1. Introduction
Fisetin (3, 7, 3', 4' tetrahydroxyflavone) is a natural flavonoid that exists in strawberries, cucumber, apples, onions and so on [
1](36558979). Early study by Pamela Maher et al showed that fisetin activated ERK and induced cAMP response element-binding protein (CREB) phosphorylation in rat hippocampal slices, facilitated long-term potentiation in rat hippocampal slices, and enhanced object recognition in mice[
2]. Fisetin also possesses neurotrophic effects by promoting rat cortical neurons cell survival and generating long neurites via promoting proteasome activity not through activation of pERK or increase glutathione[
3,
4]. Cumulative evidence of this research team indicate fisetin is a novel neuroprotective and cognition-enhancing molecule. Fisetin not only has direct antioxidant activity, but also can increase the intracellular levels of glutathione (GSH), the major intracellular antioxidant. Fisetin maintains mitochondrial function in the presence of oxidative stress. In addition, it has anti-inflammatory activity against microglial cells and inhibits the activity of 5-lipoxygenase, and consequently reducing the production of lipid peroxides and their pro-inflammatory by-products. These wide ranges of actions suggest that fisetin has potential to reduce the age-related decline in brain function[
5]. Fisetin can increase GSH level mainly by activating transcription factor 4 (ATF4) under basal conditions while activating both ATF 4 and NF-E2-related factor 2 (NRF2) under oxidative stress[
6]. In animal model, oral administration of fisetin to APPswe/PS1dE9 double transgenic Alzheimer’s disease (AD) mice from 3 to 12 months of age prevented the development of learning and memory deficits. This correlated with an increase in ERK phosphorylation and a decrease in protein carbonylation, a marker of oxidative stress. In addition, fisetin decreased the levels of p25, the cyclin-dependent kinase 5 (Cdk5) activator p35 cleavage product, in both control and AD brains. It is known elevated levels of p25 relative to p35 cause dysregulation of Cdk5 activity leading to neuroinflammation and neurodegeneration. These effects were also mediated by its anti-inflammatory effects, including changes in eicosanoid synthesis, and the maintenance of markers of synaptic function in the AD mice [
7]. Fisetin was also found to reduce cognitive deficits in old senescence-accelerated prone 8 (SAMP8) mice, a model for sporadic AD and dementia, while restoring multiple markers associated with impaired synaptic function, stress, and inflammation [
8]. Fisetin and other flavonoids also function as mitochondrial uncouplers to mitigate neurodegeneration in aged
C. elegans, possibly via a PINK1/Parkin mitophagy process [
9]. Further, fisetin boosted mental health in the aged animals. Supplementing fisetin (oral 20mg/kg/BW/day) for four weeks improved relative electroencephalograph α-power, β-power, and multi-unit activity (MUA) count in aged rats. Fisetin treated aged rats also showed significantly improved cognitive and behavioral performances than non-treated aged rats [
10]. Further, fisetin can inhibit aggregation of the tau fragment, K18, and can disaggregate tau K18 filaments in vitro and prevent the formation of tau aggregates in cells [
11].
Fisetin served as caloric restriction mimetics. Fisetin, administrated (15mg/kg b.w., orally) to young D-gal induced aged (D-gal 500mg/kg b.w subcutaneously) or naturally aged rats for 6 weeks significantly decreased the level of pro-oxidants and increased the level of antioxidants. Furthermore, fisetin also ameliorated mitochondrial membrane depolarization, apoptotic cell death and impairments in the activities of synaptosomal membrane-bound ion transporters in aged rat brain. Fisetin up-regulated the expression of autophagy genes (Atg-3 and Beclin-1), sirtuin-1 and neuronal markers (NSE and Ngb), and down-regulated the inflammatory (IL-1β and TNF-α) and Sirt-2 genes mRNA respectively in aged rat brain [
11]. Same treatment regimen also suppresses aging-induced increases in the levels of reactive oxygen species, eryptosis, lipid peroxidation, and protein oxidation in rat erythrocytes [
12]. Fisetin prevented the D-gal-mediated reactive oxygen species (ROS) accumulation, by regulating the endogenous antioxidant mechanisms, such as Sirt1/Nrf2 signaling, suppressed the activated
p-JNK/NF-kB pathway, and its downstream targets, such as inflammatory cytokines[
13]. Fisetin enhanced phosphorylation and nuclear translocation of Nrf2, which subsequently activated antioxidant enzyme heme oxygenase-1 (HO-1) in RPE cells, and therefore contributing to the amelioration of oxidative stress-induced ocular disorders [
14]. Similar effect was observed in C2C12 myoblasts cells [
15].
More recently, fisetin was found to be a potent senolytic agent. Acute or intermittent treatment of progeroid and old mice with fisetin reduced senescence markers in multiple tissues. Fisetin reduced senescence in a subset of cells in murine and human adipose tissue. Administration of fisetin to wild-type aged mice restored tissue homeostasis, reduced age-related pathology, and extended median and maximum lifespan [
16]. Senescent cells accumulate with ageing and are resistant to apoptosis and have up-regulation of anti-apoptotic pathways which defend them against their own inflammatory senescence-associated secretory phenotype (SASP), allowing them to survive, but killing neighboring cells. Because senescent cells take weeks to reaccumulate, senolytics can be administered intermittently - a 'hit-and-run' approach [
17]
. Fisetin pretreatment or treatment of old mice after COVID19 viral infection significantly reduced mortality, cellular senescence, and inflammatory markers and increased antiviral antibodies [
18]
. There may be crosstalk between fisetin’s anti-cell senescent and neuroprotection[
19]. Fisetin treatment also has been shown to restore muscle stem cells function in muscular dystrophy mice or progeria mice by targeting senescent cells in the diseased or aged muscle tissues [
20,
21]. Our recent work showed fisetin selectively attenuated markers of senescence in a dose-dependent manner while maintaining the differentiation potential of the expanded human adipose stem cells
in vitro [
22].
Fisetin also suppressed renal tubular senescence and attenuated renal fibrosis and improved tubular repair, as indicated by restoration of tubular regeneration and renal function[
23].. Fisetin treatment reduced renal fibrosis by inhibiting the phosphorylation of SMAD3, oxidative damage, inflammation, apoptotic cell death, and accumulation of profibrotic M2 macrophages in the obstructed kidneys. In cultured human proximal tubular cells, fisetin treatment inhibited TGF-β1-induced phosphorylation of SMAD3 and SMAD2[
24].
Fisetin also demonstrated the potent cardioprotective efficacy against Ang-II induced apoptosis in H9c2 cells and in spontaneous hypertension rat model. Fisetin treatment significantly reduced the apoptotic nuclei number and apoptotic proteins such as TNF- α, Fas L, FADD, Cleaved caspase-3 and Cleaved PARP and increased the cell survival and anti-apoptotic proteins like Bcl-2, Bcl-x
L, p-IGF1R, p-PI3K and p-AKT in both
in vitro and
in vivo models[
25]. Further, fisetin treatment reduced both the atherosclerosis plaque and the lipid accumulation in the aortic sinus in apoE-/- mice fed with a high-fat diet, and the expressions of PCSK9, LOX-1 and aging markers, including p53, p21 and p16 were downregulated [
26]. In vascular smooth muscle cell (VSMC) cells, fisetin inhibited cellular senescence induced by the PTEN-PKCδ-NOX1-ROS signaling pathway after hydrogen peroxide (H2O2) treatment, and this anti-aging effect was attributed to reduced ROS production caused by suppressing NOX1 activation[
27].
Taken together, fisetin has been shown broad beneficial effects on age related disease. However, majority of the previous studies used mice. Therefore, the goal of this study is to take advantage of large animal model’s translational value and investigate whether systemic fisetin treatment of aged sheep would have beneficial to multiple organs via targeting cell senescent cells.
Discussion
The goal of this study was to determine the presence of senescent cells and the effects of fisetin on cells senescent in the brain and other organs in aged sheep, a more translational model. Our results revealed wide presence of senescent cells in the 8 years old sheep (equivalent to 60-65 years old human) in both cerebral brain cortex and cerebellum as demonstrated by SA-β-Gal staining which was not reported before. Cerebral cortex gray matter has relative more senescent cells than white matter. The senescent cells are mainly large neurons in both gray and white matter of either cerebral brain cortex or cerebellum. Fisetin treatment at the current regimen (100mg/kg, two consecutive days a week for 8 weeks) did not affect general morphology of the brain. But fisetin treatment showed a trend of decrease SA-β-Gal cells in gray matter of both cerebral brain cortex and cerebellum and significantly decrease in brain cortex white matter compared to control. Furthermore, Fisetin treatment significant decreased P16+ cells in NEUN+ neurons, GFAP+ astrocytes, IBA+ microglia cells in both gray and white matter of cerebral brain cortex. Fisetin treatment also significantly decreased P16+ cells in microglia cells and a trend of decrease of P16+ cells in astrocytes in the non-CA area of hippocampus. But fisetin treatment did not change P16+ cells in the NEUN+ neurons in the CA1-4 area of hippocampus. At the mRNA level, fisetin showed a trend of decrease GLB1 in heart ventricle muscle tissue and spleen tissues but not brain and bone marrow. Fisetin treatment also showed significant or trend of decreased antioxidant gene SOD1 and increased CAT. Fisetin treatment showed variable effects in SASP and inflammasome genes in different organs.
Cell senescence was recently discovered as fundamental mechanism of aging and age-related diseases [31-34]
. Targeting cell senescent using senolytic drugs have been shown to improve SASP and frailty and enhanced metabolic function in aged mice [
35,
36]. Treatment with different senotlytic drugs such as JAK1 inhibitor Ruxolitinib or senolytic cocktail, dasatinib plus quercetin (D+Q) systemically for aged mice (20-22 months) prevented age related bone loss [
37]. Senolytic drug treatment also improved overall physical function of aged mice and extend lifespan of old mice by decreasing senescent cells [
38]. A human clinical trial showed D+Q treatment decreased senescent cells burden in humans as evidenced by reduced adipose tissue senescent cell burden with decreases in p16-and p21-expressing cells, SA-β-Gal+ cells, and adipocyte progenitors with limited replicative potential within 11 days [
39]. Most of previous studies on cell senescence used rodent animals. However, there is lacking of study to investigate the senescent burden in the brain of large animals. Our SA-β-Gal staining results indicated that senescent cells were widely present in both gray matter and white matter of 8 years old sheep brain. More senescent cells are present in the gray matter than in white matter. Further, from the morphology of the SA-β-Gal, they are mainly large neurons in these brain regions. These results indicated senescent burden in the brain is significant even in the not advanced aged sheep (equivalent to 60-65 years old human).
Alzheimer's disease (AD) is the most common form of dementia with the numbers expected to increase dramatically as populations ages. No treatments are available to cure, prevent, or delay the progression of the disease. Multiple changes associated with brain aging, including neuroinflammation and oxidative stress contributed to disease development and progression [
40]. Many studies have shown the beneficial effects fisetin on the different neurological disorders via its effects on multiple pathways. These include its anti-inflammatory and antioxidant effects as well as regulating cell death xytosis/ferroptosis pathway, the gut microbiome [
41,
42]. Fisetin was deemed as one of the promising nutraceuticals for treatment of Alzheimer’s disease. A combination of nutraceutical substances and other preventive measures could have significant clinical impact in a multi-layered therapy approach to treat AD[
43]. Fisetin treatment has also been shown to decrease senescence and SASP in aged or progeria mice and extend lifespan [
16]. But no study has investigated the effect of fisetin on brain cells senescent in large animal models. In this study, we showed that fisetin treatment in our current treatment regimen decreased SA-β-Gal positive cells in gray matter and white matter of cerebral brain cortex and gray matter in the cerebellum. However, fisetin treatment did not kill all senescent cells in the brain because many SA-β-Gal positive cells are still present in fisetin treated sheep brain regions. Furthermore, to determine which cells become senescent in the brain, we performed colocalization of P16 with neuronal marker NEUN, astrocytes marker GFAP and microglia markers IBA1. We found Fisetin treatment significantly decrease P16
+NEUN
+/Total NEUN
+, P16
+GFAP
+/Total GFAP
+ and P16
+IBA1
+/Total IBA1
+ cells percentages in the cerebral brain cortex in both gray and white matters. We revealed fisetin treatment significantly decreased P16
+/IBA1
+/Total IBA1
+ cells percentage in the non-CA area and a trend of decreased P16
+GFAP
+/Total GFAP
+ cells percentage in non-CA-area of hippocampus. But fisetin treatment has no effect on the P16
+NEUN
+/Total NEUN
+ cells percentage in the CA1-4 area of hippocampus. These results are consistent with SA-β-Gal staining results and collectively showed fisetin decreased senescent cells burden in the sheep brain for the first time. However, it is recognized that not all P16
+ cells are senescent cells although all senescent cells expressed P16 [
44]. We have attempted to use GLB1 antibody that worked for mouse and human to colocalize with neuronal marker, astrocyte marker and microglia marker, however, our antibody did not work for sheep and no GLB1 antibody was developed for sheep. Therefore, we could not determine if the P16
+ cells are all senescent cells. This is the limitation of this study. On the other hand, Fisetin may have only eliminated P16
+ senescent cells not P16
+ non-senescent cells. The reason why we see a decrease of P16
+/NEUN
+ cells /Total NEUN
+ cells percentage in cerebral brain cortex but not in the hippocampus may be due to the P16
+ cells in hippocampus are not senescent cells because nearly all CA1-4 neurons (NEUN+) are P16
+ (
Figure 4A). It might be that we used different P16 antibodies for cerebral brain cortex and hippocampus because SC1661 antibody from Santa Cruz Biotechnology did not work for hippocampus even with antigen retrieval likely due to long time formalin fixation before dissecting hippocampus. Whether other senolytic drug are more effective needs further study. Indeed, a recent clinical trial data using D+Q treatment to prevent Alzheimer’s disease showed promising results in decreasing senescent related cytokines and chemokines [
45].
It is worthy to emphasize that fisetin decreased P16
+IBA1
+/Total IBA1
+ cells percentage in microglia in both cerebral brain cortex and hippocampus although we used different P16 antibody. Microglia is macrophage lineage of cells in the brain [
46]. It is known both activated inflammatory microglia and senescent microglia secreted TNFα, IL1β and IL6 [
47]. Fisetin treatment inhibited the upregulation IL1β induced by LPS/IFN-γ- or peptidoglycan-induced inflammatory mediator in microglia cells. Fisetin also induced an endogenous anti-oxidative enzyme HO (heme oxygenase)-1 expression through the PI-3 kinase/AKT and p38 signaling pathways in microglia. Further, fisetin also significantly attenuated inflammation-related microglial activation and coordination deficit in mice in vivo[
48]. Fisetin treatment was also reported to alleviate intracerebral hemorrhage (ICH)-induced brain injury by downregulating proinflammatory cytokines and attenuating NF-κB signaling and preventing microglia activation [
49]. Hence our results that fisetin decreased P16
+ cells in the microglia added another beneficial effect of fisetin for the brain.
We also found Fisetin treatment affected the antioxidant gene SOD1 and CAT differently in different organs. We found fisetin treatment decreased SOD1 and increased CAT in both spleen and bone marrow but did not significantly change these two genes in the brain cortex and heart tissues. Fisetin is well known for its antioxidant effects by increasing glutathione (GSH) mainly through activating antioxidant transcription factor NRF2 [
4,
6]. In a vascular dementia model, fisetin treatment attenuated histological injury, malondialdehyde levels, inflammasome pathway activation, apoptosis, as well as increased brain derived neural growth factor (BDNF) expression, reduced astrocyte, microglial activation, and cognitive deficits [
50]. However, it has been reported fisetin could only reduce cell death induced by iron and copper in response to treatments that lower GSH levels, it is much less effective when the metals are combined with other inducers of oxidative stress. These effects correlated with the ability of iron but not copper to block the induction of the antioxidant transcription factor, Nrf2, by fisetin [
51]. Previous study using pressure overload induced cardiac hypertrophy model showed fisetin markedly reduced ROS by increasing expression of SOD1 and CAT and HO1[
52]. However, ours results only showed Fisetin treatment increased CAT but decreased SOD1. We did not detect other antioxidant genes due to unavailability of mRNA sequence for sheep. This inconsistence between our results and previous study might be due to different regimen of drug administration.
Finally, we found variable effect of fisetin on the expression of GLB1 in different organs. We found fisetin treatment relatively decreased GLB1 expression in spleen and bone marrow but not heart and brain cortex tissues by Q-PCR analysis. We also found different changes of SASP and inflammasome genes. It is possible this was because different organ has different working threshold of effects of fisetin. Further we used a hit-and run” strategy specific of senolytic treatment instead of regular drug treatment [
16,
39]. This treatment regimen does not produce sustained level of effective fisetin or its metabolites in the systemic level like other persistent administration regimen that used daily administration. Fisetin was shown to downregulate the activation of the NLRP3 inflammasome induced by LPS and ATP (LPS/ATP) and the subsequent maturation of IL-1β. Fisetin also activated mitophagy and prevented the accumulation of damaged mitochondria and the excessive production of mitochondrial ROS. Treatment of LPS/ATP-stimulated zebrafish model with fisetin facilitated the recovery of the impaired heart rate, decreased the recruitment of macrophage to the brain, and gradually downregulated the expression of inflammasome-related genes in a p62-dependent manner [
53]. But our study did not reveal consistent effects of fisetin on inflammasome gene NLRP3 and TREM2 at the current senolytic treatment regimen likely due to that we were not using induced inflammatory model in this study.
Author Contributions
Conceptualization, J.H. and X.G.; Methodology, C.A.H, X.G., M.D.H.,R.D.H, K.L; Software, C.A.H, X.G.; Validation, X.G., J.H.; Formal Analysis, C.A.H. X.G.; Investigation, C.A.H, X.G., M.D.H.,R.D.H, K.L.; Resources, J.H.; Data Curation, X.G.; Writing – Original Draft Preparation, C.A.H. X.G.; Writing – Review & Editing, J.H., P.J.M; Visualization, C.A.H, X.G.; Supervision, X.G, J.H.; Project Administration, J.H.; Funding Acquisition, J.H.