Next Article in Journal
Sarcopenia Is Associated with Changes in Circulating Markers of Antioxidant/Oxidant Balance and Innate Immune Response
Previous Article in Journal
Chemical Composition of Salvia fruticosa Mill. Essential Oil and Its Protective Effects on Both Photosynthetic Damage and Oxidative Stress in Conocephalum conicum L. Induced by Environmental Heavy Metal Concentrations
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Pomegranate Extract Administration Reverses Loss of Motor Coordination and Prevents Oxidative Stress in Cerebellum of Aging Mice

1
Department of Physiology, Faculty of Medicine, University of Valencia, CIBERFES, 46010 Valencia, Spain
2
Biomedical Research Institute INCLIVA, University of Valencia, 46010 Valencia, Spain
3
Central Unit for Research in Medicine (UCIM), University of Valencia, 46010 Valencia, Spain
4
Neonatal Research Group, Health Research Institute La Fe (IISLaFe), 46026 Valencia, Spain
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
These authors also contributed equally to this work.
Antioxidants 2023, 12(11), 1991; https://doi.org/10.3390/antiox12111991
Submission received: 8 October 2023 / Revised: 30 October 2023 / Accepted: 9 November 2023 / Published: 11 November 2023
(This article belongs to the Special Issue Antioxidants as Anti-Aging Interventions)

Abstract

:
The cerebellum is responsible for complex motor functions, like maintaining balance and stance, coordination of voluntary movements, motor learning, and cognitive tasks. During aging, most of these functions deteriorate, which results in falls and accidents. The aim of this work was to elucidate the effect of a standardized pomegranate extract during four months of supplementation in elderly mice to prevent frailty and improve the oxidative state. Male C57Bl/6J eighteen-month-old mice were evaluated for frailty using the “Valencia Score” at pre-supplementation and post-supplementation periods. We analyzed lipid peroxidation in the cerebellum and brain cortex and the glutathione redox status in peripheral blood. In addition, a set of aging-related genes in cerebellum and apoptosis biomarkers was measured via real-time polymerase chain reaction (RT-PCR). Our results showed that pomegranate extract supplementation improved the motor skills of C57Bl/6J aged mice in motor coordination, neuromuscular function, and monthly weight loss, but no changes in grip strength and endurance were found. Furthermore, pomegranate extract reversed the increase in malondialdehyde due to aging in the cerebellum and increased the reduced glutathione/oxidized glutathione (GSH/GSSG) ratio in the blood. Finally, aging and apoptosis biomarkers improved in aged mice supplemented with pomegranate extract in the cerebellum but not in the cerebral cortex.

1. Introduction

Aging is defined as the biological process that consists of the progressive deterioration of the organism in apparently healthy individuals [1]. The changes that occur during aging make the individual frailer. Frailty is an age-associated biological syndrome characterized by decreased biological reserves, leading to a decline and deterioration of functional properties at the cellular, tissue, and organ level [2].
During aging, there is cumulative oxidative damage to cellular structures. This can result in mitochondrial oxidative stress and even cell death [3]. Thus, free radicals act as powerful oxidizing agents and cause aging damage by combining with essential molecules, such as DNA and proteins, promoting, among other things, lipid peroxidation, forming malondialdehyde (MDA) as a product, and oxidation of reduced glutathione (GSH) to oxidized glutathione (GSSG). These parameters serve as a reference to understand the oxidative damage that exists in a certain tissue or system [4,5].
Specifically, in aging of the nervous system, a decrease in sensorimotor control and functioning is observed [6]. This leads to loss of motor coordination and eventually to falls. These declines in fine motor control, gait, and balance affect older adults’ ability to perform activities of daily living and maintain their independence. The cerebellum is one of the major areas that control complex motor functions like maintaining balance and stance and coordination of voluntary movements. In the human brain, the cerebellum represents approximately 10% of the total brain volume, and recent studies suggest that the surface area of the cerebellum is 78% of that of the cerebral cortex [7]. The cerebellum seems to be the least studied area of the brain, not only from the point of view of aging but also from a physiological and even anatomical perspective. However, it has a fundamental role in the control of cognitive processes, as well as in the control of motor coordination [8,9].
Nutrition plays a significant role in the aging process, as it can have a profound impact on overall health and well-being as people become older [10,11]. Proper nutrition can help delay the onset of age-related diseases, maintain physical and cognitive function, and improve the quality of life in older adults [12,13]. Therefore, a comprehensive approach that combines healthy eating habits, regular physical activity, stress management, and other healthy lifestyle factors is crucial to promoting healthy aging [14]. Johan Auwerx and Patrick Aebischer identified that urolithin A is able to restore the cell’s ability to recycle the components of defective mitochondria. It is the only known substance that can relaunch the mitochondrial cleaning process. It is a completely natural compound, and its effect is powerful and measurable [15].
In this sense, some studies suggest that pomegranate and its metabolites, such as punicalagin, may also have potential anti-aging properties by reducing oxidative stress and inflammation and improving cell function and longevity [16,17,18]. A recent study shows that pomegranate polyphenol punicalagin is the active molecule that effectively improved neuroinflammation, learning, and memory deficit in natural aging in a 12-month-old mice model and in a D-galactose-induced brain aging model [19]. Although there are basic studies that have shown clear neuroprotective effects of punicalagin in cell lines and mouse models, there are no medical clinical trials so far. Other researchers demonstrated that pomegranate treatment improved cognitive and functional recovery after stroke events and spent less time in the hospital than placebo controls [20]. Furthermore, there are studies on the protection that pomegranate extract exerts against neurodegenerative diseases such as Alzheimer’s disease, Parkinson’s disease, and multiple sclerosis [21,22,23,24,25]. Alzheimer’s is the most prevalent dementia in older people, and aging is their greatest risk factor.
In addition, some clinical and preclinical studies using the same pomegranate extract as ours show metabolic, cardiovascular, and neuronal protection in old individuals [26,27]. More studies on the prevention of natural aging are needed since almost all studies focus on a pathological model.
The aim of this work was to elucidate the effect of pomegranate extract (PE) during 4 months of supplementation in elderly mice from 18 months of age, in which mouse frailty begins, and this supplementation could prevent the frailty associated with natural aging.

2. Materials and Methods

2.1. Experimental Animals

The animal study was approved by the University of Valencia Ethics Committee for Research and Animal Welfare (license reference: A20201110122413). Animals were housed at the Animal House Core Facility of the University of Valencia.
Male C57BL6 (wild type, WT) eighteen-month mice were assigned to two groups: a control group (only drinking water) and another pomegranate extract (PE) supplementation (150 mg/kg/day) group (solubilized in water).
The PE dosage was adjusted to the liquid intake and to the animal’s body weights every 3 days during 16 weeks’ supplementation. The dose was chosen based on the positive effects found both in metabolic and cardiovascular function in previous clinical studies. As PE was dissolved properly in water, it was used as a vehicle in the control group.
The natural concentrated extract of whole pomegranate (PE) used in this work is Pomanox® P30, obtained through a water-based extraction process and provided by Euromed (Barcelona, Spain).
Pomanox® P30 was prepared according to the European Patent EP1967079 with a technology that utilizes only aqueous solutions, avoiding the use of organic solvents. For manufacturing Pomanox® P30, freshly harvested pomegranate fruits growing in the Spanish Mediterranean area are used. The steps of manufacturing are an extraction of water-soluble compounds, including punicalagins, in aqueous solution, a separation of aqueous solution from pomegranate paste, a step of adsorption chromatography, concentration by nanofiltration, and finally, the pomegranate aqueous extract is turned into a solid form via spray-drying.
According to the manufacturer, Pomanox® P30 (Lot 0100531601) is standardized to >50% total polyphenols and punicalagins, α + β ≥ 30% p/p, and contains α-punicalagin and β-punicalagin (174.4 g/kg and 206.8 g/kg, respectively); galloylglucose, punicalin, and ellagic acid (7.3 g/kg); ellagic acid glucoside (4.2 g/kg); ellagic acid rhamnoside (1.3 g/kg); and the anthocyanins delphinidin-3,5-diglucoside (302.9 mg/kg), cyanidin-3,5-diglucoside (425.3 mg/kg), delphinidin-3-glucoside (75.7 mg/kg), cyanidin-3-glucoside (241.8 mg/kg), and pelargonidin-3-glucoside (99.5 mg/kg). The chemical composition of PE has been characterized by Euromed, and PE polyphenol content is described in Table S1 in Supplementary Material [28].
Mice were used in a longitudinal study to evaluate the functional status using the “Valencia Score” and anthropometric determinations before and after supplementation at 18 and 22 months. After mice were euthanized, blood, plasma, and organs were obtained and stored at −80 °C. Samples of WT adult mice (10 months) were used as an aging process control in biomolecular determinations.

2.2. Hematological, Biochemical, and Body Composition Parameters

We used the veterinary "hematology element ht5" analyzer (Scil Animal Care Company, Heska group, Viernheim, Germany) located in Central Unit for Research in Medicine (UCIM), Faculty of Medicine, University of Valencia to obtain hematological parameters. Furthermore, we acquired biochemical parameters with the "skyla vb1" (cScil Animal Care Company, Heska group) located in Central Unit for Research in Medicine (UCIM), Faculty of Medicine, University of Valencia using analytical rotors (Critical-care Panel 900-330) in whole EDTA blood from 22-month-old mice without or with supplementation.
The body composition of mice (bone mineral density, lean mass, fat mass, and fat in tissue) was analyzed with simple anesthesia using the method of dual-energy X-ray absorptiometry (DXA) with InAlyzer located in Central Unit for Research in Medicine (UCIM), Faculty of Medicine, University of Valencia (Medikors Inc., Seongnam-si, Republic of Korea).

2.3. GSH and GSSG Determination

We determined GSH and GSSG concentrations in blood using the method described by [29].
A total of 10 µL of blood samples was directly added to the papers pre-soaked with 8 µmol of NEM and allowed to derivatize (GSH-NEM formation) at room temperature for 5 min. Then, they were kept at −20 °C until further processing. After, 100 μL of cold PCA solution (4%, v/v) was added to the precipitate proteins. To extract analytes, tubes with the papers were placed in a thermal mixer for 5 min at 4 °C at 1400 rpm, followed by 5 min of sonication. Finally, samples were centrifuged at 10,000× g for 15 min at 4 °C, and 40 μL of supernatants was diluted with 60 μL of H2O (0.1% FA, v/v) and 5 μL of internal standard solution containing isotopically labeled GSH-NEM and GSSG, 100 μmol/L each.

2.4. Functional Tests

We evaluated 5 parameters: grip strength (muscular force), rotarod performance test (motor coordination), ladder climbing test (neuromuscular function), treadmill (endurance), and monthly involuntary weight loss (body weight). This score is based on the construct developed by Linda Fried in humans and was termed the “Valencia score” of frailty [30].

2.4.1. Grip Strength Evaluation

To study strength, we use the grip strength test; for this, we use a dynamometer located in Central Unit for Research in Medicine (UCIM), Faculty of Medicine, University of Valencia (Grip Strength Meter, Panlab, Barcelona, Spain) adapted to rodents [31,32]. During the procedure, mice were suspended by the tail, and the force in grams of the front paws was measured. The animals were weighed on a scale to normalize the force to the weight. The animals were held by their tail and placed on the dynamometer, allowing them to grip with the front legs, leaving the animal’s body parallel to the ground. The animals were left held for 20 s, recording the maximum force that the mouse exerted. This procedure was performed 3 times with each animal, leaving 10 min of rest between determinations, and then the maximum force of the 3 tests was recorded.

2.4.2. Motor Coordination Test

To study the motor coordination of mice, rotarod equipment (Panlab, Harvard Apparatus, Holliston, MA, USA) was used [32,33,34,35]. The rotarod is a device made up of a 3 mm diameter bar, separated into 4 spaces for 4 animals, on which the animals are placed. Before the rotarod test, all mice were habituated and pretrained on the rotarod (15 rpm) for 5 min over 3 days. On the test day, the speed of rotation was accelerated gradually from 4 to 40 rpm for 300 s, and the total time spent on the rotarod was recorded. Each mouse was tested over 3 trials, with 15 min intervals between trials.

2.4.3. Endurance and Fatigue Evaluation

To study the fatigue resistance of mice, we used the Treadmill Control LE 8710 equipment (Panlab, Harvard Apparatus) [36,37]. It is a rotating belt that changes the speed of movement, divided into 5 lanes of 38 cm × 5 cm, in which the animals are placed. The distance, time, speed, and number of times the animal receives an electric shock are recorded. For this test, we used the following procedure:
Adaptation of the animals to the test is essential. This is achieved with a 10 min adaptation run. On the day of the test, a race was carried out with a gradual increase in speed. The test ends when it is considered that maximum fatigue has been reached, which is reached when the animal receives 3 electric shocks in 5 s. For the first 4 min, the animals run at 10 cm/s, and from this moment on, the speed is increased by 4 cm/s every 2 min.

2.4.4. Monthly Involuntary Weight Loss (Body Weight)

The body weight of the mice was measured with the precision balance (Gram, Barcelona, Spain, AHZ-300). Three measurements of body weight were assessed on day 0 (before starting supplementation) and every week (until the end of the supplementation program). The mean was calculated for each mouse in each group. Then, to determine individual body weight loss, the percentage of loss was defined for each mouse from 18 months of age to 22 months.

2.5. Lipid Peroxidation Determination

Lipid peroxidation determination as malondialdehyde (MDA) levels was determined in cerebellum and brain cortex samples using high-performance liquid chromatography, as described previously [38].
This method is based on the hydrolysis of lipoperoxides and the subsequent formation of an adduct between thiobarbituric acid (TBA) and MDA (TBA-MDA2). This adduct was detected using HPLC in the reverse phase and quantified at 532 nm. The chromatographic technique was performed under isocratic conditions, the mobile phase being a mixture of monopotassium phosphate at 50 mM (pH 6.8) and acetonitrile (70:30). The level of MDA in each sample was divided by the concentration of protein determined using the Lowry method.

2.6. Total RNA Extraction and Real-Time Polymerase Chain Reaction (RT-PCR) Studies

Cerebellum and brain cortex were extracted using 300 μL of TRIzol reagent. Integrity quality was performed by Nanodrop 2000 (Agilent, Santa Clara, CA, USA), and the purity was evaluated with the 260/280 ratio. RT-PCR was performed for each of the following genes, using ready-to-use primer and probe sets pre-developed by Applied Biosystems (Foster City, CA, USA, TaqMan Gene Expression Assays) using Quantum Studio v5 (QuantStudioTM Design & Analysis Software v1.4.2), establishing the proper conditions. The housekeeping gene was Gapdh (Mm99999915_g1, Applied Biosystems, NM_001289726, NM_008084.4), and the genes studied were Beta-2-Microglobulin (B2m) (Mm00437762_m1, NM_009735.3), Caspase 3 (Casp3) (Mm01195085_m1, NM_001284409.1, NM_009810.3), Caspase 8 (Casp8) (Mm01255716_m1, NM_001080126.2, NM_001277926.2, NM_009812.3), Caspase 9 (Casp9) (Mm00516563_m1, NM_001277932.1, NM_015733.5), Lysozyme C (Lzp-s) (Mm00657323_m1, NM_013590.4), Cathepsin S (Ctss) (Mm00457902_m1, NM_021281.3), Complement 4B (C4b) (Mm00437890_m1, NM_009780.2), Complement C1q A Chain (C1qa) (Mm00432142_m1, NM_007572.2), and Glial Fibrillary Acidic Protein (Gfap) (Mm00546086_m1, NM_010277.3). For the analysis of results, 2−∆∆Ct was used [39].

2.7. Statistical Methods

Values are expressed as the mean ± standard deviation (SD) or mean ± standard error (SEM). Normal distribution of the samples was assessed using the Shapiro–Wilk test. To compare two different groups, the unpaired Student’s t-test was used, or the Mann–Whitney test in case of a non-normal distribution. To study two independent variables, a two-way analysis of variance (ANOVA) test was used. For the frailty score and its criteria, differences were tested using Pearson’s χ2 test. A one-way ANOVA was used to determine differences in gene expression between groups. Statistical analysis was performed using Statistical or GraphPad Prism softwares with a significance level set at p < 0.05, and all graphs were represented with GraphPad Prism8 Software version 9.0.0.

3. Results

3.1. Effect of Pomegranate Extract (PE) on Hematological, Biochemical, and Body Composition

The PE (150 mg/Kg of body weight) does not affect the main hematological and biochemical parameters in the blood of mice (Tables S2 and S3 in Supplementary Material). These values include the blood count, urea, creatinine, and transaminases, among others. Of the 20 parameters measured, the administration of pomegranate extract only affected the lactate concentration. Similarly, the administration of PE does not affect body composition (Table S4 in Supplementary Material).

3.2. Effect of Pomegranate Extract (PE) on the Concentration of Glutathione in Mice’s Blood

Figure 1 shows the effect of PE administration on glutathione concentration and glutathione redox status in the blood of old mice. Panel A shows that there is a slight decrease in the glutathione value in older animals that is prevented when the animals have been treated with PE. Similarly, glutathione oxidation in the blood of older animals is increased, which is prevented when the animals have taken PE (see panel B). The ratio between reduced and oxidized glutathione is a good index of the redox state of the blood. Panel C shows that the redox ratio of glutathione decreases significantly with age, as expected, and that this decrease is prevented when the animals take PE.

3.3. Effect of Pomegranate Extract (PE) on Functional Studies and Body Weight

Figure 2 shows the effect of PE on functional parameters in aged mice compared with young mice. Panels A, B, and C show that the PE does not produce effects on grip strength, nor on running time or speed. However, there is a positive effect on motor coordination (Panel D, determined by the rotarod; see the Methods section) and on motor strength and coordination determined using the ladder climbing test (Panel E). Therefore, supplementation with PE is favorable to improve functional aspects, especially those related to motor coordination.
Figure 3 shows that involuntary weight loss after 4-month supplementation is diminished by PE consumption, indicating a better capacity to maintain body weight in aged mice.

3.4. Effect of Pomegranate Extract (PE) on Lipid Peroxidation Analyses

In view of these functional results, we analyzed oxidative stress by studying lipid peroxidation in the brain cortex and in the cerebellum because the latter is involved in motor coordination.
Figure 4 shows that PE has a protective effect against lipid peroxidation in the cerebellum but not in the cerebral cortex.

3.5. Effect of Pomegranate Extract (PE) on the Expression of Aging Biomarkers in Cerebellum and Brain Cortex

Prolla and his collaborators [40] found a group of genes associated with aging of the cerebellum. These genes are Complement C1q A Chain (C1qa), Cathepsin S (Ctss), Lysozyme C (Lzp-s), Glial Fibrillary Acidic Protein (Gfap), and Complement C4B (C4b).
Figure 5 shows that PE prevents the change in gene expression associated with aging in the cerebellum. The effects are highly significant and contrast with the lack of a protective effect against age-associated changes in the cerebral cortex shown in Figure 6.
We also determined the expression of B2m because it has been linked to age-associated neurodegenerative disorders [41]. Figure 7 shows that B2m expression increases with age in the cerebellum and brain cortex, but this is only prevented in the cerebellum of the supplemented group.

3.6. Effect of Pomegranate Extract (PE) on the Expression of Apoptosis Biomarkers in Cerebellum and Brain Cortex

An analysis of the relationships between the genes that change with aging and that are protected by the administration of PE showed that the main pathways and cellular processes involved convergence in the control of apoptosis (see Figure 8).
This led us to analyze the expression of genes related to apoptosis, such as Casp3, Casp8, and Casp9, and we observed that they increase with aging and that this increase is protected by the administration of PE in the cerebellum but not in the cortex (see Figure 9).

4. Discussion

Older people constitute a key sector of the population. Optimizing nutrition is an essential strategy to promote successful aging [42]. We have found that Pomanox® P30 (standardized pomegranate extract) improves oxidative stress parameters and optimizes gene expression in the cerebellum. Many previous studies have pointed to the importance of pomegranate extracts in promoting improvements ranging from sports performance, blood lipid profile, inflammation, and oxidative stress in various physiological and pathological situations. Endothelial function, and therefore also blood pressure control, also improves in patients treated with pomegranate extracts [43]. Other previous studies in preclinical models demonstrate the capacity of pomegranate and its derivatives to improve lifespan in C. elegans [15,44]. Moreover, at the clinical level, pomegranate supplementation, together with protein intake, prevents age-associated adverse events [16].
Frailty is a geriatric syndrome described by Linda Fried and her collaborators [2] that is characterized by slow gait, difficulty getting up, spontaneous weight loss, and feeling unwell. Frailty is a precursor of disability, and therefore, preventing it is a great challenge in geriatric medicine. Frailty was first described in humans. In our research group, we have described a frailty test that we have called the Valencia test [45], extrapolating to animals the changes observed in the tests used in humans. Therefore, our previous studies have made it possible to quantify age-associated frailty in laboratory mice.
Another physiological process implicated in aging and frailty is the involuntary loss of weight. Weight loss and aging are interrelated in several ways. As individuals age, they experience changes in metabolism, body composition, and lifestyle factors that can impact their ability to lose or maintain weight. Metabolism tends to slow down with age, which can make it more challenging to lose weight. This decrease in metabolic rate is partly due to a reduction in muscle mass and a decrease in physical activity levels. As a result, the body burns fewer calories at rest. Our pomegranate extract supplement is able to decrease the loss of weight in aging (see Figure 3).
Falls pose a fundamental risk for dependency and even death in older people [46]. A determining factor in these falls is the decrease in motor coordination associated with age. The fundamental role of the cerebellum in this process has been frequently ignored. Studies by Tom Prolla and his collaborators have shown that the variation of several selected genes with aging is maximum in the cerebellum, even when compared to organs like the heart [47]. We have confirmed these findings (mainly genes that are overexpressed with normal aging), and we have seen that this age-associated change is prevented by the administration of pomegranate extract.
It is now generally accepted that aging is linked to cellular disorganization due to oxidative stress caused by free radicals and other reactive oxygen species (ROS). According to theories published independently by Harman and Gerschman in the 50s of the 20th century, whose central dogma lies in analyzing how, during aerobic metabolism, radical species derived from oxygen are produced incidentally and uncontrollably, macromolecules are irreversibly damaged, damage that accumulates over time, and this results in a gradual loss of homeostatic mechanisms, interference of gene expression patterns, and loss of the cell’s functional capacity that leads to cell aging and death. In addition to the key role of the mitochondrial genome of differentiated cells as the main target of ROS, according to the oxidative stress/mitochondrial injury theory.
Taking all of this together, we have asked ourselves if long-term supplementation with pomegranate extract in 18-month-old mice could prevent a loss of functional capacities and oxidative damage associated with the physiological aging process. After 4 months of supplementation, peripheral blood was extracted and deposited on dry NEM-DBS paper to determine GSH and GSSG levels according to the described method [29]. Supplementation with pomegranate extract keeps GSH and GSSG levels similar to those levels in 10-month-old animals, avoiding the oxidation of GSH to GSSG associated with the aging process that is noted in the control aging group (see Figure 1A–C). These results are in consonance with previous preclinical [48] and clinical [49] studies, where there was an increment in GSH levels in erythrocytes, probably due to the participation of NF-κB and the Nrf2/GSH Axis.
However, the increases that occur in the cerebral cortex with aging are not prevented by the administration of this extract. A limitation of our study is that we have not studied the different areas of the brain because our results regarding phenotyping the animals fed PE led us to changes in coordination and, therefore, to investigate the cerebellum in more detail. The cerebellum, one of the major responsible organs for motor coordination, is exceptional in the large number of major phospholipids that undergo changes mainly by oxidation (with consequential changes in acyl composition) with age, whereas the motor cortex is highly resistant to change [6]. These loss of capacities are, in part, due to the accumulation of oxidative damage, as has been demonstrated by other authors [50,51] where MDA accumulation in the cerebellum and other brain zones by lipid peroxidation reduces motor capacities. For this reason, we have determined lipid peroxidation levels by measuring MDA in the cortex and cerebellum, and we have observed how these levels are lower in animals supplemented for 22 months than in the control group, specifically in cerebellar tissue (see Figure 4A). It is important to note that this reduction is specific to cerebellar tissue and does not appear in the cortex (see Figure 4B).
On the other hand, previous studies have proposed that β2-microglobulin (B2M), a subunit of primary histocompatibility complex class I (MHC I) molecules, regulates behavior, synaptic plasticity, and brain development [52,53]. B2M has been recently considered as a biomarker of Alzheimer’s disease [54,55]. The results from animal studies have emphasized that the local or systemic injection of exogenous B2M in young rats leads to neurodevelopmental and hippocampus-dependent cognitive impairments and that an increase in B2M levels was observed in aged mice, which was verified by cerebrospinal fluid or plasma from healthy populations [41,56]. Our results indicate downregulation of B2M in cerebellar tissue in the supplemented group with respect to the control aged group, which suffers an increment associated with aging (two-fold increment with respect to 10-month-old mice) (see Figure 7).
Finally, we have observed that the genes modulated by the intake of pomegranate extract converge on the apoptosis pathway. These results have been confirmed by analyzing the expression of caspase 3, 8, and 9 (see Figure 8 and Figure 9). We have previously observed that centenarians are also characterized by better control of apoptosis than people with normal aging [57]. The results shown here, together with our studies in centenarians, suggest that apoptosis plays a key role in healthy aging.
The novelty of this work focuses on the fact that pomegranate extract could be used as a preventive against the deterioration of normal aging. In our study, we have observed for the first time that the cerebellum is the area where aging is first noticed in a model without pathology and where the PE acts efficiently. Therefore, PE could be a good preventative against the risk of falls so common in aging, as well as other functional or biochemical impairments. We have observed changes in levels of oxidative stress, specifically in the cerebellum and without an underlying pathology that other authors observe in the neurodegenerative diseases [58,59]. Furthermore, it is the first study that uses 18-month-old mice, the age at which frailty begins in mice, and PE supplementation reduces part of the functional disabilities and systemic and cerebellar oxidative stress that deteriorate with age.
The main active compounds of our pomegranate extract (see Table S1) are polyphenols and punicalagins. Many articles show that punicalagin has different biological activities, such as in cancer, cardiovascular diseases, liver diseases, and inflammation [60,61,62,63]. Recently, a pharmacological study found that this substance and its polyphenols had significant neuroprotective potential against Alzheimer’s disease (AD), Parkinson’s disease (PD), stroke, and chronic mild stress [20,23,25,64,65]. It is likely that in normal aging, the main beneficial actors are also polyphenols and punicalagins that, when metabolized through the microbiota, generate beneficial active compounds at a systemic level and cross the blood–brain barrier, which would help prevent cerebellar aging and improve the redox status of the elderly.
For this reason, pomegranate extract may be a convenient nutritional intervention to promote healthy aging.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/antiox12111991/s1, Table S1: Pomegranate polyphenols content from Pomanox® P30; Table S2: Hematological parameters in the blood of old mice with or without supplementation of pomegranate extract; Table S3: Biochemical parameters in the blood of old mice with or without supplementation of pomegranate extract; Table S4: Body composition of old mice with or without supplementation of pomegranate extract.

Author Contributions

D.V. and A.V. carried out functional tests, RT-PCR studies, and all formal analyses and revised the manuscript; A.D. supervised the animal studies; A.C. and M.D. obtained the lipid peroxidation data; J.K. obtained GSH and GSSG data and revised the manuscript; E.S. and J.V. conceptualized and designed the study, coordinated and supervised data collection, and critically reviewed the manuscript for important intellectual content. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by Instituto de Salud Carlos III CB16/10/00435 (CIBERFES), (PID2019-110906RB-I00/AEI/10.13039/501100011033) from the Spanish Ministry of Innovation and Science; FGCSIC/PSLINTERREG/FEDER; PROMETEO/2019/097 de “Consellería de Sanitat de la Generalitat Valenciana” and EU Funded H2020- DIABFRAIL-LATAM (Ref: 825546); European Joint Programming Initiative “A Healthy Diet for a Healthy Life” (JPI HDHL) and of the ERA-NET Cofund ERA-HDHL (GA N° 696295 of the EU Horizon 2020 Research and Innovation Program). Part of the equipment employed in this work has been funded by Generalitat Valenciana and co-financed with ERDF funds (OP ERDF of Comunitat Valenciana 2014–2020). Support from Ramón Areces Fundation and Soria Melguizo Fundation is also acknowledged.

Institutional Review Board Statement

This study was approved by the University of Valencia Ethics Committee for Research and Animal Welfare (reference: A20201110122413; protocol code: 2021/VSC/PEA/0006 Generalitat Valenciana; date: 3 February 2021) for studies involving animals.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data is contained within the article and supplementary material.

Acknowledgments

We thank the company Euromed for providing us with the pomegranate extract used in this study.

Conflicts of Interest

The authors declare no conflict of interest. Pomanox® P30 product is from Euromed company, the Euromed company had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The Hallmarks of Aging. Cell 2013, 153, 1194–1217. [Google Scholar] [CrossRef]
  2. Fried, L.P.; Tangen, C.M.; Walston, J.; Newman, A.B.; Hirsch, C.; Gottdiener, J.; Seeman, T.; Tracy, R.; Kop, W.J.; Burke, G.; et al. Frailty in Older Adults: Evidence for a Phenotype. J. Gerontol. A Biol. Sci. Med. Sci. 2001, 56, M146–M156. [Google Scholar] [CrossRef] [PubMed]
  3. Sastre, J.; Pallardó, F.V.; Viña, J. The Role of Mitochondrial Oxidative Stress in Aging. Free Radic. Biol. Med. 2003, 35, 1–8. [Google Scholar] [CrossRef]
  4. Viña, J.; Borrás, C.; Miquel, J. Theories of Ageing. IUBMB Life 2007, 59, 249–254. [Google Scholar] [CrossRef]
  5. Viña, J.; Borras, C.; Gomez-Cabrera, M.C. A Free Radical Theory of Frailty. Free Radic. Biol. Med. 2018, 124, 358–363. [Google Scholar] [CrossRef]
  6. Hancock, S.E.; Friedrich, M.G.; Mitchell, T.W.; Truscott, R.J.W.; Else, P.L. Changes in Phospholipid Composition of the Human Cerebellum and Motor Cortex during Normal Ageing. Nutrients 2022, 14, 2495. [Google Scholar] [CrossRef] [PubMed]
  7. Sereno, M.I.; Diedrichsen, J.; Tachrount, M.; Testa-Silva, G.; d’Arceuil, H.; De Zeeuw, C. The Human Cerebellum Has Almost 80% of the Surface Area of the Neocortex. Proc. Natl. Acad. Sci. USA 2020, 117, 19538–19543. [Google Scholar] [CrossRef]
  8. Eccles, J.C. The Cerebellum as a Computer: Patterns in Space and Time. J. Physiol. 1973, 229, 1–32. [Google Scholar] [CrossRef] [PubMed]
  9. Schmahmann, J.D. The Cerebellum and Cognition. Neurosci. Lett. 2019, 688, 62–75. [Google Scholar] [CrossRef]
  10. Downer, S.; Berkowitz, S.A.; Harlan, T.S.; Olstad, D.L.; Mozaffarian, D. Food Is Medicine: Actions to Integrate Food and Nutrition into Healthcare. BMJ 2020, 369, m2482. [Google Scholar] [CrossRef]
  11. Yoshida, S.; Shiraishi, R.; Nakayama, Y.; Taira, Y. Can Nutrition Contribute to a Reduction in Sarcopenia, Frailty, and Comorbidities in a Super-Aged Society? Nutrients 2023, 15, 2991. [Google Scholar] [CrossRef]
  12. Longo, V.D.; Anderson, R.M. Nutrition, Longevity and Disease: From Molecular Mechanisms to Interventions. Cell 2022, 185, 1455–1470. [Google Scholar] [CrossRef]
  13. Di Giosia, P.; Stamerra, C.A.; Giorgini, P.; Jamialahamdi, T.; Butler, A.E.; Sahebkar, A. The Role of Nutrition in Inflammaging. Ageing Res. Rev. 2022, 77, 101596. [Google Scholar] [CrossRef] [PubMed]
  14. Ni Lochlainn, M.; Cox, N.J.; Wilson, T.; Hayhoe, R.P.G.; Ramsay, S.E.; Granic, A.; Isanejad, M.; Roberts, H.C.; Wilson, D.; Welch, C.; et al. Nutrition and Frailty: Opportunities for Prevention and Treatment. Nutrients 2021, 13, 2349. [Google Scholar] [CrossRef] [PubMed]
  15. 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]
  16. Dormal, V.; Pachikian, B.; Debock, E.; Buchet, M.; Copine, S.; Deldicque, L. Evaluation of a Dietary Supplementation Combining Protein and a Pomegranate Extract in Older People: A Safety Study. Nutrients 2022, 14, 5182. [Google Scholar] [CrossRef] [PubMed]
  17. Tan, S.; Yu, C.Y.; Sim, Z.W.; Low, Z.S.; Lee, B.; See, F.; Min, N.; Gautam, A.; Chu, J.J.H.; Ng, K.W.; et al. Pomegranate Activates TFEB to Promote Autophagy-Lysosomal Fitness and Mitophagy. Sci. Rep. 2019, 9, 727. [Google Scholar] [CrossRef]
  18. Deepika; Maurya, P.K. Ellagic Acid: Insight into Its Protective Effects in Age-Associated Disorders. 3 Biotech 2022, 12, 340. [Google Scholar] [CrossRef]
  19. Chen, P.; Chen, F.; Lei, J.; Zhou, B. Pomegranate Polyphenol Punicalagin Improves Learning Memory Deficits, Redox Homeostasis, and Neuroinflammation in Aging Mice. Phytother. Res. 2023, 37, 3655–3674. [Google Scholar] [CrossRef] [PubMed]
  20. Bellone, J.A.; Murray, J.R.; Jorge, P.; Fogel, T.G.; Kim, M.; Wallace, D.R.; Hartman, R.E. Pomegranate Supplementation Improves Cognitive and Functional Recovery Following Ischemic Stroke: A Randomized Trial. Nutr. Neurosci. 2019, 22, 738–743. [Google Scholar] [CrossRef]
  21. Mehdi, A.; Lamiae, B.; Samira, B.; Ramchoun, M.; Abdelouahed, K.; Tamas, F.; Hicham, B. Pomegranate (Punica Granatum L.) Attenuates Neuroinflammation Involved in Neurodegenerative Diseases. Foods 2022, 11, 2570. [Google Scholar] [CrossRef]
  22. Emami Kazemabad, M.J.; Asgari Toni, S.; Tizro, N.; Dadkhah, P.A.; Amani, H.; Akhavan Rezayat, S.; Sheikh, Z.; Mohammadi, M.; Alijanzadeh, D.; Alimohammadi, F.; et al. Pharmacotherapeutic Potential of Pomegranate in Age-Related Neurological Disorders. Front. Aging Neurosci. 2022, 14, 955735. [Google Scholar] [CrossRef]
  23. Khokar, R.; Hachani, K.; Hasan, M.; Othmani, F.; Essam, M.; Al Mamari, A.; Um, D.; Khan, S.A. Anti-Alzheimer Potential of a Waste by-Product (Peel) of Omani Pomegranate Fruits: Quantification of Phenolic Compounds, in-Vitro Antioxidant, Anti-Cholinesterase and in-Silico Studies. Biocatal. Agric. Biotechnol. 2021, 38, 102223. [Google Scholar] [CrossRef]
  24. Lu, X.-Y.; Han, B.; Deng, X.; Deng, S.-Y.; Zhang, Y.-Y.; Shen, P.-X.; Hui, T.; Chen, R.-H.; Li, X.; Zhang, Y. Pomegranate Peel Extract Ameliorates the Severity of Experimental Autoimmune Encephalomyelitis via Modulation of Gut Microbiota. Gut Microbes 2020, 12, 1857515. [Google Scholar] [CrossRef] [PubMed]
  25. Kujawska, M.; Jourdes, M.; Kurpik, M.; Szulc, M.; Szaefer, H.; Chmielarz, P.; Kreiner, G.; Krajka-Kuźniak, V.; Mikołajczak, P.Ł.; 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. 2019, 21, 202. [Google Scholar] [CrossRef] [PubMed]
  26. Stockton, A.; Farhat, G.; McDougall, G.J.; Al-Dujaili, E.a.S. Effect of Pomegranate Extract on Blood Pressure and Anthropometry in Adults: A Double-Blind Placebo-Controlled Randomised Clinical Trial. J. Nutr. Sci. 2017, 6, e39. [Google Scholar] [CrossRef] [PubMed]
  27. Amri, Z.; Ghorbel, A.; Turki, M.; Akrout, F.M.; Ayadi, F.; Elfeki, A.; Hammami, M. Effect of Pomegranate Extracts on Brain Antioxidant Markers and Cholinesterase Activity in High Fat-High Fructose Diet Induced Obesity in Rat Model. BMC Complement. Altern. Med. 2017, 17, 339. [Google Scholar] [CrossRef] [PubMed]
  28. Rodriguez, J.; Caille, O.; Ferreira, D.; Francaux, M. Pomegranate Extract Prevents Skeletal Muscle of Mice against Wasting Induced by Acute TNF-α Injection. Mol. Nutr. Food Res. 2017, 61, 1600169. [Google Scholar] [CrossRef]
  29. Ten-Doménech, I.; Solaz-García, Á.; Lara-Cantón, I.; Pinilla-Gonzalez, A.; Parra-Llorca, A.; Vento, M.; Quintás, G.; Kuligowski, J. Direct Derivatization in Dried Blood Spots for Oxidized and Reduced Glutathione Quantification in Newborns. Antioxidants 2022, 11, 1165. [Google Scholar] [CrossRef] [PubMed]
  30. Gomez-Cabrera, M.C.; Garcia-Valles, R.; Rodriguez-Mañas, L.; Garcia-Garcia, F.J.; Olaso-Gonzalez, G.; Salvador-Pascual, A.; Tarazona-Santabalbina, F.J.; Viña, J. A New Frailty Score for Experimental Animals Based on the Clinical Phenotype: Inactivity as a Model of Frailty. J. Gerontol. A Biol. Sci. Med. Sci. 2017, 72, 885–891. [Google Scholar] [CrossRef]
  31. Ackert-Bicknell, C.L.; Anderson, L.C.; Sheehan, S.; Hill, W.G.; Chang, B.; Churchill, G.A.; Chesler, E.J.; Korstanje, R.; Peters, L.L. Aging Research Using Mouse Models. Curr. Protoc. Mouse Biol. 2015, 5, 95–133. [Google Scholar] [CrossRef] [PubMed]
  32. Graber, T.G.; Maroto, R.; Fry, C.S.; Brightwell, C.R.; Rasmussen, B.B. Measuring Exercise Capacity and Physical Function in Adult and Older Mice. J. Gerontol. A Biol. Sci. Med. Sci. 2021, 76, 819–824. [Google Scholar] [CrossRef]
  33. Liu, H.; Graber, T.G.; Ferguson-Stegall, L.; Thompson, L.V. Clinically Relevant Frailty Index for Mice. J. Gerontol. A Biol. Sci. Med. Sci. 2014, 69, 1485–1491. [Google Scholar] [CrossRef] [PubMed]
  34. Shoji, H.; Takao, K.; Hattori, S.; Miyakawa, T. Age-Related Changes in Behavior in C57BL/6J Mice from Young Adulthood to Middle Age. Mol. Brain 2016, 9, 11. [Google Scholar] [CrossRef] [PubMed]
  35. Carter, R.J.; Morton, J.; Dunnett, S.B. Motor Coordination and Balance in Rodents. Curr. Protoc. Neurosci. 2001, 8, 8–12. [Google Scholar] [CrossRef]
  36. Marcaletti, S.; Thomas, C.; Feige, J.N. Exercise Performance Tests in Mice. Curr. Protoc. Mouse Biol. 2011, 1, 141–154. [Google Scholar] [CrossRef] [PubMed]
  37. Li, H.; Liang, A.; Guan, F.; Fan, R.; Chi, L.; Yang, B. Regular Treadmill Running Improves Spatial Learning and Memory Performance in Young Mice through Increased Hippocampal Neurogenesis and Decreased Stress. Brain Res. 2013, 1531, 1–8. [Google Scholar] [CrossRef]
  38. Wong, S.H.; Knight, J.A.; Hopfer, S.M.; Zaharia, O.; Leach, C.N.; Sunderman, F.W. Lipoperoxides in Plasma as Measured by Liquid-Chromatographic Separation of Malondialdehyde-Thiobarbituric Acid Adduct. Clin. Chem. 1987, 33, 214–220. [Google Scholar] [CrossRef]
  39. Livak, K.J.; Schmittgen, T.D. Analysis of Relative Gene Expression Data Using Real-Time Quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 2001, 25, 402–408. [Google Scholar] [CrossRef]
  40. Park, S.-K.; Kim, K.; Page, G.P.; Allison, D.B.; Weindruch, R.; Prolla, T.A. Gene Expression Profiling of Aging in Multiple Mouse Strains: Identification of Aging Biomarkers and Impact of Dietary Antioxidants. Aging Cell 2009, 8, 484–495. [Google Scholar] [CrossRef]
  41. Smith, L.K.; He, Y.; Park, J.-S.; Bieri, G.; Snethlage, C.E.; Lin, K.; Gontier, G.; Wabl, R.; Plambeck, K.E.; Udeochu, J.; et al. Β2-Microglobulin Is a Systemic pro-Aging Factor That Impairs Cognitive Function and Neurogenesis. Nat. Med. 2015, 21, 932–937. [Google Scholar] [CrossRef] [PubMed]
  42. Leitão, C.; Mignano, A.; Estrela, M.; Fardilha, M.; Figueiras, A.; Roque, F.; Herdeiro, M.T. The Effect of Nutrition on Aging-A Systematic Review Focusing on Aging-Related Biomarkers. Nutrients 2022, 14, 554. [Google Scholar] [CrossRef]
  43. Vilahur, G.; Padró, T.; Casaní, L.; Mendieta, G.; López, J.A.; Streitenberger, S.; Badimon, L. Polyphenol-Enriched Diet Prevents Coronary Endothelial Dysfunction by Activating the Akt/eNOS Pathway. Rev. Esp. Cardiol. (Engl. Ed.) 2015, 68, 216–225. [Google Scholar] [CrossRef]
  44. Zheng, J.; Heber, D.; Wang, M.; Gao, C.; Heymsfield, S.B.; Martin, R.J.; Greenway, F.L.; Finley, J.W.; Burton, J.H.; Johnson, W.D.; et al. Pomegranate Juice and Extract Extended Lifespan and Reduced Intestinal Fat Deposition in Caenorhabditis Elegans. Int. J. Vitam. Nutr. Res. 2017, 87, 149–158. [Google Scholar] [CrossRef]
  45. Martinez de Toda, I.; Garrido, A.; Vida, C.; Gomez-Cabrera, M.C.; Viña, J.; De la Fuente, M. Frailty Quantified by the “Valencia Score” as a Potential Predictor of Lifespan in Mice. J. Gerontol. A Biol. Sci. Med. Sci. 2018, 73, 1323–1329. [Google Scholar] [CrossRef] [PubMed]
  46. Zwart, L.; Germans, T.; Vogels, R.; Simsek, S.; Hemels, M.; Jansen, R. Frail Patients Who Fall and Their Risk on Major Bleeding and Intracranial Haemorrhage. Outcomes from the Fall and Syncope Registry. BMC Geriatr. 2023, 23, 422. [Google Scholar] [CrossRef] [PubMed]
  47. Park, S.-K.; Prolla, T.A. Gene Expression Profiling Studies of Aging in Cardiac and Skeletal Muscles. Cardiovasc. Res. 2005, 66, 205–212. [Google Scholar] [CrossRef]
  48. Mukherjee, S.; Ghosh, S.; Choudhury, S.; Gupta, P.; Adhikary, A.; Chattopadhyay, S. Pomegranate Polyphenols Attenuate Inflammation and Hepatic Damage in Tumor-Bearing Mice: Crucial Role of NF-κB and the Nrf2/GSH Axis. J. Nutr. Biochem. 2021, 97, 108812. [Google Scholar] [CrossRef] [PubMed]
  49. Matthaiou, C.M.; Goutzourelas, N.; Stagos, D.; Sarafoglou, E.; Jamurtas, A.; Koulocheri, S.D.; Haroutounian, S.A.; Tsatsakis, A.M.; Kouretas, D. Pomegranate Juice Consumption Increases GSH Levels and Reduces Lipid and Protein Oxidation in Human Blood. Food Chem. Toxicol. 2014, 73, 1–6. [Google Scholar] [CrossRef]
  50. Jolitha, A.B.; Subramanyam, M.V.V.; Asha Devi, S. Modification by Vitamin E and Exercise of Oxidative Stress in Regions of Aging Rat Brain: Studies on Superoxide Dismutase Isoenzymes and Protein Oxidation Status. Exp. Gerontol. 2006, 41, 753–763. [Google Scholar] [CrossRef]
  51. Al-Amin, M.M.; Akhter, S.; Hasan, A.T.; Alam, T.; Nageeb Hasan, S.M.; Saifullah, A.R.M.; Shohel, M. The Antioxidant Effect of Astaxanthin Is Higher in Young Mice than Aged: A Region Specific Study on Brain. Metab. Brain Dis. 2015, 30, 1237–1246. [Google Scholar] [CrossRef] [PubMed]
  52. Huh, G.S.; Boulanger, L.M.; Du, H.; Riquelme, P.A.; Brotz, T.M.; Shatz, C.J. Functional Requirement for Class I MHC in CNS Development and Plasticity. Science 2000, 290, 2155–2159. [Google Scholar] [CrossRef] [PubMed]
  53. Boulanger, L.M.; Shatz, C.J. Immune Signalling in Neural Development, Synaptic Plasticity and Disease. Nat. Rev. Neurosci. 2004, 5, 521–531. [Google Scholar] [CrossRef] [PubMed]
  54. Huang, Y.-M.; Ma, Y.-H.; Gao, P.-Y.; Wang, Z.-B.; Huang, L.-Y.; Hou, J.-H.; Tan, L.; Yu, J.-T. Plasma Β2-Microglobulin and Cerebrospinal Fluid Biomarkers of Alzheimer’s Disease Pathology in Cognitively Intact Older Adults: The CABLE Study. Alzheimers Res. Ther. 2023, 15, 69. [Google Scholar] [CrossRef]
  55. Barber, R.C.; Phillips, N.R.; Tilson, J.L.; Huebinger, R.M.; Shewale, S.J.; Koenig, J.L.; Mitchel, J.S.; O’Bryant, S.E.; Waring, S.C.; Diaz-Arrastia, R.; et al. Can Genetic Analysis of Putative Blood Alzheimer’s Disease Biomarkers Lead to Identification of Susceptibility Loci? PLoS ONE 2015, 10, e0142360. [Google Scholar] [CrossRef]
  56. Filiano, A.J.; Kipnis, J. Breaking Bad Blood: Β2-Microglobulin as a pro-Aging Factor in Blood. Nat. Med. 2015, 21, 844–845. [Google Scholar] [CrossRef]
  57. Borras, C.; Abdelaziz, K.M.; Gambini, J.; Serna, E.; Inglés, M.; de la Fuente, M.; Garcia, I.; Matheu, A.; Sanchís, P.; Belenguer, A.; et al. Human Exceptional Longevity: Transcriptome from Centenarians Is Distinct from Septuagenarians and Reveals a Role of Bcl-xL in Successful Aging. Aging (Albany NY) 2016, 8, 3185–3208. [Google Scholar] [CrossRef]
  58. Liang, K.J.; Carlson, E.S. Resistance, Vulnerability and Resilience: A Review of the Cognitive Cerebellum in Aging and Neurodegenerative Diseases. Neurobiol. Learn Mem. 2020, 170, 106981. [Google Scholar] [CrossRef]
  59. Lin, M.T.; Beal, M.F. Mitochondrial Dysfunction and Oxidative Stress in Neurodegenerative Diseases. Nature 2006, 443, 787–795. [Google Scholar] [CrossRef]
  60. Adams, L.S.; Seeram, N.P.; Aggarwal, B.B.; Takada, Y.; Sand, D.; Heber, D. Pomegranate Juice, Total Pomegranate Ellagitannins, and Punicalagin Suppress Inflammatory Cell Signaling in Colon Cancer Cells. J. Agric. Food Chem. 2006, 54, 980–985. [Google Scholar] [CrossRef]
  61. Cao, Y.; Chen, J.; Ren, G.; Zhang, Y.; Tan, X.; Yang, L. Punicalagin Prevents Inflammation in LPS-Induced RAW264.7 Macrophages by Inhibiting FoxO3a/Autophagy Signaling Pathway. Nutrients 2019, 11, 2794. [Google Scholar] [CrossRef] [PubMed]
  62. Zhang, Y.; Tan, X.; Cao, Y.; An, X.; Chen, J.; Yang, L. Punicalagin Protects against Diabetic Liver Injury by Upregulating Mitophagy and Antioxidant Enzyme Activities. Nutrients 2022, 14, 2782. [Google Scholar] [CrossRef] [PubMed]
  63. Alalawi, S.; Albalawi, F.; Ramji, D.P. The Role of Punicalagin and Its Metabolites in Atherosclerosis and Risk Factors Associated with the Disease. Int. J. Mol. Sci. 2023, 24, 8476. [Google Scholar] [CrossRef] [PubMed]
  64. George, N.; AbuKhader, M.; Al Balushi, K.; Al Sabahi, B.; Khan, S.A. An Insight into the Neuroprotective Effects and Molecular Targets of Pomegranate (Punica Granatum) against Alzheimer’s Disease. Nutr. Neurosci. 2023, 26, 975–996. [Google Scholar] [CrossRef] [PubMed]
  65. Naveen, S.; Siddalingaswamy, M.; Singsit, D.; Khanum, F. Anti-Depressive Effect of Polyphenols and Omega-3 Fatty Acid from Pomegranate Peel and Flax Seed in Mice Exposed to Chronic Mild Stress. Psychiatry Clin. Neurosci. 2013, 67, 501–508. [Google Scholar] [CrossRef]
Figure 1. Effect of PE administration on glutathione redox status in mice’s blood. The figure shows (A) GSH, (B) GSSG, and (C) GSH/GSSG levels in blood in mice after 18 months, 22 months, and 22 months of supplementation with PE. Results are expressed as mean ± SEM (n = 5–6 per group). * p < 0.05, ** p < 0.01.
Figure 1. Effect of PE administration on glutathione redox status in mice’s blood. The figure shows (A) GSH, (B) GSSG, and (C) GSH/GSSG levels in blood in mice after 18 months, 22 months, and 22 months of supplementation with PE. Results are expressed as mean ± SEM (n = 5–6 per group). * p < 0.05, ** p < 0.01.
Antioxidants 12 01991 g001
Figure 2. Effect of PE supplementation on functional studies. (A) Maximum force on the grip strength test; (B) Running time on treadmill; (C) Running speed on treadmill; (D) Motor coordination on the rotarod test; (E) Sensory motor evaluation on the ladder-climbing test. Results are expressed as mean ± SD (n = 8–9 per group). * p < 0.05, ** p < 0.01 and *** p < 0.005. (ns: not significant).
Figure 2. Effect of PE supplementation on functional studies. (A) Maximum force on the grip strength test; (B) Running time on treadmill; (C) Running speed on treadmill; (D) Motor coordination on the rotarod test; (E) Sensory motor evaluation on the ladder-climbing test. Results are expressed as mean ± SD (n = 8–9 per group). * p < 0.05, ** p < 0.01 and *** p < 0.005. (ns: not significant).
Antioxidants 12 01991 g002
Figure 3. Effect of PE supplementation on weight loss. Data regarding weight loss are expressed as percentage of weight loss after 4 months of supplementation with PE. Results are expressed as mean ± SEM (n = 8–9 per group). * p < 0.05.
Figure 3. Effect of PE supplementation on weight loss. Data regarding weight loss are expressed as percentage of weight loss after 4 months of supplementation with PE. Results are expressed as mean ± SEM (n = 8–9 per group). * p < 0.05.
Antioxidants 12 01991 g003
Figure 4. Effect of PE on lipid peroxidation. MDA levels (µmol/mg protein) were measured by HPLC in cerebellum homogenates (A) and in brain cortex homogenates (B) of mice at 10 and 22 months of age. Results are expressed as mean ± SEM (n = 4–7 per group). * p < 0.05. (ns: not significant).
Figure 4. Effect of PE on lipid peroxidation. MDA levels (µmol/mg protein) were measured by HPLC in cerebellum homogenates (A) and in brain cortex homogenates (B) of mice at 10 and 22 months of age. Results are expressed as mean ± SEM (n = 4–7 per group). * p < 0.05. (ns: not significant).
Antioxidants 12 01991 g004
Figure 5. The effect of PE supplementation on the expression of aging biomarkers in cerebellum. (A) C1qa, (B) Ctss, (C) Lzp-s, (D) Gfap, (E) C4b. Results are expressed as mean ± SEM (n = 7–10 per group). * p < 0.05, ** p < 0.01.
Figure 5. The effect of PE supplementation on the expression of aging biomarkers in cerebellum. (A) C1qa, (B) Ctss, (C) Lzp-s, (D) Gfap, (E) C4b. Results are expressed as mean ± SEM (n = 7–10 per group). * p < 0.05, ** p < 0.01.
Antioxidants 12 01991 g005
Figure 6. The effect of pomegranate extract supplementation on the expression of aging biomarkers in cortex. (A) C1qa, (B) Ctss, (C) Lzp-s, (D) Gfap, (E) C4b. Results are expressed as mean ± SEM (n = 5–6 per group). * p < 0.05, ** p < 0.01.
Figure 6. The effect of pomegranate extract supplementation on the expression of aging biomarkers in cortex. (A) C1qa, (B) Ctss, (C) Lzp-s, (D) Gfap, (E) C4b. Results are expressed as mean ± SEM (n = 5–6 per group). * p < 0.05, ** p < 0.01.
Antioxidants 12 01991 g006
Figure 7. The effect of PE supplementation on the expression of B2m in cerebellum (A) and brain cortex (B). Results are expressed as mean ± SEM (n = 5–6 per group). * p < 0.05.
Figure 7. The effect of PE supplementation on the expression of B2m in cerebellum (A) and brain cortex (B). Results are expressed as mean ± SEM (n = 5–6 per group). * p < 0.05.
Antioxidants 12 01991 g007
Figure 8. Connections between cell processes and six aging biomarkers. The solid line means expression, and the dashed line means regulation. Relations are colored by effect. Red represents a negative effect, and green represents a positive effect.
Figure 8. Connections between cell processes and six aging biomarkers. The solid line means expression, and the dashed line means regulation. Relations are colored by effect. Red represents a negative effect, and green represents a positive effect.
Antioxidants 12 01991 g008
Figure 9. The effect of PE supplementation on the expression of apoptosis biomarkers in cerebellum and cortex. (A) Casp3, (B) Casp8, and (C) Casp9 in cerebellum and (D) Casp3, (E) Casp8, and (F) Casp9 in cortex. Results are expressed as mean ± SEM (n = 6–8 per group). * p < 0.05, ** p < 0.01. (ns: not significant).
Figure 9. The effect of PE supplementation on the expression of apoptosis biomarkers in cerebellum and cortex. (A) Casp3, (B) Casp8, and (C) Casp9 in cerebellum and (D) Casp3, (E) Casp8, and (F) Casp9 in cortex. Results are expressed as mean ± SEM (n = 6–8 per group). * p < 0.05, ** p < 0.01. (ns: not significant).
Antioxidants 12 01991 g009
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

Verdú, D.; Valls, A.; Díaz, A.; Carretero, A.; Dromant, M.; Kuligowski, J.; Serna, E.; Viña, J. Pomegranate Extract Administration Reverses Loss of Motor Coordination and Prevents Oxidative Stress in Cerebellum of Aging Mice. Antioxidants 2023, 12, 1991. https://doi.org/10.3390/antiox12111991

AMA Style

Verdú D, Valls A, Díaz A, Carretero A, Dromant M, Kuligowski J, Serna E, Viña J. Pomegranate Extract Administration Reverses Loss of Motor Coordination and Prevents Oxidative Stress in Cerebellum of Aging Mice. Antioxidants. 2023; 12(11):1991. https://doi.org/10.3390/antiox12111991

Chicago/Turabian Style

Verdú, David, Alicia Valls, Ana Díaz, Aitor Carretero, Mar Dromant, Julia Kuligowski, Eva Serna, and José Viña. 2023. "Pomegranate Extract Administration Reverses Loss of Motor Coordination and Prevents Oxidative Stress in Cerebellum of Aging Mice" Antioxidants 12, no. 11: 1991. https://doi.org/10.3390/antiox12111991

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