Next Article in Journal
Stand Up to Stand Out: Natural Dietary Polyphenols Curcumin, Resveratrol, and Gossypol as Potential Therapeutic Candidates against Severe Acute Respiratory Syndrome Coronavirus 2 Infection
Next Article in Special Issue
The Role of One-Carbon Metabolism in Healthy Brain Aging
Previous Article in Journal
Antigingivitis and Antiplaque Effects of Oral Probiotic Containing the Streptococcus salivarius M18 Strain: A Randomized Clinical Trial
Previous Article in Special Issue
The Effects of 12 Weeks Colostrum Milk Supplementation on the Expression Levels of Pro-Inflammatory Mediators and Metabolic Changes among Older Adults: Findings from the Biomarkers and Untargeted Metabolomic Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Urolithins: A Prospective Alternative against Brain Aging

1
Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, Beijing 100048, China
2
College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
3
Rizhao Huawei Institute of Comprehensive Health Industries, Shandong Keepfit Biotech. Co., Ltd., Rizhao 276800, China
*
Author to whom correspondence should be addressed.
Nutrients 2023, 15(18), 3884; https://doi.org/10.3390/nu15183884
Submission received: 13 August 2023 / Revised: 1 September 2023 / Accepted: 2 September 2023 / Published: 6 September 2023
(This article belongs to the Special Issue Nutrition Interventions for Healthy Ageing)

Abstract

:
The impact of host–microbiome interactions on cognitive health and disease has received increasing attention. Microbial-derived metabolites produced in the gut are one of crucial mechanisms of the gut–brain axis interaction, showing attractive perspectives. Urolithins (Uros) are gut microbial-derived metabolites of ellagitannins and ellagic acid, whose biotransformation varies considerably between individuals and decreases greatly with age. Recently, accumulating evidence has suggested that Uros may have specific advantages in preventing brain aging including favorable blood–brain barrier permeability, selective brain distribution, and increasingly supporting data from preclinical and clinical studies. However, the usability of Uros in diagnosis, prevention, and treatment of neurodegenerative diseases remains elusive. In this review, we aim to present the comprehensive achievements of Uros in age-related brain dysfunctions and neurodegenerative diseases and discuss their prospects and knowledge gaps as functional food, drugs, or biomarkers against brain aging.

1. Introduction

To date, the population of dementia is approximately 45 million worldwide (this number will double if mild cognitive impairment (MCI) is included), and this population is expected to triple (approximately 130 million) by 2050 [1]. As the population of older adults is increasing, interest in the aging brain and the quest for “healthy brain aging” is growing. Modern medicine and nutrition evidence has confirmed that polyphenol consumption is an effective strategy in delaying brain aging [2]. However, the poor bioavailability of dietary polyphenols in vivo is contradictory to their health benefits [3,4]. Recently, there has been increasing evidence suggesting that the health benefits of polyphenols are mainly based on: (1) Polyphenols being either directly absorbed or in most cases converted into bioactive metabolites by gut microbiota and then get to the target tissues; (2) Polyphenols reshaping the gut microbiome during their bidirectional interaction. In addition, microbial-derived metabolites of polyphenols can be used as biomarkers to predict individual health status or even the efficacy of some drugs in vivo [5]. Over the past decade, research on gut microbiota and the gut–brain axis has brought increasingly new insights into humans [6]. Several pathways have been confirmed in the communication of the gut microbiota and the brain: (1) neuroendocrinology: the gut cells secrete a large amount of the signaling molecules in neuroendocrinology which can be influenced by gut microbiota; (2) neuroimmunity: the GI tract itself is the largest immune organ to stress response and the gut microbiota can promote immune cells to produce cytokines that can affect the brain; (3) neurotransmitters: the gut microbiota can affect the neurotransmitter secretion directly (several neurotransmitters, including acetylcholine, dopamine, noradrenaline, and serotonin can be synthesized by gut bacteria) and indirectly; and (4) microbial-derived metabolites: neuroactive metabolites mediate bidirectional interactions between the gut, gut microbiome, and the brain to modulate host neurophysiology and behavior [7,8,9].
As many of the host–microorganism interactions pertaining to human health are mediated by metabolites, interest in the microbial-derived metabolites has been increasing [10,11,12]. Among these active metabolites, urolithins (Uros), the gut bacterial metabolites of ellagitannins (ETs) and ellagic acid (EA), have shown to be beneficial in delaying many age-related diseases such as cancer (especially prostate, breast, and colorectal cancers), cardiovascular diseases, and chronic metabolic disorders (diabetes and hyperuricemia) [13,14,15,16,17,18]. Recently, growing evidence has supported the multiple health benefits of Uros against brain aging and the therapeutic potential for neurodegenerative diseases [17,19,20,21,22]. Moreover, Uros have been proposed as biomarkers of gut dysbiosis and disease stage in Parkinson’s patients [23]. Herein, we aim to review the recent achievements of Uros in brain aging and neurodegenerative diseases, with a particular emphasis on their therapeutic targets and mechanisms and discuss their prospects and knowledge gaps.

2. Overview and Advantage of Uros

Uros are gut metabolites derived from ETs and EA, which are richly available in many fruits (pomegranates, berries), nuts (walnuts), wood-aged wine, and some medicinal plants (galla chinensis, chebulae fructus, and seabuckthorn leaf) [12]. ETs and EA have very poor bioavailability in vivo and are converted into Uros by intestinal microbes in the colon, which are believed to be responsible for their biological activities [24,25]. ETs are a group of important dietary polyphenols and hydrolysable tannins that share a common core, hexahydroxydiphenoyl (HHDP), and differ in the number of monomer residues. ETs are mainly hydrolyzed to EA in the upper GI tract (stomach and small intestine), and most of the EA is converted to Uros in the lower GI tract (mainly in the colon) [25,26,27]. Uros are 6H-dibenzo [b, d] pyran-6-one derivatives which differ in their hydroxyl groups. The family includes the main end metabolites, dihydroxy derivatives UroA, Iso-UroA and monohydroxy derivative UroB, and other intermediates UroC, UroD, UroE, UroM-5, UroM-6, and UroM-7. Overall, UroA is the main metabolite produced in humans, which demonstrates the highest concentrations in plasma and urine, and remains high for days after the consumption of ET-rich food [28,29,30].
The bioconversion of Uros from ETs and EA varies considerably between individuals. Some individuals can produce high plasma concentrations of Uros (high Uro producers), while others cannot produce significant levels of Uros (low Uro producers). Additionally, Uro producers can also be classified into “metabotype A” (only high concentration of UroA is produced), and “metabotype B” (more Iso-UroA and/or UroB in comparison to UroA is produced) [31]. Therefore, the consumption of dietary ETs will produce different health benefits in high or low Uro producers, and those with different metabolic phenotypes. The different capacities for excreting Uros are mainly attributed to the variability of gut microbiota ecology, and vary with age, health status, dietary intake, obesity, and digestive organ surgery [32,33]. Notably, age is a key factor that affects the bioconversion of Uros. The bioconversion of Uros significantly decreases with aging. According to clinical observations, approximately 10% of individuals (aged 5–90) are non-Uro producers, and only 40% of elderly people (>60) can produce meaningful levels of Uros from dietary precursors [34]. Therefore, Uro supplementation may be a good alternative for certain individuals (e.g., elderly people) to meet the required healthy Uro level [35].
In comparison to precursors and live bacteria, Uros possess clear chemical structure, good bioavailability, and high safety in animals and humans [36,37]. Recently, two multi-center clinical trials showed that long-term (4-month) oral administration of UroA is safe and well tolerated [38,39]. Moreover, UroA is considered as GRAS (generally recognized as safe) for its use in food products by the Food and Drug Administration (FDA) [40]. Notably, Uros can also pass through the blood–brain barrier (BBB) and distribute in a brain-targeted manner after absorption [17,41,42], which may greatly facilitate their activities in the central nervous system (CNS), while other ET derivatives cannot cross the BBB. Additionally, as increasing evidence supporting the involvement of the gut metabolites in human health and diseases, metabolite-based treatment has been considered as a novel and promising therapeutic approach. It can overcome limitations and deficiencies of microbiome-based treatment (probiotics and prebiotics supplementation or fecal microbiome transplantation), such as colonization resistance and inter-individual variation in microbial composition [43]. Microbial-derived metabolites may be able to provide an improved efficacy (safety, stability, and individual variation) by exerting a beneficial host effect downstream of the microbiome. Collectively, these findings highlight the advantages of Uros in developing into functional food or drugs.
Many studies have investigated the biotransformations of ETs and EA to Uros. ETs are first converted into EA, facilitated by physiological pH and/or microbial enzymes such as tannin–hydrolase and lactonase. EA is then catalyzed by several microbial enzymes, including lactonase, decarboxylase, dehydroxylases, methyl esterases, and hydrolases, to produce Uros in the colon [25]. The metabolic pathways associated with these enzymes have been elucidated. Recently, several Uro-producing gut bacteria have been identified that can facilitate the conversion of EA to Uros in vitro [44,45,46]. Therefore, in addition to the chemical synthesis of Uros, the biotransformation of Uros with bacteria or enzymes in vitro in the near future can be expected [28,47].
As previously described, the consumption of dietary polyphenols is highly correlated with human health and the risk of neurodegenerative diseases [12]. Dynamic two-way communications between the gut, gut microbiome, and the CNS have been considered an increasingly vital factor for cognitive health and disease. Novel perspectives suggest that microbial-derived metabolites of polyphenols can be used as biomarkers to predict the risk of neurodegenerative diseases [48]. Therefore, in addition to the potential as functional food or drugs, Uros may be potential biomarkers of age-related cognitive decline and related disorders, which is mainly based on the following: (1) As bioactive metabolites, they exert multiple health benefits against brain aging and related diseases (which we will discuss below in detail); (2) the level of Uros in baseline is an indicator of personalized nutrition status (the consumption of dietary polyphenols in individuals, particularly for ETs and EA consumption), which is highly correlated with the risk of neurodegenerative diseases; (3) the level of Uros after consuming ET- or EA-rich food is an indicator of individual metabotypes associated with specific gut microbial ecologies (gut microbiota composition and functionality), which is tightly involved in neurodegenerative diseases. A recent study based on 52 patients and 117 healthy individuals showed that gut dysbiosis occurred during the onset and progression of Parkinson’s disease (PD) and the concentration of Uros in urine was highly associated with the severity of gut dysbiosis and PD in elderly people [23].

3. Preclinical and Clinical Studies

As UroA and UroB are the most abundant final metabolites in vivo, they are the most widely studied Uros across species, particularly UroA. Accumulating preclinical studies have demonstrated the beneficial effects of UroA and UroB on age-related brain dysfunctions. The cell studies in vitro are summarized in Table 1. Although these studies all demonstrated the neuroprotective effect of Uros in aging conditions such as oxidative stress, inflammation, and high concentrations of glucose, the treatment conditions in different laboratories and the sensitivity of different cells to Uros varied considerably. The animal studies in vivo are summarized in Table 2. Uros administration has been shown significantly improve cognition, learning, and memory in aged animals and animal models of neurodegenerative diseases.
It has been reported that ET- or EA-rich food consumption improve cognition and memory in the elderly (summarized in Table 3), whereas the effect of Uros supplementation in the elderly is still unknown. Two sequential randomized, double-blind, placebo-controlled trials in middle-aged and older adults (aged 50–75 years) with MCI showed that drinking 8 ounces (237 mL) of pomegranate juice every day for short-term 4 weeks or long-term 12 months significantly improved performance in the Brief Visuospatial Memory Test–Revised and Buschke Selective Reminding Test, and increased functional brain activation during verbal and visual memory tasks (ClinicalTrials.gov Identifier: NCT02093130) [42,49]. Moreover, cognition and memory improvement by pomegranate juice consumption is in line with the concentration of UroA–glucuronide in plasma and the plasma Trolox-equivalent antioxidant capacity. Additionally, berry and walnut consumption are related to better cognitive function and memory performance in the elderly with normal cognition or MCI [50,51,52,53]. Recently, several clinical trials have shown that UroA administration improves skeletal muscle function and mitochondrial health in elderly and middle-aged adults (ClinicalTrials.gov Identifier: NCT02655393, NCT03464500, and NCT03283462) [37,38,39]. In summary, evidence supporting the role of Uros in preventing the degeneration of brain during aging is accumulating, whereas clinical study in humans is lacking and needs further investigation.
Table 1. Preclinical studies of Uros on brain aging in vitro cell models.
Table 1. Preclinical studies of Uros on brain aging in vitro cell models.
UrosCellsPharmacological or Genetic
Interventions
Treatment
(Dosage and Time)
EffectsFindingsRefs.
UroANeuro-2a cellsH2O2 (250 μM)
for 45 min
0.5 μM, 1 μM,
2 μM, 4 μM
pretreatment for 24 h
antioxidation↑ cells viability, ↓ MAO-A and Tyrosinase, ↑ free radical (O2− and DPPH), ↓ ROS, lipid peroxidation,
↑ peroxiredoxins expression,
↑ CAT, SOD, GR, GSH-Px,
[54]
UroAPC12 cellsH2O2 (100 μM)
for 2 h
10 μg/mL, 30 μg/mL,
and 50 μg/mL
pretreatment for 24 h
antioxidation↑ cells viability, ↓ LDH release, ↓ apoptosis,
↓ caspase 3 and Bcl-2
[17]
UroASK-N-MC cellsH2O2 (300 μM)
for 18 h
1.25 μM, 2.5 μM,
and 5 μM
pretreatment for 6 h
antioxidation↑ cells viability, ↓ apoptosis, ↓ ROS, Bax/Bcl-2, PARP, cytochrome c, caspase 3/9, p38 MAPK[55]
UroASH-SY5Y cellsH2O2 (100 μM)
for 24 h
10 μM treatment
for 2 h, 6 h or 24 h
antioxidation↑ REDOX activity,
↓ cytotoxicity, ↓ ROS, ↓ apoptosis,
↓ caspase 3/8 and 9
[20]
UroASH-SY5Y cellsH2O2(200 μM)
for 6 h
5 μM, 7.5 μM,
10 μM, 15 μM
pretreatment for 12 h
antioxidation↑ cells viability, ↓ ROS, ↑ SOD, CAT, ↑ PKA/CREB/BDNF[56]
UroBNeuro-2a cellsH2O2 (250 μM)
for 2 h
20 μg/mL, 40 μg/mL,
and 60 μg/mL
pretreatment for 24 h
antioxidation↑ cells viability, ↓ ROS, ↓ apoptosis, cytotoxicity, ↓ caspase 3, ↑ Bcl-2[57]
UroBBV-2 cellsLPS (100 ng/mL) or LTA
(10 μg/mL)
or poly(I:C)
(25 μg/mL)
for 16 h
30 μM, 50 μM,
or 100 μM
pretreatment for 1 h
antioxidation,
anti-
inflamation
↓ NO, ROS, TNF-α,
IL-6, IL-1β, iNOS, COX-2, ↓ NF-κB, p-JNK, p-ERK, p-Akt, AP-1, ↑ IL-10, pAMPK, p47phox, gp91phox
[58]
UroA
mUroA
UroB
mUrOA
BV-2 cells,
SH-SY5Y cells
H2O2 (100 μM)
for 6 h;
LPS (1 μg/mL)
for 24 h
0.1 μM, 0.5 μM, 5 μM, 10 μM pretreatment for 1 h, 24 h, or 48 hantioxidation,
anti-
inflammation
↓ apoptosis, ↓ NO, TNF-α,NO, COX-2, IL-1, IL-6, PGE2, ↓ caspase 3/7 and 9, ↓ oxidative stress[19]
UroABV-2 cellsLPS (500 ng/mL) for 3 h, 12 h, 24 h; IL-4 (100 ng/mL),
IL-13 (10 ng/mL) for 24 h
10 μM
pretreatment for 12 h
anti-
inflammation
↓ IL-6, IL-1β, TNF-α, ↓ NOS,
↓ JNK/c-Jun,
↑ M2 microglia polarization
[59]
UroABV-2 cellsLPS (1 μg/mL) for 6 h, 12 h or 24 h2.5 μM, 5 μM, and 10μM pretreatment for 2 hanti-
inflammation
improved
mitochondrial function
↓ IL-1β, iNOS, COX-2, ↓ ROS, ↑ MMP, ↑ p62, ↓ LC3-II, ↑ Parkin, PINK, ↓ caspase 1, NLRP3, ↓ TOM20, Tim23,↑ mitophagy, ↑ OXPHOS[14]
UroA, UroB, UroC, mUroA
dmUroC
BV-2 cellsLPS
(100 ng/mL) for 30 min,
16 h or 24 h
3 μM, 10 μM,
30 μM
treatment for 30 min, 1 h, 16 h or 24 h
anti-
inflammation
↓ NO, TNF-α, IL-6, IL-1β, iNOS, COX-2, ↓ pAkt,↓ pERK1/2, p38 MAPK, ↓ NF-κB[60]
UroABV-2 cells,
ReNcell VM cells
LPS (100 ng/mL) for 1 h, 24 h,
or transfected with APPSwe
2 μM, 5 μM, 10 μM
treatment for 30 min, 6 h or 48 h
anti-
inflammation, anti-Aβ
↑ cells viability, ↓ NO, TNFα, IL-6, ↓ Aβ, ↑ SIRT1, ↓ NF-Κb,
↑ induction of autophagic flux
[21]
UroASH-SY5Y cells,
iPSC-ND cells
D-glucose 25 mM for 24 h, 48 h,
and 72 h,
(1–42) for 24 h, 48 h, and 72 h
100 nM
pretreatment for
30 min
anti-Aβ,
improved
mitochondrial function
↓ APP, BACE1, TGM2, Aβ(1–42), mitochondrial calcium influx, AhR, mtROS, ↓ LDH release[61]
UroASH-SY5Y cellstransfected with the APP 6951 µM,10 µM
treatment for
1 h or 24 h
improved
mitochondrial function
↑ MMP, ATP, ROS, OXPHOS, mitochondrial biogenesis[62]
UroAPC12 cells6-OHDA
(150 μM)
for 18 h or 24 h
2.5 μM, 5 μM,
10 mM treatment for 2 h
improved
mitochondrial function
↑ cells viability, MPP PGC-1α, SIRT1, TFAM, ↓ apoptosis, ↑ APP, ↓ ROS, ↓ mitochondria damage[63]
UroA
DHA+
LUT+ UroA
BE(2)-M17 cellsoligomeric Aβ1–42 (20 μM)
for 72 h
5 μM to 40 μM
pretreatment for 24 h, 5 μM (combination) pretreatment for 24 h
anti-Aβ↑ cells viability,
↓ LDH release
[64]
UroA, UroA+
EGCG
HT22 cellstransfected with APP cDNA for
24 h
no concentration mentioned, treatment for 24 himproved
mitochondrial function
↑ mitochondrial
respiration
[65]
UroA, UroA+
EGCG
HT22 cellstransfected with Tau cDNA for
24 h
1 μM or 10 mM
treatment for
24 h
improved
mitochondrial function
↓ Drp1 and Fis1,
↑ PGC-1α, Nrf1, Nrf2,
↑ TFAM, PINK1, Parkin,
↑ Mfn1, Mfn2, and Opa1
[66]
UroAHT22 cellstransfected with APP cDNA for
24 h
1 μM, 2 μM, 5 μM,
10 mM treatment
for 24 h
improved
mitochondrial function
↓ Drp1 and Fis1,
↑ PGC-1α, Nrf1, Nrf2,
↑ TFAM, PINK1, Parkin,
↑ Mfn1, Mfn2, and Opa1
[67]
Abbreviations: dmUroC: 8,9-dimethyl-O-Urolithin C, MAO: monoamine oxidase, DPPH: 1,1-diphenyl-2-picrylhydrazyl, ROS: reactive oxygen species, CAT: catalase, GR: glutathione reductase, GSH-Px: glutathione peroxidase, LDH: Lactic dehydrogenase, Bcl-2: B-cell lymphoma-2, Bax: Bcl-2-associated X, TNF-α: tumor necrosis factor-α, iNOS: inducible nitric oxide synthase, IL-6: interleukin-6, IL-1β: interleukin-1β, IL-1: interleukin-1, COX-2: cyclooxygenase-2, PARP: poly ADP-ribose polymerase, MAPK: mitogen-activated protein kinase, REDOX: mitochondrial oxidation-reduction, PKA: protein kinase A, CREB: cAMP-response element binding protein, BDNF: brain derived neurotrophic factor, NLRP3: negative regulation of NLR family pyrin domain containing 3, PGE2: prostaglandin E2, JNK: c-Jun N-terminal kinase, LC3-II: protein light chain 3-II, PINK1: PTEN induced kinase 1, MMP: mitochondrial membrane potential, OXPHOS: oxidative phosphorylation, APP: amyloid precursor protein, TGM2: transglutaminase type 2, PGC-1α: peroxisome proliferator-activated receptor-gamma coactivator-1-alpha, AhR: aryl hydrocarbon receptor, TFAM: transcription factor A, ERK1/2: extracellular signal-regulated kinase 1/2, SIRT1: silent information regulator of transcription 1, BACE1: β-site APP cleaving enzyme-1, poly(I:C): polyinosinic-polycytidylic acid, Drp1: dynamin-related protein 1, Fis1: fission mitochondrial 1; Nrf1: nuclear respiratory factor 1, Nrf2: nuclear respiratory factor 2, Mfn1: mitofusin, Mfn2: mitofusin 2, Opa1: optic atrophy 1.
Table 2. Preclinical studies of Uros on brain aging in vivo animal models.
Table 2. Preclinical studies of Uros on brain aging in vivo animal models.
UrosAnimalPharmacological or Genetic
Interventions
Route of AdministrationTreatment
(Dosage and Time)
EffectsFindingsRefs.
UroAmale
ICR mice
(4–6 weeks,
18–22 g)
D-gal
150 mg/kg/d s.c. for 8 weeks
i.g.50, 100, 150 mg/kg b.w./day
for 8 weeks
anti-brain
aging,
anti-
inflammation,
antioxidation
↑ spontaneous locomotion, object recognition learning, ↓ AchE, MAO, ↑ SOD, CAT, GSH-Px, ↓ p53/p21, TEAC, ↑ SIRT1, ↓ TNF-α, IL-1β, and IL-6, ↑ Bcl-2, ↓ caspase 3, mTOR, ↓ dysfunctional autophagy, astrocyte activation, ↓ apoptosis[17]
UroBmale C57BL/6 mice
(6–8 weeks,
18–22 g)
D-gal
150 mg/kg/d s.c. for 8 weeks
i.g.50, 100, 150 mg/kg b.w./day for 8 weeksanti-brain
aging,
anti-
inflammation, antioxidation
↓ cognitive deficits, ↑ pAkt, ↑ hippocampal LTP,↑ CAT, GSH-Px, TEAC, SOD, ↓ MDA, ↓ TNF-α, IL-6, IL-1β, AGEs, cytotoxicity, ↓ the activation of microglia and astrocytes, ↓ AchE, ↑ number of neuron, ↓ MAO[57]
UroAfemale APP/PS1 transgenic mice
(28 weeks)
transgenic AD micei.g. 300 mg/kg b.w./day for 14 daysanti-
inflammation
↓ spatial learning deficits, ↑ neurogenesis, ↓ neuronal apoptosis, ↓ reactive gliosis, ↓ Aβ, IL-1β, TNF-α, ↑ AMPK, ↓ p-P65, NF-κB, p-P38, MAPK, BACE1[22]
UroAAPP/PS1
transgenic mice
(13 months)
C.elegans
transgenic AD micei.g.200 mg/kg b.w./day for
1 month or
0.1 mM
(C.elegans)
anti-
inflammation
↑ learning and memory retention, ↑ OCR, ↓ ROS, ↓ Aβ1–42,
1–40, ↑ IL-10, ↓ autophagy,
↓ IL-6, TNF-α, ↓ NLRP3, IL-1β, ↓ p-tau, caspase 1
[68]
UroACX3CR1-
Cre mice
MPTP 15 mg/kg/d, i.p. 4 times a day every2 hi.g. 20 mg/kg b.w./day for 7 daysanti-
inflammation
↓ motor deficits, ↑ TH,
↓ caspase 1, NLRP3,
↓ astrogliosis
[14]
UroBMale ICR mice
(7 weeks, 2–37 g)
LPS
5 mg/kg/d, i.p.
i.p. 50 mg/kg b.w./day for4 daysanti-
inflammation
↓ microglia activation, ↓ NADPH, Akt, JNK, ERK, ↑ AMPK, HO-1[58]
UroA male C57BL/6J mice
(8–10 weeks)
6-OHDA
9 µg
i.p.10 mg/kg b.w./day for 7 daysimproved
mitochondria function
↓ neurotoxicity, mitochondria damage, OXPHOS, ↑ PGC-1α, TFAM, ↑ SIRT1[63]
UroAmiceSTZ 75 mg/kg/d i.p. for 3 daysi.p. 2.5 mg/kg b.w./day for 8 weeksanti-Aβ,
improved
mitochondria function
↓ APP, BACE1, p-tau, Aβ(1–42), TGM2[61]
UroA
UroB
mUroA
mUroB
C. eleganstransgenic AD
C. elegans (CL4176)
feeding 10 μg/mL pretreatment for 20 hanti-AβC. elegans survival and mobility[69]
UroA UroA + EGCGhAbKI mice
(3 months)
humanized
homozygous Aβ knockin (hAbKI)
AD mice
i.p.UroA 2.5 mg/kg b.w.,
EGCG 25 mg/kg b.w.,
3 times per week for 4 months
anti-Aβ,
improved
mitochondria function
↑ mitochondrial fusion, synaptic, ↓ Aβ(1–40) and Aβ(1–42), ↑ mitophagy, autophagy genes,
↓ mitochondrial fission genes, mitochondrial dysfunction, ↑ dendritic spines, ↓ fragmented mitochondria number, ↑ mitochondrial length, mitophagosomal formations
[65]
UroAmale C57BL/6J mice
(5 weeks,
18–22 g)
STZ 30 mg/kg b.w./day i.p. for 4 daysi.g. 200 mg/kg b.w.anti-brain
aging,
anti-
inflammation
↓ hyperglycemia, ↑ learning and memory, ↓ IL-6, IL-1β, TNF-α,
IL-1β, COX-2, iNOS-2,
↑ IL-10, ↓ NLRP3
[70]
Abbreviations: i.p.: intraperitoneal, s.c.: subcuta, i.g.: intragastrical, b.w.: body weight, AchE: acetylcholinesterase, MAO: monoamine oxidase, SOD: superoxide dismutase, CAT: catalase, GSH-Px: glutathione peroxidase, AGEs: advanced glycation end products, NADPH: triphosphopyridine nucleotide, TEAC: Trolox-equivalent antioxidant capacity, SIRT1: silent information regulator of transcription 1, TNF-α: tumor necrosis factor-α, iNOS: inducible nitric oxide synthase, IL-6: interleukin-6, IL-1β: interleukin-1β, IL-1: interleukin-1, COX-2: cyclooxygenase-2, Bcl-2: B-cell lymphoma-2, LTP: long-term potentiation, NLRP3: negative regulation of NLR family pyrin domain containing 3, MDA: malondialdehyde, BACE1: β-site APP cleaving enzyme-1, OCR: oxygen consumption rate, TH: tyrosine hydroxylase, HO-1: heme oxygenase 1, TFAM: transcription factor A, STZ: streptozotocin, TGM2: transglutaminase type 2.
Table 3. Clinical studies of Uro precursors on brain aging.
Table 3. Clinical studies of Uro precursors on brain aging.
SourceSubjectsClinical Trial
Procedure
Treatment
(Dosage and Time)
EffectsFundingsRefs
Pomegranate juice Age: 54–72 years;
Cognition and memory:
age-related memory decline; Other heath state: no neurological, psychiatric and major medical conditions
Randomized,
placebo
controlled,
double blind trial
Dosage:
240 mL/day of pomegranate juice (n = 15) or placebo drink (n = 13)
Time: 4 weeks
anti-age-related memory decline↑ fMRI activity during verbal and visual memory tasks, ↑ memory ability,
↑ plasma antioxidant status
[42]
Pomegranate juiceAge: 50–75 years;
Cognition and memory:
age-related memory
decline;
other heath state:
no cerebrovascular disease, neurological or physical illnesses associated with cognitive
deterioration
Randomized,
placebo
controlled,
double blind trial
Dosage:
236.5 mL/day of pomegranate juice (n = 98) or placebo drink
(n = 102)
Time: 48 weeks
anti-age-related memory decline↑ visual memory,
↑ visual learning and recall, ↑ verbal memory,
words recall
[49]
NutsAge: 55–80 years;
Cognition and memory: healthy;
other heath state: no diabetes, smoking, hypertension,
dyslipidemia, overweight and cardiovascular disease
Randomized,
placebo controlled
trial
Dosage:
MedDiet + EVOO 1 L/week (n = 224);
MedDiet + nuts 30 g/day (n = 166); or low-fat diet (n = 132)
Time: 6.5 years
anti-age-related memory decline↑ orientation to time and place, ↑ registration, attention and calculation, ↑ recall, language, and visual construction,
↑ visuospatial abilities, working memory, attention, ↑ abstract thinking, language comprehension
[71]
WalnutsAge: 63–79 years;
Cognition and memory: healthy;
other heath state: no
neurodegenerative disease, stroke, head trauma, brain surgery, psychiatric illness, depression, obesity, diabetes, hypertension and chemotherapy
Randomized
controlled trial
Dosage: Walnuts 30–60 g/day (n = 336) or control diet (abstention from walnuts)
(n = 321)
Time: 2 years
anti-age-related memory decline↑ global cognition and
perception
[51]
StrawberryAge: 60–75 years;
Cognition and memory: age-related motor and cognitive decline;
other heath state: BMI (18.5–29.9), no psychological or psychiatric disorders and chronic disease
Randomized,
placebo controlled,
double blind trial
Dosage:
Strawberry
24 g/day (n = 18) or placebo (n = 19)
Time: 45 or 90 days
anti-age-related memory decline↑ words recalled,
verbal learning
[72]
Mixture of
berries
Age: 50–70 years;
Cognition and memory: healthy;
other heath state:
no metabolic
disorders, food allergies and, gastrointestinal disorder
Randomized
cross-over trial
Dosage:
mixture of berries
(150 g blueberries, 50 g blackcurrant, 50 g elderberry,
50 g lingonberries, 50 g strawberry, and 100 g tomatoes/day) (n = 20);
or placebo drink
(n = 21)
Time: 5 weeks
anti-age-related memory decline↑ verbal working memory, ↑ selective attention,
↓ total- and LDL cholesterol,
↑ insulin concentrations
[73]
Grape and blueberry extractAge: 60–70 years;
Cognition and memory: age-related memory
decline;
other heath state:
BMI (20–30)
Randomized,
placebo
controlled,
double blind trial
Dosage: grape and blueberry extract 600 mg/day (n = 91)
or placebo
(n = 98)
Time: 6 months
anti-age-related memory decline↑ verbal episodic,
↑ recognition memory,
↑ working memory
[74]
Blueberry and blueberry extractAge: 65–80 years;
Cognition and memory: age-related memory
decline;
other heath state:
no metabolic
disorders and
diabetes
Randomized,
placebo
controlled,
double blind trial
Dosage: blueberry 500 mg/day
(n = 28); blueberry 1000 mg/day
(n = 29); blueberry extract 111 mg/day
(n = 28); or placebo
(n = 27)
Time: 6 months
anti-age-related memory decline↑ word recognition,
↑ total number of sequences correctly recalled, ↓ systolic blood pressure
[75]
BlueberryAge: >65 years;
Cognition and memory: age-related memory
decline
other heath state: healthy
Pilot,
single-blind,
one-arm trial
Dosage: blueberry
444 mL/day (weighing
54–64 kg);
532 mL/day (weighing
54–64 kg);
621 mL/day (weighing
77–91 kg); (n = 9)
Time: 12 weeks
anti-age-related memory decline; antidepressant↑ paired associate learning, ↑ word list recall,
↓ depressive symptoms,
↓ fasting glucose levels
[50]
BlueberryAge: <65 years;
Cognition and memory: healthy
other heath state: no
contraindications to fMRI
Randomized,
placebo
controlled,
double blind trial
Dosage: blueberry
30 mL/day
(n = 12) or
placebo drink
(n = 14)
Time: 12 weeks
anti-age-related memory decline↑ brain perfusion and activation, ↑ psychomotor function, visual processing, executive function, verbal and spatial memory[52]
BlueberryAge: 62–80 years;
Cognition and memory: age-related memory decline;
other heath state:
no diabetes,
kidney disease, liver disease, hematological coagulation disorder
Randomized,
parallel groups, placebo controlled,
double blind trial
Dosage: fish oil
(1.6 g EPA + 0.8 g DHA/day) (n = 15);
blueberry 25 g/day (n = 16); fish oil + blueberry 24 g/day (n = 17); or placebo (n = 17)
Time: 24 weeks
anti-age-related memory decline↑ psychomotor speed, working memory,
↑ lexical access,
↑ long-term memory
[53]
Frozen blueberryAge: 60–75 years;
Cognition and memory: healthy;
other heath state: BMI (18.5–29.9), no smoking or use of medications
Randomized,
placebo controlled,
double blind trial
Dosage:
frozen blueberry 24 g/day (n = 19) or placebo (n = 19)
Time: 90 days
anti-age-related memory decline↑ executive function,
↑ long-term memory, short term memory,
↑ spatial cognition, and attention
[76]
Frozen blueberryAge: 68–92 years;
Cognition and memory: age-related memory decline;
other heath state: no serious psychiatric disorder,
substance abuse, and claustrophobia
Randomized,
placebo controlled,
double blind trial
Dosage:
frozen blueberry
25 g/day (n = 8)
or placebo
(n = 8)
Time: 16 weeks
anti-age-related memory decline↑ working memory,
accuracy,
↑ blood oxygen level dependent activation
[77]
Frozen blueberryAge: >68 years;
Cognition and memory: age-
related memory decline;
other heath state: no dementia, serious psychiatric condition, substance abuse
Randomized,
placebo controlled,
double blind trial
Dosage:
frozen blueberry 24 g/day (n = 16) or placebo 20 g/day (n =21)
Time: 16 weeks
anti-age-related memory decline↑ lexical access for semantic information,
↑ speed of processing and working memory,
↑ verbal and nonverbal long-term memory
[78]
Abbreviations: fMRI: functional magnetic resonance imaging, BMI: body mass index, EVOO: extra-virgin olive oil, MedDiet: Mediterranean diet, LDL: low-density lipoprotein, EPA: eicosapentaenoic acid, DHA: docosahexaenoic acid.

4. Mechanisms of Action

The beneficial effects of Uros on brain function during aging are associated with multi-target actions that involve relieving chronic oxidative stress and inflammation, promoting mitophagy and mitochondrial function, inhibiting amyloid-β (Aβ) and tau pathology, and regulating tryptophan (Trp) metabolism. The mechanisms are summarized in Figure 1.

4.1. Antioxidant Activity in CNS

As the brain consumes more than one-fifth of the total oxygen, the oxidative stress exerted by reactive oxygen species (ROS) and related degeneration is particularly severe in the brain owing to aging. Damage to neural cells occurs when ROS production overwhelms the antioxidant defense mechanisms. Endogenous antioxidant defenses in the brain are relatively low compared to those in other vital organs [79,80]. This makes the protection of antioxidants, which can pass through the BBB, particularly important for the progression of brain aging. Although ETs and EA cannot cross the BBB, Uros are potent antioxidants with good BBB permeability [36,69].
First, UroA and UroB are evidenced as direct radical scavengers (details listed in Table 4) [54,57]. Furthermore, cell models have shown that UroA and UroB protected neuron-like cells from direct H2O2-induced damage, including PC12 cells, SK-N-MC cells, SH-SY5Y cells, and Neuro-2a cells, in which they effectively inhibited ROS formation and lipid peroxidation [17,20,54,55,57,81]. Moreover, Uros can boost endogenous antioxidant defenses in neuronal cells and brains of aged mice. Data from an array of studies have shown that UroA and UroB increased the activity of antioxidant enzymes, including catalase, superoxide dismutase, glutathione reductase, and glutathione peroxidase [58,82]. UroA increases the expression of cytoprotective peroxiredoxins 1 and 3 in Neuro-2a cells [54]. A study on UroB demonstrated that pretreatment with UroB significantly decreased the mRNA expression of nicotinamide adenine dinucleotide phosphate oxidase subunits (p47phox and gp91phox) and increased the antioxidant hemeoxygenase-1 expression via nuclear factor erythroid-2 related factor 2/antioxidant response element signaling in lipopolysaccharide (LPS)-treated BV2 cells [58].
Additionally, Uros are inhibitors of several oxidases (pro-oxidant enzymes) that can promote ROS formation. It is shown that Uros inhibited the activity of monoamine oxidase (MAO), which is responsible for the metabolism of monoamine neurotransmitters such as serotonin and dopamine [54,57,83]. A recent study demonstrated that Uros (A, B, and C) were selective inhibitors of human MAO-A (rather than MAO-B), and UroB was the strongest inhibitor with the IC50 of 0.88 ± 0.24 μM (the IC50 of UroA and UroC was 5.88 ± 0.69 μM and 29.6 ± 1.8 μM, respectively), whereas EA had no effect on MAO-A [83]. Clinically, MAO inhibitors may alleviate the symptoms of depression and Parkinson’s disease (PD). Therefore, these findings may suggest the potential benefits of Uros for related disorders. Saha et al. reported that UroA significantly inhibited heme peroxidase activity, including myeloperoxidase and lactoperoxidase in in vitro and in vivo models [84], which may provide a better understanding of the peroxidase inhibitory and anti-inflammatory activities of UroA.
The antioxidant effects of Uros are beyond the traditional antioxidant activities of their precursors. ETs and EA are typical antioxidants, which are mainly attributed to their potent free radical scavenging activity, including a wide variety of ROS and reactive nitrogen species [85,86]. As the bioavailability of ETs and EA is poor, the high antioxidant capacity of dietary ETs and EA may be important and restricted mostly to local actions in the GI tract [25,87]. González-Sarrías et al. suggested a lower neuroprotective activity of Uros against oxidative stress-induced cell death than that of their precursors [20,59]. It appears that Uros are not as potent antioxidants as their precursors, at least in vitro, and may systemically exert their antioxidant activity. Recently, we found that UroA (5, 10 μM) treatment significantly increased protein kinase A (PKA)/cAMP-response element binding protein (CREB)/brain derived neurotrophic factor (BDNF) neurotrophic signaling pathway in H2O2-treated SH-SY5Y cells, and pretreatment with PKA inhibitor H89 abolished the protective effects of UroA in H2O2-treated SH-SY5Y cells [56]. These results indicated that PKA/CREB/BDNF neurotrophic signaling pathway might involve the neuroprotective effect of UroA against oxidative stress.

4.2. Mitigation of Neuroinflammatioin

Chronic inflammation caused by innate immune cells such as glia is thought to play a key role in brain aging and related diseases. Glial activation and the release of inflammatory molecules such as proinflammatory cytokines are hallmarks of chronic inflammation in the brain. Mitigating neuroinflammation is one of the most important mechanisms underlying the health benefits of Uros.
It has been well documented that UroA and UroB significantly attenuated the inflammation induced by LPS in mouse microglia BV2 cells, including inhibiting the production of NO and the expression of inducible nitric oxide synthase (iNOS) and cyclo-oxygen-ase-2 (COX-2), regulating the levels of proinflammatory cytokines such as interleukin (IL)-6, IL-1β, tumor necrosis factor-α (TNF-α), and anti-inflammatory cytokines such as IL-10 [19,21,58,60,88]. In addition to LPS, Lee et al. reported that UroB significantly inhibited lipoteichoic acid (LTA)- and polyinosinic-polycytidylic acid (poly(I:C))-induced inflammation in BV2 cells, which are known as toll-like receptor (TLR) 2 and TLR3 agonists, respectively [58]. These results suggested that the anti-inflammatory effect of UroB is not confined to LPS stimulation. Moreover, UroA can induce M2 polarization from the M1 state in microglia and other macrophage-like cells which can subsequently inhibit pro-inflammatory cytokines generation and promote neuroprotection [59,89]. Animal studies have shown that the administration of UroA and UroB could significantly improve behavioral deficits and neuroinflammation in D-galactose-induced aged mice, transgenic APP/PS1 mice, LPS-brain-injected mice, and 1-methyl-4-pheny1-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mice, including decreasing the level of pro-inflammatory cytokines in the brain and inhibiting the activation of microglia and astrocytes in the hippocampus and cortex [14,17,22,57,58]. In addition, UroA was evidenced to reduce the expression and activity of the NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome in LPS-treated BV2 cells and mouse models (APP/PS1 mice and MPTP-treated mice) [30,68].
Multiple proteins and signaling pathways such as adenosine monophosphate (AMP)-dependent kinase (AMPK), mitogen-activated protein kinase (MAPK), nuclear factor-κB (NF-κB), and silent information regulator of transcription 1 (SIRT1) are considered to participate in the anti-inflammatory effects of Uros [58,68,88,89,90,91,92,93,94]. The NF-κB pathway is the best-studied inflammatory signaling pathway and is thought to be critical for the synthesis and effects of pro-inflammatory cytokines. An experiment in LPS-induced BV2 cells showed that UroA and UroB significantly inhibited NF-κB p65 expression and nuclear translocation in a dose-dependent manner and that UroB displayed more potent inhibitory activity than UroA [60]. AMPK, a central regulator of energy and metabolism involved in the pathophysiology of aging and age-related diseases, plays an important role in chronic inflammation. AMPK activation exerts anti-inflammatory effects by regulating cytokine synthesis and multiple inflammatory signaling pathways, such as NF-κB and MAPK pathways [95,96,97]. Moreover, AMPK signaling regulates NLRP3 inflammasome formation and activation during aging [98]. In addition to its established role in inflammation, AMPK activation has recently been implicated in promoting microglial M2 polarization, thereby relieving brain inflammation [99]. Experiments in cell and animal models have indicated that AMPK activation is tightly involved in the anti-inflammatory effects of Uros. In LPS-treated BV2 cells, UroB increases AMPK phosphorylation and decreases the phosphorylation of its downstream molecules Akt, JNK, and ERK [58,60]. In APP/PS1 mice, Zhuo et al. found that UroA treatment significantly upregulated AMPK signaling and downregulated NF-κB and MAPK signaling in the cortex and hippocampus, which was responsible for the improvement of memory and neuroinflammation [22]. SIRT1, a histone deacetylase, plays a key role in regulating neuroinflammation and the release of pro-inflammatory cytokines [100,101,102,103]. SIRT1 activation inhibits NF-κB signaling by involving deacetylation of the p65 subunit [104]. A study on UroA showed that treatment with UroA (5 and 10 μM) significantly increased the level (nuclear) and the activity (cell) of SIRT1 in BV2 cells, and the anti-inflammatory effect of UroA was abolished in the presence of EX527, a SIRT1 inhibitor, suggesting that the activation of SIRT1 is required for the anti-inflammatory effect of UroA [21].
Additionally, to compare the anti-inflammatory effect of UroA and its precursors, Ashley et al. investigated the difference between whole red raspberry polyphenols (RRW) and UroA in BV-2 cells under 3 h, 12 h, and 24 h inflammatory conditions (LPS and ATP treatment). The results demonstrated that RRW only inhibited the inflammation induced by 3 h of LPS stimulation, whereas UroA inhibited the inflammation caused by LPS treatments (3, 12, and 24 h). Moreover, the anti-inflammatory effects of RRW and UroA were both mediated by downregulation of the JNK/c-Jun pathway [59].

4.3. Promotion of Mitophagy and Mitochondrial Function

The brain possesses high mitochondrial activity to meet its relatively high energy demands. Mitochondrial dysfunction, including the abnormalities of mitochondrial bioenergetics, mitochondrial biogenesis, and autophagy in neurons, accumulate during aging. An increasing number of studies have indicated that the maintenance of mitochondrial homeostasis is particularly important for brain function in the elderly and may be a potential therapeutic strategy for age-related neurodegenerative diseases. Accumulating evidence suggests that UroA is an important regulator of mitochondrial homeostasis, particularly a robust mitophagy inducer, which seems to be one of the crucial mechanisms underlying its health benefits against aging [61,105]. Ryu et al. were the first to report that UroA extended the lifespan of C. elegans and improved muscle function in mice through mitophagy activation [106]. Recently, an increasing number of clinical trials have confirmed the impact of UroA on mitophagy in humans. Results from two clinical trials in elderly individuals demonstrated that supplementation with UroA at doses of 500 mg and 1000 mg daily for 4 weeks or 4 months significantly improved mitophagy and mitochondrial biogenesis in skeletal muscles [37]. Moreover, another recent clinical trial in middle-aged adults showed that supplementation with UroA increased mitophagy proteins and reduced inflammation [38]. Fang et al. conducted a systematic study on the promotion of mitophagy by UroA and found that mitophagy induction underlies the multiple actions of UroA in the brain, including the improvement of cognition and memory deficits, the alleviation of chronic neuroinflammation, and the inhibition of Aβ and tau pathology [68,107]. In vitro study demonstrated that UroA increased the levels of a series of mitophagy-related proteins, including phosphatase and tensin homolog deleted on chromosome ten induced kinase 1 (PINK1), Parkinson’s disease-related-1, beclin-1, Bcl-2-like protein 13, activating molecule in BECN1-regulated autophagy protein 1, and serine/threonine-protein kinase ULK1 (ULK1) in human neuronal SH-SY5Y cells. Moreover, UroA inhibited the phosphorylation of tau (p-tau) in a mitophagy-dependent manner, and the inhibitory effect was diminished in PINK1 or ULK1 knockdown cells. Consistently, in vivo studies showed that UroA activated neuronal mitophagy and reduced mtROS levels and mitochondrial damage in the hippocampus and prefrontal cortex of APP/PS1 mice, and mitophagy induction was also required for memory improvement of UroA in tau transgenic C. elegans and mice [68]. More recently, a study in humanized homozygous Aβ knockin (hAbKI) mice of late-onset AD demonstrated that UroA administration for 4 months significantly improved phenotypic behavior changes and mitochondrial dysfunction, including mitochondrial bioenergetics, biogenesis, and mitophagy [65].
Similar to neuronal cells, UroA promotes mitophagy and improves mitochondrial dysfunction in microglial cells. Treatment with the autophagy inhibitor 3-methyladenine abolished the effect of UroA on mitochondrial dysfunction and inflammation in LPS-treated BV2 cells [14,21]. Studies in animals demonstrated that mitophagy in microglial cells decreased by 60% in the hippocampus of AD mice, whereas UroA treatment normalized the decreased mitophagy, thereby increasing the phagocytic efficiency of microglia and the removal of Aβ plaques [68]. Similarly, UroA reduced the elevated expression and activity of NLRP3 and related neuroinflammation in AD mice by inducing mitophagy in the microglia. Moreover, using Atg5flox/flox:CX3CR1-Cre mice to block microglial mitophagy in vivo, the improvement in behavioral deficits and neuroinflammation induced by UroA in PD mice was diminished [14].
However, studies on the autophagy and mitophagy of UroA have also yielded conflicting results. Research conducted by Esselun et al. revealed that UroA did not affect mitophagy in SH-SY5Y-APP695 cells but moderately promoted mitochondrial biogenesis [62]. Another recent study indicated UroA induced robust neuronal mitophagy but did not increase neuronal macroautophagy in transgenic nematodes [68]. Additionally, Ahsan et al. demonstrated that UroA activated autophagy but not mitophagy in ischemia/reperfusion injured neuronal cells and mice [108].
Studies have indicated that UroA activated autophagy and mitophagy by regulating several key signaling molecules, including AMPK, peroxisome proliferator-activated receptor-gamma coactivator-1-alpha (PGC-1α), and SIRT, and inhibition of the mammalian target of rapamycin (mTOR). These signaling pathways stimulate mitophagy and mitochondrial biogenesis and are regulated by Uros. As discussed in the anti-inflammatory section, AMPK is one of the most important targets in aging and has been well documented in age-related mitochondrial dysfunction [109,110,111]. The downregulation of AMPK activity during aging impairs metabolic regulation, increases oxidative stress, and reduces autophagic clearance [98]. Activated AMPK increases PGC-1α level, which directly increases mitochondrial biogenesis [112]. The key autophagy/mitophagy effectors, such as SIRT, mTOR, and ULK1, are all downstream molecules of AMPK signaling [113,114,115]. Moreover, SIRT can deacetylate and activate liver kinase B1 (LKB1), an upstream kinase that activates AMPK [116]. Recent studies have demonstrated that Uros activate AMPK activity in a variety of cell types in vitro, including microglia, macrophages, and nucleus pulposus cells [22,58,117], as well as in multiple tissues in vivo, including the brain, muscle, and pancreas [92,118,119], indicating that Uros may be an AMPK activator. Therefore, AMPK may be a crucial target underlying the beneficial effects of Uros on aging, which requires further investigation.
Additionally, UroA activates SIRT1 and SIRT3. UroA stimulates SIRT3 promoter activity in Caco-2 cells [120] and increases ATP and NAD+ levels, leading to activation of the SIRT1-PGC-1α pathway in murine muscle [121]. Moreover, Chen et al. reported that UroA improved impaired autophagy in D-galactose-induced mice by upregulating SIRT1 signaling and downregulating mTOR signaling, and these effects appear to be mediated by the activation of miR-34a [17]. Treatment with both SIRT1 inhibitor EX527 and autophagy inhibitor chloroquine abolished the neuroprotective effect of UroA in APPSwe-transfected ReNcell VM human neural cells, as well as the anti-inflammatory effect of UroA in LPS-treated BV2 cells [21]. SIRT1 increases mitofusin 2 (Mfn2) and Parkin levels during mitophagy activation [122,123], and UroA has been shown to increase Mfn2 and Parkin expression in humans after 4 weeks of administration [37]. In addition, UroA activates autophagy by inhibiting endoplasmic reticulum (ER) stress in cell and mouse models of ischemic neuronal injury [62]. In summary, mitophagy deficiency and subsequent mitochondrial dysfunctions are early and pivotal events in brain aging and neurodegenerative diseases, and mitophagy or autophagy induction seems to be a crucial mechanism in the effects of Uros.

4.4. Inhibition of Aβ and Tau Pathology

Abnormal accumulation of Aβ and neurofibrillary tangles of p-tau protein are hallmark features of AD. It has been well documented that Aβ deposition in the brain occurs approximately two decades before the onset of the disease. Several studies have shown that Uros inhibited Aβ deposition and p-tau generation during brain aging [22,61,68,69]. The thioflavin T (ThT) binding assay in vitro showed that UroA, UroB, methyl-UroA (mUroA), and mUroB inhibited Aβ fibrillation including Aβ fibril content, β-sheet formation, and peptide oligomerization, whereas only mUroB reversed the neurotoxicity and muscular paralysis in the Aβ1–42-induced C. elegans model of AD [69]. A more recent study in vitro demonstrated that UroA inhibited Aβ1–42-induced toxicity in human neuroblastoma BE(2)-M17 cells [64]. In transgenic APP/PS1 AD mice, immunohistochemistry and ELIAS results demonstrated that UroA treatment significantly decreased Aβ1–40 and Aβ1–42 plaques in the cortex and hippocampus [22]. Moreover, UroA administration for 4 months significantly reduced the levels of Aβ1–40 and Aβ1–42, and improved inflammation, synaptic structure, and function in the brain of hAbKI AD mice [65]. In a further study conducted by Fang et al., UroA treatment restored memory impairment in both Aβ nematodes expressing pan-neuronal human Aβ1–42 (CL2355) protein and tau nematodes expressing pan-neuronal tau fragments (BR5270) [68]. Aβ plaque formation is a result of imbalance between Aβ clearance and its production from amyloid precursor protein (APP), and cleavage of APP by β-site APP cleaving enzyme-1 (BACE-1) is responsible for Aβ production. Although the precursors of Uros, such as EA and punicalagin, have been reported to inhibit the activity of BACE1 [124,125], UroA was shown to decrease only the expression of BACE1 in mice, and the effects of UroA on APP expression are inconsistent [61,62]. Additionally, microglia play a crucial role in the clearance of extracellular Aβ plaques [126,127]. In a study by Fang, UroA administration enhanced the removal of insoluble Aβ1–42 and Aβ1–40 proteins in the hippocampus of APP/PS1 mice by increasing the phagocytic efficiency of microglia, whereas no significant effect on the levels of APP cleavage intermediates was observed, suggesting that APP proteolysis may not be the target of UroA. Furthermore, the study also revealed that UroA treatment inhibited the phosphorylation of tau at Thr181, Ser202/Thr205, and Ser262 in tau-overexpressing human SH-SY5Y cells. Consistently, UroA administration inhibited p-tau and improved memory impairment in the 3×Tg AD mice [68]. More recently, Tu et al. showed that dual-specific tyrosine phosphorylation-regulated kinase 1A (DYRK1A) is the main target of UroA in its anti-AD effect. UroA significantly inhibited the activity of DYRK1A, which led to the de-phosphorylation of tau and further stabilized microtubule polymerization [128].
Hyperglycemia and diabetes are risk factors for neurodegeneration and critical contributors to amyloidogenesis. Uros were shown to improve diabetes-associated neurodegeneration. Lee et al. systematically studied and reported that UroA attenuated the amyloidogenesis and neurodegeneration in a diabetes model. Their experiments in vitro showed that pretreatment with UroA significantly decreased high glucose-induced Aβ1–42 formation, as well as APP and BACE1 expression in both SH-SY5Y cells and human iPSC-derived neuronal differentiated cells. Furthermore, the experiments in vivo showed that UroA injection at dose of 2.5 mg/kg for 8 weeks improved cognitive impairment in streptozotocin-induced diabetic mice and decreased the levels of Aβ1–42, APP, BACE1, and p-tau (Ser262 and Ser396) in the prefrontal cortex and hippocampus [61]. In addition, Chen et al. reported that UroB supplementation improved learning and memory impairment by inhibiting the accumulation of advanced glycation end products in the brain of aged mice [57]. Xiao et al. found that UroA relieved diabetes-associated cognitive impairment and neuroinflammation by alleviating intestinal barrier dysfunction via N-glycan biosynthesis pathway [70].

4.5. Regulation of Trp Metabolism

Recent studies have indicated that the regulation of Trp metabolism may be a novel mechanism underlying the health benefits of UroA against brain aging. Trp metabolism is an important communication strategy in the “microbiome–gut–brain” axis, in which both host and gut microbiota are involved. Trp metabolites can serve as neurotransmitters and signaling molecules in CNS. Although it is well known that Trp is used for the synthesis of serotonin, a large majority of Trp (>95%) is metabolized by the kynurenine (Kyn) pathway, thus producing bioactive metabolites with distinct activities in CNS. In humans, the activity of Kyn pathway increases with age [129]. Several lines of evidence suggest that the imbalance in Trp metabolism is associated with various neurodegenerative and neurological diseases [130,131]. Trp metabolites can control the pathogenic activities of microglia and astrocytes via aryl hydrocarbon receptors (AhRs), and further inhibit neuroinflammation and neurodegeneration [132,133]. A recent study showed that oral administration of EA and UroA for 8 weeks significantly regulated the microbial composition and Trp metabolism in DBA/2J mice fed with a high-fat and high-sucrose diet. Both UroA and EA supplementation reduced the Kyn pathway, and UroA significantly decreased the level of indole sulfate in serum [134]. Additionally, UroA elevated Trp hydroxylase-2 transcription, the rate-limiting enzyme in Trp metabolism to serotonin, and subsequently increased serotonin production in differentiated rat serotonergic raphe cells [135]. Therefore, the regulation of UroA on Trp metabolism may be an important mechanism underlying its beneficial effect, but this has not been clarified and deserves further research.

4.6. Others

Uros have been evidenced to exert neuroprotection by direct action on estrogen receptors (ERs) [136,137] and AhRs [138] in the brain. They are considered as “enterophytoestrogens”, which are microflora-derived metabolites with estrogenic and/or antiestrogenic activities [139,140]. Furthermore, UroA has demonstrated high selectivity on ERα and has recently been reported to regulate ERα-dependent gene expression in endometrial cancer cells [141]. AhRs are ligand-activated transcription factors involved in multiple physiological and pathological processes. In addition to indirectly regulating AhRs through Trp metabolites, UroA has been evidenced as a human-selective AhR antagonist [142,143], which is considered to mediate its anti-inflammatory activity. Additionally, UroA can alleviate BBB dysfunction [144].
Although these studies have clarified the effects of Uros from different aspects, they are relatively isolated from each other. The key targets and intracellular signaling pathways underlying the effects of Uros on aging brain remain to be elucidated. Recently, pharmaceutical studies have been conducted on Uros. Several natural and synthetic Uro analogs exhibit inhibitory effects on various enzymes, including cholinesterases, MAO-B, and cyclooxygenases [145,146]. Therefore, Uros may be multi-target agents against neurodegenerative and neurological diseases.

5. Knowledge Gaps

First, clinical evidence for Uros or UroA on age-related cognitive decline or neurodegenerative diseases is still unclear. UroA has been designated as GRAS by FDA in 2018 and sold as an ingredient of anti-aging products in USA. Growing clinical evidence has demonstrated that precursors of Uros including pomegranate, blueberry, and walnut supplementation significantly improved age-related brain dysfunctions (Table 3). The specific advantages of UroA in preventing brain aging is mainly based on (i) good safety and tolerance, (ii) favorable blood–brain barrier permeability and selective brain distribution, (iii) the accumulating evidence from preclinical studies, (iv) the different capacities for producing Uros between individuals and the decreased capacities in the elderly, and (v) overcoming limitations and deficiencies of microbiome-based treatment. Therefore, clinical trials of UroA supplementation on cognitive and memory decline in the elderly should be carried out as soon as possible, which seem to be the crucial obstacle on the way to its application.
Second, the pharmacokinetic features of UroA (pharmacokinetic research of Uros is mainly focused on UroA) in the brain have not been clarified. UroA is easily absorbed after oral administration and mainly metabolized to UroA glucuronide, UroA sulfate, and mUroA. Studies in rodents have shown that UroA and mUroA are detectable in the brain after administration of UroA or pomegranate juice [147]. The Uro metabolites identified in the brain are summarized in Table 5. A recent study depicted a more detailed profile of UroA in the brain, in which the concentration of UroA increased slowly after 0–3 h of oral administration (UroA, 200 mg/kg) and reached the maximum concentration (Cmax) at approximately 28 ng/g and 35 ng/g in the cortex and hippocampus, respectively, after 4 h (Tmax) of oral administration. In contrast, the Cmax and Tmax of UroA in plasma were 15 ng/mL and 2 h, respectively [148]. This study is the first to report that UroA was detectable in the specific brain regions (cortex and hippocampus) and remained at high levels for hours after a single oral administration. However, more detailed pharmacokinetic parameters should be determined and investigated.
Third, the therapeutic targets and molecular mechanisms underlying the effects of UroA against brain aging remain to be elucidated. Although UroA has been found to activate anti-aging signaling pathways such as AMPK, SIRTs, and mTOR in vivo, the direct actions of UroA on these targets are currently unclear.
Fourth, the biotransformation of Uros in vitro is essential for their further application. Although there are anti-aging products containing high purity UroA of chemical synthesis on sale, biotransformation may greatly improve producing efficiency. This process is a two-step bioconversion. In the first step, the conversion of ETs to EA has been clarified and achieved in vitro [150]. Elucidating the second step and achieving the transformation of EA into Uros in vitro could yield promising application.
Fifth, it has been reported that UroA can attenuate diabetes-associated cognitive impairment by ameliorating intestinal barrier dysfunction [70]. However, the impact of UroA supplementation on gut microbiota ecology is unclear. The role of UroA supplementation on the gut microbiota composition and other neuroactive metabolites, e.g., SCFAs need further investigation.
Last, recent studies in animals and cells indicated that the combination of UroA with other food functional factors such as docosahexaenoic acid (DHA) and egpigallocatechin gallate (EGCG) produced significantly synergistic effects against brain aging and AD [64,65]. The combined treatments may have referential value for UroA to develop into related functional food and drugs. However, the synergistic efficacy and mechanisms warrant further elaboration.

Author Contributions

Conceptualization: L.A.; Investigation: L.A. and Q.L.; Formal analysis: L.A.; Writing original draft: L.A.; Review and editing: L.A., Q.L. and K.W.; Visualization: L.A.; Supervision: Y.W.; Project administration: Y.W.; Funding acquisition: Y.W. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Natural Science Foundation of China (31972127; 31471626), the Science and Technology Program of the Beijing Municipal Education Commission (KZ201910011013), and the Natural Science Foundation of Rizhao (202117).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created.

Acknowledgments

We would like to show our deepest gratitude to all persons who have made substantial contributions to the work reported in the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Livingston, G.; Huntley, J.; Sommerlad, A.; Ames, D.; Ballard, C.; Banerjee, S.; Brayne, C.; Burns, A.; Cohen-Mansfield, J.; Cooper, C.; et al. Dementia prevention, intervention, and care: 2020 report of the Lancet Commission. Lancet 2020, 396, 413–446. [Google Scholar] [CrossRef] [PubMed]
  2. Wahl, D.; Cogger, V.C.; Solon-Biet, S.M.; Waern, R.V.; Gokarn, R.; Pulpitel, T.; Cabo, R.; Mattson, M.P.; Raubenheimer, D.; Simpson, S.J.; et al. Nutritional strategies to optimise cognitive function in the aging brain. Ageing Res. Rev. 2016, 31, 80–92. [Google Scholar] [CrossRef]
  3. D’Archivio, M.; Filesi, C.; Varì, R.; Scazzocchio, B.; Masella, R. Bioavailability of the polyphenols: Status and controversies. Int. J. Mol. Sci. 2010, 11, 1321–1342. [Google Scholar] [CrossRef] [PubMed]
  4. Walle, T.; Hsieh, F.; DeLegge, M.H.; Oatis, J.E., Jr.; Walle, U.K. High absorption but very low bioavailability of oral resveratrol in humans. Drug Metab. Dispos. Biol. Fate Chem. 2004, 32, 1377–1382. [Google Scholar] [CrossRef] [PubMed]
  5. Gentile, C.L.; Weir, T.L. The gut microbiota at the intersection of diet and human health. Science 2018, 362, 776–780. [Google Scholar] [CrossRef]
  6. Pistollato, F.; Cano, S.S.; Elio, I.; Vergara, M.M.; Giampieri, F.; Battino, M. Role of gut microbiota and nutrients in amyloid formation and pathogenesis of Alzheimer disease. Nutr. Rev. 2016, 74, 624–634. [Google Scholar] [CrossRef] [PubMed]
  7. Cryan, J.F.; O’Riordan, K.J.; Cowan, C.S.M.; Sandhu, K.V.; Bastiaanssen, T.F.S.; Boehme, M.; Codagnone, M.G.; Cussotto, S.; Fulling, C.; Golubeva, A.V.; et al. The Microbiota-Gut-Brain Axis. Physiol. Rev. 2019, 99, 1877–2013. [Google Scholar] [CrossRef] [PubMed]
  8. Megur, A.; Baltriukienė, D.; Bukelskienė, V.; Burokas, A. The Microbiota-Gut-Brain Axis and Alzheimer’s Disease: Neuroinflammation Is to Blame? Nutrients 2020, 13, 37. [Google Scholar] [CrossRef]
  9. Jie, Z.; Xia, H.; Zhong, S.L.; Feng, Q.; Li, S.; Liang, S.; Zhong, H.; Liu, Z.; Gao, Y.; Zhao, H.; et al. The gut microbiome in atherosclerotic cardiovascular disease. Nat. Commun. 2017, 8, 845. [Google Scholar] [CrossRef]
  10. Luca, S.V.; Macovei, I.; Bujor, A.; Miron, A.; Skalicka-Woźniak, K.; Aprotosoaie, A.C.; Trifan, A. Bioactivity of dietary polyphenols: The role of metabolites. Crit. Rev. Food Sci. Nutr. 2020, 60, 626–659. [Google Scholar] [CrossRef]
  11. Reddy, V.P.; Aryal, P.; Robinson, S.; Rafiu, R.; Obrenovich, M.; Perry, G. Polyphenols in Alzheimer’s Disease and in the Gut-Brain Axis. Microorganisms 2020, 8, 199. [Google Scholar] [CrossRef]
  12. Aravind, S.M.; Wichienchot, S.; Tsao, R.; Ramakrishnan, S.; Chakkaravarthi, S. Role of dietary polyphenols on gut microbiota, their metabolites and health benefits. Food Res. Int. 2021, 142, 110189. [Google Scholar] [CrossRef]
  13. Hasheminezhad, S.H.; Boozari, M.; Iranshahi, M.; Yazarlu, O.; Sahebkar, A.; Hasanpour, M.; Iranshahy, M. A mechanistic insight into the biological activities of urolithins as gut microbial metabolites of ellagitannins. Phytother. Res. 2022, 36, 112–146. [Google Scholar] [CrossRef]
  14. Qiu, J.; Chen, Y.; Zhuo, J.; Zhang, L.; Liu, J.; Wang, B.; Sun, D.; Yu, S.; Lou, H. Urolithin A promotes mitophagy and suppresses NLRP3 inflammasome activation in lipopolysaccharide-induced BV2 microglial cells and MPTP-induced Parkinson’s disease model. Neuropharmacology 2022, 207, 108963. [Google Scholar] [CrossRef]
  15. D’Amico, D.; Andreux, P.A.; Valdes, P.; Singh, A.; Rinsch, C.; Auwerx, J. Impact of the Natural Compound Urolithin A on Health, Disease, and Aging. Trends Mol. Med. 2021, 27, 687–699. [Google Scholar] [CrossRef]
  16. Adachi, S.-i.; Sasaki, K.; Kondo, S.; Komatsu, W.; Yoshizawa, F.; Isoda, H.; Yagasaki, K. Antihyperuricemic Effect of Urolithin A in Cultured Hepatocytes and Model Mice. Molecules 2020, 25, 5136. [Google Scholar] [CrossRef]
  17. Chen, P.; Chen, F.; Lei, J.; Li, Q.; Zhou, B. Activation of the miR-34a-Mediated SIRT1/mTOR Signaling Pathway by Urolithin A Attenuates d-Galactose-Induced Brain Aging in Mice. Neurotherapeutics 2019, 16, 1269–1282. [Google Scholar] [CrossRef]
  18. Lv, M.Y.; Shi, C.J.; Pan, F.F.; Shao, J.; Feng, L.; Chen, G.; Ou, C.; Zhang, J.F.; Fu, W.M. Urolithin B suppresses tumor growth in hepatocellular carcinoma through inducing the inactivation of Wnt/β-catenin signaling. J. Cell. Biochem. 2019, 120, 17273–17282. [Google Scholar] [CrossRef]
  19. DaSilva, N.A.; Nahar, P.P.; Ma, H.; Eid, A.; Wei, Z.; Meschwitz, S.; Zawia, N.H.; Slitt, A.L.; Seeram, N.P. Pomegranate ellagitannin-gut microbial-derived metabolites, urolithins, inhibit neuroinflammation in vitro. Nutr. Neurosci. 2019, 22, 185–195. [Google Scholar] [CrossRef]
  20. Gonzalez-Sarrias, A.; Angeles Nunez-Sanchez, M.; Tomas-Barberan, F.A.; Carlos Espin, J. Neuroprotective Effects of Bioavailable Polyphenol-Derived Metabolites against Oxidative Stress-Induced Cytotoxicity in Human Neuroblastoma SH-SY5Y Cells. J. Agric. Food Chem. 2017, 65, 752–758. [Google Scholar] [CrossRef]
  21. Velagapudi, R.; Lepiarz, I.; El-Bakoush, A.; Katola, F.O.; Bhatia, H.; Fiebich, B.L.; Olajide, O.A. Induction of autophagy and activation of SIRT-1 deacetylation mechanisms mediate neuroprotection by the pomegranate metabolite urolithin A in BV2 microglia and differentiated 3D human neural progenitor cells. Mol. Nutr. Food Res. 2019, 63, 1801237. [Google Scholar] [CrossRef]
  22. Gong, Z.; Huang, J.; Xu, B.; Ou, Z.; Zhang, L.; Lin, X.; Ye, X.; Kong, X.; Long, D.; Sun, X.; et al. Urolithin A attenuates memory impairment and neuroinflammation in APP/PS1 mice. J. Neuroinflammation 2019, 16, 62. [Google Scholar] [CrossRef]
  23. Romo-Vaquero, M.; Fernández-Villalba, E.; Gil-Martinez, A.L.; Cuenca-Bermejo, L.; Espín, J.C.; Herrero, M.T.; Selma, M.V. Urolithins: Potential biomarkers of gut dysbiosis and disease stage in Parkinson’s patients. Food Funct. 2022, 13, 6306–6316. [Google Scholar] [CrossRef]
  24. Sanchez-Gonzalez, C.; Ciudad, C.J.; Noe, V.; Izquierdo-Pulido, M. Health benefits of walnut polyphenols: An exploration beyond their lipid profile. Crit. Rev. Food Sci. Nutr. 2017, 57, 3373–3383. [Google Scholar] [CrossRef]
  25. Garcia-Munoz, C.; Vaillant, F. Metabolic Fate of Ellagitannins: Implications for Health, and Research Perspectives for Innovative Functional Foods. Crit. Rev. Food Sci. Nutr. 2014, 54, 1584–1598. [Google Scholar] [CrossRef]
  26. Gonzalez-Barrio, R.; Edwards, C.A.; Crozier, A. Colonic Catabolism of Ellagitannins, Ellagic Acid, and Raspberry Anthocyanins: In Vivo and In Vitro Studies. Drug Metab. Dispos. 2011, 39, 1680–1688. [Google Scholar] [CrossRef]
  27. Tomas-Barberan, F.A.; Garcia-Villalba, R.; Gonzalez-Sarrias, A.; Selma, M.V.; Espin, J.C. Ellagic Acid Metabolism by Human Gut Microbiota: Consistent Observation of Three Urolithin Phenotypes in Intervention Trials, Independent of Food Source, Age, and Health Status. J. Agric. Food Chem. 2014, 62, 6535–6538. [Google Scholar] [CrossRef]
  28. Piwowarski, J.P.; Granica, S.; Stefanska, J.; Kisst, A.K. Differences in Metabolism of Ellagitannins by Human Gut Microbiota Ex Vivo Cultures. J. Nat. Prod. 2016, 79, 3022–3030. [Google Scholar] [CrossRef]
  29. Nunez-Sanchez, M.A.; Garcia-Villalba, R.; Monedero-Saiz, T.; Garcia-Talavera, N.V.; Gomez-Sanchez, M.B.; Sanchez-Alvarez, C.; Garcia-Albert, A.M.; Rodriguez-Gil, F.J.; Ruiz-Marin, M.; Pastor-Quirante, F.A.; et al. Targeted metabolic profiling of pomegranate polyphenols and urolithins in plasma, urine and colon tissues from colorectal cancer patients. Mol. Nutr. Food Res. 2014, 58, 1199–1211. [Google Scholar] [CrossRef]
  30. Cerdá, B.; Espín, J.C.; Parra, S.; Martínez, P.; Tomás-Barberán, F.A. The potent in vitro antioxidant ellagitannins from pomegranate juice are metabolised into bioavailable but poor antioxidant hydroxy-6H-dibenzopyran-6-one derivatives by the colonic microflora of healthy humans. Eur. J. Nutr. 2004, 43, 205–220. [Google Scholar] [CrossRef]
  31. Tomas-Barberan, F.A.; Gonzalez-Sarrias, A.; Garcia-Villalba, R.; Nunez-Sanchez, M.A.; Selma, M.V.; Garcia-Conesa, M.T.; Espin, J.C. 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. 2017, 61. [Google Scholar] [CrossRef] [PubMed]
  32. Selma, M.V.; Gonzalez-Sarrias, A.; Salas-Salvado, J.; Andres-Lacueva, C.; Alasalvar, C.; Orem, A.; Tomas-Barberan, F.A.; Espin, J.C. The gut microbiota metabolism of pomegranate or walnut ellagitannins yields two urolithin-metabotypes that correlate with cardiometabolic risk biomarkers: Comparison between normoweight, overweight-obesity and metabolic syndrome. Clin. Nutr. 2018, 37, 897–905. [Google Scholar] [CrossRef] [PubMed]
  33. Zhang, M.; Cui, S.; Mao, B.; Zhang, Q.; Zhao, J.; Zhang, H.; Tang, X.; Chen, W. Ellagic acid and intestinal microflora metabolite urolithin A: A review on its sources, metabolic distribution, health benefits, and biotransformation. Crit. Rev. Food Sci. Nutr. 2022; online ahead of print. [Google Scholar] [CrossRef]
  34. Cortes-Martin, A.; Garcia-Villalba, R.; Gonzalez-Sarrias, A.; Romo-Vaquero, M.; Loria-Kohen, V.; Ramirez-de-Molina, A.; Tomas-Barberan, F.A.; Selma, M.V.; Espin, J.C. The gut microbiota urolithin metabotypes revisited: The human metabolism of ellagic acid is mainly determined by aging. Food Funct. 2018, 9, 4100–4106. [Google Scholar] [CrossRef] [PubMed]
  35. Singh, A.; D’Amico, D.; Andreux, P.A.; Dunngalvin, G.; Kern, T.; Blanco-Bose, W.; Auwerx, J.; Aebischer, P.; Rinsch, C. Direct supplementation with Urolithin A overcomes limitations of dietary exposure and gut microbiome variability in healthy adults to achieve consistent levels across the population. Eur. J. Clin. Nutr. 2022, 76, 297–308. [Google Scholar] [CrossRef] [PubMed]
  36. Kujawska, M.; Jourdes, M.; Kurpik, M.; Szulc, M.; Szaefer, H.; Chmielarz, P.; Kreiner, G.; Krajka-Kuzniak, V.; Mikolajczak, P.L.; Teissedre, P.-L.; et al. Neuroprotective Effects of Pomegranate Juice against Parkinson’s Disease and Presence of Ellagitannins-Derived Metabolite-Urolithin A-In the Brain. Int. J. Mol. Sci. 2020, 21, 202. [Google Scholar] [CrossRef] [PubMed]
  37. Andreux, P.A.; Blanco-Bose, W.; Ryu, D.; Burdet, F.; Ibberson, M.; Aebischer, P.; Auwerx, J.; Singh, A.; Rinsch, C. The mitophagy activator urolithin A is safe and induces a molecular signature of improved mitochondrial and cellular health in humans. Nat. Metab. 2019, 1, 595–603. [Google Scholar] [CrossRef]
  38. Singh, A.; D’Amico, D.; Andreux, P.A.; Fouassier, A.M.; Blanco-Bose, W.; Evans, M.; Aebischer, P.; Auwerx, J.; Rinsch, C. Urolithin A improves muscle strength, exercise performance, and biomarkers of mitochondrial health in a randomized trial in middle-aged adults. Cell Rep. Med. 2022, 3, 100633. [Google Scholar] [CrossRef]
  39. Liu, S.; D’Amico, D.; Shankland, E.; Bhayana, S.; Garcia, J.M.; Aebischer, P.; Rinsch, C.; Singh, A.; Marcinek, D.J. Effect of Urolithin A Supplementation on Muscle Endurance and Mitochondrial Health in Older Adults A Randomized Clinical Trial. JAMA Netw. Open 2022, 5, e2144279. [Google Scholar] [CrossRef]
  40. García-Villalba, R.; Giménez-Bastida, J.A.; Cortés-Martín, A.; Ávila-Gálvez, M.; Tomás-Barberán, F.A.; Selma, M.V.; Espín, J.C.; González-Sarrías, A. Urolithins: A Comprehensive Update on their Metabolism, Bioactivity, and Associated Gut Microbiota. Mol. Nutr. Food Res. 2022, 66, e2101019. [Google Scholar] [CrossRef]
  41. Gasperotti, M.; Passamonti, S.; Tramer, F.; Masuero, D.; Guella, G.; Mattivi, F.; Vrhovsek, U. Fate of microbial metabolites of dietary polyphenols in rats: Is the brain their target destination? ACS Chem. Neurosci. 2015, 6, 1341–1352. [Google Scholar] [CrossRef]
  42. Bookheimer, S.Y.; Renner, B.A.; Ekstrom, A.; Li, Z.; Henning, S.M.; Brown, J.A.; Jones, M.; Moody, T.; Small, G.W. Pomegranate juice augments memory and FMRI activity in middle-aged and older adults with mild memory complaints. Evid.-Based Complement. Altern. Med. 2013, 2013, 946298. [Google Scholar] [CrossRef] [PubMed]
  43. Suez, J.; Elinav, E. The path towards microbiome-based metabolite treatment. Nat. Microbiol. 2017, 2, 17075. [Google Scholar] [CrossRef]
  44. Gaya, P.; Peiroten, A.; Medina, M.; Alvarez, I.; Landete, J.M. Bifidobacterium pseudocatenulatum INIA P815: The first bacterium able to produce urolithins A and B from ellagic acid. J. Funct. Foods 2018, 45, 95–99. [Google Scholar] [CrossRef]
  45. Selma, M.V.; Beltran, D.; Garcia-Villalba, R.; Espin, J.C.; Tomas-Barberan, F.A. Description of urolithin production capacity from ellagic acid of two human intestinal Gordonibacter species. Food Funct. 2014, 5, 1779–1784. [Google Scholar] [CrossRef] [PubMed]
  46. Selma, M.V.; Beltran, D.; Luna, M.C.; Romo-Vaquero, M.; Garcia-Villalba, R.; Mira, A.; Espin, J.C.; Tomas-Barberan, F.A. Isolation of Human Intestinal Bacteria Capable of Producing the Bioactive Metabolite Isourolithin A from Ellagic Acid. Front. Microbiol. 2017, 8, 1521. [Google Scholar] [CrossRef] [PubMed]
  47. Watanabe, H.; Kishino, S.; Kudoh, M.; Yamamoto, H.; Ogawa, J. Evaluation of electron-transferring cofactor mediating enzyme systems involved in urolithin dehydroxylation in Gordonibacter urolithinfaciens DSM 27213. J. Biosci. Bioeng. 2020, 129, 552–557. [Google Scholar] [CrossRef]
  48. Connell, E.; Le Gall, G.; Pontifex, M.G.; Sami, S.; Cryan, J.F.; Clarke, G.; Müller, M.; Vauzour, D. Microbial-derived metabolites as a risk factor of age-related cognitive decline and dementia. Mol. Neurodegener. 2022, 17, 43. [Google Scholar] [CrossRef]
  49. Siddarth, P.; Li, Z.; Miller, K.J.; Ercoli, L.M.; Merril, D.A.; Henning, S.M.; Heber, D.; Small, G.W. Randomized placebo-controlled study of the memory effects of pomegranate juice in middle-aged and older adults. Am. J. Clin. Nutr. 2020, 111, 170–177. [Google Scholar] [CrossRef] [PubMed]
  50. Krikorian, R.; Shidler, M.D.; Nash, T.A.; Kalt, W.; Vinqvist-Tymchuk, M.R.; Shukitt-Hale, B.; Joseph, J.A. Blueberry supplementation improves memory in older adults. J. Agric. Food Chem. 2010, 58, 3996–4000. [Google Scholar] [CrossRef] [PubMed]
  51. Sala-Vila, A.; Valls-Pedret, C.; Rajaram, S.; Coll-Padrós, N.; Cofán, M.; Serra-Mir, M.; Pérez-Heras, A.M.; Roth, I.; Freitas-Simoes, T.M.; Doménech, M.; et al. Effect of a 2-year diet intervention with walnuts on cognitive decline. The Walnuts And Healthy Aging (WAHA) study: A randomized controlled trial. Am. J. Clin. Nutr. 2020, 111, 590–600. [Google Scholar] [CrossRef]
  52. Bowtell, J.L.; Aboo-Bakkar, Z.; Conway, M.E.; Adlam, A.R.; Fulford, J. Enhanced task-related brain activation and resting perfusion in healthy older adults after chronic blueberry supplementation. Appl. Physiol. Nutr. Metab. 2017, 42, 773–779. [Google Scholar] [CrossRef]
  53. McNamara, R.K.; Kalt, W.; Shidler, M.D.; McDonald, J.; Summer, S.S.; Stein, A.L.; Stover, A.N.; Krikorian, R. Cognitive response to fish oil, blueberry, and combined supplementation in older adults with subjective cognitive impairment. Neurobiol. Aging 2018, 64, 147–156. [Google Scholar] [CrossRef]
  54. Cásedas, G.; Les, F.; Choya-Foces, C.; Hugo, M.; López, V. The Metabolite Urolithin-A Ameliorates Oxidative Stress in Neuro-2a Cells, Becoming a Potential Neuroprotective Agent. Antioxidants 2020, 9, 177. [Google Scholar] [CrossRef]
  55. Kim, K.B.; Lee, S.; Kim, J.H. Neuroprotective effects of urolithin A on H2O2-induced oxidative stress-mediated apoptosis in SK-N-MC cells. Nutr Res Prac. 2020, 14, 3–11. [Google Scholar] [CrossRef]
  56. An, L.; Li, M.; Zou, C.; Wang, K.; Zhang, W.; Huang, X.; Wang, Y. Walnut polyphenols and the active metabolite urolithin A improve oxidative damage in SH-SY5Y cells by up-regulating PKA/CREB/BDNF signaling. Food Funct. 2023, 14, 2698–2709. [Google Scholar] [CrossRef]
  57. Chen, P.; Chen, F.; Lei, J.; Wang, G.; Zhou, B. The Gut Microbiota Metabolite Urolithin B Improves Cognitive Deficits by Inhibiting Cyt C-Mediated Apoptosis and Promoting the Survival of Neurons through the PI3K Pathway in Aging Mice. Front. Pharmacol. 2021, 12, 768097. [Google Scholar] [CrossRef]
  58. Lee, G.; Park, J.S.; Lee, E.J.; Ahn, J.H.; Kim, H.S. Anti-inflammatory and antioxidant mechanisms of urolithin B in activated microglia. Phytomedicine Int. J. Phytother. Phytopharm. 2019, 55, 50–57. [Google Scholar] [CrossRef]
  59. Toney, A.M.; Albusharif, M.; Works, D.; Polenz, L.; Schlange, S.; Chaidez, V.; Ramer-Tait, A.E.; Chung, S. Differential Effects of Whole Red Raspberry Polyphenols and Their Gut Metabolite Urolithin A on Neuroinflammation in BV-2 Microglia. Int. J. Environ. Res. Public Health 2020, 18, 68. [Google Scholar] [CrossRef]
  60. Xu, J.; Yuan, C.; Wang, G.; Luo, J.; Ma, H.; Xu, L.; Mu, Y.; Li, Y.; Seeram, N.P.; Huang, X.; et al. Urolithins Attenuate LPS-Induced Neuroinflammation in BV2Microglia via MAPK, Akt, and NF-κB Signaling Pathways. J. Agric. Food Chem. 2018, 66, 571–580. [Google Scholar] [CrossRef]
  61. Lee, H.J.; Jung, Y.H.; Choi, G.E.; Kim, J.S.; Chae, C.W.; Lim, J.R.; Kim, S.Y.; Yoon, J.H.; Cho, J.H.; Lee, S.-J.; et al. Urolithin A suppresses high glucose-induced neuronal amyloidogenesis by modulating TGM2-dependent ER-mitochondria contacts and calcium homeostasis. Cell Death Differ. 2021, 28, 184–202. [Google Scholar] [CrossRef]
  62. Esselun, C.; Theyssen, E.; Eckert, G.P. Effects of Urolithin A on Mitochondrial Parameters in a Cellular Model of Early Alzheimer Disease. Int. J. Mol. Sci. 2021, 22, 8333. [Google Scholar] [CrossRef] [PubMed]
  63. Liu, J.; Jiang, J.; Qiu, J.; Wang, L.; Zhuo, J.; Wang, B.; Sun, D.; Yu, S.; Lou, H. Urolithin A protects dopaminergic neurons in experimental models of Parkinson’s disease by promoting mitochondrial biogenesis through the SIRT1/PGC-1 alpha signaling pathway. Food Funct. 2022, 13, 375–385. [Google Scholar] [CrossRef] [PubMed]
  64. Jayatunga, D.P.W.; Hone, E.; Fernando, W.M.A.D.B.; Garg, M.L.; Verdile, G.; Martins, R.N. A Synergistic Combination of DHA, Luteolin, and Urolithin A Against Alzheimer’s Disease. Front. Aging Neurosci. 2022, 14, 780602. [Google Scholar] [CrossRef]
  65. Kshirsagar, S.; Alvir, R.V.; Pradeepkiran, J.A.; Hindle, A.; Vijayan, M.; Ramasubramaniam, B.; Kumar, S.; Reddy, A.P.; Reddy, P.H. A Combination Therapy of Urolithin A plus EGCG Has Stronger Protective Effects than Single Drug Urolithin A in a Humanized Amyloid Beta Knockin Mice for Late-Onset Alzheimer’s Disease. Cells 2022, 11, 2660. [Google Scholar] [CrossRef]
  66. Kshirsagar, S.; Sawant, N.; Morton, H.; Reddy, A.P.; Reddy, P.H. Mitophagy enhancers against phosphorylated Tau-induced mitochondrial and synaptic toxicities in Alzheimer disease. Pharmacol. Res. 2021, 174, 105973. [Google Scholar] [CrossRef] [PubMed]
  67. Kshirsagar, S.; Sawant, N.; Morton, H.; Reddy, A.P.; Reddy, P.H. Protective effects of mitophagy enhancers against amyloid beta-induced mitochondrial and synaptic toxicities in Alzheimer disease. Hum. Mol. Genet. 2022, 31, 423–439. [Google Scholar] [CrossRef]
  68. Fang, E.F.; Hou, Y.; Palikaras, K.; Adriaanse, B.A.; Kerr, J.S.; Yang, B.; Lautrup, S.; Hasan-Olive, M.M.; Caponio, D.; Dan, X.; et al. Mitophagy inhibits amyloid-β and tau pathology and reverses cognitive deficits in models of Alzheimer’s disease. Nat. Neurosci. 2019, 22, 401–412. [Google Scholar] [CrossRef]
  69. Yuan, T.; Ma, H.; Liu, W.; Niesen, D.B.; Shah, N.; Crews, R.; Rose, K.N.; Vattem, D.A.; Seeram, N.P. Pomegranate’s Neuroprotective Effects against Alzheimer’s Disease Are Mediated by Urolithins, Its Ellagitannin-Gut Microbial Derived Metabolites. ACS Chem. Neurosci. 2016, 7, 26–33. [Google Scholar] [CrossRef]
  70. Yao, X.; Kailin, L.; Ji, B.; Hang, L.; Xiaotong, Z.; El-Omar, E.; Lin, H.; Lan, G.; Min, W. Urolithin a attenuates diabetes-associated cognitive impairment by ameliorating intestinal barrier dysfunction via n-glycan biosynthesis pathway. Mol. Nutr. Food Res. 2022, 66, 2100863. [Google Scholar] [CrossRef]
  71. Martínez-Lapiscina, E.H.; Clavero, P.; Toledo, E.; Estruch, R.; Salas-Salvadó, J.; San Julián, B.; Sanchez-Tainta, A.; Ros, E.; Valls-Pedret, C.; Martinez-Gonzalez, M. Mediterranean diet improves cognition: The PREDIMED-NAVARRA randomised trial. J. Neurol. Neurosurg. Psychiatry 2013, 84, 1318–1325. [Google Scholar] [CrossRef]
  72. Miller, M.G.; Thangthaeng, N.; Rutledge, G.A.; Scott, T.M.; Shukitt-Hale, B. Dietary strawberry improves cognition in a randomised, double-blind, placebo-controlled trial in older adults. Br. J. Nutr. 2021, 126, 253–263. [Google Scholar] [CrossRef] [PubMed]
  73. Nilsson, A.; Salo, I.; Plaza, M.; Björck, I. Effects of a mixed berry beverage on cognitive functions and cardiometabolic risk markers; A randomized cross-over study in healthy older adults. PLoS ONE 2017, 12, e0188173. [Google Scholar] [CrossRef] [PubMed]
  74. Bensalem, J.; Dudonné, S.; Etchamendy, N.; Pellay, H.; Amadieu, C.; Gaudout, D.; Dubreuil, S.; Paradis, M.E.; Pomerleau, S.; Capuron, L.; et al. Polyphenols From Grape and Blueberry Improve Episodic Memory in Healthy Elderly with Lower Level of Memory Performance: A Bicentric Double-Blind, Randomized, Placebo-Controlled Clinical Study. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 2019, 74, 996–1007. [Google Scholar] [CrossRef]
  75. Whyte, A.R.; Cheng, N.; Fromentin, E.; Williams, C.M. A Randomized, Double-Blinded, Placebo-Controlled Study to Compare the Safety and Efficacy of Low Dose Enhanced Wild Blueberry Powder and Wild Blueberry Extract (ThinkBlue™) in Maintenance of Episodic and Working Memory in Older Adults. Nutrients 2018, 10, 660. [Google Scholar] [CrossRef] [PubMed]
  76. Miller, M.G.; Hamilton, D.A.; Joseph, J.A.; Shukitt-Hale, B. Dietary blueberry improves cognition among older adults in a randomized, double-blind, placebo-controlled trial. Eur. J. Nutr. 2018, 57, 1169–1180. [Google Scholar] [CrossRef]
  77. Boespflug, E.L.; Eliassen, J.C.; Dudley, J.A.; Shidler, M.D.; Kalt, W.; Summer, S.S.; Stein, A.L.; Stover, A.N.; Krikorian, R. Enhanced neural activation with blueberry supplementation in mild cognitive impairment. Nutr. Neurosci. 2018, 21, 297–305. [Google Scholar] [CrossRef]
  78. Krikorian, R.; Kalt, W.; McDonald, J.E.; Shidler, M.D.; Summer, S.S.; Stein, A.L. Cognitive performance in relation to urinary anthocyanins and their flavonoid-based products following blueberry supplementation in older adults at risk for dementia. J. Funct. Foods 2020, 64, 103667. [Google Scholar] [CrossRef]
  79. Halliwell, B. Reactive oxygen species and the central nervous system. J. Neurochem. 1992, 59, 1609–1623. [Google Scholar] [CrossRef]
  80. Shohami, E.; Beit-Yannai, E.; Horowitz, M.; Kohen, R. Oxidative stress in closed-head injury: Brain antioxidant capacity as an indicator of functional outcome. J. Cereb. Blood Flow Metab. 1997, 17, 1007–1019. [Google Scholar] [CrossRef]
  81. Shi, P.Z.; Wang, J.W.; Wang, P.C.; Han, B.; Lu, X.H.; Ren, Y.X.; Feng, X.M.; Cheng, X.F.; Zhang, L. Urolithin a alleviates oxidative stress-induced senescence in nucleus pulposus-derived mesenchymal stem cells through SIRT1/PGC-1α pathway. World J. Stem Cells 2021, 13, 1928–1946. [Google Scholar] [CrossRef]
  82. Mazumder, M.K.; Choudhury, S.; Borah, A. An in silico investigation on the inhibitory potential of the constituents of Pomegranate juice on antioxidant defense mechanism: Relevance to neurodegenerative diseases. IBRO Rep. 2019, 6. [Google Scholar] [CrossRef]
  83. Singh, R.; Chandrashekharappa, S.; Vemula, P.K.; Haribabu, B.; Jala, V.R. Microbial Metabolite Urolithin B Inhibits Recombinant Human Monoamine Oxidase A Enzyme. Metabolites 2020, 10, 258. [Google Scholar] [CrossRef] [PubMed]
  84. Saha, P.; Yeoh, B.S.; Singh, R.; Chandrasekar, B.; Vemula, P.K.; Haribabu, B.; Vijay-Kumar, M.; Jala, V.R. Gut Microbiota Conversion of Dietary Ellagic Acid into Bioactive Phytoceutical Urolithin A Inhibits Heme Peroxidases. PLoS ONE 2016, 11, e0156811. [Google Scholar] [CrossRef] [PubMed]
  85. Priyadarsini, K.I.; Khopde, S.M.; Kumar, S.S.; Mohan, H. Free radical studies of ellagic acid, a natural phenolic antioxidant. J Agric. Food Chem. 2002, 50, 2200–2206. [Google Scholar] [CrossRef] [PubMed]
  86. Galano, A.; Francisco Marquez, M.; Pérez-González, A. Ellagic acid: An unusually versatile protector against oxidative stress. Chem. Res. Toxicol. 2014, 27, 904–918. [Google Scholar] [CrossRef] [PubMed]
  87. Ríos, J.L.; Giner, R.M.; Marín, M.; Recio, M.C. A Pharmacological Update of Ellagic Acid. Planta Medica 2018, 84, 1068–1093. [Google Scholar] [CrossRef] [PubMed]
  88. Fu, X.; Gong, L.F.; Wu, Y.F.; Lin, Z.; Jiang, B.J.; Wu, L.; Yu, K.H. Urolithin A targets the PI3K/Akt/NF-κB pathways and prevents IL-1β-induced inflammatory response in human osteoarthritis: In vitro and in vivo studies. Food Funct. 2019, 10, 6135–6146. [Google Scholar] [CrossRef]
  89. Yan, C.; Ma, Z.; Ma, H.; Li, Q.; Zhai, Q.; Jiang, T.; Zhang, Z.; Wang, Q. Mitochondrial Transplantation Attenuates Brain Dysfunction in Sepsis by Driving Microglial M2 Polarization. Mol. Neurobiol. 2020, 57, 3875–3890. [Google Scholar] [CrossRef]
  90. Lin, X.H.; Ye, X.J.; Li, Q.F.; Gong, Z.; Cao, X.; Li, J.H.; Zhao, S.T.; Sun, X.D.; He, X.S.; Xuan, A.G. Urolithin A Prevents Focal Cerebral Ischemic Injury via Attenuating Apoptosis and Neuroinflammation in Mice. Neuroscience 2020, 448, 94–106. [Google Scholar] [CrossRef]
  91. Komatsu, W.; Kishi, H.; Yagasaki, K.; Ohhira, S. Urolithin A attenuates pro-inflammatory mediator production by suppressing PI3-K/Akt/NF-κB and JNK/AP-1 signaling pathways in lipopolysaccharide-stimulated RAW264 macrophages: Possible involvement of NADPH oxidase-derived reactive oxygen species. Eur. J. Pharmacol. 2018, 833, 411–424. [Google Scholar] [CrossRef]
  92. Zhang, Y.; Aisker, G.; Dong, H.; Halemahebai, G.; Zhang, Y.; Tian, L. Urolithin A suppresses glucolipotoxicity-induced ER stress and TXNIP/NLRP3/IL-1β inflammation signal in pancreatic β cells by regulating AMPK and autophagy. Phytomed. Int. J. Phytother. Phytopharm. 2021, 93, 153741. [Google Scholar] [CrossRef]
  93. Ding, S.L.; Pang, Z.Y.; Chen, X.M.; Li, Z.; Liu, X.X.; Zhai, Q.L.; Huang, J.M.; Ruan, Z.Y. Urolithin a attenuates IL-1β-induced inflammatory responses and cartilage degradation via inhibiting the MAPK/NF-κB signaling pathways in rat articular chondrocytes. J. Inflamm. 2020, 17, 13. [Google Scholar] [CrossRef] [PubMed]
  94. Toney, A.M.; Fan, R.; Xian, Y.; Chaidez, V.; Ramer-Tait, A.E.; Chung, S. Urolithin A, a Gut Metabolite, Improves Insulin Sensitivity Through Augmentation of Mitochondrial Function and Biogenesis. Obesity 2019, 27, 612–620. [Google Scholar] [CrossRef] [PubMed]
  95. Cordero, M.D.; Williams, M.R.; Ryffel, B. AMP-Activated Protein Kinase Regulation of the NLRP3 Inflammasome during Aging. Trends Endocrinol. Metab. TEM 2018, 29, 8–17. [Google Scholar] [CrossRef] [PubMed]
  96. Salt, I.P.; Palmer, T.M. Exploiting the anti-inflammatory effects of AMP-activated protein kinase activation. Expert Opin. Investig. Drugs 2012, 21, 1155–1167. [Google Scholar] [CrossRef]
  97. Carling, D. AMPK signalling in health and disease. Curr. Opin. Cell Biol. 2017, 45, 31–37. [Google Scholar] [CrossRef] [PubMed]
  98. Salminen, A.; Kaarniranta, K. AMP-activated protein kinase (AMPK) controls the aging process via an integrated signaling network. Ageing Res. Rev. 2012, 11, 230–241. [Google Scholar] [CrossRef]
  99. Wang, Y.; Huang, Y.; Xu, Y.; Ruan, W.; Wang, H.; Zhang, Y.; Saavedra, J.M.; Zhang, L.; Huang, Z.; Pang, T. A Dual AMPK/Nrf2 Activator Reduces Brain Inflammation After Stroke by Enhancing Microglia M2 Polarization. Antioxid. Redox Signal. 2018, 28, 141–163. [Google Scholar] [CrossRef]
  100. Jęśko, H.; Wencel, P.; Strosznajder, R.P.; Strosznajder, J.B. Sirtuins and Their Roles in Brain Aging and Neurodegenerative Disorders. Neurochem. Res. 2017, 42, 876–890. [Google Scholar] [CrossRef]
  101. Cho, S.H.; Chen, J.A.; Sayed, F.; Ward, M.E.; Gao, F.; Nguyen, T.A.; Krabbe, G.; Sohn, P.D.; Lo, I.; Minami, S.; et al. SIRT1 deficiency in microglia contributes to cognitive decline in aging and neurodegeneration via epigenetic regulation of IL-1β. J. Neurosci. 2015, 35, 807–818. [Google Scholar] [CrossRef]
  102. Ye, J.; Liu, Z.; Wei, J.; Lu, L.; Huang, Y.; Luo, L.; Xie, H. Protective effect of SIRT1 on toxicity of microglial-derived factors induced by LPS to PC12 cells via the p53-caspase-3-dependent apoptotic pathway. Neurosci. Lett. 2013, 553, 72–77. [Google Scholar] [CrossRef] [PubMed]
  103. Rizzi, L.; Roriz-Cruz, M. Sirtuin 1 and Alzheimer’s disease: An up-to-date review. Neuropeptides 2018, 71, 54–60. [Google Scholar] [CrossRef] [PubMed]
  104. Yang, L.; Zhang, J.; Yan, C.; Zhou, J.; Lin, R.; Lin, Q.; Wang, W.; Zhang, K.; Yang, G.; Bian, X.; et al. SIRT1 regulates CD40 expression induced by TNF-α via NF-ĸB pathway in endothelial cells. Cell. Physiol. Biochem. 2012, 30, 1287–1298. [Google Scholar] [CrossRef] [PubMed]
  105. Fonseca, É.; Marques, C.C.; Pimenta, J.; Jorge, J.; Baptista, M.C.; Gonçalves, A.C.; Pereira, R. Anti-Aging Effect of Urolithin A on Bovine Oocytes In Vitro. Animals 2021, 11, 2048. [Google Scholar] [CrossRef] [PubMed]
  106. Ryu, D.; Mouchiroud, L.; Andreux, P.A.; Katsyuba, E.; Moullan, N.; Nicolet-Dit-Félix, A.A.; Williams, E.G.; Jha, P.; Lo Sasso, G.; Huzard, D.; et al. Urolithin A induces mitophagy and prolongs lifespan in C. elegans and increases muscle function in rodents. Nat. Med. 2016, 22, 879–888. [Google Scholar] [CrossRef]
  107. Fivenson, E.M.; Lautrup, S.; Sun, N.; Scheibye-Knudsen, M.; Stevnsner, T.; Nilsen, H.; Bohr, V.A.; Fang, E.F. Mitophagy in neurodegeneration and aging. Neurochem. Int. 2017, 109, 202–209. [Google Scholar] [CrossRef]
  108. Ahsan, A.; Zheng, Y.R.; Wu, X.L.; Tang, W.D.; Liu, M.R.; Ma, S.J.; Jiang, L.; Hu, W.W.; Zhang, X.N.; Chen, Z. Urolithin A-activated autophagy but not mitophagy protects against ischemic neuronal injury by inhibiting ER stress in vitro and in vivo. CNS Neurosci. Ther. 2019, 25, 976–986. [Google Scholar] [CrossRef]
  109. Iorio, R.; Celenza, G.; Petricca, S. Mitophagy: Molecular Mechanisms, New Concepts on Parkin Activation and the Emerging Role of AMPK/ULK1 Axis. Cells 2022, 11, 30. [Google Scholar] [CrossRef]
  110. Wang, Y.; Xu, E.; Musich, P.R.; Lin, F. Mitochondrial dysfunction in neurodegenerative diseases and the potential countermeasure. CNS Neurosci. Ther. 2019, 25, 816–824. [Google Scholar] [CrossRef]
  111. Wang, S.; Kandadi, M.R.; Ren, J. Double knockout of Akt2 and AMPK predisposes cardiac aging without affecting lifespan: Role of autophagy and mitophagy. Biochim. Biophys. Acta. Mol. Basis Dis. 2019, 1865, 1865–1875. [Google Scholar] [CrossRef]
  112. Atherton, P.J.; Babraj, J.A.; Smith, K.; Singh, J.; Rennie, M.J.; Wackerhage, H. Selective activation of AMPK-PGC-1 alpha or PKB-TSC2-mTOR signaling can explain specific adaptive responses to endurance or resistance training-like electrical muscle stimulation. FASEB J. 2005, 19, 786–788. [Google Scholar] [CrossRef] [PubMed]
  113. Laker, R.C.; Drake, J.C.; Wilson, R.J.; Lira, V.A.; Lewellen, B.M.; Ryall, K.A.; Fisher, C.C.; Zhang, M.; Saucerman, J.J.; Goodyear, L.J.; et al. Ampk phosphorylation of Ulk1 is required for targeting of mitochondria to lysosomes in exercise-induced mitophagy. Nat. Commun. 2017, 8, 548. [Google Scholar] [CrossRef] [PubMed]
  114. Egan, D.F.; Shackelford, D.B.; Mihaylova, M.M.; Gelino, S.; Kohnz, R.A.; Mair, W.; Vasquez, D.S.; Joshi, A.; Gwinn, D.M.; Taylor, R.; et al. Phosphorylation of ULK1 (hATG1) by AMP-Activated Protein Kinase Connects Energy Sensing to Mitophagy. Science 2011, 331, 456–461. [Google Scholar] [CrossRef] [PubMed]
  115. Kim, J.; Kundu, M.; Viollet, B.; Guan, K.-L. AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat. Cell Biol. 2011, 13, 132–141. [Google Scholar] [CrossRef] [PubMed]
  116. Pillai, V.B.; Sundaresan, N.R.; Kim, G.; Gupta, M.; Rajamohan, S.B.; Pillai, J.B.; Samant, S.; Ravindra, P.V.; Isbatan, A.; Gupta, M.P. Exogenous NAD Blocks Cardiac Hypertrophic Response via Activation of the SIRT3-LKB1-AMP-activated Kinase Pathway. J. Biol. Chem. 2010, 285, 3133–3144. [Google Scholar] [CrossRef]
  117. Lin, J.; Zhuge, J.; Zheng, X.; Wu, Y.; Zhang, Z.; Xu, T.; Meftah, Z.; Xu, H.; Wu, Y.; Tian, N.; et al. Urolithin A-induced mitophagy suppresses apoptosis and attenuates intervertebral disc degeneration via the AMPK signaling pathway. Free Radic. Biol. Med. 2020, 150, 109–119. [Google Scholar] [CrossRef]
  118. Han, Q.-a.; Su, D.; Shi, C.; Liu, P.; Wang, Y.; Zhu, B.; Xia, X. Urolithin A attenuated ox-LDL-induced cholesterol accumulation in macrophages partly through regulating miR-33a and ERK/AMPK/SREBP1 signaling pathways. Food Funct. 2020, 11, 3432–3440. [Google Scholar] [CrossRef]
  119. Rodriguez, J.; Pierre, N.; Naslain, D.; Bontemps, F.; Ferreira, D.; Priem, F.; Deldicque, L.; Francaux, M. Urolithin B, a newly identified regulator of skeletal muscle mass. J. Cachexia Sarcopenia Muscle 2017, 8, 583–597. [Google Scholar] [CrossRef]
  120. Zhao, C.; Sakaguchi, T.; Fujita, K.; Ito, H.; Nishida, N.; Nagatomo, A.; Tanaka-Azuma, Y.; Katakura, Y. Pomegranate-Derived Polyphenols Reduce Reactive Oxygen Species Production via SIRT3-Mediated SOD2 Activation. Oxidative Med. Cell. Longev. 2016, 2016, 2927131. [Google Scholar] [CrossRef]
  121. Ghosh, N.; Das, A.; Biswas, N.; Gnyawali, S.; Singh, K.; Gorain, M.; Polcyn, C.; Khanna, S.; Roy, S.; Sen, C.K. Urolithin A augments angiogenic pathways in skeletal muscle by bolstering NAD+ and SIRT1. Sci. Rep. 2020, 10, 20184. [Google Scholar] [CrossRef]
  122. Sebastián, D.; Sorianello, E.; Segalés, J.; Irazoki, A.; Ruiz-Bonilla, V.; Sala, D.; Planet, E.; Berenguer-Llergo, A.; Muñoz, J.P.; Sánchez-Feutrie, M.; et al. Mfn2 deficiency links age-related sarcopenia and impaired autophagy to activation of an adaptive mitophagy pathway. EMBO J. 2016, 35, 1677–1693. [Google Scholar] [CrossRef] [PubMed]
  123. Qiao, H.; Ren, H.; Du, H.; Zhang, M.; Xiong, X.; Lv, R. Liraglutide repairs the infarcted heart: The role of the SIRT1/Parkin/mitophagy pathway. Mol. Med. Rep. 2018, 17, 3722–3734. [Google Scholar] [CrossRef] [PubMed]
  124. Kwak, H.M.; Jeon, S.Y.; Sohng, B.H.; Kim, J.G.; Lee, J.M.; Lee, K.B.; Jeong, H.H.; Hur, J.M.; Kang, Y.H.; Song, K.S. Beta-Secretase (BACE1) inhibitors from pomegranate (Punica granatum) husk. Arch. Pharmacal Res. 2005, 28, 1328–1332. [Google Scholar] [CrossRef] [PubMed]
  125. Youn, K.; Jun, M. In Vitro BACE1 inhibitory activity of geraniin and corilagin from Geranium thunbergii. Planta Medica 2013, 79, 1038–1042. [Google Scholar] [CrossRef]
  126. Iaccarino, H.F.; Singer, A.C.; Martorell, A.J.; Rudenko, A.; Gao, F.; Gillingham, T.Z.; Mathys, H.; Seo, J.; Kritskiy, O.; Abdurrob, F.; et al. Gamma frequency entrainment attenuates amyloid load and modifies microglia. Nature 2016, 540, 230–235. [Google Scholar] [CrossRef]
  127. Keren-Shaul, H.; Spinrad, A.; Weiner, A.; Matcovitch-Natan, O.; Dvir-Szternfeld, R.; Ulland, T.K.; David, E.; Baruch, K.; Lara-Astaiso, D.; Toth, B.; et al. A Unique Microglia Type Associated with Restricting Development of Alzheimer’s Disease. Cell 2017, 169, 1276–1290.e1217. [Google Scholar] [CrossRef]
  128. Tu, H.J.; Su, C.J.; Peng, C.S.; Lin, T.E.; HuangFu, W.C.; Hsu, K.C.; Hwang, T.L.; Pan, S.L. Urolithin A exhibits a neuroprotective effect against Alzheimer’s disease by inhibiting DYRK1A activity. J. Food Drug Anal. 2023, 31, 358–370. [Google Scholar] [CrossRef]
  129. Frick, B.; Schroecksnadel, K.; Neurauter, G.; Leblhuber, F.; Fuchs, D. Increasing production of homocysteine and neopterin and degradation of tryptophan with older age. Clin. Biochem. 2004, 37, 684–687. [Google Scholar] [CrossRef]
  130. Platten, M.; Nollen, E.A.A.; Röhrig, U.F.; Fallarino, F.; Opitz, C.A. Tryptophan metabolism as a common therapeutic target in cancer, neurodegeneration and beyond. Nat. Rev. Drug Discov. 2019, 18, 379–401. [Google Scholar] [CrossRef]
  131. Roager, H.M.; Licht, T.R. Microbial tryptophan catabolites in health and disease. Nat. Commun. 2018, 9, 3294. [Google Scholar] [CrossRef]
  132. Rothhammer, V.; Mascanfroni, I.D.; Bunse, L.; Takenaka, M.C.; Kenison, J.E.; Mayo, L.; Chao, C.C.; Patel, B.; Yan, R.; Blain, M.; et al. Type I interferons and microbial metabolites of tryptophan modulate astrocyte activity and central nervous system inflammation via the aryl hydrocarbon receptor. Nat. Med. 2016, 22, 586–597. [Google Scholar] [CrossRef] [PubMed]
  133. Rothhammer, V.; Borucki, D.M.; Tjon, E.C.; Takenaka, M.C.; Chao, C.C.; Ardura-Fabregat, A.; de Lima, K.A.; Gutiérrez-Vázquez, C.; Hewson, P.; Staszewski, O.; et al. Microglial control of astrocytes in response to microbial metabolites. Nature 2018, 557, 724–728. [Google Scholar] [CrossRef]
  134. Yang, J.; Guo, Y.; Lee, R.; Henning, S.M.; Wang, J.; Pan, Y.; Qing, T.; Hsu, M.; Nguyen, A.; Prabha, S.; et al. Pomegranate Metabolites Impact Tryptophan Metabolism in Humans and Mice. Curr. Dev. Nutr. 2020, 4, nzaa165. [Google Scholar] [CrossRef] [PubMed]
  135. Livingston, S.; Mallick, S.; Lucas, D.A.; Sabir, M.S.; Sabir, Z.L.; Purdin, H.; Nidamanuri, S.; Haussler, C.A.; Haussler, M.R.; Jurutka, P.W. Pomegranate derivative urolithin A enhances vitamin D receptor signaling to amplify serotonin-related gene induction by 1,25-dihydroxyvitamin D. Biochem. Biophys. Rep. 2020, 24, 100825. [Google Scholar] [CrossRef] [PubMed]
  136. Green, P.S.; Simpkins, J.W. Neuroprotective effects of estrogens: Potential mechanisms of action. Int. J. Dev. Neurosci. 2000, 18, 347–358. [Google Scholar] [CrossRef] [PubMed]
  137. Brann, D.W.; Dhandapani, K.; Wakade, C.; Mahesh, V.B.; Khan, M.M. Neurotrophic and neuroprotective actions of estrogen: Basic mechanisms and clinical implications. Steroids 2007, 72, 381–405. [Google Scholar] [CrossRef]
  138. Barroso, A.; Mahler, J.V.; Fonseca-Castro, P.H.; Quintana, F.J. The aryl hydrocarbon receptor and the gut-brain axis. Cell. Mol. Immunol. 2021, 18, 259–268. [Google Scholar] [CrossRef]
  139. Larrosa, M.; Gonzalez-Sarrias, A.; Garcia-Conesa, M.T.; Tomas-Barberan, F.A.; Espin, J.C. Urolithins, ellagic acid-derived metabolites produced by human colonic microflora, exhibit estrogenic and antiestrogenic activities. J. Agric. Food Chem. 2006, 54, 1611–1620. [Google Scholar] [CrossRef]
  140. Skledar, D.G.; Tomasic, T.; Dolenc, M.S.; Masic, L.P.; Zega, A. Evaluation of endocrine activities of ellagic acid and urolithins using reporter gene assays. Chemosphere 2019, 220, 706–713. [Google Scholar] [CrossRef]
  141. Zhang, W.; Chen, J.-H.; Aguilera-Barrantes, I.; Shiau, C.-W.; Sheng, X.; Wang, L.-S.; Stoner, G.D.; Huang, Y.-W. Urolithin A suppresses the proliferation of endometrial cancer cells by mediating estrogen receptor-alpha-dependent gene expression. Mol. Nutr. Food Res. 2016, 60, 2387–2395. [Google Scholar] [CrossRef]
  142. Shen, P.-X.; Li, X.; Deng, S.-Y.; Zhao, L.; Zhang, Y.-Y.; Deng, X.; Han, B.; Yu, J.; Li, Y.; Wang, Z.-Z.; et al. Urolithin A ameliorates experimental autoimmune encephalomyelitis by targeting aryl hydrocarbon receptor. Ebiomedicine 2021, 64, 103227. [Google Scholar] [CrossRef] [PubMed]
  143. Muku, G.E.; Murray, I.A.; Espín, J.C.; Perdew, G.H. Urolithin A Is a Dietary Microbiota-Derived Human Aryl Hydrocarbon Receptor Antagonist. Metabolites 2018, 8, 86. [Google Scholar] [CrossRef] [PubMed]
  144. Gong, Q.-Y.; Cai, L.; Jing, Y.; Wang, W.; Yang, D.-X.; Chen, S.-W.; Tian, H.-L. Urolithin A alleviates blood-brain barrier disruption and attenuates neuronal apoptosis following traumatic brain injury in mice. Neural Regen. Res. 2022, 17, 2007–2013. [Google Scholar] [CrossRef]
  145. Shukur, K.T.; Ercetin, T.; Luise, C.; Sippl, W.; Sirkecioglu, O.; Ulgen, M.; Coskun, G.P.; Yarim, M.; Gazi, M.; Gulcan, H.O. Design, synthesis, and biological evaluation of new urolithin amides as multitarget agents against Alzheimer’s disease. Arch. Pharm. 2021, 354, e2000467. [Google Scholar] [CrossRef] [PubMed]
  146. Noshadi, B.; Ercetin, T.; Luise, C.; Yuksel, M.Y.; Sippl, W.; Sahin, M.F.; Gazi, M.; Gulcan, H.O. Synthesis, Characterization, Molecular Docking, and Biological Activities of Some Natural and Synthetic Urolithin Analogs. Chem. Biodivers. 2020, 17, e2000197. [Google Scholar] [CrossRef]
  147. Seeram, N.P.; Aronson, W.J.; Zhang, Y.; Henning, S.M.; Moro, A.; Lee, R.-P.; Sartippour, N.; Harris, D.M.; Rettig, M.; Suchard, M.A.; et al. Pomegranate ellagitannin-derived metabolites inhibit prostate cancer growth and localize to the mouse prostate gland. J. Agric. Food Chem. 2007, 55, 7732–7737. [Google Scholar] [CrossRef]
  148. Yao, X.; Kailin, L.; Haiyan, Z.; Yunlong, L.; Lin, H.; Hang, L.; Min, W. The profile of buckwheat tannins based on widely targeted metabolome analysis and pharmacokinetic study of ellagitannin metabolite urolithin A. LWT Food Sci. Technol. 2022, 156, 113069. [Google Scholar] [CrossRef]
  149. Kujawska, M.; Jourdes, M.; Witucki, Ł.; Karaźniewicz-Łada, M.; Szulc, M.; Górska, A.; Mikołajczak, P.; Teissedre, P.L.; Jodynis-Liebert, J. Pomegranate Juice Ameliorates Dopamine Release and Behavioral Deficits in a Rat Model of Parkinson’s Disease. Brain Sci. 2021, 11, 1127. [Google Scholar] [CrossRef]
  150. Aguilera-Carbo, A.; Augur, C.; Prado-Barragan, L.A.; Favela-Torres, E.; Aguilar, C.N. Microbial production of ellagic acid and biodegradation of ellagitannins. Appl. Microbiol. Biotechnol. 2008, 78, 189–199. [Google Scholar] [CrossRef]
Figure 1. Outline of effects of UroA on brain aging (actions and mechanisms). The mechanisms of UroA on aging brain mainly focus on promoting mitophagy and mitochondrial function and relieving neuroinflammation. Other mechanisms include mitigating oxidative stress, inhibiting Aβ and tau pathology, and regulating Trp metabolism. Although UroA has been shown to activate anti-aging signaling pathways such as AMPK, SIRTs, and mTOR in vivo, the direct or key targets of UroA have not been fully elucidated.
Figure 1. Outline of effects of UroA on brain aging (actions and mechanisms). The mechanisms of UroA on aging brain mainly focus on promoting mitophagy and mitochondrial function and relieving neuroinflammation. Other mechanisms include mitigating oxidative stress, inhibiting Aβ and tau pathology, and regulating Trp metabolism. Although UroA has been shown to activate anti-aging signaling pathways such as AMPK, SIRTs, and mTOR in vivo, the direct or key targets of UroA have not been fully elucidated.
Nutrients 15 03884 g001
Table 4. IC50 of Uros and corresponding references on synthetic and oxygen free radicals.
Table 4. IC50 of Uros and corresponding references on synthetic and oxygen free radicals.
SampleSuperoxide Radical 1DPPH RadicalPeroxyl
Radicals 2
ABTS RadicalHydroxyl
Radical
UroA5.01 ± 5.01 μM152.66 ± 35.01 μM13.1 μM------
Gallic acid0.26 ± 0.21 μM3.10 ± 3.11 μM---------
Ascorbic acid---14.81 ± 14.90 μM---------
Pomegranate
extracts
------0.49 μM------
UroB495.32 ± 3.28 mM295.41 ± 2.36 mM---316.18 ± 1.85 mM306.28 ± 4.61 mM
Ascorbic acid874.39 ± 1.48 mM446.25 ± 1.78 mM---526.24 ± 3.18 mM540.16 ± 2.52 mM
IC50 is expressed as mean ± SD, and data for peroxyl radicals are expressed as µmol Trolox equivalents per mg of sample. 1 UroA scavenges superoxide radicals generated by the xanthine/xanthine oxidase system. 2 The capacity of UroA to scavenge peroxyl radicals was measured by the oxygen radical antioxidant capacity (ORAC).
Table 5. Summary of Uros metabolites identified in the brain.
Table 5. Summary of Uros metabolites identified in the brain.
AnimalsRoute of
Administration
TreatmentBrain TissueIdentified
Metabolites
in Brain
Plasma
Concentratio
Refs
male C57BL/6 mice
(7 months, 25–30 g)
i.g.UroA
0.3 mg/mouse,
single administration
brain tissuesmUroA: 8 ng/g---[147]
male C57BL/6 mice
(6 weeks)
i.g.UroA 200 mg/kg b.w.,
single administration
cortexUroA: 28 ng/g15 ng/mL[148]
hippocampusUroA: 35 ng/g
male rats
(12 weeks, 288 ± 20 g)
i.v.Polyphenol metabolites
(12.5 μg UroA + 5.3 μgUroB) 2.7 µmol/rat/day for 2 days
brain tissuesUroA: 2.2 ng/g---[41]
UroB: 0.5 ng/g
male albino
Wistar rats
(6 weeks, 250–300 g)
i.g.Pomegranate juice
500 mg/kg b.w./day
for 10 days
brain tissuesUroA: 1.68 ± 0.25 ng/g18.75 ± 3.21 ng/mL[36]
male albino
Wistar rats
(6 weeks, 250–300 g)
i.g.Pomegranate juice
500 mg/kg b.w./day
for 45 days
brain tissuesUroA: 2.068 ± 0.274 ng/g---[149]
Abbreviations: i.v.: intravenous, i.g.: intragastrical, b.w.: body weight.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

An, L.; Lu, Q.; Wang, K.; Wang, Y. Urolithins: A Prospective Alternative against Brain Aging. Nutrients 2023, 15, 3884. https://doi.org/10.3390/nu15183884

AMA Style

An L, Lu Q, Wang K, Wang Y. Urolithins: A Prospective Alternative against Brain Aging. Nutrients. 2023; 15(18):3884. https://doi.org/10.3390/nu15183884

Chicago/Turabian Style

An, Lei, Qiu Lu, Ke Wang, and Yousheng Wang. 2023. "Urolithins: A Prospective Alternative against Brain Aging" Nutrients 15, no. 18: 3884. https://doi.org/10.3390/nu15183884

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop