Skip to main content

Proanthocyanidins supplemented diet alter anti-aging-markers and improved lifespan in Drosophila melanogaster model

Abstract

Background

It is unequivocally believed that phenolics and flavonoids from fruits and vegetables hold robust prevention potentials against age-related disease development through their abundant hydroxyl groups. This study explored the potential neuromuscular enhancement and anti-aging effects of dietary supplemented proanthocyanidins-rich fraction from Tamarindus indica on Drosophila melanogaster model. One- to three-day-old male and female D. melanogaster were fed with a proanthocyanidins-rich fraction-supplemented diet for 7 days at two different concentrations. Following the effective dose determination, longevity assay (rate of survival), behavioral assay (negative geotaxis and eclosion), and biochemical assays (aging and antioxidant enzymes activities) were conducted to assess the fraction’s longevity, antioxidant, and anti-aging effects on D. melanogaster model.

Result

The results showed a significant (p < 0.05) improvement in the rate of emergence and lifespan of the flies fed with proanthocyanidins-rich fraction-supplemented diet at both concentrations (1.5 mg/g and 2.5 mg/g) compared to the normal control. A significant decrease in acetylcholinesterase (AChE) activity and the level of caspase-3 and caspase-9 were observed in the D. melanogaster flies fed with the fraction-containing diet when compared with the normal control. The supplemented diet also significantly increases the activity of catalase, superoxide dismutase (SOD), and glutathione-s-transferase (GST) in a concentration-dependent manner but not nicotinamide quinone oxidoreductase one (NQO1) in D. melanogaster upon comparison with the normal control.

Conclusion

The observable changes in the experiment were attributed to the T. indica-derived proanthocyanidins, flavonoids with robust biological activities. The flavonoid-rich fraction proved its potential by enhancing the antioxidant system in D. melanogaster via the increase in the activities of some of the phase II antioxidant enzymes. The present study provides more insights into the wider perspectives of societies on the use of plant-derived natural compounds as the potential approach toward prevention against aging and age-related morbidities which enhance wellness and the quality of life in humans and animals.

1 Background

Despite its complexity, aging is considered a gradual accumulation of changes over time that are associated with increased vulnerability to morbidities and mortality [1]. As the global population ages, neurodegenerative diseases like Alzheimer’s and Parkinson’s become increasingly common, posing a serious threat to human health [2, 3]. Mitochondria play a pivotal role in generating free radicals such as reactive oxygen (ROS) and nitrogen species (RNS) that alter the redox status of the body [3]. Progressive oxidation of macromolecules generates large amounts of ROS to pathological levels that trigger mitochondrial damage through oxidative stress with consequent apoptotic cell death [3, 4]. The production of ROS and the subsequent response to oxidative stress have been established as important factors in the determination of longevity [5, 6].

The process of aging is a multifaceted molecular phenomenon that is influenced by a variety of molecular pathways and biochemical occurrences, which are influenced by both genetic and environmental factors [7]. Aging can be specifically described as a gradual decrease in functional capacity and stress resistance over time, which is accompanied by an increased likelihood of experiencing illness and death [8]. The effects relate to the progressive accumulation of stressors associated with aging, leading to the gradual deterioration of biomolecules and subsequent disruption of cellular homeostasis [9]. However, previous studies demonstrated that genetic or dietary interventions have the potential to extend the lifespan of a wide range of organisms, suggesting that it is possible to delay mortality through such interventional approaches [10,11,12].

Natural compounds are a vast collection of structurally diverse scaffolds that hold great potential as candidate chemical entities for addressing the significant healthcare challenge of extending health-spans and/or slowing down the aging process [13, 14]. One of the medicinal plants rich in bioactive phytochemicals is Tamarindus indica commonly called Tamarin tree. T. indica holds significant dietary importance in sub-Saharan Africa and possesses considerable medicinal attributes [15, 16]. The fruit contains a variety of bioactive phytochemicals, such as alkaloids, phenolic, and bioflavonoids [16]. The primary focus of this research has been on proanthocyanidins, the principal bioflavonoid found in the fruit part of the plant. Several studies have investigated its medicinal effect in various model organisms and reported its potential to be considered for clinical trials [17, 18].

Drosophila melanogaster is among the most influential model organism in biomedical research, and it has been used extensively for biochemical research such as molecular mechanisms that underline human diseases [19,20,21]. The model has revealed a noteworthy resemblance in neurotoxicity between Homo sapiens (humans) and D. melanogaster (fruit flies) [22]. Researchers took advantage of the flies’ simple neural network for the exploration of antioxidant activities of bioactive compounds with potential neuroprotective activities [23]. These and other reasons strengthen the use of the fly model to unveil the mystery of life at a molecular level and screen the potential therapeutic agents [24]. In this study, we focused on evaluating the anti-aging activity of proanthocyanidins-rich fraction from T. indica and its ability to enhance longevity in a D. melanogaster model.

2 Methods

2.1 Collection of sample and preparation

A sample of the whole fruit of Tamarin was collected from Zaria main market of Kaduna State Nigeria and was identified by a botanist with a Boucher number 5451. Upon drying and removal of the shelves, the pulp parts were crushed into finely powdered particles using a stainless steel blender. The powdered sample was soaked in ethanol for 72 h. The filtrate was reduced to dryness by a rotary evaporator, and the percentage yield was 8.2% w/w, which was kept in the refrigerator until needed.

2.2 Stocking and culturing of Drosophila melanogaster

Drosophila melanogaster (Harwich strain) was donated by the College of Medicine, University of Ibadan, Nigeria. The flies originated from the National Species Stock Center, Bowling Green, Ohio, USA, and were grown in the Drosophila Research Laboratory, Department of Biochemistry, Kaduna State University. They were maintained at the respective standard temperature (24 ± 2 °C) and relative humidity (60 – 70%), under 12 h of lightness/darkness cycle conditions on a cornmeal diet containing 0.08% w/v methylparaben, 1% w/v agar–agar, 1% w/v brewer’s yeast, and 2% w/v sucrose.

2.3 Experimental design

Two- to three-day-old male and female flies were placed into three separate groups, namely normal control (diet without fraction), treatment group 1 (1.5 mg proanthocyanidins / g diet) and treatment group 2 (2.5 mg proanthocyanidins / g diet) as the most effective doses of the fraction. Each group comprised three replicates containing 100 flies and were fed for seven days.

2.4 Behavioral assays

2.4.1 Longevity assay

To ascertain the impact of proanthocyanidins-rich fraction on the lifespan of experimental D. melanogaster, a total of 100 flies per vial in triplicates were subjected to seven days treatment with or without a proanthocyanidins-rich fraction at 1.5 and 2.5 mg/g diet. Daily mortality of the flies was observed and recorded for seventy-seven (77) days. The survival rate was analyzed using GraphPad Prism and presented in the result section.

2.4.2 Negative geotaxis assay

Locomotor activity of D. melanogaster supplemented with proanthocyanidins-rich fraction at two concentrations (1.5 and 2.5 mg/g diet) were evaluated using a negative geotaxis assay as described by [25] with modifications. Briefly, following anesthesia, a total of twenty (20) flies out of the 100 from each experimental vial were picked and placed into a graduated column of 15 cm in height and 1.5 cm in diameter. The 8 cm of the column was considered a threshold and was marked, and the number of flies that traversed the line and those that remained at the bottom within 8 s were recorded. The experiment was conducted three times per vial at the interval of 1 min between readings, and the data were analyzed and presented.

2.4.3 Emergence rate determination

The rate of flies’ offspring emergence in the ADFP fraction treatment group was evaluated as previously described by [26].

2.5 Evaluation of aging-related markers in Drosophila melanogaster

2.5.1 Determination of total protein and estimation of Caspase-3 and Caspase-9 levels

The levels of caspase-3 and caspase-9 were estimated spectrophotometrically using a GenScript colorimetric assay kit (GenScript, Piscataway, NJ, USA). Upon completion of the experimental period, flies were homogenized in ice-cold PBS in a ratio of 1 fly to 10 µL PBS. To lyse the cells of the flies, about 50 µL cold lysis buffer containing 0.25 µL phenylmethanesulfonyl fluoride (PMSF) and 0.5 µL Dithiothreitol (DTT) was added to tubes containing the homogenate. The tubes were kept on ice for an hour with a thorough vortex at intervals of 8–12 min. The tube content was centrifuged at 10,000 rpm under 4 °C for one minute, and protein concentrations were measured using the Bradford assay in the collected supernatant thereafter as highlighted by [27]. Then, 200 µg of protein was added to a tube containing 50 µL reaction buffer comprising 0.25 µL PMSF and 0.5 µL DTT, and the contents were vortexed and allowed to stay on ice. The suspension was transferred to 96-well plates upon the addition of 5 µL caspases substrates. The plate was covered with aluminum foil and placed in the dark at physiological temperature for four (4) hours. The levels of caspase-3 and caspase-9 were estimated spectrophotometrically at 405 nm using a microplate reader (Universal Microplate Reader; Biotech, Inc).

2.5.2 Estimation of acetylcholinesterase activities

AChE activity was measured using the modified method of [28]. Briefly, a reaction mixture comprising 135 µL dH2O, 20 µL 10 mM DTNB, 20 µL 100 mM potassium phosphate buffer (pH 7.4), 5 µL homogenate sample, and 20 µL 8 mM ACh substrate was shaken vigorously. The activity of acetylcholinesterase was observed using a UV/visible spectrophotometer for 5 min (at an interval of 15 s) at 412 nm. The resulting data were corrected using protein content upon calculation with blank and sample blank.

2.6 Evaluation of antioxidants related markers Drosophila melanogaster

2.6.1 Estimation of catalase activity

A modified method reported by [29] was used to measure catalase activity. A reaction vessel containing 1800 µL of 50 mM phosphate buffer (pH 7.0), 20 µL of homogenate sample (1:50 dilution), and 180 µL of 300 mM H2O2 substrate. The disappearance of the substrate was monitored for 2 min at an interval of 10 s using a UV/visible spectrophotometer at 240 nm. The results were expressed as µmol of hydrogen peroxide (H2O2) consumed/min/mg of protein.

2.6.2 Estimation of superoxide dismutase (SOD) activity

The SOD activity was evaluated according to [29] methods with slight modification, by reducing nitrite formation in 40 min at 37 °C. The test was based on the SOD-mediated inhibition of nitrite formation from hydroxyl ammonium in the presence of O2 generators. The activity was measured spectrophotometrically at 550 nm, and the results were presented as the unit of the enzyme’s activity/mg of protein.

2.6.3 Estimation of glutathione-s-transferase activity

As demonstrated in [30], the activity of glutathione-s-transferase was determined by careful monitoring of the increase in the absorbance at 340 nm wavelength. The sample (50 µL) was added to the tube containing 20 µM each of 1-chloro-2,4-dinitrobenzene (CDNB) and a reduced form of glutathione. Optical density was taken at 406 nm for three minutes, and the result was expressed as the quantity of protein necessary to inhibit half of the quercetin auto-oxidation.

2.6.4 Estimation of N-quinone oxidoreductase one (NQO1) Activity

The activity of NQO1 enzyme was estimated spectrophotometrically using 2,6-Dichlorophenolindophenol (DCPIP) reduction method as described by [31] with some modifications. Briefly, the experiment was conducted kinetically on a microplate reader (Universal Microplate Reader; Biotech, Inc) at 600 nm, employing the enzyme’s substrate (DCPIP) and its inhibitor (dicumarol). To calculate the activity, the mean value of DCPIP reduction from 0 to 1 min in the presence of dicumarol was subtracted from the mean value of the reduction without the inhibitor. The NQO1 activity was expressed as mole DCPIP reduced/min/mg protein.

2.7 Statistical analysis

The data were presented as means ± standard deviations, and statistical analysis was performed using one-way analysis of variance (ANOVA) followed by Tukey post hoc test on GraphPad Prism (v 6) (San Diego, CA, USA). Differences in the results were considered statistically significant (p < 0.05) at 95% confidence level. All experiments were conducted in three replications (n = 3).

3 Results

3.1 Effect of proanthocyanidins-rich fraction-supplemented diet on longevity in D. melanogaster

The life span in D. melanogaster was improved significantly upon supplemented with a proanthocyanidins-rich fraction of T. indica for thirty days compared to the normal control that received only diet. The effect was in a concentration-dependent manner as the flies supplemented with 2.5 mg of the fraction per gram of diet showed a higher increase in the lifespan than those that received 1.5 mg/g diet, though the difference between the two concentrations was not remarkable as seen in Fig. 1.

Fig. 1
figure 1

Effect of proanthocyanidins-rich fraction-supplemented diet on longevity in D. melanogaster. All experiments were conducted in three replications (n = 3)

3.2 Effect of proanthocyanidins fraction on emergence rate and locomotor function in D. melanogaster

Figure 2 shows the effect of proanthocyanidins-rich fraction-supplemented diet on the rate of flies emergence and locomotor activities in D. melanogaster. The fraction enhanced significantly (p < 0.05) the emergence of new flies supplemented at both concentrations (1.5 and 2.5 mg fraction/g diet) compared to the normal control. The effects seemed to be in concentration-dependent manner as 2.5 mg/g appeared to be more effective than 1.5 mg/g as seen in Fig. 2a. Furthermore, the locomotor activity of the flies supplemented with proanthocyanidins-rich fraction increased significantly (p < 0.05) in a dose-dependent manner compared to the normal control group (Fig. 2b).

Fig. 2
figure 2

Effect of proanthocyanidins-rich fraction-supplemented diet on rate of emergence (a) and locomotor function (b) in D. melanogaster. The result for emergence rate was presented as percent pupation of new flies with and without the fraction and also that of negative geotaxis assay was presented as mean ± SD of the number of flies that cross an 8 cm in 8 s within a column. Differences in the results were considered statistically significant (p < 0.05) at 95% confidence level. All experiments were conducted in three replications (n = 3)

3.3 Effect of proanthocyanidins-rich fraction dietary supplement on aging-related enzymes’ activities and quantity in D. melanogaster

The effect of the fraction was further evaluated on aging-related enzymes where the fraction showed inhibitory activity on acetylcholinesterase in D. melanogaster. Only 2.5 mg of fraction/ g of diet revealed a significant (p < 0.05) difference but not the 1.5 mg/g when compared to the normal control (Fig. 3a). Likewise, Caspase-3 level dropped significantly (p < 0.05) in the flies that received proanthocyanidins-rich fraction at both concentrations. Again, the increase in the level of the enzyme occurs in a concentration-dependent manner (Fig. 3b). Similarly, the level of Caspase-9 was lowered by the fraction at 2.5 mg/g when compared to the normal control. However, the 1.5 mg/g fraction did not show any effect on the level of Capspase-9 in the flies fed with the supplemented diet (Fig. 3c).

Fig. 3
figure 3

Effect of proanthocyanidins-rich fraction-supplemented diet on the activity and level of acetylcholinesterase (a), Caspase-3 (b), and Caspase-9 (c) in D. melanogaster. The results were expressed as mean ± SD of the activity and quantity of the enzymes. Differences in the results were considered statistically significant (p < 0.05) at 95% confidence level. All experiments were conducted in three replications (n = 3)

3.4 Effect of proanthocyanidins-rich fraction supplement on antioxidants and age-related enzymes’ activities in D. melanogaster

Figure 4 shows the effect of proanthocyanidins-rich fraction dietary supplemented diet on selected antioxidant markers in D. melanogaster. A 2.5 mg/g of the fraction effectuated a significant (p < 0.05) increase in catalase activities in the flies when compared with the normal control group (flies that were fed with only diet without the fraction). However, there was no significant effect on the activity when the flies were fed with 1.5 mg/g supplemented diet (Fig. 4a). The result also revealed a significant (p < 0.05) increase in the activity of superoxide dismutase (SOD) in the flies upon fed with the fraction-supplemented diet compared to the normal control flies. The two concentrations of the supplemented fraction showed a similar pattern of effect on the SOD activity since there was no observable difference in their means (Fig. 4b). Similarly, we observed a significant (p < 0.05) increase in the activity of glutathione-s-transferase by the fraction in a concentration-dependent manner when compared with the normal control group. The increasing effect of 1.5 mg/g fraction on the enzyme’s activity was significantly lowered (p < 0.05) when the two were compared statistically (Fig. 4c). In contrast, the fraction-supplemented diet had no effect on the activity of nicotinamide quinone oxidoreductase one (NQO1) at both concentrations (Fig. 4d).

Fig. 4
figure 4

Effect of proanthocyanidins-rich fraction supplement on some selected antioxidant and anti-aging enzymes activities; Catalase (a), superoxide dismutase (b), glutathione-s-transferase (c) and quinone oxygenase one (d) in D. melanogaster. Differences in the results were considered statistically significant (p < 0.05) at 95% confidence level. All experiments were conducted in three replications (n = 3)

4 Discussion

The effects of various flavonoids and polyphenols on lifespan extension, health improvement, and aging-related morbidities have been investigated using Drosophila melanogaster model [32,33,34]. However, the specific impact of proanthocyanidins on longevity, redox status, and aging conditions remained to be elucidated. Proanthocyanidins demonstrated numerous health benefits on various model organisms [18, 35]. The role of oxidative stress has been suggested in the process of aging and the development of various age-related diseases [36]. Oxidative stress is characterized by a disparity between the generation of ROS and RNS, and the ability of the cellular antioxidant defense system to counteract the generated species [37]. This condition arises when there is an elevation in ROS/RNS levels or a decline in antioxidant capacity [37]. Meanwhile, the aging process is influenced by the detrimental effects on lipids, proteins, and DNA in different tissues [38]. According to the findings of this study, the inclusion of proanthocyanidins-rich fraction in the diet has been shown to improve the lifespan of the flies above those that have not received the fraction (Fig. 1). The current findings align with prior research that demonstrated the life-extending effects of a phenolic and other forms of flavonoids from fruits [39,40,41]. Phenolics and flavonoids are robust bioactive compounds found primarily in vegetables and several other fruits including T. indica [42]. Their demonstrated antioxidant properties are attributed to the hydroxyl groups on their aromatic ring structures and the presence of highly activated carbon atom between the two methoxyphenol rings [34, 42].

On the other hand, several studies have shown that the cholinergic system and intrinsic mitochondrial pathway play an important role in the pathophysiology of aging and other neurodegenerative diseases [43,44,45]. Acetylcholine (ACh), a cholinergic neurotransmitter, plays a crucial role in modulating cholinergic functions such as learning, memory, and locomotor activity [46, 47]. AChE, a serine protease, however, hydrolyses acetylcholine to choline and acetate, affecting cholinergic neurotransmission [47]. The cholinergic marker enzyme (AChE) is specific for the active state of cholinergic neurons, and it is crucial for maintaining acetylcholine levels at cholinergic neurons and responsible for acetylcholine degradation in the synaptic cleft [48]. AChE activities have been linked to other neurodegenerative diseases and the aging process [49]. When compared to the control, the proanthocyanidins-rich fraction-supplemented diet resulted in a significant decrease in AChE activity (Fig. 3a) and an increase in climbing activity (Fig. 2b), translating to neuromuscular strength enhancement in the experimental flies. These findings are consistent with previous reports on in vitro and in vivo findings [50,51,52,53,54]. Thus, in our study, the decrease in AChE activity after dietary proanthocyanidins supplementation could lead to an increase in acetylcholine levels in the synaptic cleft and, as a result, increase cholinergic neurotransmission efficiency in the flies.

Furthermore, the impact of the fraction on caspase-3 and caspase-9 levels in the flies also signified its key role in mitigating the development of age-related diseases. Caspase-3 and -9 are the primary markers for the mitochondrial intrinsic pathway that contribute to cellular senescence with eventual cell death, leading to aging and its associated complications. Study has shown that phenolics enhance the longevity of organisms by decreasing the level of caspases and consequent disruption of the intrinsic pathway [55]. Our findings revealed a significant decreasing effect on the two caspases level by proanthocyanidins-rich fraction-supplemented diet in D. melanogaster (Fig. 3b, c). The present findings are in line with documented evidences that reported the inhibitory effects of phenolic on caspases level, activities, and their expression level in pathophysiological age-related morbidities [56,57,58].

The current investigation demonstrates that the inclusion of proanthocyanidins-rich fraction in the diet also leads to an enhancement in the antioxidant status of D. melanogaster, as depicted in Fig. 4. Multiple studies have demonstrated the antioxidative properties of phenolics, flavonoids and other important plant-derived active compounds [55, 59,60,61]. The effective strategies employed by organisms to mitigate the harmful effects of reactive oxygen and nitrogen species (ROS and RNS) involve the enzymatic activity of numerous markers including catalase, superoxide dismutase (SOD), glutathione-s-transferase, nicotinamide quinone oxidoreductase (NQO1) and others [16, 41, 62]. SOD aids in the transformation of superoxide anion into less harmful compounds, which are subsequently converted into water through the catalytic activity of catalase [63]. The significance of this mechanism in the lifespan of D. melanogaster has been described in several reports [63, 64]. Previous studies have demonstrated that the genome of D. melanogaster contains individual single regions that exhibit the ability to enhance SOD and catalase activities, along with four regions that possess the capacity to suppress their respective activity as well [65, 66].

It is currently observed that feeding D. melanogaster with proanthocyanidins-rich fraction results in a significant alteration in the activities of SOD and catalase in comparison with the control group (Fig. 4a, b). Again, our findings coincide with the results presented in [67], which reported an increase in SOD and catalase activities in fruit flies fed with polyphenolic (curcumin) compared to those on the control diet. Another important antioxidant marker is glutathione-s-transferase (GST) which represents a phase II group of multifunctional enzymes characterized by the presence of cysteine-rich domains [68]. The catalytic activity of GST in the conjugation of glutathione (GSH) with electrophilic molecules is a crucial process in the detoxification of xenobiotics that could disrupt redox status in living organisms [69]. The present findings demonstrated the positive effect of proanthocyanidins-rich fraction on GST activities in flies fed with the fraction compared to the normal control group. GST activity was increased significantly by curcumin even within a toxic environment. It also neutralized the noxious effect of the ecotoxic agent that alters redox status of an organism [67]. However, the present findings revealed the ineffectiveness of the supplemented diet on the activities of NQO1, which is contrary to the previous reports on the impact of the enzyme, where NQO1 and other phase II enzymes demonstrated strong antioxidant activities [70, 71]. Our finding strengthened the existence of isoforms of the protein in some organisms and their respective physiological role in different model organisms.

5 Conclusion

Collectively, our findings indicate that proanthocyanidins-rich fraction slows down aging process in flies. It is posited that the anti-aging capacity of the flavonoid-rich fraction can be attributed to its antioxidative properties, as indicated by the observed increase in the activities of some phase II antioxidant enzymes with the consequent decrease in acetylcholinesterase activity and level of mitochondrial intrinsic pathway caspases in D. melanogaster. Therefore, proanthocyanidins-rich fraction of T. indica origin could be considered a potential anti-aging intervention and may provide protection against neurological-related disorders particularly those associated with oxidative stress, such as Parkinson’s and Alzheimer’s diseases. Moreover, the findings of the current study provide additional evidence supporting the effectiveness of Drosophila melanogaster as a valuable model organism for exploring potential therapeutic interventions that hold promise in the management of neurodegenerative disorders.

Availability of data and materials

All data are available in the present manuscript.

Abbreviations

AChE:

Acetylcholinesterase

Ach:

Acetylcholine

GST:

Glutathione-s-transferase

SOD:

Superoxide dismutase

NQO1:

Nicotinamide quinone oxidoreductase one

RNS:

Reactive oxygen species

ROS:

Reactive oxygen species

PMSF:

Phenylmethanesulfonyl fluoride

DTT:

Dithiothreitol

UV:

Ultraviolet

H2O2 :

Hydrogen peroxide

O2 :

Oxygen

DCPIP:

Dichlorophenolindophenol

ANOVA:

Analysis of variance

References

  1. Chung KW, Kim DH, Jung HJ, Arulkumar R, Chung HY, Yu BP (2023) Chronic inflammation as an underlying mechanism of ageing and ageing-related diseases. Subcell Biochem 103:31–44. https://doi.org/10.1007/978-3-031-26576-1_3

    Article  PubMed  Google Scholar 

  2. Bouvier DS, Fixemer S, Heurtaux T, Jeannelle F, Frauenknecht KBM, Mittelbronn M (2022) The multifaceted neurotoxicity of astrocytes in ageing and age-related neurodegenerative diseases: a translational perspective. Front Physiol 13:814889. https://doi.org/10.3389/fphys.2022.814889

    Article  PubMed  PubMed Central  Google Scholar 

  3. Bazzani V, Equisoain Redin M, McHale J, Perrone L, Vascotto C (2022) Mitochondrial DNA repair in neurodegenerative diseases and ageing. Int J Mol Sci. https://doi.org/10.3390/ijms231911391

    Article  PubMed  PubMed Central  Google Scholar 

  4. Zheng WQ, Zhang JH, Li ZH, Liu X, Zhang Y, Huang S, Li J, Zhou B, Eriani G, Wang ED, Zhou XL (2023) Mammalian mitochondrial translation infidelity leads to oxidative stress-induced cell cycle arrest and cardiomyopathy. Proc Natl Acad Sci U S A 120:e2309714120. https://doi.org/10.1073/pnas.2309714120

    Article  CAS  PubMed  Google Scholar 

  5. Vagasi CI, Vincze O, Patras L, Osvath G, Penzes J, Haussmann MF, Barta Z, Pap PL (2019) Longevity and life history coevolve with oxidative stress in birds. Funct Ecol 33:152–161. https://doi.org/10.1111/1365-2435.13228

    Article  PubMed  Google Scholar 

  6. Amorim JA, Coppotelli G, Rolo AP, Palmeira CM, Ross JM, Sinclair DA (2022) Mitochondrial and metabolic dysfunction in ageing and age-related diseases. Nat Rev Endocrinol 18:243–258. https://doi.org/10.1038/s41574-021-00626-7

    Article  PubMed  PubMed Central  Google Scholar 

  7. Sharma R, Diwan B (2023) Lipids and the hallmarks of ageing: from pathology to interventions. Mech Ageing Dev. https://doi.org/10.1016/j.mad.2023.111858

    Article  PubMed  Google Scholar 

  8. Zhang G, Devo P, O’Leary VB, Ovsepian SV (2023) Ageing perspective on cognitive outcomes from reproductive hormone adjustments. Heliyon 9:e19050. https://doi.org/10.1016/j.heliyon.2023.e19050

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Brandl C, Finger RP, Heid IM, Mauschitz MM (2023) Age-related macular degeneration in an ageing society: current epidemiological research. Klin Monbl Augenheilkd 240:1052–1059. https://doi.org/10.1055/a-2105-1064

    Article  PubMed  Google Scholar 

  10. Yuan R, Musters CJM, Zhu Y, Evans TR, Sun Y, Chesler EJ, Peters LL, Harrison DE, Bartke A (2020) Genetic differences and longevity-related phenotypes influence lifespan and lifespan variation in a sex-specific manner in mice. Aging Cell 19:e13263. https://doi.org/10.1111/acel.13263

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Molon M, Dampc J, Kula-Maximenko M, Zebrowski J, Molon A, Dobler R, Durak R, Skoczowski A (2020) Effects of temperature on lifespan of Drosophila melanogaster from different genetic backgrounds: links between metabolic rate and longevity. Insects. https://doi.org/10.3390/insects11080470

    Article  PubMed  PubMed Central  Google Scholar 

  12. Jin K, Wilson KA, Beck JN, Nelson CS, Brownridge GW 3rd, Harrison BR, Djukovic D, Raftery D, Brem RB, Yu S, Drton M, Shojaie A, Kapahi P, Promislow D (2022) Correction: Genetic and metabolomic architecture of variation in diet restriction-mediated lifespan extension in Drosophila. PLoS Genet 18:e1010199. https://doi.org/10.1371/journal.pgen.1010199

    Article  PubMed  PubMed Central  Google Scholar 

  13. Herath H, Taki AC, Sleebs BE, Hofmann A, Nguyen N, Preston S, Davis RA, Jabbar A, Gasser RB (2021) Advances in the discovery and development of anthelmintics by harnessing natural product scaffolds. Adv Parasitol 111:203–251. https://doi.org/10.1016/bs.apar.2020.10.002

    Article  PubMed  Google Scholar 

  14. Gomez-Garcia A, Medina-Franco JL (2022) Progress and impact of Latin American natural product databases. Biomolecules. https://doi.org/10.3390/biom12091202

    Article  PubMed  PubMed Central  Google Scholar 

  15. Abdelnaby A, Abdel-Aleem N, Mansour A, Abdelkader A, Ibrahim AN, Sorour SM, Elgendy E, Bayoumi H, Abdelrahman SM, Ibrahim SF, Alsaati I, Abdeen A (2022) The combination of tamarindus indica and coenzyme q10 can be a potential therapy preference to attenuate cadmium-induced hepatorenal injury. Front Pharmacol 13:954030. https://doi.org/10.3389/fphar.2022.954030

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Saxena M, Prabhu SV, Mohseen M, Pal AK, Alarifi S, Gautam N, Palanivel H (2022) Antidiabetic effect of Tamarindus indica and Momordica charantia and downregulation of tet-1 gene expression by Saroglitazar in glucose feed adipocytes and their involvement in the type 2 diabetes-associated inflammation in vitro. Biomed Res Int 2022:9565136. https://doi.org/10.1155/2022/9565136

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Xie Y, Deng Q, Guo M, Li X, Xian D, Zhong J (2023) Proanthocyanidins: a novel approach to Henoch–Schonlein purpura through balancing immunity and arresting oxidative stress via TLR4/MyD88/NF-kappaB signaling pathway (Review). Exp Ther Med 25:300. https://doi.org/10.3892/etm.2023.11999

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Chen Y, Yang Z, He X, Zhu W, Wang Y, Li J, Han Z, Wen J, Liu W, Yang Y, Zhang K (2023) Proanthocyanidins inhibited colorectal cancer stem cell characteristics through Wnt/beta-catenin signaling. Environ Toxicol. https://doi.org/10.1002/tox.23924

    Article  PubMed  Google Scholar 

  19. Mishra E, Thakur MK (2023) Mitophagy: a promising therapeutic target for neuroprotection during ageing and age-related diseases. Br J Pharmacol 180:1542–1561. https://doi.org/10.1111/bph.16062

    Article  CAS  PubMed  Google Scholar 

  20. Mishra S, Sharma N, Lone SR (2023) Understanding the impact of sociosexual interactions on sleep using Drosophila melanogaster as a model organism. Front Physiol 14:1220140. https://doi.org/10.3389/fphys.2023.1220140

    Article  PubMed  PubMed Central  Google Scholar 

  21. Baenas N, Wagner AE (2022) Drosophila melanogaster as a model organism for obesity and type-2 diabetes mellitus by applying high-sugar and high-fat diets. Biomolecules. https://doi.org/10.3390/biom12020307

    Article  PubMed  PubMed Central  Google Scholar 

  22. Rouka E, Gourgoulianni N, Lupold S, Hatzoglou C, Gourgoulianis KI, Zarogiannis SG (2022) Prediction and enrichment analyses of the Homo sapiens-Drosophila melanogaster COPD-related orthologs: potential for modeling of human COPD genomic responses with the fruit fly. Am J Physiol Regul Integr Comp Physiol 322:R77–R82. https://doi.org/10.1152/ajpregu.00092.2021

    Article  CAS  PubMed  Google Scholar 

  23. Deolankar SC, Najar MA, Ramesh P, Kanichery A, Kudva AK, Raghu SV, Prasad TSK (2023) Discovery of molecular networks of neuroprotection conferred by brahmi extract in abeta(42)-induced toxicity model of Drosophila melanogaster using a quantitative proteomic approach. Mol Neurobiol 60:303–316. https://doi.org/10.1007/s12035-022-03066-0

    Article  CAS  PubMed  Google Scholar 

  24. Luna EM, Freitas TS, Campina FF, Costa MS, Rocha JE, Cruz RP, Sena Junior DL, Silveira ZS, Macedo NS, Pinheiro JCA, Pereira-Junior FN, Lisboa MAN, Cruz GV, Calixto Junior JT, Teixeira AMR, Coutinho HDM (2021) Evaluation of phytochemical composition, toxicity in Drosophila melanogaster and effects on antibiotics modulation of Plathymenia reticulata Benth extract. Toxicol Rep 8:732–739. https://doi.org/10.1016/j.toxrep.2021.03.020

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Nichols CD, Becnel J, Pandey UB (2012) Methods to assay Drosophila behavior. J Vis Exp 7:e3795. https://doi.org/10.3791/3795

    Article  Google Scholar 

  26. Arias AM (2008) Drosophila melanogaster and the development of biology in the 20th century. Drosophila Methods Protocols. https://doi.org/10.1007/978-1-59745-583-1_1

    Article  Google Scholar 

  27. Kruger NJ (2009) The Bradford method for protein quantitation. The protein protocols handbook, 17–24. https://doi.org/10.1385/0-89603-268-X:9

  28. Worek F, Mast U, Kiderlen D, Diepold C, Eyer P (1999) Improved determination of acetylcholinesterase activity in human whole blood. Clin Chim Acta 288:73–90. https://doi.org/10.1016/S0009-8981(99)00144-8

    Article  CAS  PubMed  Google Scholar 

  29. Vives-Bauza C, Starkov A, Garcia-Arumi E (2007) Measurements of the antioxidant enzyme activities of superoxide dismutase, catalase, and glutathione peroxidase. Methods Cell Biol 80:379–393. https://doi.org/10.1016/S0091-679X(06)80019-1

    Article  CAS  PubMed  Google Scholar 

  30. Prohaska JR (1980) The glutathione peroxidase activity of glutathione S-transferases. Biochim Biophys Acta BBA Enzymol 611:87–98. https://doi.org/10.1016/0005-2744(80)90045-5

    Article  CAS  Google Scholar 

  31. Siegel D, Kepa JK, Ross D (2007) Biochemical and genetic analysis of NAD (P) H: quinone oxidoreductase 1 (NQO1). Curr Protocols Toxicol 32:1–21. https://doi.org/10.1002/0471140856.tx0422s32

    Article  Google Scholar 

  32. Guo C, Zhang H, Guan X, Zhou Z (2019) The anti-aging potential of neohesperidin and its synergistic effects with other citrus flavonoids in extending chronological lifespan of saccharomyces cerevisiae BY4742. Molecules. https://doi.org/10.3390/molecules24224093

    Article  PubMed  PubMed Central  Google Scholar 

  33. Panchenko AV, Tyndyk ML, Fedoros EI, Maydin MA, Semenov AL, Gubareva EA, Golubev AG, Anisimov VN (2019) Comparative analysis of experimental data about the effects of various polyphenols on lifespan and aging. Adv Gerontol 32:325–330

    CAS  PubMed  Google Scholar 

  34. Wang S, Xue J, Zhang S, Zheng S, Xue Y, Xu D, Zhang X (2020) Composition of peony petal fatty acids and flavonoids and their effect on Caenorhabditis elegans lifespan. Plant Physiol Biochem 155:1–12. https://doi.org/10.1016/j.plaphy.2020.06.029

    Article  CAS  PubMed  Google Scholar 

  35. Zhang X, Song X, Hu X, Chen F, Ma C (2023) Health benefits of proanthocyanidins linking with gastrointestinal modulation: an updated review. Food Chem 404:134596. https://doi.org/10.1016/j.foodchem.2022.134596

    Article  CAS  PubMed  Google Scholar 

  36. Kristiani L, Kim Y (2023) The interplay between oxidative stress and the nuclear lamina contributes to laminopathies and age-related diseases. Cells. https://doi.org/10.3390/cells12091234

    Article  PubMed  PubMed Central  Google Scholar 

  37. Korovesis D, Rubio-Tomas T, Tavernarakis N (2023) Oxidative stress in age-related neurodegenerative diseases: an overview of recent tools and findings. Antioxidants. https://doi.org/10.3390/antiox12010131

    Article  PubMed  PubMed Central  Google Scholar 

  38. Adeyemi KD, Shittu RM, Sabow AB, Ebrahimi M, Sazili AQ (2016) Influence of diet and Postmortem ageing on oxidative stability of lipids, myoglobin and myofibrillar proteins and quality attributes of Gluteus Medius muscle in goats. PLoS ONE 11:e0154603. https://doi.org/10.1371/journal.pone.0154603

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Wang TH, Tseng WC, Leu YL, Chen CY, Lee WC, Chi YC, Cheng SF, Lai CY, Kuo CH, Yang SL, Yang SH, Shen JJ, Feng CH, Wu CC, Hwang TL, Wang CJ, Wang SH, Chen CC (2022) The flavonoid corylin exhibits lifespan extension properties in mouse. Nat Commun 13:1238. https://doi.org/10.1038/s41467-022-28908-2

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Warnsmann V, Hainbuch S, Osiewacz HD (2018) Quercetin-induced lifespan extension in Podospora anserina requires methylation of the flavonoid by the o-methyltransferase PaMTH1. Front Genet 9:160. https://doi.org/10.3389/fgene.2018.00160

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Yang F, Xiu M, Yang S, Li X, Tuo W, Su Y, He J, Liu Y (2021) Extension of Drosophila lifespan by Astragalus polysaccharide through a mechanism dependent on antioxidant and insulin/igf-1 signaling. Evid Based Complement Alternat Med 2021:6686748. https://doi.org/10.1155/2021/6686748

    Article  PubMed  PubMed Central  Google Scholar 

  42. Fagbemi KO, Aina DA, Adeoye-Isijola MO, Naidoo KK, Coopoosamy RM, Olajuyigbe OO (2022) Bioactive compounds, antibacterial and antioxidant activities of methanol extract of Tamarindus indica Linn. Sci Rep 12:9432. https://doi.org/10.1038/s41598-022-13716-x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Abate G, Vezzoli M, Sandri M, Rungratanawanich W, Memo M, Uberti D (2020) Mitochondria and cellular redox state on the route from ageing to Alzheimer’s disease. Mech Ageing Dev 192:111385. https://doi.org/10.1016/j.mad.2020.111385

    Article  CAS  PubMed  Google Scholar 

  44. Bamshad C, Najafi-Ghalehlou N, Pourmohammadi-Bejarpasi Z, Tomita K, Kuwahara Y, Sato T, Feizkhah A, Roushnadeh AM, Roudkenar MH (2023) Mitochondria: How eminent in ageing and neurodegenerative disorders? Hum Cell 36:41–61. https://doi.org/10.1007/s13577-022-00833-y

    Article  CAS  PubMed  Google Scholar 

  45. Booth LK, Redgrave RE, Tual-Chalot S, Spyridopoulos I, Phillips HM, Richardson GD (2023) Heart disease and ageing: the roles of senescence, mitochondria, and telomerase in cardiovascular disease. Subcell Biochem 103:45–78. https://doi.org/10.1007/978-3-031-26576-1_4

    Article  PubMed  Google Scholar 

  46. Graur A, Sinclair P, Schneeweis AK, Pak DT, Kabbani N (2023) The human acetylcholinesterase C-terminal T30 peptide activates neuronal growth through alpha 7 nicotinic acetylcholine receptors and the mTOR pathway. Sci Rep 13:11434. https://doi.org/10.1038/s41598-023-38637-1

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Nascimento LA, Nascimento ECM, Martins JBL (2022) In silico study of tacrine and acetylcholine binding profile with human acetylcholinesterase: docking and electronic structure. J Mol Model 28:252. https://doi.org/10.1007/s00894-022-05252-2

    Article  CAS  PubMed  Google Scholar 

  48. Taylor P, Shyong YJ, Samskey N, Ho KY, Radic Z, Fenical W, Sharpless KB, Kovarik Z, Camacho-Hernandez GA (2021) Ligand design for human acetylcholinesterase and nicotinic acetylcholine receptors, extending beyond the conventional and canonical. J Neurochem 158:1217–1222. https://doi.org/10.1111/jnc.15335

    Article  CAS  PubMed  Google Scholar 

  49. Lista S, Vergallo A, Teipel SJ, Lemercier P, Giorgi FS, Gabelle A, Garaci F, Mercuri NB, Babiloni C, Gaire BP, Koronyo Y, Koronyo-Hamaoui M, Hampel H, Nistico R, Neurodegeneration Precision Medicine I (2023) Determinants of approved acetylcholinesterase inhibitor response outcomes in Alzheimer’s disease: relevance for precision medicine in neurodegenerative diseases. Ageing Res Rev 84:101819. https://doi.org/10.1016/j.arr.2022.101819

    Article  CAS  PubMed  Google Scholar 

  50. Tallini LR, da Silva CR, Jung T, Alves EO, Baldin SL, Apel M, Timmers L, Rico EP, Bastida J, Zuanazzi JAS (2023) Acetylcholinesterase inhibition activity of Hippeastrum papilio (Ravenna) Van Scheepen (Amaryllidaceae) using zebrafish brain homogenates. Life. https://doi.org/10.3390/life13081721

    Article  PubMed  PubMed Central  Google Scholar 

  51. Durmaz L, Erturk A, Akyuz M, Polat Kose L, Uc EM, Bingol Z, Saglamtas R, Alwasel S, Gulcin I (2022) Screening of carbonic anhydrase, acetylcholinesterase, butyrylcholinesterase, and alpha-glycosidase enzyme inhibition effects and antioxidant activity of coumestrol. Molecules. https://doi.org/10.3390/molecules27103091

    Article  PubMed  PubMed Central  Google Scholar 

  52. Amirahmadi S, Hosseini M, Ahmadabady S, Akbarian M, Abrari K, Vafaee F, Rajabian A (2021) Folic acid attenuated learning and memory impairment via inhibition of oxidative damage and acetylcholinesterase activity in hypothyroid rats. Metab Brain Dis 36:2393–2403. https://doi.org/10.1007/s11011-021-00815-3

    Article  CAS  PubMed  Google Scholar 

  53. Cardoso ADS, Santos EGG, Lima ADS, Temeyer KB, Perez de Leon AA, Costa LMJ, Soares A (2020) Terpenes on Rhipicephalus (Boophilus) microplus: acaricidal activity and acetylcholinesterase inhibition. Vet Parasitol 280:109090. https://doi.org/10.1016/j.vetpar.2020.109090

    Article  CAS  PubMed  Google Scholar 

  54. Ferreira J, Santos S, Pereira H (2020) In Vitro screening for acetylcholinesterase inhibition and antioxidant activity of Quercus suber cork and corkback extracts. Evid Based Complement Alternat Med 2020:3825629. https://doi.org/10.1155/2020/3825629

    Article  PubMed  PubMed Central  Google Scholar 

  55. Suchowilska E, Bienkowska T, Stuper-Szablewska K, Wiwart M (2020) Concentrations of phenolic acids, flavonoids and carotenoids and the antioxidant activity of the grain, flour and bran of Triticum polonicum as compared with three cultivated wheat Species. Agriculture 10:591

    Article  CAS  Google Scholar 

  56. Ogunsuyi OB, Olagoke OC, Afolabi BA, Loreto JS, Ademiluyi AO, Aschner M, Oboh G, Barbosa NV, da Rocha JBT (2022) Effect of Solanum vegetables on memory index, redox status, and expressions of critical neural genes in Drosophila melanogaster model of memory impairment. Metab Brain Dis 37:729–741. https://doi.org/10.1007/s11011-021-00871-9

    Article  CAS  PubMed  Google Scholar 

  57. Kushalan S, D’Souza LC, Aloysius K, Sharma A, Hegde S (2022) Toxicity assessment of Curculigo orchioides leaf extract using drosophila melanogaster: a preliminary study. Int J Environ Res Public Health. https://doi.org/10.3390/ijerph192215218

    Article  PubMed  PubMed Central  Google Scholar 

  58. Ogunsuyi OB, Olagoke OC, Afolabi BA, Oboh G, Ijomone OM, Barbosa NV, da Rocha JBT (2022) Dietary inclusions of Solanum vegetables mitigate aluminum-induced redox and inflammation-related neurotoxicity in Drosophila melanogaster model. Nutr Neurosci 25:2077–2091. https://doi.org/10.1080/1028415X.2021.1933331

    Article  CAS  PubMed  Google Scholar 

  59. Muflihah YM, Gollavelli G, Ling YC (2021) Correlation study of antioxidant activity with phenolic and flavonoid compounds in 12 Indonesian indigenous herbs. Antioxidants. https://doi.org/10.3390/antiox10101530

    Article  PubMed  PubMed Central  Google Scholar 

  60. Pauliuc D, Dranca F, Oroian M (2020) Antioxidant activity, total phenolic content, individual phenolics and physicochemical parameters suitability for romanian honey authentication. Foods. https://doi.org/10.3390/foods9030306

  61. Jaafaru MS, Abd Karim NA, Enas ME, Rollin P, Mazzon E, Abdull Razis AF (2018) Protective effect of glucosinolates hydrolytic products in neurodegenerative diseases (NDDs). Nutrients. https://doi.org/10.3390/nu10050580

    Article  PubMed  PubMed Central  Google Scholar 

  62. Qiu D, Song S, Chen N, Bian Y, Yuan C, Zhang W, Duan H, Shi Y (2023) NQO1 alleviates renal fibrosis by inhibiting the TLR4/NF-kappaB and TGF-beta/Smad signaling pathways in diabetic nephropathy. Cell Signal 108:110712. https://doi.org/10.1016/j.cellsig.2023.110712

    Article  CAS  PubMed  Google Scholar 

  63. Forman HJ, Zhang H (2021) Targeting oxidative stress in disease: promise and limitations of antioxidant therapy. Nat Rev Drug Discov 20:689–709. https://doi.org/10.1038/s41573-021-00233-1

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. He J, Li X, Yang S, Li Y, Lin X, Xiu M, Li X, Liu Y (2021) Gastrodin extends the lifespan and protects against neurodegeneration in the Drosophila PINK1 model of Parkinson’s disease. Food Funct 12:7816–7824. https://doi.org/10.1039/d1fo00847a

    Article  CAS  PubMed  Google Scholar 

  65. Liu JK (2022) Antiaging agents: safe interventions to slow aging and healthy life span extension. Nat Prod Bioprospect 12:18. https://doi.org/10.1007/s13659-022-00339-y

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Deepashree S, Shivanandappa T, Ramesh SR (2022) Genetic repression of the antioxidant enzymes reduces the lifespan in Drosophila melanogaster. J Comp Physiol B 192:1–13. https://doi.org/10.1007/s00360-021-01412-7

    Article  CAS  PubMed  Google Scholar 

  67. Abolaji AO, Fasae KD, Iwezor CE, Aschner M, Farombi EO (2020) Curcumin attenuates copper-induced oxidative stress and neurotoxicity in Drosophila melanogaster. Toxicol Rep 7:261–268. https://doi.org/10.1016/j.toxrep.2020.01.015

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Semaniuk UV, Gospodaryov DV, Strilbytska OM, Kucharska AZ, Sokół-Łętowska A, Burdyliuk NI, Storey KB, Bayliak MM, Lushchak O (2022) Chili-supplemented food decreases glutathione-S-transferase activity in Drosophila melanogaster females without a change in other parameters of antioxidant system. Redox Rep 27:221–229. https://doi.org/10.1080/13510002.2022.2123884

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Potega A (2022) Glutathione-mediated conjugation of anticancer drugs: an overview of reaction mechanisms and biological significance for drug detoxification and bioactivation. Molecules. https://doi.org/10.3390/molecules27165252

    Article  PubMed  PubMed Central  Google Scholar 

  70. El-Akad RH, Abou Zeid AH, El-Rafie HM, Kandil ZA, Farag MA (2021) Comparative metabolites profiling of Caryota mitis & Caryota urens via UPLC/MS and isolation of two novel in silico chemopreventive flavonoids. J Food Biochem 45:e13648. https://doi.org/10.1111/jfbc.13648

    Article  CAS  PubMed  Google Scholar 

  71. Zhao T, Zheng F, Liu Y, Khan A, Wang Z, Cheng G (2023) A comparative analysis of chemical constituents and antioxidant effects of Dendrobium fimbriatum hook fractions with different polarities. Int J Mol Sci. https://doi.org/10.3390/ijms241612646

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The author acknowledged the unwavering support from the chief technologist, all the technologists, technicians, and laboratory attendants in Drosophila and Molecular Biology Research Laboratories, Department of Biochemistry, Kaduna State University toward achieving this success.

Funding

The present study did not receive any specific financial support from public, commercial, or profit funding organizations.

Author information

Authors and Affiliations

Authors

Contributions

JMS, ZKM and SAM conceived and designed the experiment; JMS, ZKM and AY conducted the experiment and analyzed the data; JMS, ASM and AFAR drafted and proofread the manuscript. All the authors read and approved the final draft.

Corresponding author

Correspondence to Mohammed Sani Jaafaru.

Ethics declarations

Ethics approval and consent to participation

Not applicable.

Consent for publication

All the authors have read the manuscript and gave their consent for publication.

Competing interests

The author declared that there is no competing interest in the present study.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jaafaru, M.S., Muhammad, S.A., Mohammed, Z.K. et al. Proanthocyanidins supplemented diet alter anti-aging-markers and improved lifespan in Drosophila melanogaster model. Beni-Suef Univ J Basic Appl Sci 13, 11 (2024). https://doi.org/10.1186/s43088-024-00469-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s43088-024-00469-x

Keywords