ArticlePDF Available

The mitophagy activator urolithin A is safe and induces a molecular signature of improved mitochondrial and cellular health in humans

Authors:

Abstract and Figures

Urolithin A (UA) is a natural dietary, microflora-derived metabolite shown to stimulate mitophagy and improve muscle health in old animals and in preclinical models of aging¹. Here, we report the results of a first-in-human clinical trial in which we administered UA, either as a single dose or as multiple doses over a 4-week period, to healthy, sedentary elderly individuals. We show that UA has a favourable safety profile (primary outcome). UA was bioavailable in plasma at all doses tested, and 4 weeks of treatment with UA at doses of 500 mg and 1,000 mg modulated plasma acylcarnitines and skeletal muscle mitochondrial gene expression in elderly individuals (secondary outcomes). These observed effects on mitochondrial biomarkers show that UA induces a molecular signature of improved mitochondrial and cellular health following regular oral consumption in humans. © 2019, The Author(s), under exclusive licence to Springer Nature Limited.
| uA impacts markers of mitochondrial function after 28 d of treatment. a, Comparison of mRNA levels of autophagy/mitophagy, mitochondrial biogenesis and fatty acid oxidation markers as measured by qPCR in vastus lateralis of subjects who received placebo, UA 500 or 1,000 mg for 28 d (n = 9 biologically independent samples). Results are expressed as a ratio over the placebo group for better readability. b, Change in mitochondrial abundance as measured by qPCR in vastus lateralis skeletal muscle of subjects who received placebo, UA 500 or 1,000 mg for 28 d (n = 9 biologically independent samples). All data are means ± s.e.m. # 0.05 < P < 0.15; *P < 0.05; **P < 0.01; ***P < 0.001 after a one-way ANOVA followed by Dunnett's post-hoc test (a,b). c, Graphical representation of GSEA results. Bars represent the normalized enrichment score for the mitochondrial gene sets that are significantly upregulated with FDR < 0.1 in the vastus lateralis skeletal muscle of subjects following UA treatment at 500 mg and 1,000 mg for 28 d compared with placebo. FDR is the estimated probability that a gene set with a given enrichment score (normalized for gene set size) represents a false positive finding. The first three gene sets are upregulated by both UA 500 mg and 1,000 mg, and the others are upregulated by 1,000 mg with the 500 mg being not significant (NS; FDR > 0.1). Mb: membrane. d,e, Genes within the GO_MITOCHONDRION gene set that are upregulated (see Methods) in vastus lateralis skeletal muscle of subjects following UA treatment at 500 or 1,000 mg for 28 d compared with placebo (c, n = 9) and in the vastus lateralis skeletal muscle of pre-frail sedentary or active healthy elderly individuals (NCT02472340) (d, n = 11). Heat map represents change in expression over time (day 28 versus pre-dose) (c) or as difference between active healthy and sedentary pre-frail (d) as Z scores. Related to Tables 1 and 2.
… 
UA phase 1 study design, pharmacokinetic analysis and impact on plasma acylcarnitines in elderly individuals a, Simplified schema of the clinical study design. The dose escalation was designed to progress from the lowest to the highest UA dose investigated in both parts of the study. Dose escalation to the next higher UA dose was always twofold higher than the previous dose. During part A, UA was administered as a single ascending dose, ranging from 250 to 2,000 mg on fasting, and at 500 and 1,000 mg in a fed state with a high-protein yogurt food matrix. Muscle biopsies were collected at pre-dose and 8 h after oral administration of UA only in the 2,000 mg group. During part B, UA was administered once daily in the morning on fasting for 28 d. Plasma and muscle biopsies were collected at pre-dose and at day 28 for biomarker activity measurements (see arrows). The corresponding CONSORT diagram is represented in Supplementary Fig. 1. b, Dose-dependent increase in plasma UA, UA-glucuronide and UA-sulfate maximum concentrations and exposure during the 96-h sampling period following its administration on the last day of the 28-d treatment period for 250, 500 and 1,000 mg doses (n = 9 biologically independent samples). Data represent mean ± s.e.m. c, Change in plasma levels of acylcarnitines compared to baseline (day 28 (D28) versus pre-dose (D –1)) (n = 9 biologically independent samples). Data represent geometric mean ± 95% confidence interval. #0.05 < P < 0.15; *P < 0.05; **P < 0.01 after a two-way, repeated-measures ANOVA. Related to Supplementary Figs. 1 and 2 and Supplementary Tables 1–5.
… 
Content may be subject to copyright.
Letters
https://doi.org/10.1038/s42255-019-0073-4
1Amazentis SA, EPFL Innovation Park, Bâtiment C, Lausanne, Switzerland. 2Laboratory for Integrative and Systems Physiology, Ecole Polytechnique Fédérale
de Lausanne, Lausanne, Switzerland. 3Vital-IT Group, SIB Swiss Institute of Bioinformatics, Quartier Sorge, Bâtiment Génopode, Lausanne, Switzerland.
4Neurodegenerative Diseases Laboratory, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland. 5Present address: Department of Molecular
Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea. 6These authors jointly supervised this work: Anurag Singh, Chris
Rinsch. *e-mail: contact@amazentis.com
Urolithin A (UA) is a natural dietary, microflora-derived
metabolite shown to stimulate mitophagy and improve mus-
cle health in old animals and in preclinical models of aging1.
Here, we report the results of a first-in-human clinical trial in
which we administered UA, either as a single dose or as mul-
tiple doses over a 4-week period, to healthy, sedentary elderly
individuals. We show that UA has a favourable safety profile
(primary outcome). UA was bioavailable in plasma at all doses
tested, and 4 weeks of treatment with UA at doses of 500 mg
and 1,000 mg modulated plasma acylcarnitines and skeletal
muscle mitochondrial gene expression in elderly individuals
(secondary outcomes). These observed effects on mitochon-
drial biomarkers show that UA induces a molecular signature
of improved mitochondrial and cellular health following regu-
lar oral consumption in humans.
During aging, there is progressive decline in the cell’s capacity to
eliminate its dysfunctional elements by autophagy2. Accumulating
evidence has highlighted the decrease in the specific autophagy,
or recycling, of dysfunctional mitochondria, known as mitophagy,
in aging skeletal muscle3. This can result in poor mitochondrial
function in the skeletal muscle, and has been closely linked to
slow walking speed and poor muscle strength in elderly individu-
als4,5. Consequently, improving mitochondrial function in elderly
people by restoring levels of mitophagy represents a promising
approach to halt or delay the development of age-related decline in
muscle health.
UA is a first-in-class natural food metabolite that stimulates
mitophagy and prevents the accumulation of dysfunctional mito-
chondria with age, thereby maintaining mitochondrial biogenesis
and respiratory capacity in cells, and, in the nematode Caenorhabditis
elegans, improving mobility and extending lifespan1. In rodents,
UA improves endurance capacity in young rats and in old mice
either fed a healthy diet or placed under conditions of metabolic
challenge1. Recently, UA was shown to have a favourable safety
profile following a battery of standardized toxicological tests,
including subchronic exposure for 90 d in rodent models6,
and received a favourable review by the US Food and Drug
Administration under the agency’s generally recognized as safe
(GRAS) notification program7.
In this report, we detail the outcome of a first-in-human, ran-
domized, double-blind, placebo-controlled clinical study with UA
in healthy, sedentary elderly individuals, and describe its safety,
bioavailability and beneficial impact on key biomarkers of mito-
chondrial health (NCT02655393). Physiological endpoints were
not evaluated as part of this study, as the 4-week intervention was
considered too short in comparison to the extended protocols (min-
imum 3 months) deemed necessary to improve muscle strength or
physical performance parameters in elderly individuals8.
This phase 1 study was a two-part study, with a single ascend-
ing dose (part A) followed by a multiple ascending dose (part B).
As the first objective of the study was safety assessment, the dose
escalation was designed to progress from the lowest to the highest
UA dose investigated in both parts of the study. Dose escalation to
the next higher UA dose was always twofold higher than the previ-
ous dose (see Methods and Supplementary Table 1 for the decision
tree and stopping rule criteria to advance to the next higher UA
dose). During part A of the study, three cohorts of eight subjects
each (24 subjects) received either placebo or UA in a two-period
design separated by a minimum 3-week wash-out period and at
single ascending doses of 250, 500, 1,000 or 2,000 mg, either in soft
gels or admixed with food (Fig. 1a, also the CONSORT diagram
in Supplementary Fig. 1). In part B of the study, three cohorts of
12 elderly subjects were given either placebo or UA at 250, 500 or
1,000 mg once daily in soft gels for 28 d (Fig. 1a and Supplementary
Fig. 1). The lowest dose of 250 mg was chosen on the basis
of preclinical studies, where the equivalent daily dosing of 50 mg per
kg (mpk) of body weight in mice demonstrated efficacy on mito-
chondrial and muscle function after a 6-week oral intervention1.
Clinical study treatment groups were evenly matched for age, sex
and body mass index, and all of the subjects were sedentary at the
time of inclusion in the study (Supplementary Tables 2 and 3). All
enrolled subjects completed the study, there were no major devia-
tions in the clinical protocol or in product intake, and no subjects
were excluded in the final analysis for the main study endpoints
(Supplementary Fig. 1).
As the study was a single and multiple dose escalation phase 1
study, designed according to guidelines and recommendations for
first-in-human studies9 and following standard dose escalation safety
trial design10,11, it was powered to meet the primary outcome of safety
and tolerability of UA in elderly humans to provide sufficient infor-
mation on human safety and pharmacokinetic profile and to allow
dose selection for future phase 2 efficacy trials (see also Methods).
The mitophagy activator urolithin A is safe
and induces a molecular signature of improved
mitochondrial and cellular health in humans
Pénélope A. Andreux1, William Blanco-Bose1, Dongryeol Ryu 1,2,5, Frédéric Burdet3, Mark Ibberson3,
Patrick Aebischer4, Johan Auwerx2, Anurag Singh1,6 and Chris Rinsch 1,6*
NATURE METABOLISM | VOL 1 | JUNE 2019 | 595–603 | www.nature.com/natmetab 595
Letters Nature MetabolisM
In each part of the study, subjects underwent physical examinations
and electrocardiogram (ECG) evaluations and were monitored for
adverse events. A battery of laboratory safety tests (serum biochem-
istry, haematology and urinalysis) were conducted before and after
dosing. The primary outcome was successfully met, and no serious
adverse events and no product-related non-serious adverse events
were reported during both part A and part B of the study. All other
non-serious adverse events were of mild to moderate intensity and
resolved during the course of the study (Supplementary Tables 4 and
5). No clinically relevant abnormal laboratory test values from the
study baseline were observed for any of the biochemistry tests assess-
ing liver and kidney function, or for any of the haematology and uri-
nalysis tests at any of the doses investigated during the course of the
study. No abnormal and clinically notable findings were observed
b
c
a
Part A : single ascending dose
Part B : multiple ascending dose
Period 1
Cohort 1A (week 1) Cohort 2A (week 2)
Period 2
Placebo (n = 2)
250 mg UA (n = 6)
Placebo (n = 2)
500 mg UA (n = 6)
Placebo (n = 2)
1,000 mg UA (n = 6)
Placebo (n = 2)
2,000 mg UA (n = 6)
°muscle biopsy
Placebo (n = 2)
500 mg UA
in food (n = 6) Placebo (n = 2)
1,000 mg UA
in food (n = 6)
3-week
washout
period
Placebo (n = 3)
250 mg UA (n = 9)
Placebo (n = 3)
500 mg UA (n = 9)
Placebo (n = 3)
1,000 mg UA (n = 9)
Muscle biopsy
Plasma
28 days
****
#
** **
****
##
UA sulfate (nM)
1,000 mg UA
250 mg UA
500 mg UA
0.0
0.5
1.0
1.5
2.0
2
3
4
5Placebo
0.0
0.5
1.0
1.5
2.0
UA 500 mg UA 1,000 mg
C6-DC
C8
C8-DC
C10
C10:1
C12
C14
C14:1
C20
C20:1
C22
C22:1
C6-DC
C8
C8-DC
C10
C10:1
C12
C14
C14:1
C20
C20:1
C22
C22:1
C6-DC
C8
C8-DC
C10
C10:1
C12
C14
C14:1
C20
C20:1
C22
C22:1
Fold change
(D28 over D –1)
Fold change
(D28 over D –1)
0.0
0.5
1.0
1.5
2.0
Fold change
(D28 over D –1)
##
*
#
Length of the carbon chain (acyl group) bound to carnitine
0
2
4
6
8
10
0
1,000
2,000
3,000
0
200
400
600
Time after oral UA administration (h)
Time after oral UA administration
(h)
12 18 24 48 72 96
12 18 24 48 72 96
06 06
Time after oral UA administration (h)
12 18 24 48 72 96
06
UA glucuronide (nM)
UA (nM)
Cohort 3A (week 3) Cohort 1A (week 7) Cohort 2A (week 8) Cohort 3A (week 9)
Cohort 1b (week 1-4) Cohort 2B (week 6-9) Cohort 3B (week 10-13)
28 days
28 days
Dose escalation
Dose escalation
Fig. 1 | UA phase 1 study design, pharmacokinetic analysis and impact on plasma acylcarnitines in elderly individuals. a, Simplified schema of the
clinical study design. The dose escalation was designed to progress from the lowest to the highest UA dose investigated in both parts of the study. Dose
escalation to the next higher UA dose was always twofold higher than the previous dose. During part A, UA was administered as a single ascending dose,
ranging from 250 to 2,000 mg on fasting, and at 500 and 1,000 mg in a fed state with a high-protein yogurt food matrix. Muscle biopsies were collected
at pre-dose and 8 h after oral administration of UA only in the 2,000 mg group. During part B, UA was administered once daily in the morning on fasting
for 28 d. Plasma and muscle biopsies were collected at pre-dose and at day 28 for biomarker activity measurements (see arrows). The corresponding
CONSORT diagram is represented in Supplementary Fig. 1. b, Dose-dependent increase in plasma UA, UA-glucuronide and UA-sulfate maximum
concentrations and exposure during the 96-h sampling period following its administration on the last day of the 28-d treatment period for 250, 500
and 1,000 mg doses (n= 9 biologically independent samples). Data represent mean ± s.e.m. c, Change in plasma levels of acylcarnitines compared to
baseline (day 28 (D28) versus pre-dose (D –1)) (n= 9 biologically independent samples). Data represent geometric mean ± 95% confidence interval.
#0.05 <P< 0.15; *P< 0.05; **P< 0.01 after a two-way, repeated-measures ANOVA. Related to Supplementary Figs. 1 and 2 and Supplementary Tables 1–5.
NATURE METABOLISM | VOL 1 | JUNE 2019 | 595–603 | www.nature.com/natmetab
596
Letters
Nature MetabolisM
for ECG findings for any subjects taking active intervention at any
of the doses during the course of the study. This follows the favour-
able safety profile observed in preclinical toxicology studies, which
showed no toxic effect at the highest doses tested (3,451 and 3,826
mpk in male and female rodents, respectively)6.
As another key outcome, the pharmacokinetic profile of UA was
characterized in humans. We have developed robust, precise and
validated methods to measure the individual concentrations of the
parent UA and its detectable metabolites in both plasma and in the
human skeletal muscle. Validation of the methods for the measure-
ments of UA aglycone and its glucuronide and sulfate metabolites
was performed following guidance on method validation12,13. The
levels of total UA (UA and its metabolites) observed in plasma
before dosing in the enrolled subjects ranged from undetectable
(69%) to low (17%), moderate (11%) and high (3%), demonstrat-
ing the substantial variability of UA exposure, probably due to dif-
ferences in diet and to potential variations in the composition of
the gut microflora14 (Supplementary Fig. 2a). UA was bioavailable
in plasma at all doses tested (250–2,000 mg) in the single ascend-
ing part A of the study, and there was no food effect when UA
was administered in a high-protein yogurt food matrix (data not
shown). Similarly, in part B of the study, where pharmacokinetics
were assessed following the last dosing on day 28, there was a dose-
dependent increase in maximum plasma concentrations (Cmax) and
total exposure (AUC) when escalating UA oral administration from
250 to 1,000 mg. UA was detectable in the plasma in the form of the
parent compound and its two major metabolites, UA-glucuronide
and UA-sulfate, with the levels of the conjugated UA metabolites in
plasma being higher than those of the parent UA (Fig. 1b). UA and
its conjugate metabolites (that is, UA-glucuronide and UA-sulfate)
exhibited similar kinetics, with concentrations peaking in plasma
at 6 h (Tmax) post-dosing (Fig. 1b). The half-life (t1/2) of the par-
ent UA compound and UA-glucuronide was in the range 17–22 h,
with UA-sulfate being slightly longer at 25–58 h. Both UA and its
bioavailable metabolites were eliminated from plasma circulation
72–96 h after the last intake (Fig. 1b). A dose-dependent increase in
total UA steady-state levels from 250 to 1,000 mg was also observed
during the 4-week UA administration (Supplementary Fig. 2b–d).
No accumulation of UA in plasma was seen when comparing the
single and multiple-dosing pharmacokinetics (data not shown).
Altogether, these pharmacokinetic data indicate a favourable bio-
available profile for UA. Following unblinding of the study, all sub-
jects receiving UA showed consistent levels of UA, highlighting
the high compliance of elderly subjects with the UA intervention
(Supplementary Fig. 2b–d). The presence of the conjugated forms
of UA, that is, UA-glucuronide and UA-sulfate, in human plasma
indicates that UA undergoes phase 2 conjugation metabolism in the
liver and active enterohepatic recirculation. UA was detectable in
the skeletal muscle tissue 8 h after a single oral dosing at 2,000 mg,
primarily in its parent state (Supplementary Fig. 2e). The skeletal
muscle tissue of only two out of the six participants showed trace
levels of UA-glucuronide, whereas UA-sulfate was not detected in
any of the subjects (data not shown).
To assess the impact of UA on mitochondria in humans, we tested
several surrogate molecular markers for mitochondrial health, both
in the plasma and in the skeletal muscle of the elderly participants.
While this study was powered for safety, the effects observed on
mitochondria-related biomarkers were significant and showed a
global impact on mitochondrial health following 28 d of UA oral
administration at doses of 500 and 1,000 mg. Dosing UA at 250 mg
showed no significant improvement in mitochondrial biomarkers
(data not shown). There is likely to be a dose–duration relationship
in the pharmacodynamics of UA in humans, with longer treatments
and larger sample sizes possibly being required to observe the ben-
efits of UA at lower doses. Therefore, the results included here focus
on UA doses of 500 mg and 1,000 mg.
In the plasma compartment, we observed a dose-dependent
decrease of acylcarnitine levels (C8 to C14 and >C20) in the 500
and 1,000 mg groups (Fig. 1c). Comparing relative plasma levels of
acylcarnitines in subjects before and after 28 d of dosing, no dif-
ferences were observed in the placebo and 250 mg groups, while
participants receiving 500 mg or 1,000 mg UA experienced a sig-
nificant reduction in acylcarnitine levels compared with baseline.
Acylcarnitines are the form in which fatty acids enter into the mito-
chondrion to undergo fatty acid oxidation. The impact of UA was
especially dramatic on the shorter chain acylcarnitines (C8, C10,
C12, C14:1) (Fig. 1c), that is, intermediates of the fatty acid oxi-
dation process, which signifies a better efficiency of the fatty acid
oxidation process15. It is also important to highlight that free carni-
tine levels were not changed (data not shown), which makes it more
likely that the decrease in acylcarnitines is a primary event and not a
secondary event in response to changes in free carnitine availability.
These systemic results in plasma support that UA administration
improves fatty acid oxidation in humans, one key function of the
mitochondrion, at the level of the whole body.
Available literature shows that plasma levels of acylcarnitines
are inversely correlated with mitochondrial function and/or exer-
cise levels of subjects. Elevated plasma acylcarnitines are used as
diagnostic biomarkers for mitochondrial diseases characterized by
a defect in fatty acid oxidation16 and they are also longitudinally
increased with poor metabolic health and with the aging process,
by a magnitude of about 1.5–2-fold over time17. On the other hand,
in middle-aged male subjects, a 10-week aerobic exercise regimen
known to stimulate mitochondrial function led to a decrease in
plasma acylcarnitine levels, similar to that observed with 4-week
UA intervention (a decrease in the range of 20–50%)18.
The direct impact of UA at the level of the skeletal muscle (vastus
lateralis) was evaluated by gene expression analysis, using a series
of genes related to autophagy/mitophagy, mitochondrial biogenesis
and fatty acid oxidation selected on the basis of previous preclini-
cal efficacy data1. A general pattern of dose-dependent upregulation
of gene expression in the human muscle, similar to that observed
previously in preclinical models, was seen after 28 d of UA treat-
ment at 500 and 1,000 mg, with some reaching statistical signifi-
cance (GABARAPL1, FABP3) (Fig. 2a). Mitochondrial abundance
was also evaluated by measuring the ratio of mitochondrial DNA
to nuclear DNA (mtDNA/nuDNA) by qualitative PCR (qPCR). The
mtDNA/nuDNA ratio tended to increase, although this did not
reach statistical significance (Fig. 2b).
To determine more broadly if mitochondrial gene expression was
altered, microarray analysis was performed on the messenger RNA
from the vastus lateralis skeletal muscle and analysed using gene-
set enrichment analysis (GSEA), to look for over-representation of
known pathways and gene functional categories. GSEA is designed
to detect subtle gene expression changes at the level of a biological
process or pathway19. Treatment with UA at 500 and 1,000 mg was
seen to upregulate several mitochondrial gene sets with a false dis-
covery rate (FDR) < 0.1, including the GO_MITOCHONDRION
gene set (Fig. 2c,d and Table 1). Consequently, this unbiased
approach indicates that 28-d administration of UA upregulates the
transcription of mitochondrial genes. Taken together, these data
further substantiate the results on gene expression and mtDNA
measured by qPCR and demonstrate that UA stimulates mitochon-
drial biogenesis in the skeletal muscle of humans. Similar observa-
tions have been made in other studies on the impact of different
exercise regimens and their related effects on the human muscle
transcriptome2022. In particular, 12-week, high-intensity aero-
bic interval training induced upregulation of both mitochondria
gene and protein expression in the skeletal muscle of young and
older subjects22.
We have also compared the UA-induced transcriptional signa-
ture in the skeletal muscle to the natural transcription signatures
NATURE METABOLISM | VOL 1 | JUNE 2019 | 595–603 | www.nature.com/natmetab 597
Letters Nature MetabolisM
observed in age-matched pre-frail (that is, elderly with low muscle
strength) and active aged-matched elderly individuals (non-inter-
ventional study (NIS); NCT02472340), investigated previously23.
As described, GSEA showed a clear downregulation of multiple
mitochondrial gene sets in the skeletal muscle of pre-frail subjects
compared with active subjects, with the top ten downregulated gene
sets being only related to mitochondria, highlighting a decline in
mitochondrial biogenesis in pre-frail muscle23. In total, there were
16 gene sets that were both downregulated in pre-frail versus active
subjects, and upregulated in subjects who received UA at 1,000 mg
for 28 d (Table 2). Of these, 13 were related to mitochondrion
organelle or function. The 63 genes that were the most induced
within the GO_MITOCHONDRION gene set (Fig. 2d), follow-
ing 28-d treatment with UA, were extracted from the published
NIS NCT02472340 dataset and plotted as a heat map (Fig. 2e).
This graphical representation of the GSEA results shows that the
molecular signature in skeletal muscle of the pre-frail individuals is
marked by a downregulation of mitochondrial gene expression. In
comparison, UA significantly upregulates the transcription of the
same mitochondrial gene set in skeletal muscle, providing encour-
aging evidence that UA may provide a benefit for age-related decline
in muscle mitochondrial health. This comparison is particularly rel-
evant, as the participants from phase 1 and the published NIS study
are matched for age and body mass index (BMI), and are all seden-
tary, except those in the active healthy elderly group.
The global impact on plasma acylcarnitines and the specific
effect on muscle transcriptomics in subjects receiving daily doses
of either 500 mg or 1,000 mg of UA revealed an improved systemic
Autophagy/mitophagy
Mitochondrial
biogenesis and dynamics
PIK3C3 ULK1 BECN1 SQSTM1 LC3B GABA-
RAPL1
PARK2
mRNA (ratio of placebo)
#
*
#
mRNA (ratio of placebo)
CPT1B FABP3
#
*
ATP2A1
MYOM2
COX6A2
DUSP26
KCNJ11
PLN
CKMT2
GOT1
UQCRC1
CISD1
RTN4IP1
NNT
PHYH
VDAC3
GBAS
POLDIP2
ALDH4A1
SDHA
LYRM2
ACSL4
PTCD3
IMMP1L
PTS
NDUFS1
MRPS9
BCS1L
BCO2
SFXN4
SLC25A38
AGPAT5
COQ2
TOMM40L
TARS2
GPT2
GATM
MECR
AKAP1
ATP5SL
VARS
PARK2
MLYCD
LIG3
CHCHD7
CECR5
NUDT2
SIRT5
GLRX5
PDHB
HADHB
NUDT19
MTRF1L
NDUFA3
NDUFA12
TRMT2B
DLD
MRPS17
MRPL27
ECSIT
MCEE
ATP5F1
UQCRHL
FASTKD2
ALDH1B1
Placebo UA 500 mg UA 1,000 mg
–4 4
Row z score
(day 28 – pre-dose) –4 4
Row z score
(active healthy –
sedentary pre-frail)
ATP2A1
MYOM2
COX6A2
DUSP26
KCNJ11
PLN
CKMT2
GOT1
UQCRC1
CISD1
RTN4IP1
NNT
PHYH
VDAC3
GBAS
POLDIP2
ALDH4A1
SDHA
LYRM2
ACSL4
PTCD3
IMMP1L
NDUFS1
MRPS9
BCS1L
BCO2
SFXN4
SLC25A3
8
AGPAT5
COQ2
TOMM40L
TARS2
GPT2
GATM
MECR
AKAP1
ATP5SL
VARS
PARK2
MLYCD
LIG3
CHCHD7
CECR5
NUDT2
SIRT5
GLRX5
PDHB
HADHB
NUDT19
MTRF1L
NDUFA3
NDUFA12
TRMT2B
DLD
MRPS17
MRPL27
ECSIT
MCEE
ATP5F1
UQCRHL
FASTKD2
ALDH1B1
PTS
Sedentary
pre-frail
Active
healthy
de
Fatty acid oxidation
PPPARA ESRRA PGC1A MFN2
#
a
Placebo UA 500 mg UA 1,000 mg
b
mtDNA/nuDNA ratio
(ratio of placebo)
Mitochondrial part
Mitochondrial matrix
Mitochondrion
Mitochondrial envelope
Mitochondrion
organization
Mitochondrial mb part
Mitochondrial protein
complex
Inner mitochondrial mb
protein complex
c
Normalized enrichment
score
Placebo
UA 500 mg
UA 1,000 mg
UA 500 mg UA 1,000 mg
0123
NS
NS
NS
NS
NS
0
2
4
10
15
0
2
4
6
8
10
15
25
0
2
4
6
8
12
22
0
2
4
6
8
20
60
100
mRNA (ratio of placebo)
0
1
2
3
4
Fig. 2 | UA impacts markers of mitochondrial function after 28 d of treatment. a, Comparison of mRNA levels of autophagy/mitophagy, mitochondrial
biogenesis and fatty acid oxidation markers as measured by qPCR in vastus lateralis of subjects who received placebo, UA 500 or 1,000 mg for 28 d (n= 9
biologically independent samples). Results are expressed as a ratio over the placebo group for better readability. b, Change in mitochondrial abundance
as measured by qPCR in vastus lateralis skeletal muscle of subjects who received placebo, UA 500 or 1,000 mg for 28 d (n= 9 biologically independent
samples). All data are means ± s.e.m. #0.05 <P< 0.15; *P< 0.05; **P< 0.01; ***P< 0.001 after a one-way ANOVA followed by Dunnett’s post-hoc test
(a,b). c, Graphical representation of GSEA results. Bars represent the normalized enrichment score for the mitochondrial gene sets that are significantly
upregulated with FDR < 0.1 in the vastus lateralis skeletal muscle of subjects following UA treatment at 500 mg and 1,000 mg for 28 d compared with
placebo. FDR is the estimated probability that a gene set with a given enrichment score (normalized for gene set size) represents a false positive finding.
The first three gene sets are upregulated by both UA 500 mg and 1,000 mg, and the others are upregulated by 1,000 mg with the 500 mg being not
significant (NS; FDR > 0.1). Mb: membrane. d,e, Genes within the GO_MITOCHONDRION gene set that are upregulated (see Methods) in vastus lateralis
skeletal muscle of subjects following UA treatment at 500 or 1,000 mg for 28 d compared with placebo (c, n= 9) and in the vastus lateralis skeletal
muscle of pre-frail sedentary or active healthy elderly individuals (NCT02472340) (d, n= 11). Heat map represents change in expression over time (day 28
versus pre-dose) (c) or as difference between active healthy and sedentary pre-frail (d) as Z scores. Related to Tables 1 and 2.
NATURE METABOLISM | VOL 1 | JUNE 2019 | 595–603 | www.nature.com/natmetab
598
Letters
Nature MetabolisM
mitochondrial health, an enhanced fatty acid oxidation rate and a
gene expression profile consistent with mitochondrial biogenesis
in the muscle. Future clinical trials may allow a more in-depth
characterization of UA’s impact on mitochondrial function in tis-
sues. Given the wide range of health benefits of UA that have been
recently reported in the brain24,25 and intestine26, this evidence sug-
gests that UA will have benefits on mitochondrial health in tissues
other than skeletal muscle.
The present study reveals that UA induces a molecular signature
response, in both the plasma and skeletal muscle of humans, resem-
bling that observed as a consequence of a regular exercise regimen.
It is important to highlight that our earlier work revealed that the
stimulation of mitophagy by UA led to an induction of mitochondrial
biogenesis and an enhancement of mitochondrial function, resulting
in improved aerobic endurance and higher muscle strength in treated
rodents1. In humans, endurance exercise is well known to trigger
mitochondrial biogenesis27 and fatty acid oxidation in the skeletal
muscle28 to optimize efficient production of ATP by skeletal muscle
cells under aerobic conditions. It has also been shown that exercise is
a natural means of triggering mitophagy29,30, making it particularly
important to maintain an active lifestyle during aging, as it ultimately
results in improved mitochondrial function in the muscle4,5.
The research community has shown considerable interest in
the therapeutic potential of stimulating the mitophagy pathway,
as it may be the key to treating many of the conditions and dis-
eases associated with a decline in mitochondrial function linked to
aging. Aside from the present study, the most advanced published
developments in stimulating mitophagy have been at the preclini-
cal stage, where the approach of targeting deubiquitylating enzymes
appears to be promising31.
Table 1 | List of mitochondrial gene sets that are enriched in human skeletal muscle after 28 d of UA administration
Gene sets Number of genes
per gene set 500mg change over time
normalized over placebo 1,000mg change over time
normalized over placebo
NES FDR NES FDR
GO_MITOCHONDRIAL_ENVELOPE 691 NA NA 1.762903 0.0387385
GO_MITOCHONDRIAL_MATRIX 412 2.525320 0.0020414 1.987229 0.0069189
GO_MITOCHONDRIAL_PART 953 2.078387 0.0751073 2.090689 0.0021444
GO_MITOCHONDRION 1,633 2.192505 0.0432333 1.939153 0.0102209
GO_MITOCHONDRION_ORGANIZATION 594 NA NA 1.757141 0.0398731
GO_INNER_MITOCHONDRIAL_MEMBRANE_PROTEIN_COMPLEX 106 NA NA 1.639517 0.0855748
GO_MITOCHONDRIAL_MEMBRANE_PART 173 NA NA 1.697246 0.0621488
GO_MITOCHONDRIAL_PROTEIN_COMPLEX 136 NA NA 1.668623 0.0728421
NES, normalized enrichment score; FDR, the estimated probability that a gene set with a given enrichment score (normalized for gene set size) represents a false positive finding; NA, not available (no
significant enrichment).
Table 2 | List of gene sets that are enriched in human skeletal muscle after 28 d of UA administration at 1,000mg and downregulated
in human skeletal muscle of pre-frail compared to active elderly subjects (NIS; NCT02472340)
Gene sets Number of genes
per gene set Downregulated in pre-frail
compared to active elderly
(NIS; NCT02472340)
1,000mg change over
time normalized over
placebo
NES FDR NES FDR
GO_GENERATION_OF_PRECURSOR_METABOLITES_AND_ENERGY 292 3.0632 <1 × 1062.144 0.0011
GO_MITOCHONDRIAL_PART 953 3.4394 <1 × 1062.0907 0.0021
GO_MITOCHONDRIAL_MATRIX 412 3.4099 <1 × 1061.9872 0.0069
GO_ENERGY_DERIVATION_BY_OXIDATION_OF_ORGANIC_COMPOUNDS 217 3.2779 <1 × 1061.9484 0.0096
GO_MITOCHONDRION 1,633 3.0257 <1 × 1061.9392 0.0102
GO_ORGANELLE_INNER_MEMBRANE 525 3.5479 <1 × 1061.9058 0.0136
GO_MITOCHONDRIAL_ENVELOPE 691 3.3754 <1 × 1061.7629 0.0387
GO_MITOCHONDRION_ORGANIZATION 594 2.6828 <1 × 1061.7571 0.0399
GO_CELLULAR_PROTEIN_COMPLEX_DISASSEMBLY 122 2.7963 <1 × 1061.7302 0.0494
GO_MITOCHONDRIAL_MEMBRANE_PART 173 3.6959 <1 × 1061.6972 0.0621
GO_MITOCHONDRIAL_PROTEIN_COMPLEX 136 3.6668 <1 × 1061.6686 0.0728
GO_INNER_MITOCHONDRIAL_MEMBRANE_PROTEIN_COMPLEX 106 3.55 <1 × 1061.6395 0.0856
GO_CELLULAR_RESPIRATION 143 3.81365 <1 × 1061.6263 0.0924
GO_LYASE_ACTIVITY 179 1.6667 0.0962 1.7771 0.0352
GO_ALPHA_AMINO_ACID_CATABOLIC_PROCESS 95 1.8396 0.0357 1.6945 0.0621
GO_ORGANIC_ACID_METABOLIC_PROCESS 953 1.847 0.034 1.6783 0.0686
In total, there were 16 gene sets that are both downregulated in skeletal muscle of pre-frail versus active elderly, and upregulated in subjects who received UA at 1,000 mg for 28 d. Of these 16 gene sets,
13 are clearly linked to mitochondrion organelle and/or function (shown first in this table). The three other gene sets (last ones in this table) are related to more specific enzymatic (GO_LYASE_ACTIVITY)
and metabolic processes (GO_ALPHA_AMINO_ACID_CATABOLIC_PROCESS and GO_ORGANIC_ACID_METABOLIC_PROCESS).
NATURE METABOLISM | VOL 1 | JUNE 2019 | 595–603 | www.nature.com/natmetab 599
Letters Nature MetabolisM
This report of a clinical investigation of an activator of mitoph-
agy demonstrates the successful translation of the benefits of the
natural food metabolite UA to humans, particularly the combina-
tion of its positive biological effects on mitochondrial health and,
importantly, its favourable safety and bioavailability profile. These
promising findings support an approach of dietary supplementa-
tion with UA as a nutritional intervention to assist in managing the
declining mitochondrial function that accompanies aging and to
promote healthy muscle function throughout life.
Methods
Trial design. e phase 1 clinical trial was designed to investigate the safety of
the food ingredient UA in elderly adults, as well as its impact on biomarkers of
mitochondrial health. e trial was conducted as a single centre (in a phase 1
clinical trial unit), randomized, double-blind, placebo-controlled study in 60
healthy male and female elderly volunteers. e study was divided into two parts
(Supplementary Fig. 1): part A involved administering a single ascending dose
(250 mg, 500 mg, 1,000 mg and 2,000 mg of UA delivered orally) to 24 healthy
elderly male and female volunteers, where each participant was randomized into
two subsequent doses in three cohorts. To minimize risk, administration of the
investigational product in each dose group of the study was done sequentially
within each cohort (maximum of four subjects each day, respectively). For the
study, a 3:1 ratio was followed in part A, with each cohort receiving six active
doses and two placebo doses. Four participants (3 active, 1 placebo) received the
specied dose at the start of the week and, in the absence of adverse events, the
remaining four participants (3 active, 1 placebo) in the cohort received their doses
later in the week. Dose escalation to the next higher dose tested was always twofold
higher than the previous dose. A decision tree was employed (also visually depicted
in Fig. 1a) and followed for the single-dosing part A of the study. In period 1, the
following doses of UA or placebo were tested:
• Cohort 1 (6 active + 2 placebo) was orally administered the lowest dose of UA
(250 mg). Aer safety of this dose was documented, the study proceeded to the
next higher dosing.
• Cohort 2 (6 active + 2 placebo) was orally administered the next higher dose
of UA (500 mg). Once safety was documented, the study proceeded to the next
higher dosing.
• Cohort 3 (6 active + 2 placebo) was orally administered the next higher single
dose of UA (1,000 mg). Once safety was documented, the study proceeded to
the next higher dosing.
Following a minimum 3-week wash-out period, in period 2, the following
doses of UA or placebo were tested:
• Cohort 1 (6 active + 2 placebo) was orally administered the highest single dose
of UA (2,000 mg) examined in the study.
• Cohorts 2 and 3 were administered single doses of UA at 500 mg and 1,000 mg,
respectively, which were admixed in a high-protein yogurt.
Once safety was documented for a given UA dose, only in the subsequent
week was the UA dosing escalated to the next higher dosing. Safety (adverse
events, serum biochemistry, urinalysis, ECG and physical examination) was
documented for each dose before escalation to the next higher dose. This
evaluation was overseen by a safety monitoring committee that included two
qualified physicians at the clinical site. At the end of each dose level, an interim
safety report was issued by the study medical investigator. A dose escalation
meeting was held, and the decision to proceed (for example, to the next higher
dose) was taken on the basis of a blind safety data review. The same decision tree
was employed for the 4-week multiple dosing in part B of the study, starting
from lowest to highest UA dosing. Part B consisted of administering multiple
ascending doses (250 mg, 500 mg and 1,000 mg) to 36 healthy elderly male and
female volunteers, who were orally administered placebo or UA-containing soft
gels for 4 weeks. In both parts, placebo or UA were administered in the form of
soft gels containing either 250 mg UA or placebo. The analytical laboratories,
the study investigator and the team and the subject were blinded for the duration
of the clinical study. Each subject was randomized to receive either UA or placebo
in soft gels that looked identical in appearance. The subjects were recruited from
the volunteers database of the clinical unit and all participants were Caucasian.
The randomization list was generated by the clinical site (Eurofins Optimed)
using SAS statistical software (v.9.3). All clinical data were recorded electronically
on a web-based electronic case report form (eCRF-RDC 4.6, a validated
Electronic Records/Electronic Signature-compliant (21 CFR Part 11) application
of Oracle Clinical 4.6). The study was carried out in accordance with the
Declaration of Helsinki as modified in Fortaleza (2013), the recommendations
on Good Clinical Practice (GCP) (ICH E6) and applicable local regulatory
requirement(s). The clinical study was approved by both the Ethics Committee
‘Comité de Protection des Personnes’ and by the French/National Health
Authorities ‘Agence Nationale de sécurité du médicament et des produits de santé’
for the use of urolithin A as a food ingredient. The clinical study is registered in
clinicaltrials.gov as NCT02655393.
Sample size. The sample size was chosen on the basis of feasibility to allow
preliminary characterization of safety, tolerability and pharmacokinetics and to
explore pharmacodynamic measures of the UA intervention. Following guidelines
and recommendations9 and similar standard phase 1 study dose escalation
designs10,11, the single-dosing cohorts consisted of 6 + 2 (6 active and 2 placebo)
receiving subjects per dose at randomization, while the multiple-dosing cohorts
were 9 + 3 (9 active and 3 placebo) receiving subjects per dose at randomization.
Placebo samples from each dose level were grouped during analysis, resulting in
six placebo subjects in the single-dosing cohorts, and nine placebo subjects in the
multiple-dosing cohorts. This is consistent with most phase 1 pharmacokinetic
trials designed to provide sufficient information about human safety and the
bioactive pharmacokinetics profile and to allow dose selection and powering of the
design of future phase 2 efficacy trials.
Inclusion and exclusion criteria. Subjects were healthy, sedentary elderly people
who were included in the study on meeting the inclusion and exclusion criteria,
as reviewed by the study Principle Investigator. If volunteers agreed to enter the
study, they signed the informed consent form. The participants agreed to refrain
from consuming dietary supplements that could potentially impact either muscle
or mitochondrial function, such as resveratrol, pomegranate and ellagitannins,
nicotinamide riboside, whey protein, leucine, iso-leucine, -carnitine, creatinine,
coenzyme Q10, vitamin A, niacin, folic acids, vitamin C, vitamin E and probiotic
foods and supplements, during the 2 weeks before inclusion and throughout the
study. Concomitant medications were recorded during the course of the study.
Participants were requested to follow a stable lifestyle throughout the duration
of the trial with no sports and exercise activity. Elderly subjects were medically
screened up to 21 d before study enrolment for eligibility. General inclusion criteria
included an age of 61 y to 85 y; BMI 18–32 kg m–2 and demonstrated sedentary
behaviour, that is, having an activity level <600 MET (metabolic equivalent
unit—minutes per week as assessed by the International Physical Activity
Questionnaire (IPAQ)).
General exclusion criteria included any presence of cardiovascular, pulmonary,
gastrointestinal, hepatic, renal, metabolic, haematological, neurological,
psychiatric, systemic or infectious disease; inability to abstain from muscular
and physical activity >20 min each day during the course of the study; history
or presence of drug or alcohol abuse (alcohol consumption >40 grams per day);
positive hepatitis B surface antigen or anti-hepatitis C virus antibody, or positive
results for human immunodeficiency virus-1 or -2 tests; inability to refrain from
smoking more than half a pack of cigarettes (or similar for other tobacco products)
per day during the course of the study; excessive consumption of beverages with
xanthine bases (>4 cups or glasses per day) and blood donation within 2 months
before the start of the clinical study product administration. Adverse events and
concomitant medications were continuously registered throughout the entire
clinical study period.
Study schedule and product intake. Subjects were screened for eligibility up to
21 d before study enrolment. The level of activity was assessed only during
screening. Once the subjects were confirmed to have met the inclusion and
exclusion criteria by the medical investigator and had signed the informed
consent form, they were admitted to the clinical research unit for the part A single
dosing that was conducted in two periods, separated by at least a 3-week wash-
out period. UA was synthesized to a high purity (>99%) for this study and was
formulated into soft gels containing 250-mg doses. The placebo soft gels were
indistinguishable in appearance from UA-containing soft gels. UA was also directly
admixed into a high-protein yogurt (17 grams perserving) at doses of 500 mg
and 1,000 mg, to study the effect of food on bioavailability. Placebo yogurts were
colour-matched and indistinguishable to the UA admixed yogurt. These products
were administered orally during fasting (that is, before breakfast) in the morning.
Following single dosing, plasma samples were collected at frequent intervals up
to 96 h following dosing to establish UA pharmacokinetics. In the highest single
dosing of UA at 2,000 mg, muscle biopsies were performed 8 h after dosing to
detect UA levels in skeletal muscle in the subjects. For part B of the study, repeated
administrations of UA were performed in the morning.
During part A, treatments containing UA or placebo were administered under
the supervision of the investigator in a clinical pharmacology unit (Eurofins
Optimed) at around 8:00. For part B, subjects were provided with the study
product to take during each ambulatory visit and were given the necessary
supply for administration at home between visits. Subjects were given a diary to
record the number of capsules taken and the time of intake. The actual time of
product administration was documented in the individual eCRF of the clinical
study. All unused capsules were returned by the subjects at the end of the study
intervention. Muscle tissue collection and blood sampling for biological markers of
mitochondrial function were performed at the day 1 visit (that is, before the start
of the 4-week dosing in part B of the study) and on day 28, following the
last UA dosing.
IPAQ. The short version of the IPAQ was used to estimate an individual’s
level of physical activity in the domains of household and yard work activities,
occupational activity, self-powered transport and leisure-time physical activity, as
NATURE METABOLISM | VOL 1 | JUNE 2019 | 595–603 | www.nature.com/natmetab
600
Letters
Nature MetabolisM
well as sedentary activity. The questionnaire was taken only during screening and
was self-assessed by the subjects. The scores were recorded in the eCRF.
Safety and bioavailability assessment of UA. Adverse events were recorded and
coded according to the Medical Dictionary for Regulatory Activity (MedDRA).
The safety events were classified as serious adverse events and adverse events by the
study medical principal investigator. The relationship to the study product was also
assessed and assigned to the following categories: related, unrelated, unlikely (to be
related) and possible (could be related but a doubt exists). Twelve-lead ECGs were
recorded in supine position (Cartouch Cardionics Device). A physical examination
was conducted on all subjects by the study medical principal investigator at
the start and end of study treatments and including evaluation of main body
systems/regions, including: skin and mucous, ears/nose/throat, pulmonary,
cardiac, gastrointestinal and neurological systems. A panel of haematological
(haemoglobin, haematocrit, red blood cells, white blood cells, differential count,
platelet count, mean corpuscular volume, mean corpuscular haemoglobin and
mean corpuscular haemoglobin concentration); serum biochemistry (creatinine,
uric acid, alanine serine transferase, alanine leucine transferase, gamma glutamyl
transferase, and total and conjugated bilirubin); and urinalysis (pH, ketone bodies,
proteins, glucose and blood) safety tests were also performed at the start and end of
UA or placebo treatment, to compare the safety profile of UA in elderly subjects.
Plasma concentrations of UA and its metabolites, UA-glucuronide and
UA-sulfate, were analysed in plasma and muscle biopsy samples. UA levels in
plasma were assessed at the following time points during both the single and
following the last dose of the 4-week multiple-dosing oral intervention with UA
(pre-dosing, 1, 2, 4, 6, 8, 12, 24, 72 and 96 h except for the 250 mg and 500 mg
single dosing where the plasma was sampled up to the 36 h time point) for
bioavailability assessments. Plasma samples were also collected for assessment of
UA steady-state concentrations during the 4-week UA study at the following time
points (day 1, day 7, day 14, day 28, day 29, day 31, day 32). UA levels in skeletal
muscle biopsies were also assessed at pre-dosing and at 8 h post-dosing.
We have developed robust, precise and validated methods to measure the
individual concentrations of the parent urolithin A (UA) and its detectable
metabolites in both plasma and in human skeletal muscle. Validation of the
methods for the measurements of urolithin A aglycone and its glucuronide
and sulfate metabolites was performed following both the guidance on method
validation from the Food and Drug Administration13 and the European Medicines
Agency12. The limit of quantification was 5.00 pg ml1 for UA in plasma and
5.00 ng ml1 for UA-glucuronide and UA-sulfate in plasma, 5.00 pg ml1 for UA in
skeletal muscle and 5 ng ml1 for UA-glucuronide and UA-sulfate in skeletal muscle.
For mean value calculations, all values below the limit of quantification were set
to zero. Concentrations were converted to molarity values using the following
molecular weight: M(UA) = 228.2 g mol1; M(UA-glucuronide) = 404.3 g mol1;
M(UA-sulfate) = 325.3 g mol1. The pharmacokinetic variables were calculated
on the basis of the actual sampling times. Non-compartmental pharmacokinetic
analysis was performed using Phoenix WinNonlin v.6.3 (Pharsight Corporation).
Muscle biopsy procedure. Muscle tissue was collected from the vastus lateralis
skeletal muscle of the right leg using a 4.5-mm Bergström muscle biopsy needle.
The subjects were in fasting condition before the collection of the muscle biopsy
sample. Subjects were placed in a semi-supine position with the knees supported
and slightly flexed. The lateral side of the leg was palpated to determine the
location of biopsy, which was 10 cm proximal of the upper pole of the patella on
a line between the patella and the anterior superior iliac spine. After disinfecting
the skin, the skin and muscle fascia were locally anaesthetized with 5–10 ml
lidocaine 5% solution. Additional lidocaine was administered when the anaesthetic
effect was not sufficient. A sterile cloth with a hole was placed on the leg, keeping
the biopsy site exposed. A small incision of 5 mm was made in the skin and the
muscle fascia was incised minimally, just wide enough for the biopsy needle to
pass through. The biopsy needle was introduced via the skin and fascia into the
muscle. Considering the length of the needle, the depth of the collection could be
estimated to be around 4–5 cm below the skin and, therefore, in the skeletal muscle.
Moreover, the passing of the fascia lata was always perceptible. The minimal
amount of each muscle sample was at least 30 mg. After collecting the required
amount of muscle tissue, the wound was closed with a single, non-absorbable skin
suture and pressure was applied by an elastic bandage. Subjects were instructed not
to perform strenuous physical activity with the right leg for 2 d. Tissue collected
for RNA and DNA expression was snap-frozen in liquid nitrogen within 30 min of
collection and stored at 80 °C.
Biomarker analysis in skeletal muscle biopsies. mtDNA abundance. Muscle
samples were incubated overnight in 360 µl of buer ATL and 40 µl proteinase
(Qiagen) at 55 °C in a thermomixer set at 300 r.p.m. Cell debris was removed
by centrifugation and 200 µl of clear lysates was placed in the QIAsymphony SP
workstation (Qiagen). DNA was extracted with the QIAsymphony DNA Mini
kit (Qiagen, catalogue no. 937236) following the manufacturer’s procedures.
Quantitative PCR was performed on the Fluidigm Biomark system following the
Fluidigm Specic Target Amplication Quick Reference (Fluidigm). Samples were
loaded as technical triplicates. e real-time PCR data were analysed using the
Linear Derivative baseline correction and User (detector) Ct threshold method
on the latest version of the Fluidigm Biomark soware (v.4.1.3). Quantication of
mtDNA was performed using two customized Taqman assays targeted against a
nuDNA sequence (18S) and a conserved region of mtDNA (MTND1)32. Relative
mtDNA copy number was determined comparing MTND1 to 18S signal. All
quantications were determined using the 2−∆∆Ct method and the mean Ct of the
technical triplicates.
mRNA extraction from muscle biopsies. Approximately 10–15 mg of muscle samples
was homogenized in 800 µl of buffer RLT Plus (Qiagen) plus two steel balls using a
Tissue Lyser (Qiagen). Cell debris was removed by centrifugation and clear lysates
were placed in the QIAsymphony SP workstation (Qiagen). RNA was extracted
with the QIAsymphony RNA kit (Qiagen, catalogue no. 931636) following the
manufacturer’s procedures. RNA was quantified and checked for purity on a
Nanodrop-8000. RNA integrity was controlled using RNA 6000 Nano LabChip
kit (Agilent Technologies, catalogue no. 5065-4476) on an Agilent Bioanalyzer
(Agilent Technologies).
Gene expression using qPCR. Complementary DNA was synthesized using an ABI
High Capacity cDNA kit using 50 ng of RNA or the highest quantities isolated.
Quantitative PCR was performed on the Fluidigm Biomark system following the
Fluidigm Specific Target Amplification Quick Reference (Fluidigm). Samples were
loaded as technical triplicates. The real-time PCR data were analysed using the
Linear Derivative baseline correction and User (detector) Ct threshold method on
the latest version of the Fluidigm Biomark software (v.4.1.3).
Gene expression using microarray. Two nanograms of total RNA was processed
using the Human Clariom D microarray following the Affymetrix GeneChip
WT Pico Reagent Kit Guide. The data were normalized with the signal space
transformation-robust multi-array average (SST-RMA) method.
Gene set enrichment analysis (GSEA). GSEA was employed to look for over-
representation of known pathways and gene functional categories among the
regulated genes19. First, a ranked list of differentially expressed genes was
generated using the R package limma (R v.3.3.2, limma v.3.30.11). A design
matrix was created using the status (dose + day as factors). Comparisons were
generated using a contrast matrix. A linear model was then fitted to each gene.
A moderated t-test was used to compute the t-statistics, moderated F-statistic
and log-odds of differential expression using the empirical Bayes method.
Genes were ranked by log2(fold change) (high to low) and were filtered for an
unadjusted P value of 0.1 before using them in the GSEA analysis to reduce the
risk of false positives. Gene sets defined by the Gene Ontology Consortium
(http://www.geneontology.org/) were downloaded from the Broad Institute’s
MSigDB website (http://software.broadinstitute.org/gsea/msigdb), using the C5:
GO gene sets (MSigDB v.5.2). A total of 6,166 gene sets were used as an input for
all GSEA comparisons. The genes contributing the most to the enrichment of
GO_MITOCHONDRION were identified using the leading edge analysis available
in the GSEA software (v.2.2.3).
Global metabolomics in plasma. A 6-ml blood sample was withdrawn into a
K2 EDTA-coated tube. The blood samples were gently inverted a few times for
complete mixing with the anticoagulant. The exact time of sample collection
was recorded on the eCRF. Within 30 min following blood collection, each blood
sample was centrifuged at 1500g for 10 min at 4 °C. At 30 min after centrifugation,
the top layer of human plasma was transferred into two pre-labelled polypropylene
tubes, containing approximately 1,500 µl of plasma. Tubes were capped
immediately and the plasma was frozen in an upright position at approximately
80 °C for storage.
Metabolomics of plasma was performed by Metabolon according to published
methods33. In brief, sample preparation was conducted using a proprietary series
of organic and aqueous extractions to remove the protein fraction while allowing
maximum recovery of small molecules. The extracted samples were split into
equal parts for analysis on the gas chromatography–mass spectroscopy (GC–MS)
and liquid chromatography–tandem mass spectroscopy (LC–MS/MS) platforms.
For LC–MS/MS, samples were split in two aliquots that were either analysed in
positive (acidic solvent) or negative (basic solvent) ionization mode. GC–MS was
performed on bis(trimethylsilyl)triflouroacetamide-derivatized samples in a 5%
phenyl GC column.
Statistical analysis. The SAS statistical software (v.9.3) was used to analyse all
the clinical safety endpoints and the qPCR data (RNA and DNA). This is a first-
in-human phase 1 study of a nutritional ingredient that follows the standard dose
escalating design (single and multiple) employed in phase 1 trials, with stopping
rule criteria for safety analysis; that is, all the doses have the same number of
subjects assigned until experimentation is stopped, starting from the lowest to the
maximum tolerated dose. We chose a 6 + 2 (6 active and 2 placebo) design for the
single-dosing cohorts and a 9 + 3 (9 active and 3 placebo) design for the multiple-
dosing cohorts to permit a robust statistical characterization of UA in the healthy
elderly population for endpoints of safety, tolerability and pharmacokinetics and
NATURE METABOLISM | VOL 1 | JUNE 2019 | 595–603 | www.nature.com/natmetab 601
Letters Nature MetabolisM
to explore pharmacodynamic parameters. For pharmacodynamics analysis, the
alpha level was set at a standard level of 5% and all statistical tests are bilateral. For
each gene assessed via qPCR the following analyses were performed: descriptive
statistics on normalized Ct values for each assessment by dose group; descriptive
statistics on mean fold change values at each assessment by dose group and
analyses of variance (ANOVA) performed on mean fold change values. The
following model was performed on mean fold change values in gene expression:
model included dose (placebo/250 mg/500 mg/1,000 mg) as a fixed factor. In
case of a significant dose effect (alpha risk: 0.05), pairwise comparisons between
the active dose groups and the placebo were carried out from ANOVA using a
Dunnett’s test. If residuals from ANOVA were not normally distributed, a natural
log (ln) transformation was applied to the data. If the normal hypothesis was not
demonstrated from the ln transformation data, rank data were retained.
For metabolomics data, statistical analyses were performed in ArrayStudio on
log-transformed data. A two-way, repeated-measures ANOVA was used where
one factor was applied to each subject and the second factor was a time point. The
model took into account the repeated measures, that is, the treatments are given to
the same subject over time, to determine whether there was a significant effect of
the compound over time.
All the samples that were measured and all the data points were included in the
analysis. There was no sample or data point exclusion.
Non-interventional study (NIS) comparing pre-frail sedentary with active
healthy elderly subjects. The study was conducted as described elsewhere23
(NCT02472340). Briefly, 11 pre-frail (6 males and 5 females) subjects aged
70.2 ± 5.8 y and 11 active (6 males and 5 females) subjects aged 70.0 ± 6.7 y
participated in this study, with subjects having a mean BMI of 25.7 ± 4.2 versus
24.6 ± 3.9 kg m2, respectively. Subjects were considered pre-frail when fulfilling
at least one to two of the three criteria for frailty34. According to the IPAQ
questionnaire, pre-frail subjects had a mean daily energy expenditure of
392 MET minutes per week, corresponding to less than 20 min of walking per day,
while the active group had a score of 6,508 MET minutes per week, corresponding
to 1 h of vigorous exercise per day. Muscle biopsy was collected under fasted state
in the morning and processed for RNA extraction, as described above. The HTA
2.0 microarray chip from Affymetrix was used to measure mRNA expression
levels of 42,935 reporters/probes associated with 33,804 annotated transcripts
or genes (mRNA). The raw gene expression values were normalized with the
SST-RMA algorithm. The genes were ordered in a ranked list according to the
magnitude and direction of their differential expression between pre-frail and
active groups with the R library limma (R v.3.3.2). This list was used to perform
the GSEA analysis using the same gene sets as for the phase 1 study (see ‘GSEA’
section above).
Reporting Summary. Further information on research design is available in the
Nature Research Reporting Summary linked to this article.
Data availability
The gene expression data are deposited at the European Genome-phenome Archive
under accession code EGAS00001003638 and can be accessed subject to signing a
data access agreement.
Received: 15 January 2019; Accepted: 3 May 2019;
Published online: 14 June 2019
References
1. Ryu, D. etal. Urolithin A induces mitophagy and prolongs lifespan in
C. elegans and increases muscle function in rodents. Nat. Med. 22,
879–888 (2016).
2. Choi, A. M., Ryter, S. W. & Levine, B. Autophagy in human health and
disease. N. Engl. J. Med. 368, 651–662 (2013).
3. Drake, J. C. & Yan, Z. Mitophagy in maintaining skeletal muscle
mitochondrial proteostasis and metabolic health with ageing. J. Physiol. 595,
6391–6399 (2017).
4. Coen, P. M. etal. Skeletal muscle mitochondrial energetics are associated
with maximal aerobic capacity and walking speed in older adults. J. Gerontol.
A 68, 447–455 (2013).
5. Zane, A. C. etal. Muscle strength mediates the relationship between
mitochondrial energetics and walking performance. Aging Cell 16,
461–468 (2017).
6. Heilman, J., Andreux, P., Tran, N., Rinsch, C. & Blanco-Bose, W. Safety
assessment of Urolithin A, a metabolite produced by the human gut
microbiota upon dietary intake of plant derived ellagitannins and ellagic acid.
Food Chem. Toxicol. 108, 289–297 (2017).
7. Keefe, D. M. GRAS Notice No. GRN 000791 (Food and Drug
Administration, 2018).
8. Yoshimura, Y. etal. Interventions for treating sarcopenia: a systematic review
and meta-analysis of randomized controlled studies. J. Am. Med. Dir. Assoc.
18, 553 e551–553.e516 (2017).
9. Guideline on Strategies to Identify and Mitigate Risks for First-in-Human
Clinical Trials with Investigational Medicinal Products EMEA/CHMP/
SWP/28367/07 (European Medicines Agency, 2007).
10. Yang, H. etal. Phase 1 single- and multiple-ascending-dose randomized
studies of the safety, pharmacokinetics, and pharmacodynamics of AG-348, a
rst-in-class allosteric activator of pyruvate kinase R, in healthy volunteers.
Clin. Pharmacol. Drug Dev. 8, 246–259 (2019).
11. Chandorkar, G., Zhan, Q., Donovan, J., Rege, S. & Patino, H.
Pharmacokinetics of surotomycin from phase 1 single and multiple
ascending dose studies in healthy volunteers. BMC Pharmacol. Toxicol. 18,
24 (2017).
12. Guideline on Bioanalytical Method Validation EMEA/CHMP/
EWP/192217/2009 (European Medicines Agency, 2011).
13. Guidance for Industry: Bioanalytical Method Validation (US Department of
Health and Human Services, Food and Drug Administration, Center for
Drug Evaluation and Research and, Center for Veterinary Medicine, 2001).
14. Tomas-Barberan, F. A. etal. Urolithins, the rescue of ‘old’ metabolites to
understand a ‘new’ concept: metabotypes as a nexus among phenolic
metabolism, microbiota dysbiosis, and host health status. Mol. Nutr. Food Res.
61, 1500901 (2017).
15. Schooneman, M. G., Vaz, F. M., Houten, S. M. & Soeters, M. R.
Acylcarnitines: reecting or inicting insulin resistance? Diabetes 62,
1–8 (2013).
16. Mitochondrial Medicine Society’s Committee on Diagnosis etal.e in-depth
evaluation of suspected mitochondrial disease. Mol. Genet. Metab. 94,
16–37 (2008).
17. Lum, H. etal. Plasma acylcarnitines are associated with physical performance
in elderly men. J. Gerontol. A 66, 548–553 (2011).
18. Felder, T. K. etal. Specic circulating phospholipids, acylcarnitines, amino
acids and biogenic amines are aerobic exercise markers. J. Sci. Med. Sport 20,
700–705 (2017).
19. Subramanian, A. etal. Gene set enrichment analysis: a knowledge-based
approach for interpreting genome-wide expression proles. Proc. Natl Acad.
Sci. USA 102, 15545–15550 (2005).
20. Mahoney, D. J., Parise, G., Melov, S., Safdar, A. & Tarnopolsky, M. A. Analysis
of global mRNA expression in human skeletal muscle during recovery from
endurance exercise. FASEB J. 19, 1498–1500 (2005).
21. Lammers, G. etal. Expression of genes involved in fatty acid transport
and insulin signaling is altered by physical inactivity and exercise training
in human skeletal muscle. Am. J. Physiol. Endocrinol. Metab. 303,
E1245–E1251 (2012).
22. Robinson, M. M. etal. Enhanced protein translation underlies improved
metabolic and physical adaptations to dierent exercise training modes in
young and old humans. Cell Metab. 25, 581–592 (2017).
23. Andreux, P. A. etal. Mitochondrial function is impaired in the skeletal
muscle of pre-frail elderly. Sci. Rep. 8, 8548 (2018).
24. Gong, Z. etal. Urolithin A attenuates memory impairment and
neuroinammation in APP/PS1 mice. J. Neuroinammation 16,
62 (2019).
25. Fang, E. F. etal. Mitophagy inhibits amyloid-beta and tau pathology and
reverses cognitive decits in models of Alzheimer’s disease. Nat. Neurosci. 22,
401–412 (2019).
26. Singh, R. etal. Enhancement of the gut barrier integrity by a
microbial metabolite through the Nrf2 pathway. Nat. Commun. 10,
89 (2019).
27. Olesen, J., Kiilerich, K. & Pilegaard, H. PGC-1alpha-mediated adaptations in
skeletal muscle. Pugers Arch. 460, 153–162 (2010).
28. Jeppesen, J. etal. Enhanced fatty acid oxidation and FATP4 protein
expression aer endurance exercise training in human skeletal muscle.
PLoS ONE 7, e29391 (2012).
29. Laker, R. C. etal. Ampk phosphorylation of Ulk1 is required for targeting of
mitochondria to lysosomes in exercise-induced mitophagy. Nat. Commun. 8,
548 (2017).
30. Vainshtein, A., Tryon, L. D., Pauly, M. & Hood, D. A. Role of PGC-1alpha
during acute exercise-induced autophagy and mitophagy in skeletal muscle.
Am. J. Physiol., Cell Physiol. 308, C710–C719 (2015).
31. Harrigan, J. A., Jacq, X., Martin, N. M. & Jackson, S. P. Deubiquitylating
enzymes and drug discovery: emerging opportunities. Nat. Rev. Drug Discov.
17, 57–78 (2018).
32. Spendi, S. etal. Mitochondrial DNA deletions in muscle satellite cells:
implications for therapies. Hum. Mol. Genet. 22, 4739–4747 (2013).
33. Milburn, M. V. & Lawton, K. A. Application of metabolomics to diagnosis of
insulin resistance. Annu. Rev. Med. 64, 291–305 (2013).
34. Fried, L. P. etal. Frailty in older adults: evidence for a phenotype. J. Gerontol.
A Biol. Sci. Med. Sci. 56, M146–M156 (2001).
Acknowledgements
We would like to thank the volunteers for their participation in the study and Eurofins
Optimed study staff for subject recruitment, data collection and processing, and
NATURE METABOLISM | VOL 1 | JUNE 2019 | 595–603 | www.nature.com/natmetab
602
Letters
Nature MetabolisM
S. Houten for his insight on the acylcarnitine data. Grant support by the Fondation
Suisse de Recherche sur les Maladies Musculaires and the Fondation Marcel Levaillant
is acknowledged.
Author contributions
A.S., W.B., P.A.A., P.A. and C.R. contributed to the design of the study. P.A.A., A.S., C.R.
and J.A. wrote the manuscript with the help of the other co-authors. A.S., W.B., P.A.A.
and C.R. collected all the ex vivo data. P.A.A. and D.R. analysed the metabolomics data.
F.B., M.I. and P.A.A. analysed the microarray data. All authors reviewed the manuscript.
Competing interests
The authors declare the following competing interests: A.S., P.A.A., W.B. and C.R. are
employees; P.A. and C.R. are board members; and J.A. and P.A. are members of the
Scientific Advisory Board of Amazentis SA, the sponsor of this clinical study. The other
authors declare no competing interests.
Additional information
Supplementary information is available for this paper at https://doi.org/10.1038/
s42255-019-0073-4.
Reprints and permissions information is available at www.nature.com/reprints.
Correspondence and requests for materials should be addressed to C.R.
Journal peer review information: Primary Handling Editor: Christoph Schmitt.
Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in
published maps and institutional affiliations.
© The Author(s), under exclusive licence to Springer Nature Limited 2019
NATURE METABOLISM | VOL 1 | JUNE 2019 | 595–603 | www.nature.com/natmetab 603
1
nature research | reporting summary April 2018
Corresponding author(s): Chris Rinsch
Reporting Summary
Nature Research wishes to improve the reproducibility of the work that we publish. This form provides structure for consistency and transparency
in reporting. For further information on Nature Research policies, see Authors & Referees and the Editorial Policy Checklist.
Statistical parameters
When statistical analyses are reported, confirm that the following items are present in the relevant location (e.g. figure legend, table legend, main
text, or Methods section).
n/a Confirmed
The exact sample size (n) for each experimental group/condition, given as a discrete number and unit of measurement
An indication of whether measurements were taken from distinct samples or whether the same sample was measured repeatedly
The statistical test(s) used AND whether they are one- or two-sided
Only common tests should be described solely by name; describe more complex techniques in the Methods section.
A description of all covariates tested
A description of any assumptions or corrections, such as tests of normality and adjustment for multiple comparisons
A full description of the statistics including central tendency (e.g. means) or other basic estimates (e.g. regression coefficient) AND
variation (e.g. standard deviation) or associated estimates of uncertainty (e.g. confidence intervals)
For null hypothesis testing, the test statistic (e.g. F, t, r) with confidence intervals, effect sizes, degrees of freedom and P value noted
Give P values as exact values whenever suitable.
For Bayesian analysis, information on the choice of priors and Markov chain Monte Carlo settings
For hierarchical and complex designs, identification of the appropriate level for tests and full reporting of outcomes
Estimates of effect sizes (e.g. Cohen's d, Pearson's r), indicating how they were calculated
Clearly defined error bars
State explicitly what error bars represent (e.g. SD, SE, CI)
Our web collection on statistics for biologists may be useful.
Software and code
Policy information about availability of computer code
Data collection No software was used.
Data analysis The real-time PCR data were analyzed using the Linear Derivative baseline correction and User (detector) Ct threshold method on the
latest version of the Fluidigm BioMark software (ver. 4.1.3). The commercial SAS statistical software (version 9.3) was used to analyze all
the clinical related endpoints and qPCR data (RNA and DNA). Microarray was analyzed using the publicly available softwares Gene-Set
Enrichment Analysis (2.2.3), the gene sets collection from the Broad Institute’s MSigDB website (v5.2) and R package limma (R version
3.3.2, limma version 3.30.11). For metabolomics data, statistical analyses were performed in ArrayStudio. Non-compartmental
pharmacokinetic analysis was performed using Phoenix WinNonlin Version 6.3.
For manuscripts utilizing custom algorithms or software that are central to the research but not yet described in published literature, software must be made available to editors/reviewers
upon request. We strongly encourage code deposition in a community repository (e.g. GitHub). See the Nature Research guidelines for submitting code & software for further information.
2
nature research | reporting summary April 2018
Data
Policy information about availability of data
All manuscripts must include a data availability statement. This statement should provide the following information, where applicable:
- Accession codes, unique identifiers, or web links for publicly available datasets
- A list of figures that have associated raw data
- A description of any restrictions on data availability
The genomic dataset generated during the current study will be made publicly available in an appropriate genomic data repository.
Field-specific reporting
Please select the best fit for your research. If you are not sure, read the appropriate sections before making your selection.
Life sciences Behavioural & social sciences Ecological, evolutionary & environmental sciences
For a reference copy of the document with all sections, see nature.com/authors/policies/ReportingSummary-flat.pdf
Life sciences study design
All studies must disclose on these points even when the disclosure is negative.
Sample size The sample size has been chosen based on feasibility to allow preliminary characterization of safety, tolerability and pharmacokinetics and to
explore pharmacodynamic measures of the UA intervention.
Data exclusions All the samples that were measured and all the datapoints were included in the analysis. There was no sample or datapoint exclusion.
Replication Experimental replication was not attempted.
Randomization The randomization list was generated by the clinical site (Eurofins Optimed, Grenoble, France) using SAS statistical software (version 9.3).
Blinding The analytical laboratories, the study investigator and the team and the subject were blinded for the duration of the clinical study.
Reporting for specific materials, systems and methods
Materials & experimental systems
n/a Involved in the study
Unique biological materials
Antibodies
Eukaryotic cell lines
Palaeontology
Animals and other organisms
Human research participants
Methods
n/a Involved in the study
ChIP-seq
Flow cytometry
MRI-based neuroimaging
Human research participants
Policy information about studies involving human research participants
Population characteristics Subjects were healthy, sedentary elderly males and females all of Caucasian origin. General inclusion criteria included an age of
61 years to 85 years; body mass index 18-32 kg/m2 and demonstrated sedentary behavior, i.e., having an activity category of 1
with an activity level < 600 MET (metabolic equivalent unit – minutes per week as assessed by the International Physical Activity
Questionnaire (IPAQ).
Recruitment The subjects were recruited from the volunteers’ database of the clinical unit and based on population characteristics described
above.
... In 2018, urolithin A was officially recognized as a safe substance and approved for human studies [130,131]. Additionally, in 2019, for the first time, a study of urolithin A was conducted in the elderly to assess the safety profile [86]. Thus, urolithin A is the most promising compound for the role of a drug for the prevention and treatment of neurodegenerative diseases (see Table 1). ...
... approved for human studies [130,131]. Additionally, in 2019, for the first time, a study of urolithin A was conducted in the elderly to assess the safety profile [86]. Thus, urolithin A is the most promising compound for the role of a drug for the prevention and treatment of neurodegenerative diseases (see Table 1). ...
... A marker of the degree of mitophagy is needed to select the dosage of the drug. In a placebo-controlled study of urolithin A, a thigh muscle biopsy was taken from the subjects and markers associated with the activation of mitochondrial genes under the influence of urolithin A were measured [86]. In clinical practice, this method can be considered barbaric; therefore, methods for determining mitophagy markers directly in plasma are currently being developed [175,176]. ...
Article
Full-text available
Neurodegenerative diseases, such as Alzheimer’s disease or Parkinson’s disease, significantly reduce the quality of life of patients and eventually result in complete maladjustment. Disruption of the synapses leads to a deterioration in the communication of nerve cells and decreased plasticity, which is associated with a loss of cognitive functions and neurodegeneration. Maintaining proper synaptic activity depends on the qualitative composition of mitochondria, because synaptic processes require sufficient energy supply and fine calcium regulation. The maintenance of the qualitative composition of mitochondria occurs due to mitophagy. The regulation of mitophagy is usually based on several internal mechanisms, as well as on signals and substances coming from outside the cell. These substances may directly or indirectly enhance or weaken mitophagy. In this review, we have considered the role of some compounds in process of mitophagy and neurodegeneration. Some of them have a beneficial effect on the functions of mitochondria and enhance mitophagy, showing promise as novel drugs for the treatment of neurodegenerative pathologies, while others contribute to a decrease in mitophagy.
... Because mitochondrial dysfunction is a feature of many neurological diseases, pharmacological compounds that increase the elimination of defective mitochondria are expected to have important therapeutic value(Lou et al., 2020;Swerdlow & Wilkins, 2020). Various pharmacological compounds, either natural bioactive compounds or synthetic drugs, are used for the preservation of mitochondrial integrity and enhancement of mitochondrial biogenesis by inducing mitophagy(Andreux et al., 2019;Jayatunga et al., 2021;Lou et al., 2020). These modest chemical therapies, in addition to nutritional management and regular exercise, help to restore energy metabolism in several model species, resulting in mitophagy-related neuroprotection and anti-ageing effects. ...
... IL-10 induces mitophagy by suppressing inflammasome activation and decreasing the mTOR via AMPK stimulation. In 60 healthy aged people, oral ingestion of UA (up to 1 gÁday À1 ) was non-toxic for 4 weeks and induced skeletal muscle mitochondrial and plasma acylcarnitine gene expression(Andreux et al., 2019). ...
Article
Mitochondria and mitochondria‐mediated signaling pathways are known to control synaptic signaling as well as long‐lasting changes in neuronal structure and function. Mitochondrial impairment is linked to synaptic dysfunction in normal aging and age‐associated neurodegenerative ailments including Parkinson's disease (PD) and Alzheimer's disease (AD). Both proteolysis and mitophagy perform a major role in neuroprotection by maintaining a healthy mitochondrial population during aging. Mitophagy, a highly evolutionarily conserved cellular process, helps in the clearance of damaged mitochondria and thereby maintains the mitochondrial and metabolic balance, energy supply, neuronal survival, and neuronal health. Besides the maintenance of brain homeostasis, hippocampal mitophagy also helps in synapse formation, axonal development, dopamine release, and long‐term depression. In contrast, defective mitophagy contributes to aging and age‐related neurodegeneration by promoting the accumulation of damaged mitochondria leading to cellular dysfunction. Exercise, stress management, maintaining healthy mitochondrial dynamics, and administering natural or synthetic pharmacological compounds are some of the strategies used for neuroprotection during aging and age‐related neurological diseases. The current review discusses the impact of defective mitophagy in aging and age‐associated neurodegenerative conditions, the underlying molecular pathways, and potential therapies based on recently elucidated mitophagy‐inducing strategies.
... Protocatechuic acid has significant potential in the inhibition of the NLRP3 inflammasome. Urolithin A is reported to improve mitochondrial and neuronal cell health [36,200,[215][216][217]. ...
Article
Full-text available
Flavonoids are a biodiverse family of dietary compounds that have antioxidant, anti-inflammatory, antiviral, and antibacterial cell protective profiles. They have received considerable attention as potential therapeutic agents in biomedicine and have been widely used in traditional complimentary medicine for generations. Such complimentary medical herbal formulations are extremely complex mixtures of many pharmacologically active compounds that provide a therapeutic outcome through a network pharmacological effects of considerable complexity. Methods are emerging to determine the active components used in complimentary medicine and their therapeutic targets and to decipher the complexities of how network pharmacology provides such therapeutic effects. The gut microbiome has important roles to play in the generation of bioactive flavonoid metabolites retaining or exceeding the antioxidative and anti-inflammatory properties of the intact flavonoid and, in some cases, new antitumor and antineurodegenerative bioactivities. Certain food items have been identified with high prebiotic profiles suggesting that neutraceutical supplementation may be beneficially employed to preserve a healthy population of bacterial symbiont species and minimize the establishment of harmful pathogenic organisms. Gut health is an important consideration effecting the overall health and wellbeing of linked organ systems. Bioconversion of dietary flavonoid components in the gut generates therapeutic metabolites that can also be transported by the vagus nerve and systemic circulation to brain cell populations to exert a beneficial effect. This is particularly important in a number of neurological disorders (autism, bipolar disorder, AD, PD) characterized by effects on moods, resulting in depression and anxiety, impaired motor function, and long-term cognitive decline. Native flavonoids have many beneficial properties in the alleviation of inflammation in tissues, however, concerns have been raised that therapeutic levels of flavonoids may not be achieved, thus allowing them to display optimal therapeutic effects. Dietary manipulation and vagal stimulation have both yielded beneficial responses in the treatment of autism spectrum disorders, depression, and anxiety, establishing the vagal nerve as a route of communication in the gut-brain axis with established roles in disease intervention. While a number of native flavonoids are beneficial in the treatment of neurological disorders and are known to penetrate the blood–brain barrier, microbiome-generated flavonoid metabolites (e.g., protocatechuic acid, urolithins, γ-valerolactones), which retain the antioxidant and anti-inflammatory potency of the native flavonoid in addition to bioactive properties that promote mitochondrial health and cerebrovascular microcapillary function, should also be considered as potential biotherapeutic agents. Studies are warranted to experimentally examine the efficacy of flavonoid metabolites directly, as they emerge as novel therapeutic options.
... Considering that mitophagy is the process of selective degradation of damaged or unnecessary mitochondria within the cells, recent studies have suggested that mitochondrial defects may be associated with the pathogenesis of sarcopenia [225] and that mitophagy may play a protective role against the development of sarcopenia [226], attenuating age-related loss of muscle mass and strength [227]. Enhancing mitophagy through various methods, such as the activation of specific signaling pathways or the use of mitochondria-targeted drugs that induce mitophagy [228,229], may be a potential strategy to counteract sarcopenia. ...
Article
Full-text available
Mitochondria are cellular organelles that play an essential role in generating the chemical energy needed for the biochemical reactions in cells. Mitochondrial biogenesis, i.e., de novo mitochondria formation, results in enhanced cellular respiration, metabolic processes, and ATP generation, while autophagic clearance of mitochondria (mitophagy) is required to remove damaged or useless mitochondria. The balance between the opposing processes of mitochondrial biogenesis and mitophagy is highly regulated and crucial for the maintenance of the number and function of mitochondria as well as for the cellular homeostasis and adaptations to metabolic demands and extracellular stimuli. In skeletal muscle, mitochondria are essential for maintaining energy homeostasis, and the mitochondrial network exhibits complex behaviors and undergoes dynamic remodeling in response to various conditions and pathologies characterized by changes in muscle cell structure and metabolism, such as exercise, muscle damage, and myopathies. In particular, the involvement of mitochondrial remodeling in mediating skeletal muscle regeneration following damage has received increased attention, as modifications in mitophagy-related signals arise from exercise, while variations in mitochondrial restructuring pathways can lead to partial regeneration and impaired muscle function. Muscle regeneration (through myogenesis) following exercise-induced damage is characterized by a highly regulated, rapid turnover of poor-functioning mitochondria, permitting the synthesis of better-functioning mitochondria to occur. Nevertheless, essential aspects of mitochondrial remodeling during muscle regeneration remain poorly understood and warrant further characterization. In this review, we focus on the critical role of mitophagy for proper muscle cell regeneration following damage, highlighting the molecular mechanisms of the mitophagy-associated mitochondrial dynamics and network reformation.
... The 4-week clinical trial in which up to 2000 mg oral Uro A doses were administered to elderly volunteers confirmed that the treatment had no adverse health effects. The observed 31 unfavorable effects were determined to be unrelated to the compound tested [48]. On the basis of the above findings, the US Food and Drug Administration already issued a favorable review for using Uro A as a food ingredient [49]. ...
Article
Mitophagy is a fundamental quality control mechanism of mitochondria. Its regulatory mechanisms and pathological implications remain poorly understood. Here, via a mitochondria-targeted genetic screen, we found that knockout (KO) of FBXL4, a mitochondrial disease gene, hyperactivates mitophagy at basal conditions. Subsequent counter screen revealed that FBXL4-KO hyperactivates mitophagy via two mitophagy receptors BNIP3 and NIX. We determined that FBXL4 functions as an integral outer-membrane protein that forms an SCF-FBXL4 ubiquitin E3 ligase complex. SCF-FBXL4 ubiquitinates BNIP3 and NIX to target them for degradation. Pathogenic FBXL4 mutations disrupt SCF-FBXL4 assembly and impair substrate degradation. Fbxl4-/- mice exhibit elevated BNIP3 and NIX proteins, hyperactive mitophagy, and perinatal lethality. Importantly, knockout of either Bnip3 or Nix rescues metabolic derangements and viability of the Fbxl4-/- mice. Together, beyond identifying SCF-FBXL4 as a novel mitochondrial ubiquitin E3 ligase restraining basal mitophagy, our results reveal hyperactivated mitophagy as a cause of mitochondrial disease and suggest therapeutic strategies.
Preprint
Mitochondrial calcium (mtCa2+) uptake via the Mitochondrial Calcium Uniporter (MCU) couples the regulation of calcium homeostasis to energy production. mtCa2+ uptake is rate-limiting for mitochondrial activation during muscle contraction, but how MCU is affected during physiopathology and whether it can be stimulated therapeutically remains largely uncharacterized. By profiling human and preclinical aging of skeletal muscle, we discovered a conserved down-regulation of MCUR1 during aging that decreases mtCa2+ uptake and drives sarcopenia. Through a screen of 5000 bioactive nutrients, we identify the natural polyphenol Oleuropein as a specific MCU activator that stimulates mitochondrial respiration via binding to MICU1. Oleuropein activates mtCa2+ uptake and oxidative energy metabolism to enhance endurance and limit fatigue in vivo both in young and aged. These effects of Oleuropein are mediated by an MCU-dependent mechanism in skeletal muscle as they are lost upon muscle-specific MCU KO. Our work demonstrates that impaired mtCa2+ uptake causes mitochondrial dysfunction during aging and establishes Oleuropein as a novel nutrient that specifically targets MCU to stimulate mitochondrial bioenergetics and muscle performance.
Article
Excessive fructose consumption exacerbates the progression of nonalcoholic fatty liver disease (NAFLD) by disrupting hepatic lipid homeostasis. This study sought to evaluate the efficacy of urolithin A (UroA) in a fructose-induced NAFLD mouse model. UroA was administered in the high-fructose-fed mice to investigate the antisteatotic effects in vivo. Fructose-stimulated HepG2 cells and primary hepatocytes were established for in vitro mechanistic assessment. The results suggested that UroA ameliorated fructose-induced hepatic steatosis in mice. Mechanistically, UroA impaired lipogenesis and enhanced β-oxidation in the livers of fructose-fed mice. Notably, UroA facilitated hepatic lipophagy through the AMPK/ULK1 pathway both in vivo and in vitro, degrading lipid droplets for fueling β-oxidation. This study indicates that UroA alleviates excessive lipid accumulation and restores lipid homeostasis in the livers of fructose-fed mice by suppressing lipid metabolic reprogramming and triggering lipophagy. Therefore, dietary supplementation of UroA or ellagitannins-rich foods may be beneficial for NAFLD individuals with high fructose intake.
Chapter
Primary mitochondrial diseases are some of the most common and complex inherited inborn errors of metabolism. Their molecular and phenotypic diversity has led to difficulties in finding disease-modifying therapies and clinical trial efforts have been slow due to multiple significant challenges. Lack of robust natural history data, difficulties in finding specific biomarkers, absence of well-validated outcome measures, and small patient numbers have made clinical trial design and conduct difficult. Encouragingly, new interest in treating mitochondrial dysfunction in common diseases and regulatory incentives to develop therapies for rare conditions have led to significant interest and efforts to develop drugs for primary mitochondrial diseases. Here, we review past and present clinical trials and future strategies of drug development in primary mitochondrial diseases.
Article
Full-text available
Background Alzheimer’s disease (AD) is a progressive neurodegenerative disorder characterized by an abnormal accumulation of amyloid-β (Aβ) plaques, neuroinflammation, and impaired neurogenesis. Urolithin A (UA), a gut-microbial metabolite of ellagic acid, has been reported to exert anti-inflammatory effects in the brain. However, it is unknown whether UA exerts its properties of anti-inflammation and neuronal protection in the APPswe/PS1ΔE9 (APP/PS1) mouse model of AD. Methods Morris water maze was used to detect the cognitive function. Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay was performed to detect neuronal apoptosis. Immunohistochemistry analyzed the response of glia, Aβ deposition, and neurogenesis. The expression of inflammatory mediators were measured by enzyme-linked immunosorbent assay (ELISA) and quantitative real-time polymerase chain reaction (qRT-PCR). The modulating effects of UA on cell signaling pathways were assayed by Western blotting. Results We demonstrated that UA ameliorated cognitive impairment, prevented neuronal apoptosis, and enhanced neurogenesis in APP/PS1 mice. Furthermore, UA attenuated Aβ deposition and peri-plaque microgliosis and astrocytosis in the cortex and hippocampus. We also found that UA affected critical cell signaling pathways, specifically by enhancing cerebral AMPK activation, decreasing the activation of P65NF-κB and P38MAPK, and suppressing Bace1 and APP degradation. Conclusions Our results indicated that UA imparted cognitive protection by protecting neurons from death and triggering neurogenesis via anti-inflammatory signaling in APP/PS1 mice, suggesting that UA might be a promising therapeutic drug to treat AD.
Article
Full-text available
Accumulation of damaged mitochondria is a hallmark of aging and age-related neurodegeneration, including Alzheimer’s disease (AD). The molecular mechanisms of impaired mitochondrial homeostasis in AD are being investigated. Here we provide evidence that mitophagy is impaired in the hippocampus of AD patients, in induced pluripotent stem cell-derived human AD neurons, and in animal AD models. In both amyloid-β (Aβ) and tau Caenorhabditis elegans models of AD, mitophagy stimulation (through NAD⁺ supplementation, urolithin A, and actinonin) reverses memory impairment through PINK-1 (PTEN-induced kinase-1)-, PDR-1 (Parkinson’s disease-related-1; parkin)-, or DCT-1 (DAF-16/FOXO-controlled germline-tumor affecting-1)-dependent pathways. Mitophagy diminishes insoluble Aβ1–42 and Aβ1–40 and prevents cognitive impairment in an APP/PS1 mouse model through microglial phagocytosis of extracellular Aβ plaques and suppression of neuroinflammation. Mitophagy enhancement abolishes AD-related tau hyperphosphorylation in human neuronal cells and reverses memory impairment in transgenic tau nematodes and mice. Our findings suggest that impaired removal of defective mitochondria is a pivotal event in AD pathogenesis and that mitophagy represents a potential therapeutic intervention.
Article
Full-text available
The importance of gut microbiota in human health and pathophysiology is undisputable. Despite the abundance of metagenomics data, the functional dynamics of gut microbiota in human health and disease remain elusive. Urolithin A (UroA), a major microbial metabolite derived from polyphenolics of berries and pomegranate fruits displays anti-inflammatory, anti-oxidative, and anti-ageing activities. Here, we show that UroA and its potent synthetic analogue (UAS03) significantly enhance gut barrier function and inhibit unwarranted inflammation. We demonstrate that UroA and UAS03 exert their barrier functions through activation of aryl hydrocarbon receptor (AhR)-nuclear factor erythroid 2-related factor 2 (Nrf2)-dependent pathways to upregulate epithelial tight junction proteins. Importantly, treatment with these compounds attenuated colitis in pre-clinical models by remedying barrier dysfunction in addition to anti-inflammatory activities. Cumulatively, the results highlight how microbial metabolites provide two-pronged beneficial activities at gut epithe-lium by enhancing barrier functions and reducing inflammation to protect from colonic diseases.
Article
Full-text available
Aging is accompanied by a gradual decline in both muscle mass and strength over time, which can eventually lead to pathologies, such as frailty and sarcopenia. While these two conditions are well characterized, further investigation of the early biological signs present in pre-frail elderly is still needed to help identify strategies for preventative therapeutic intervention. The goal of the present clinical study was to evaluate the level of mitochondrial (dys)function in a well-defined population of pre-frail elderly (>60 years of age). Pre-frail elderly were compared with an age-matched population of active elderly. Muscle mitochondrial function was assessed in vivo using phosphorus magnetic resonance spectroscopy (31P-MRS) and a comprehensive set of biological biomarkers were measured ex vivo in vastus lateralis muscle biopsies. In pre-frail subjects, phosphocreatine recovery was impaired and mitochondrial respiratory complex protein and activity levels were significantly lower when compared with active elderly. Analysis of microarray data showed that mitochondrial genes were also significantly down-regulated in muscle of pre-frail compared to active elderly. These results show that mitochondrial impairment is a hallmark of pre-frailty development and the onset of decline in muscle function in the elderly.
Article
Full-text available
Mitochondrial health is critical for skeletal muscle function and is improved by exercise training through both mitochondrial biogenesis and removal of damaged/dysfunctional mitochondria via mitophagy. The mechanisms underlying exercise-induced mitophagy have not been fully elucidated. Here, we show that acute treadmill running in mice causes mitochondrial oxidative stress at 3–12 h and mitophagy at 6 h post-exercise in skeletal muscle. These changes were monitored using a novel fluorescent reporter gene, pMitoTimer, that allows assessment of mitochondrial oxidative stress and mitophagy in vivo, and were preceded by increased phosphorylation of AMP activated protein kinase (Ampk) at tyrosine 172 and of unc-51 like autophagy activating kinase 1 (Ulk1) at serine 555. Using mice expressing dominant negative and constitutively active Ampk in skeletal muscle, we demonstrate that Ulk1 activation is dependent on Ampk. Furthermore, exercise-induced metabolic adaptation requires Ulk1. These findings provide direct evidence of exercise-induced mitophagy and demonstrate the importance of Ampk-Ulk1 signaling in skeletal muscle.
Article
Pyruvate kinase deficiency is a chronic hemolytic anemia caused by mutations in PK-R, a key glycolytic enzyme in erythrocytes. These 2 phase 1 randomized, placebo-controlled, double-blind healthy-volunteer studies assessed the safety, tolerability, and pharmacokinetics/pharmacodynamics of AG-348, a first-in-class allosteric PK-R activator. Twelve sequential cohorts were randomized 2:6 to receive oral placebo or AG-348, respectively, as a single dose (30-2500 mg) in the single-ascending-dose (SAD) study (ClinicalTrials.gov: NCT02108106) or 15-700 mg every 12 hours or 120 mg every 24 hours, for 14 days in the multiple-ascending-dose (MAD) study (ClinicalTrials.gov: NCT02149966). All 48 subjects completed the fasted SAD part; 44 of 48 completed the MAD (2 discontinued because of adverse events [AEs], 2 withdrew consent). The most common treatment-related AEs in AG-348-treated subjects were headache (16.7% [SAD] and 13.9% [MAD]) and nausea (13.9%, both studies). AE frequency increased at AG-348 doses ≥ 700 mg (SAD) and at 700 mg every 12 hours (MAD); 1 grade ≥ 3 AE occurred in the latter cohort. Pharmacokinetics were favorable with low variability. Dose-dependent changes in blood glycolytic intermediates consistent with glycolytic pathway activation were observed at all MAD doses, supporting future trials investigating the potential of AG-348 for treating PK deficiency or other anemias.
Article
More than a decade after a Nobel Prize was awarded for the discovery of the ubiquitin–proteasome system and clinical approval of proteasome and ubiquitin E3 ligase inhibitors, first-generation deubiquitylating enzyme (DUB) inhibitors are now approaching clinical trials. However, although our knowledge of the physiological and pathophysiological roles of DUBs has evolved tremendously, the clinical development of selective DUB inhibitors has been challenging. In this Review, we discuss these issues and highlight recent advances in our understanding of DUB enzymology and biology as well as technological improvements that have contributed to the current interest in DUBs as therapeutic targets in diseases ranging from oncology to neurodegeneration.
Article
Skeletal muscle is important for overall functionality and health. Aging is associated with an accumulation of damage to mitochondria DNA and proteins. In particular, damage to mitochondrial proteins in skeletal muscle, which is a loss of mitochondrial proteostasis, contributes to tissue dysfunction and negatively impacts systemic health. Therefore, understanding the mechanisms underlying the regulation of mitochondrial proteostasis and how those mechanisms change with age is important for the development of interventions to promote healthy aging. Herein, we examine how impairment in the selective degradation of damaged/dysfunctional mitochondria through mitophagy may play a central role in the loss of mitochondrial proteostasis in skeletal muscle aging as well as its broader implications for systemic health. Further, we explore how stimulating mitophagy through exercise may promote healthy aging.
Article
Urolithins are metabolites produced in the gut following consumption of ellagitannin and ellagic acid rich foods such as pomegranate, nuts and certain berries. Urolithin A (UA) is one of the predominant isoforms of urolithins in humans and has demonstrated interesting biological activities, suggesting potential benefits of direct consumption of UA. However, an evaluation of the safety of direct administration of UA has not yet been published. The aim of this study was to investigate for the first time the genotoxicity, toxicokinetics, and repeated dose safety of orally administered synthetic UA in rats. The battery of genotoxicity assays demonstrated that UA is not genotoxic. The ADME study showed that glucuronidated and sulfonated forms of UA are the predominant metabolites following both oral and i.v. administration. The 28-day (0, 0.175, 1.75, and 5.0% UA mixed in diet) and 90-day studies (0, 1.25, 2.5, and 5.0% UA mixed in diet) did not demonstrate any alterations in clinical parameters, blood chemistry, or hematology, nor indicate any target organs, nor any specific toxic mechanisms. The NOAEL was the highest dose tested, 5% UA by weight in the diet, or 3451 mg/kg bw/day in males and 3826 mg/kg bw/day in females in the 90-day oral study.
Article
Background: Much interest has been focused on interventions for treating sarcopenia; however, the effects have gained little evidence. Objective: To analyze the effectiveness of exercise, nutritional, drug, and combinational interventions for treating sarcopenia in older people. Method: We systematically searched MEDLINE via PubMed, the Cochrane Library of Cochrane Reviews and Cochrane Central Register of Controlled Trials, and Ichushi-Web for randomized controlled trials (RCTs) from January 2000 to December 2016. We have assessed the type of intervention, the cohort used, the way sarcopenia was diagnosed, the outcomes, and the quality of evidence. We meta-analyzed the outcomes with the net difference between-group treatment from baseline to the end of the study. Results: We screened a total of 2668 records and included seven RCTs that investigated the effects of exercise (4 RCTs), nutrition (5 RCTs), drug (1 RCT), and combination (4 RCTs) on muscle mass, strength, and function in older people with sarcopenia. Very low to low-quality evidence suggests that (1) exercise interventions may play a role in improving muscle mass, muscle strength, and walking speed in 3 months of intervention; (2) nutritional interventions may be effective in improving muscle strength in 3 months of intervention; (3) as drug intervention, selective androgen receptor modulator had no clear effect on muscle mass, strength, and physical function; and (4) a combined intervention of exercise and nutrition may have positive effects in improving the walking speed in 3 months of intervention. Conclusion: Our systematic review and meta-analysis showed some positive effects of exercise and nutritional interventions for treating sarcopenia in older people, although the quality of the evidence was low. Future high-quality RCTs should be implemented to strengthen the results.