RS 2000, RAD source) using a Seahorse Mito Stress Test. Briefly, c-Kit+ hematopoietic progeni-
24 h before transplant. The dose was split in two doses of 4.25 Gy sepa- tor cells were sorted and suspended in Seahorse XF basic medium with
rated by a 4–6-h interval. Mice were injected alternatively with 2,000 11 mM glucose, 1 mM sodium pyruvate and 2 mM glutamine (pH 7.4, at
donor HSC progeny postculture or 200 donor HSCs freshly isolated or 37 °C) (100,000 cells per well). The injection ports were loaded with
150,000 total BM cells freshly isolated derived from C57BL/6 Ly5.1 mice 1 μM oligomycin, 2 μM carbonyl cyanide-p-trifluoromethoxyphenyl-
(young or old) and 150,000 competitor cells derived from C57BL/6 hydrazone and 0.5 μM rotenone/antimycin. During sensor calibration,
Ly5.1/5.2 mice, via tail vein injection. Peripheral blood was collected cells were incubated in a 37 °C non-CO2 incubator for 45 min.
every 4 weeks to determine the percentage of chimerism by FACS
analysis. Spleen and BM were analyzed at the endpoints. For secondary Splenocyte culture
transplants, adult mice (8–12 weeks old) were lethally irradiated with Splenocytes from old mice were stimulated in vitro with plate-bound
the same dose 24 h before transplant and each mouse was injected with anti-CD3 (3 μg ml−1, catalog no. 16-0037-85, Thermo Fisher Scientific)
3 million whole BM cells from a donor mouse. and soluble anti-CD28 (2 μg ml−1, catalog no. 102116, BioLegend) for
3 days and expanded in medium containing 10 ng ml−1 human inter-
Colony-forming unit assay leukin-2 (catalog no. 200-02, PeproTech) for 2 days. UA was supple-
A colony-forming unit (CFU) assay with human HSCs was carried mented at 5 μM. To assess cytokine production, cells were restimulated
out using MethoCult H4434 (STEMCELL Technologies) according to with plate-bound anti-CD3 (3 μg ml−1) for 4 h in presence of GolgiStop
the manufacturer’s instructions. One thousand cells from each well (catalog no. 554724, BioLegend) and brefeldin A (catalog no. 420601,
were plated in duplicate at different dilutions. Colonies were counted BioLegend) according to the manufacturer’s instruction. For mito-
15 days after plating using Stem Vision (STEMCELL Technologies). For chondrial mass and mitochondrial potential, cells were cultured at
secondary CFU assays, all cells were recovered from the primary CFU 37 °C for 30 min with 25 nM MitoTracker Green and 100 nM TMRM
plates and plated at serial dilutions (10–500 K per plate). Plates with before flow analysis.
fewer than 150 colonies were scored to determine the normalized
counts per 1,000 HSCs plated in the primary CFU assay. Mice model for mitophagy
Mito-QC mice in the C57BL/6J background provided by P.C. Ho and bred
Food preparation for in vivo feeding at the Ludwig Institute for Cancer Research (LICR), Lausanne were used
The UA diet was prepared by mixing custom synthesized (by Novalix) as reporter mice for mitophagy. Mitophagy induction was analyzed by
UA dissolved in DMSO with a 2916 powder diet (Charles River Labo- flow cytometry based on the shift on the GFP signal. Mx1-Cre mice were
ratories) and air-dried into pellets under sterile conditions. The UA provided by F. Radtke from École Polytechnique Fédérale de Lausanne
dosage in the mix was calculated by considering the average mouse (EPFL) while PARK2loxP/loxP mice were provided by P.C. Ho. The cross of
food intake per day (5 g food per mouse per day) for a calculated intake these two strains to achieve PARK2 deletion in the hematopoietic line-
of 50 mg UA per mouse per day). An equivalent amount of DMSO was age was performed at LICR.
added in the control food. Weekly food consumption was monitored
in all cages and no difference was found in the feeding behavior of mice qPCR
on UA supplementation in the diet. HSCs were FACS-sorted (Lin−c-Kit+Sca1+CD150+CD48−) and cultured in
vitro for 3 days in the presence or absence of UA (20 mM). After incu-
Viral infections and staining bation, total RNA was extracted from HSCs using the RNA Microprep
Mice were infected intraperitoneally with 2 × 105 plaque-forming units Kit (Zymo Research), according to the manufacturer’s instructions.
of LCMV 53b Armstrong strain. To determine viral titers, spleens from RNA was retrotranscribed to complementary DNA (cDNA) with the
LCMV-infected mice were ‘shock-frozen’. Diluted spleen suspensions PrimeScript cDNA Synthesis Kit (Takara Bio). qPCR with reverse tran-
were then used to infect Vero cells, and viral titers were determined by scription reactions was performed using 1.5 μl cDNA, 5 μl Power SYBR
an LCMV focus-forming assay, as described elsewhere36. Green Master Mix (Applied Biosystems) and 200 nM of primers, and
Cell suspensions from the spleen were obtained by mashing analyzed on the 7900HT system (Applied Biosystems). The relative
through a 40-μM nylon cell strainer, followed by red blood cell lysis expression of Actb was used to normalize gene expression across sam-
using ammonium-chloride-potassium buffer. Surface staining was ples. The following primers were used: Atg5 (forward: GGAGAGAAGAG-
performed with mAbs for 20 min at 4 °C in PBS supplemented with 2% GAGCCAGGT; reverse: GCTGGGGGACAATGCTAATA); Park2 (forward:
FCS (FACS buffer). CCGAATCACCTGACGGTTCA; reverse: TCTGGCTGCTTCTGAATCCC);
For tetramer staining, cell suspensions were incubated with anti- Becn1 (forward: CCGCGGTAGAACGAGCC; reverse: AAGTAATGGAGCT
CD16/32 (clone 2.4G2) hybridoma supernatant before staining for GTGAGTTCCT); Pik3c3 (forward: GTGAAGTACCCTGACCTGCC; reverse:
90 min at 4 °C with APC-conjugated major histocompatibility complex AGTCATGCATTCCTTGGCGA); p62 (forward: GCTGAAGGAAGCTGCC
class I tetramers. CTAT; reverse: TTGGTCTGTAGGAGCCTGGT); Actb (forward: GAGAC
For intranuclear staining, cells were surface-stained before fixa- CTTCAACACCCC; reverse: GTGGTGGTGAAGCTGTAGCC).
tion and permeabilization using the Foxp3 Transcription Factor Stain-
ing Kit (catalog no. 00-5523, eBioscience) followed by intranuclear Statistic and reproducibility
staining in permeabilization buffer 1× (permeabilization buffer). All experiments were reproduced at least once. Data were analyzed
For the detection of cytokine production, splenocytes were res- using two-sided or one-sided Student’s t-tests or a two-way ANOVA or
timulated in vitro with LCMV gp33-41 (gp33) or np396-404 (1 μM) Mann–Whitney U-test with a 95% confidence interval.
peptide for 5 h in the presence of brefeldin A (5 μg ml−1) for the last No statistical method was used to predetermine sample size. Sample
4.5 h. Cells were then surface-stained before fixation and permeabili- sizes were chosen based on previous studies18,37. The investigators were
zation (using the Intracellular Fixation & Permeabilization Buffer Set, not blinded to allocation and outcome assessment. Some data points

Nature Aging | Volume 3 | September 2023 | 1057–1066 1064


Letter https://doi.org/10.1038/s43587-023-00473-3

were excluded from the analyses based on an outlier Grubbs statistical 19. Mohrin, M. et al. Stem cell aging. A mitochondrial UPR-mediated
test or when an animal developed splenomegaly. Data distribution was metabolic checkpoint regulates hematopoietic stem cell aging.
assumed to be normal but this was not formally tested. Science 347, 1374–1377 (2015).
20. Ho, T. T. et al. Aged hematopoietic stem cells are refractory to
Reporting summary bloodborne systemic rejuvenation interventions. J. Exp. Med. 218,
Further information on research design is available in the Nature Port- e20210223 (2021).
folio Reporting Summary linked to this article. 21. Cheng, C.-W. et al. Prolonged fasting reduces IGF-1/PKA to
promote hematopoietic-stem-cell-based regeneration and
Data availability reverse immunosuppression. Cell Stem Cell 14, 810–823 (2014).
All data and materials are available from the corresponding author 22. Sun, X. et al. Nicotinamide riboside attenuates age-associated
to any researcher for the purpose of reproducing or extending the metabolic and functional changes in hematopoietic stem cells.
analyses. Source data are provided with this paper. Nat. Commun. 12, 2665 (2021).
23. Larrosa, M., García-Conesa, M. T., Espín, J. C. & Tomás-Barberán,
References F. A. Ellagitannins, ellagic acid and vascular health. Mol. Aspects
1. Morrison, S. J., Wandycz, A. M., Akashi, K., Globerson, A. & Med. 31, 513–539 (2010).
Weissman, I. L. The aging of hematopoietic stem cells. Nat. Med. 24. Ryu, D. et al. Urolithin A induces mitophagy and prolongs lifespan
2, 1011–1016 (1996). in C. elegans and increases muscle function in rodents. Nat. Med.
2. Pang, W. W. et al. Human bone marrow hematopoietic stem 22, 879–888 (2016).
cells are increased in frequency and myeloid-biased with age. 25. Luan, P. et al. Urolithin A improves muscle function by inducing
Proc. Natl Acad. Sci. USA 108, 20012–20017 (2011). mitophagy in muscular dystrophy. Sci. Transl. Med. 13, eabb0319
3. Ho, T. T. et al. Autophagy maintains the metabolism and function (2021).
of young and old stem cells. Nature 543, 205–210 (2017). 26. Andreux, P. A. et al. The mitophagy activator urolithin A is safe and
4. Geiger, H., de Haan, G. & Florian, M. C. The ageing haematopoietic induces a molecular signature of improved mitochondrial and
stem cell compartment. Nat. Rev. Immunol. 13, 376–389 (2013). cellular health in humans. Nat. Metab. 1, 595–603 (2019).
5. Chandel, N. S., Jasper, H., Ho, T. T. & Passegué, E. Metabolic 27. D’Amico, D. et al. Impact of the natural compound urolithin A on
regulation of stem cell function in tissue homeostasis and health, disease, and aging. Trends Mol. Med. 27, 687–699 (2021).
organismal ageing. Nat. Cell Biol. 18, 823–832 (2016). 28. Beerman, I. et al. Functionally distinct hematopoietic stem cells
6. Hine, C. & Mitchell, J. R. Saying no to drugs: fasting protects modulate hematopoietic lineage potential during aging by a
hematopoietic stem cells from chemotherapy and aging. Cell mechanism of clonal expansion. Proc. Natl Acad. Sci. USA 107,
Stem Cell 14, 704–705 (2014). 5465–5470 (2010).
7. Flach, J. et al. Replication stress is a potent driver of functional 29. Ho, Y. H. et al. Remodeling of bone marrow hematopoietic stem
decline in ageing haematopoietic stem cells. Nature 512, 198–202 cell niches promotes myeloid cell expansion during premature or
(2014). physiological aging. Cell Stem Cell 25, 407–418 (2019).
8. Beerman, I., Seita, J., Inlay, M. A., Weissman, I. L. & Rossi, D. J. 30. Decman, V. et al. Defective CD8 T cell responses in aged mice
Quiescent hematopoietic stem cells accumulate DNA damage are due to quantitative and qualitative changes in virus-specific
during aging that is repaired upon entry into cell cycle. Cell Stem precursors. J. Immunol. 188, 1933–1941 (2012).
Cell 15, 37–50 (2014). 31. Ucar, D. et al. The chromatin accessibility signature of human
9. Beerman, I. & Rossi, D. J. Epigenetic regulation of hematopoietic immune aging stems from CD8+ T cells. J. Exp. Med. 214,
stem cell aging. Exp. Cell Res. 329, 192–199 (2014). 3123–3144 (2017).
10. Chambers, S. M. et al. Aging hematopoietic stem cells decline in 32. McWilliams, T. G. et al. mito-QC illuminates mitophagy and
function and exhibit epigenetic dysregulation. PLoS Biol. 5, e201 mitochondrial architecture in vivo. J. Cell Biol. 214, 333–345 (2016).
(2007). 33. Klionsky, D. J. et al. Guidelines for the use and interpretation of
11. Sahin, E. & Depinho, R. A. Linking functional decline of telomeres, assays for monitoring autophagy (4th edition)1. Autophagy 17,
mitochondria and stem cells during ageing. Nature 464, 520–528 1–382 (2021).
(2010). 34. Mansell, E. et al. Mitochondrial potentiation ameliorates age-
12. Nakada, D., Saunders, T. L. & Morrison, S. J. Lkb1 regulates cell related heterogeneity in hematopoietic stem cell function.
cycle and energy metabolism in haematopoietic stem cells. Cell Stem Cell 28, 241–256 (2021).
Nature 468, 653–658 (2010). 35. Jovaisaite, V., Mouchiroud, L. & Auwerx, J. The mitochondrial
13. Gurumurthy, S. et al. The Lkb1 metabolic sensor maintains unfolded protein response, a conserved stress response pathway
haematopoietic stem cell survival. Nature 468, 659–663 (2010). with implications in health and disease. J. Exp. Biol. 217, 137–143
14. Gan, B. et al. Lkb1 regulates quiescence and metabolic (2014).
homeostasis of haematopoietic stem cells. Nature 468, 701–704 36. Battegay, M. et al. Quantification of lymphocytic choriomeningitis
(2010). virus with an immunological focus assay in 24- or 96-well plates.
15. Zhang, J. et al. UCP2 regulates energy metabolism and J. Virol. Methods 33, 191–198 (1991).
differentiation potential of human pluripotent stem cells. EMBO J. 37. Vannini, N. et al. Specification of haematopoietic stem cell fate
30, 4860–4873 (2011). via modulation of mitochondrial activity. Nat. Commun. 7, 13125
16. Suda, T., Takubo, K. & Semenza, G. L. Metabolic regulation of (2016).
hematopoietic stem cells in the hypoxic niche. Cell Stem Cell 9,
298–310 (2011). Acknowledgements
17. Ito, K. et al. Self-renewal of a purified Tie2+ hematopoietic stem We thank G. Coukos (LICR, University of Lausanne (UNIL), Centre
cell population relies on mitochondrial clearance. Science 354, Hospitalier Universitaire Vaudois) for critical feedback and F. Derouet
1156–1160 (2016). (UNIL) for animal care. Flow cytometry analysis and sorting was
18. Vannini, N. et al. The NAD-booster nicotinamide riboside potently performed at the Flow Cytometry Facility at UNIL with the help of
stimulates hematopoiesis through increased mitochondrial R. Bedel, F. Sala de Oyanguren, K. Blackney and A. Wilson. We thank
clearance. Cell Stem Cell 24, 405–418 (2019). Nestlé Health Science for their suggestions about the study.

Nature Aging | Volume 3 | September 2023 | 1057–1066 1065


Letter https://doi.org/10.1038/s43587-023-00473-3

M.G. was partially funded by a collaborative Jebsen Foundation of patent no. 19152437.0 of which N.V. and M.G. are inventors. The
grant (EPFL/UNIL) to N.V. and O.N. The N.V. laboratory is supported remaining authors declare no competing interests.
by the Swiss Cancer Research Foundation (KFS-4993-02-2020-R)
and San Salvatore Foundation. The J.A. laboratory is supported by Additional information
grants from EPFL, the European Research Council (ERC-AdG-787702), Extended data is available for this paper at
the Swiss National Science Foundation (SNSF) (31003A_179435) https://doi.org/10.1038/s43587-023-00473-3.
and a Global Research Laboratory grant of the National Research
Foundation of Korea (2017K1A1A2013124). F.S. is supported by the Supplementary information The online version
SNSF (323530_183986). The O.N. laboratory was supported by SNSF contains supplementary material available at
grant nos. PP00P3_183725 and CRSII5-186271. P.-C.H. is funded in part https://doi.org/10.1038/s43587-023-00473-3.
by the European Research Council Staring Grant (802773-MitoGuide),
an SNSF project grant (no. 31003A_182470), the Cancer Research Correspondence and requests for materials should be addressed to
Institute (Lloyd J. Old STAR Award) and LICR. W.H. is funded in part by Nicola Vannini.
a grant from the SNSF (no. 310030_200898). All the funders had no
role in study design, data collection and analysis, decision to publish Peer review information Nature Aging thanks Saghi Ghaffari and the
or preparation of the manuscript. other, anonymous, reviewer(s) for their contribution to the peer review
of this work.
Author contributions
M.G., Y-H.C. and N.V. conceived the ideas, designed the experiments Reprints and permissions information is available at
and analyzed the results. M.G., Y-H.C., N.V. and F.S. performed the HSC www.nature.com/reprints.
experiments. M.C., H.C.H. and W.H. performed and interpreted the
infection model experiments. P.G. evaluated the impact of UA dietary Publisher’s note Springer Nature remains neutral with regard
supplementation on secondary lymphoid tissues and performed and to jurisdictional claims in published maps and institutional
interpreted the cell respiration analyses. J.A. and O.N. conceived affiliations.
the ideas and provided key reagents and samples. S.C. provided the
human samples for HSC purification. O.N. and C.B. performed the Springer Nature or its licensor (e.g. a society or other partner) holds
human HSC experiments. F.F., Y-R.Y., H.G. and P.-C.H. provided the exclusive rights to this article under a publishing agreement with
animal model and helped with data interpretation. N.V. wrote the the author(s) or other rightsholder(s); author self-archiving of the
manuscript. All authors edited and reviewed the final manuscript. accepted manuscript version of this article is solely governed by the
terms of such publishing agreement and applicable law.
Competing interests
J.A. is a scientific advisor to Amazentis, a company that develops © The Author(s), under exclusive licence to Springer Nature America,
UA as a therapeutic agent. Some elements of this work are part Inc. 2023

Nature Aging | Volume 3 | September 2023 | 1057–1066 1066


Letter https://doi.org/10.1038/s43587-023-00473-3

Extended Data Fig. 1 | Old HSCs have higher mitochondria content. Mitochondrial DNA (mtDNA) quantification by QPCR expressed as fold change compared to
young HSCs. Young values are normalized on their mean value (P-value = 0.0571) (n = 4; values are mean ± s.e.m.; two-tailed Mann-Whitney test; * P ≤ 0.05, ** P ≤ 0.01,
*** P ≤ 0.001).

Nature Aging
Letter https://doi.org/10.1038/s43587-023-00473-3

Extended Data Fig. 2 | Donor chimerism analysis in the bone marrow. of primary transplant. d, Analysis of donor derived HSCs at the endpoint of
a, Lymphoid vs myeloid compartments of primary transplants in peripheral secondary transplant (n = 10; values are mean ± s.e.m.; two-tailed Student’s t test;
blood at the indicated timepoints. b, Analysis of donor derived contribution * P ≤ 0.05, ** P ≤ 0.01, *** P ≤ 0.001, **** P ≤ 0.0001). e, HSCs isolated from bone
for lymphoid and myeloid lineages in bone marrow at the endpoint of primary marrow of young mice were cultured for three days in the presence of 20μM UA
transplant (Lymphoid: young ctrl vs old ctrl P-value = 0.0003; old ctrl vs old and transplanted in lethally irradiated primary recipient together with bone
UA P-value = 0.0011; myeloid: young ctrl vs old ctrl P-value = 0.0002; old ctrl vs marrow derived from competitor mice. Blood donor chimerism analyses of
old UA P-value = 0.0006;). c, Analysis of donor derived HSCs at the endpoint primary transplant at indicated timepoints (n = 10; values are mean ± s.e.m.).

Nature Aging
Letter https://doi.org/10.1038/s43587-023-00473-3

Extended Data Fig. 3 | UA improves Colony forming capacity (CFU assay) of (n = 14; values are mean ± s.e.m.; two-tailed Student’s t test; * P ≤ 0.05, ** P ≤ 0.01,
old human HSCs. a, 1000 CD34+CD38−CD45RA- cells derived from the bone *** P ≤ 0.001). c, Assessment of long-term hematopoietic capacity by secondary
marrow of anonymous old adults (58-77 years old) were treated for 3 days with CFUs analysis (P-value = 0.0042) (n = 8; values are mean ± s.e.m.; two-tailed
20μM UA, and colony formations was measured at 15 days post seeding and Student’s t test; * P ≤ 0.05, ** P ≤ 0.01, *** P ≤ 0.001).
replated for secondary CFUs. b, analysis of primary CFUs (P-value < 0.0001)

Nature Aging
Letter https://doi.org/10.1038/s43587-023-00473-3

Extended Data Fig. 4 | Peripheral blood analyses of UA fed mice. Peripheral counter and normalized to day 0 (WBC: white blood cell; Lym: Lymphocytes; Gra:
blood cell analyses of old mice fed with ctrl (old ctrl) or UA enriched diet (old Granulocytes; Mon: Monocytes; RBC: red blood cells; HGB: hemoglobin) (n = 8;
UA). Analyses were performed at the indicated time points with blood cell values are mean ± s.e.m.;).

Nature Aging
Letter https://doi.org/10.1038/s43587-023-00473-3

Extended Data Fig. 5 | Hematopoietic stem and progenitor analyses of old KLS–FcR–CD34–). b, Analyses of lymphoid vs myeloid compartments of primary
mice supplemented with UA. a, Gating strategy to identify hematopoietic transplants at the indicated timepoints (n = 10; values are mean ± s.e.m). c, Donor
stem and progenitor populations from the BM for endpoint analysis. HSCs: chimerism analysis of secondary recipient mice in peripheral blood (Total:
Lineage−cKit+Sca1+ (LKS) CD150+CD48−; Multipotent progenitors (MPPs): 4 weeks P-value = 0.0013; 8 weeks P-value < 0.0001; 12 weeks P-value = 0.0003;
LKS CD150−, Common lymphoid progenitor (CLP): CD127+ cKitlow Sca1low; Lymphoid: 4 weeks P-value < 0.0001; 8 weeks P-value < 0.0001; 12 weeks
Committed progenitors (cKit+) include common myeloid progenitors (CMPs: P-value = 0.0002; Myeloid: 4 weeks P-value = 0.0089; 8 weeks P-value = 0.0049;
Lineage−cKit+Sca1–(KLS–) FcRlowCD34+), granulocyte–macrophage progenitors 12 weeks P-value = 0.0153) (n = 5; values are mean ± s.e.m.; two-tailed Student’s
(GMPs: KLS-FcRlowCD34 + ), and megakaryocyte-erythroid progenitors (MEPs: t test; * P ≤ 0.05, ** P ≤ 0.01, *** P ≤ 0.001).

Nature Aging
Letter https://doi.org/10.1038/s43587-023-00473-3

Extended Data Fig. 6 | Expression of inhibitory receptors markers and P-value < 0.0001; young ctrl vs old UA P-value = 0.0125; old ctrl vs old UA
cytokines by virus-specific CD8 + T cells. a, Expression of IFNγ (gp33 + : young P-value = 0.0146 np396+:young ctrl vs old ctrl P-value < 0.0001; young ctrl vs old
ctrl vs old UA P-value = 0.0112; np396+: young ctrl vs old UA P-value = 0.0034; UA P-value = 0.0025; old ctrl vs old UA P-value = 0.0159)and Lag3 (gp33 + :young
old ctrl vs old UA P-value = 0.0160), TNFα (gp33 + :young ctrl vs old ctrl ctrl vs old ctrl P-value < 0.0001; young ctrl vs old UA P-value = 0.0007; old ctrl
P-value < 0.0001; young ctrl vs old UA P-value = 0.0003; np396+:young ctrl vs old UA P-value = 0.0173; np396+:young ctrl vs old ctrl P-value < 0.0001; young
vs old ctrl P-value < 0.0001; young ctrl vs old UA P-value < 0.0001) and IL-2 ctrl vs old UA P-value < 0.0001; old ctrl vs old UA P-value = 0.0301) by gp33+ and
by CD8+ T cells in response to restimulation with gp33 or np396 peptides. np396 CD44+ CD8+ T cells (n = 10; values are mean ± s.e.m.; two-way ANOVA;
b, Expression of co- inhibitory receptors PD-1 (gp33 + :young ctrl vs old ctrl * P ≤ 0.05, ** P ≤ 0.01, *** P ≤ 0.001, **** P ≤ 0.0001).

Nature Aging
Letter https://doi.org/10.1038/s43587-023-00473-3

Extended Data Fig. 7 | Viral response of CD8 + T cells derived from old np396+:young ctrl vs old ctrl P-value = 0.0298). b, Proportion of activated KLRG1+
UA treated HSCs. a, Quantification of donor derived (CD45.1+) virus specific within donor derived virus specific CD8+ T. (n = 5; values are mean ± s.e.m.;
(gp33+ or np396+) CD8+ T cells (gp33 + :young ctrl vs old ctrl P-value = 0.0407; two-way ANOVA; * P ≤ 0.05, ** P ≤ 0.01, *** P ≤ 0.001, **** P ≤ 0.0001).

Nature Aging
Letter https://doi.org/10.1038/s43587-023-00473-3

Extended Data Fig. 8 | UA in vitro treatment modulates T cell function and Green) in CD8+ and CD4 + T cells measured as mean fluorescence intensity (MFI).
mitochondrial profiles. a, Splenocytes derived from old mice were activated c, Analyses of the proportion of cells INFγ+TNFα+ upon restimulation with CD3/
at day0 and restimulated at day5 with CD3/CD28 beads cultured with 5 μM UA. CD28 beads at day 5 (n = 4; values are mean ± s.e.m.; two-tailed paired Student’s
b, Analyses of mitochondrial activity (TMRM) and mass (MTG, mitotracker t test; * P ≤ 0.05, ** P ≤ 0.01, *** P ≤ 0.001).

Nature Aging
Letter https://doi.org/10.1038/s43587-023-00473-3

Extended Data Fig. 9 | UA modulates mitochondrial recycling in HSCs. Mx1-PARK2 mice (CD45.2) were transplanted in lethally irradiated recipient mice
a, Analysis of mitophagy induction in HSCs purified from mito-QC mice (CD45.1/2) together with total bone marrow competitor cells (CD45.1).d, Blood
measured as MFI ratio between GFP and mCherry signals(P-value = 0.0037). donor chimerism of primary transplant at the indicated timepoint. e, Lymphoid
(n = 4; two tailed paired Student’s t test; * P ≤ 0.05, ** P ≤ 0.01, *** P ≤ 0.001). vs myeloid compartments of primary transplants at the indicated timepoints
b, Expression of CD150 measured by mean fluorescence intensity (MFI) in young (Ctrl vs PARK2-/- P-value = 0.0002; Ctrl vs PARK2-/- UA P-value = 0.0006) (n = 10;
and old HSCs (P-value = 0.0009)(n = 4; values are mean ± s.e.m.; two-tailed values are mean ± s.e.m.; two-way ANOVA; * P ≤ 0.05, ** P ≤ 0.01, *** P ≤ 0.001).
Student’s t test; * P ≤ 0.05, ** P ≤ 0.01, *** P ≤ 0.001). c, HSCs derived from

Nature Aging

You might also like