Review article

Oxidative stress and senescence in aging kidneys: the protective role of SIRT1

Waleed Hassan Almalki1[*], Salem Salman Almujri2

1Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia

2Department of Pharmacology, College of Pharmacy, King Khalid University, Abha 61421, Aseer, Saudi Arabia

EXCLI J 2024;23:Doc1030

 

Abstract

Aging leads to a gradual decline in kidney function, making the kidneys increasingly vulnerable to various diseases. Oxidative stress, together with cellular senescence, has been established as paramount in promoting the aging process of the kidney. Oxidative stress, defined as an imbalance between ROS formation and antioxidant defense mechanisms, has been implicated in the kidney's cellular injury, inflammation, and premature senescence. Concurrently, the accumulation of SCs in the kidney also exacerbates oxidative stress via the secretion of pro-inflammatory and tissue-damaging factors as the senescence-associated secretory phenotype (SASP). Recently, SIRT1, a nicotinamide adenine dinucleotide (NAD)-dependent deacetylase, has been pivotal in combating oxidative stress and cellular senescence in the aging kidney. SIRT1 acts as a potential antioxidant molecule through myriad pathways that influence diverse transcription factors and enzymes essential in maintaining redox homeostasis. SIRT1 promotes longevity and renal health by modulating the acetylation of cell cycle and senescence pathways. This review covers the complex relationship between oxidative stress and cellular senescence in the aging kidney, emphasizing the protective role of SIRT1.

See also the graphical abstract(Fig. 1).

Keywords: SIRT1, SASP, oxidative stress, cellular senescence, homeostasis

Introduction

Aging is a complex process characterized by progressive physiological declines in multiple organs, including the kidney (Fan et al., 2024[104]; Glassock and Rule, 2016[126]). The kidneys act to maintain homeostasis by filtering metabolic waste products, regulating electrolyte and fluid balance, and controlling blood pressure (Li and Wang, 2018[206]). However, as one age, kidney function gradually declines, signaling renal aging or age-associated kidney dysfunction (Minami et al., 2023[238]; Yang et al., 2019[375]). On a structural level, nephrons, the units of the kidney, are lost and responsible for reducing renal mass (Kimura et al., 2017[186]). This nephron loss is associated with glomerulosclerosis, or the hardening of the glomeruli, and tubulointerstitial fibrosis, characterized by an increased connective tissue deposition in the kidney interstitial (Hussain et al., 2024[155]; Tang et al., 2020[317]). These alterations explain the reduction in GFR, an important parameter for the kidney (Denic et al., 2022[91]; Qiu et al., 2018[280]). In a simplified format, functionally, old kidneys poorly conserve sodium and water, cannot concentrate urine to full potential, and do not respond as robustly to various metabolic hormone regulation (Cailleaux and Cohen-Solal, 2022[59]). Such modifications increase older people's susceptibility to dehydration, electrolyte disturbances and high blood pressure (Denic et al., 2016[90]; Satturwar and Parwani, 2023[296]). Older individuals are also highly vulnerable to more acute types of kidney injury, such as AKI/CKD, as aging kidneys have a poorer ability to repair following acute injury (Fang et al., 2020[107]; Shen et al., 2024[302]). Various mechanisms at a cellular level in the kidney drive the aging process, such as oxidative stress, cellular senescence, and inflammation (Epstein, 1996[100]). Oxidative stress arises when an increased ROS production exceeds the capacity of the antioxidant defense system, leading to cellular damage (Bridges and Zalups, 2017[56]). The irrecoverable cell growth inhibition causes cellular senescence, tissue damage and fibrosis. Additionally, inflammation contributes to both the progression of renal injury and fibrosis (Aaseth et al., 2021[1]; Chen et al., 2023[71]).

Role of oxidative stress and senescence in age-related kidney dysfunction

Oxidative stress and cellular senescence are the major factors involved in the progression of age-related changes in the kidney (Gekle, 2017[122]). Oxidative stress results from an imbalance between the production of potentially hazardous ROS and the body's antioxidant defenses, leading to oxidative damage to cellular proteins, lipids, and DNA (Bahl et al., 2024[23]; Sobamowo and Prabhakar, 2017[311]). This oxidative damage further exacerbates the deterioration of renal function by inducing inflammation, fibrosis and apoptosis of renal cells in aging kidneys (Hu et al., 2023[149]; Shankland et al., 2021[300]). Senescent cells, which have irreversibly exited the cell cycle, are another important contributor to renal aging (Chen et al., 2019[69]). Cellular senescence is associated with a secretion of proinflammatory cytokines, growth factors, and proteases collectively known as the SASP that eventually leads to accumulation of senescent cells in the kidney with age (Hommos et al., 2017[146]). SASP promotes tissue inflammation and fibrosis, aggravating renal injury. Therefore, oxidative stress and senescence form a vicious cycle in the aging kidney (Frassetto et al., 2020[116]; Zhu et al., 2024[396]). Cellular senescence is a new attractive target for both anti-oxidative stress treatment and anti-inflammatory therapy, whereas these therapies directly or indirectly converge to each other due to oxidative stress directly induces senescence, and senescence secretes proinflammatory cytokines to exaggerate oxidative damage (Akasaka-Manya et al., 2016[5]; Modi et al., 2024[241]). This dynamic promotes renal structural and functional impairment as aging progresses and may further aggravate CKD and other age-related renal diseases (Cheng et al., 2024[76]; Rodríguez-Castro and Córdova, 2011[285]).

Oxidative Stress and Kidney Aging

Reactive oxygen species (ROS) and oxidative stress

ROS is always naturally formed, even in cellular pathways (Wakino and Itoh, 2016[349]). They are important fuels of aerobic organisms as they are produced as by-products of aerobic metabolism in numerous cell organelles, such as mitochondria, chloroplasts, peroxisomes, cytosol and plasma membrane (Lyulcheva-Bennett et al., 2023[226]; Mir et al., 2024[239]; Musso et al., 2016[249]). ROS are produced when O2 acquire one electron at a time, forming reactive species, such as O2, H2O2, and OH (van den Anker et al., 2018[340]). ROS have dual roles in cells based on their concentrations (Alharbi et al., 2022[8]; Ferenbach and Bonventre, 2015[110]). At high enough levels, they will tend to react around everything. They can lead to oxidative damage to proteomics, DNA/RNA, and lipids and, in turn, cellular dysfunction or even death (Alharbi et al., 2022[9]; Kanasaki et al., 2012[170]). Lower concentrations of ROS, in contrast, can play positive roles, regulating cellular signaling pathways and modulating gene expression (Kooman et al., 2015[192]). Red light can be used to prevent oxidative stress, which occurs when the generation of ROS exceeds the reducing capabilities of the cell, in which free radicals are not generated in very high numbers as a result of an imbalance state of redox (Alharbi et al., 2021[10]; Morigi et al., 2018[245]; Zheng et al., 2022[392]). This imbalance can attack cellular components and lead to different diseases, including varicocele, in which ROS formation is responsible for testicular tissue damage and decreased fertility (Bauer et al., 1991[31]). In this regard, we have determined that the leptin-stimulated generation of ROS in cultured SMCs from obese-hyperleptinemic rats is partially resistant in this context of leptin resistance (Karam and Tuazon, 2013[172]). These changes were accompanied by higher protein expression of p22phox and gp91phox subunits of the NADPH oxidase complex and protein markers of leptin resistance, i.e. pSTAT3, PTP1B and SOCS3 (Alharbi et al., 2022[11]; Huber et al., 2012[152]). These studies suggest that ROS may play a critical role in several other physiological processes, including brain oxidative stress during most likely hypotensive sepsis, but are ameliorated with simvastatin pre-treatment (Bouarich et al., 2021[52]). In this regard, knowledge of ROS generation and consequent oxidative stress is important for developing treatments that can limit their negative effects, and help the maintenance of cell health (Alvis and Hughes, 2015[13]; Bhat et al., 2024[41]).

Sources of ROS in aging kidneys

Sources of ROS in aging kidneys are multifactorial and linked to different cellular processes (Bitzer and Wiggins, 2016[48]). XD enzyme complex serves as a major source of ROS that oxidizes hypoxanthine to form xanthine and produces uric acid (Bhat et al., 2022[44]; Zhou et al., 2008[394]). XDH is converted to XOD in aging kidneys, inferring more ROS generation (Lin et al., 2022[211]). AGEs, generated by non-enzymatic glycation and accumulate with aging in tissues, represent another major source of ROS (Bhat et al., 2024[45]; Sato and Yanagita, 2018[294]; Zhang et al., 2023[387]). AGEs trigger the RAGE to increase oxidative stress, causing ROS generation by NADPH oxidase in mitochondria (Choudhury and Levi, 2011[77]). In aging kidneys, mitochondrial dysfunction is also an important factor in ROS generation (Vlassara et al., 2009[345]). Increased ROS production increases oxidative damage at the cellular level due to mitochondrial dysfunction (Baylis, 2012[33]). This area of investigation assesses the regulation of renal mitochondrial bioenergetics and ROS generative pathways by sex hormones (Bhat et al., 2023[46]; Perico et al., 2024[270]). Estrogen, although not excellent for all renal diseases, is generally considered a protective agent, and its depletion can potentially accelerate the deterioration of renal mitochondrial function and enhance ROS production (Baylis, 2005[33]). The aging-related sources of ROS in kidneys are diverse but result from multiple cellular mechanisms (Bolignano et al., 2014[50]). It is important to understand these mechanisms to develop therapeutic strategies to protect and not damage kidney integrity in aging by producing effective elimination and maintaining healthy cells, reducing/abolishing proteinuria through protection against injury, and maintaining cell integrity and homeostasis under sublethal requirement oxidative stress with ROS, insignificant injury (Gupta et al., 2021[134]; Kaysen and Myers, 1985[176]).

Consequences of oxidative stress on kidney function

Aging-aged human kidneys are affected by oxidative stress (Wang et al., 2014[357]). Oxidative stress, the imbalance between ROS production and its scavenging by antioxidant defenses, results in peroxidation of lipids, damage to DNA, and oxidation of proteins (Pannarale et al., 2010[265]; Tufail et al., 2024[333]). The harm is done to proper cellular function and becomes a key pathogenic mechanism of many kidney diseases (Hayashi et al., 2019[140]; Hussain et al., 2023[153]). One major hormonal effect of oxidative stress in the kidneys is inflammation. ROS stimulate inflammatory signaling pathways, releasing chemotactic signals such as cytokines and other pro-inflammatory cytokines to attract immune cells to the injured region (Jo et al., 2023[167]; Uddin et al., 2021[335]). This further leads to progressive glomerulosclerosis, tubulointerstitial fibrosis, and excessive extracellular matrix deposition in the renal interstitium (Wei et al., 2020[359]). These two illnesses limit the filterability of the kidneys, and subsequently, the GFR goes down (Dybiec et al., 2022[99]; Hussain et al., 2023[154]). In addition, oxidative stress can lead renal cells to apoptosis (Hu et al., 2020[148]). Losing a portion of the cells that perform specific functions further impairs the kidney's ability to maintain homeostasis - the balance that your body requires to process fluids, electrolyte/salt regulation, and waste disposal (Nguyen et al., 2018[254]). Furthermore, oxidative stress can reduce the ability of renal cells to regenerate, thus rendering the kidneys less responsive to acute injury recovery (Gava et al., 2011[121]; Rohilla et al., 2023[286]).

Role of Epigenetic Processes in Regulating SIRT1 Expression

Epigenetic mechanisms greatly contribute to the regulation of gene expression independently from changes in DNA sequence (Alves-Fernandes and Jasiulionis, 2019[12]). These mechanisms involve DNA methylation, histone modifications, and the role of non-coding RNAs acting to modulate expression levels of important homeostatic genes such as SIRT1 (Ashapkin et al., 2017[18]). On the other hand, DNA methylation is one of the common epigenetic modifications that involves adding a methyl group to cytosine residues at position 5 nucleotide and has an important function as a gene silencing process (Ayissi et al., 2014[20]). Hypermethylation in its promoter region can lead to transcriptional silencing, while hypomethylation could promote its expression. Moreover, the methylation of the SIRT 1 promoter in response to environmental stresses such as oxidative stress may affect expression levels (D'Onofrio et al., 2018[97]; Thapa et al., 2024[323]). The changes in DNA methylation in the SIRT1 promoter region were related to the increased expression level and activity of SIRT1 during a stress reaction. Histone proteins possess several post-translational modifications, such as acetylation and methylation, phosphorylation, and ubiquitination (Fernandes et al., 2017[111]). These modifications alter chromatin structure and may influence gene expression. Specifically, histone acetylation, commonly linked to active gene expression, occurs when the acetyl group is transferred to lysine residues of histone tails (Fledderus et al., 2021[112]). This formation reduces affinity between DNA and histone tails, making chromatin more open and relaxed. It can also condense chromatin by working as a target gene inhibitor. SIRT1 is that type of inhibitor (Garcia-Peterson and Li, 2021[120]; Thapa et al., 2023[325]). This protein serves as an NAD + - dependent deactylase, meaning it removes acetyl groups from histones. Sirtuin 1 deacetylates histones, leading to chromatin compaction and target gene transcriptional inhibition (Jia et al., 2019[163]).

On the contrary, SIRT1 expression levels may increase if the latest undergoes the same end. The situation is then that the SIRT1 promoter region will behave in the form of higher acetylation (Nakatani and Inagi, 2016[252]). These steps constitute a cycle that changes target gene expression depending on the internal cellular signals. MiRNAs and lncRNAs are two non-coding RNAs that regulate gene expression post-transcriptional stage. As is known, miRNAs are capable of binding to the 3'UTR of SIRT1 mRNA, which later leads to its degradation or translational repression.

On the other hand, some lncRNAs may act as a reservoir for miRNAs in the form of molecular sponges. Thus, these miRNAs will not interact with the SIRT1 mRNA sequence (Pyo et al., 2020[278]). If UTR is not bonded with miR-34a, it cannot destroy it. This situation will lead to mRNA stabilization that results in enhanced SIRT1 synthesis. The miRNAs and lncRNAs system create a highly balanced approach to SIRT1 expression levels (Tao et al., 2023[320]).

Cellular Senescence in Aging Kidneys

Essence and features of cellular senescence

Cellular senescence is a state of irreversible cell cycle arrest triggered by various stress stimuli such as DNA damage, oxidative stress, and oncogene activation (Islam et al., 2024[159]; Musso and Oreopoulos, 2011[250]; Thapa et al., 2023[327]). It protectively prevents the generation of cellulite and, in the end, helps to avoid the risk of hopefully getting malignant cells (Lonergan, 1988[222]). Nevertheless, senescent cells are far from inert, characterized by their idiosyncratic phenotype, which entails massive cellular morphological and functional alterations (Esposito and Dal Canton, 2010[102]). A large and flat morphology characterizes senescent cells, enhanced SA-β-gal activity, and production of a plethora of pro-inflammatory cytokines, chemokines, growth factors and proteases known as SASP (Sato and Yanagita, 2019[295]). In normal cells, the SASP can enhance the senescence growth arrest and modulate the tissue microenvironment; additionally, the SASP can provoke inflammation and tissue remodeling, thereby promoting aging and age-related pathologies (Buemi et al., 2005[57]). Such cells can be arrested or removed to the benefit of the tissue. Still, their presence in vivo mandates further research to learn how they are regulated during aging and to identify the causal factors that provoke them in human pathologies (Sands, 2012[292]).

Mechanisms of senescence induction in kidney cells

Cellular senescence in kidney cells is induced by various types of stress and signaling pathways (Jassal and Oreopoulos, 1998[162]). Among these is damage to genetic material, especially DNA, frequently arising from oxidative stress (Baylis and Corman, 1998[34]). DNA damage is also the result of other factors; ROS are impermanent, high-reactive compounds that have the potential to produce harm in biological macromolecules such as DNA, proteins, and lipids and can initiate the DDR (Artioli et al., 2019[16]; Jiang et al., 2017[164]; Li et al., 2024[204]). DDR activation at this moment results in cell cycle arrest mainly through the p53/p21 and p16INK4a/Rb pathways, controlling important elements of senescence (Khan et al., 2017[178]; Wang et al., 2018[355]). It also turned out that oxidative stress was a potent inducer of senescence in kidney cells (Wang et al., 2021[358]). ROS helps to disturb cellular homeostasis and leads to oxidative damage and mitochondrial impairment (Franzin et al., 2020[115]; Presta et al., 2012[276]). Due to ROS and a vicious circle where ROS production is exacerbated by the loss of the ability to access functioning mitochondria, cellular aging and senescence may be accelerated (Schmitt et al., 2015[297]). Telomere shortening is another mechanism leading to senescence, which occurs through normal cellular replication (Nguyen and Goldfarb, 2012[256]). More importantly, critically short telomeres are considered DSB, which triggers DDR pathways. Senescence in kidney cells is also driven by inflammation. Sustained exposure to inflammatory cytokines and growth factors, as seen in chronic inflammatory conditions, can also promote senescence via paracrine signaling (Hirakawa et al., 2017[145]; Speeckaert et al., 2014[312]). This phenomenon is commonly called inflammaging, and it promotes the damage and senescence of tissues (Wiggins, 2009[363]).

Senescence induction and epigenetic changes

Events such as histone methylation and acetylation that induce chromatin changes in gene expression profiles necessary for senescence-associated growth arrest and the SASP can also be modified by miRNAs (Han et al., 2022[137]; Kuro-o, 2010[195]). Understanding these mechanisms is important for developing targeted interventions to prevent or ameliorate renal senescence, preserving kidney function and delaying the onset of age-related kidney pathologies (Meng et al., 2020[231]).

Telomere shortening

Telomere shortening is a critical mechanism for cellular senescence and aging (Li et al., 2021[207]). Telomeres are sequences of repetitive nucleotides found at both ends of the chromosome and which function to protect the ends from degradation and fusion with the ends of other chromosomes (Mohamad Kamal et al., 2020[242]; Obas and Vasan, 2018[260]). During cell division, the telomeres are shortened because the DNA polymerases only partially replicate the DNA ends of the linear chromosomes, with each proliferation narrowing the telomeres (Liu, 2022[220]; Ogrodnik, 2021[261]). Like all somatic cells, renal cells encounter this programmed shortening. When this results in the shortest telomere in the cell, the cell recognizes that a critical length threshold affecting homologous recombination and generating significant loss of genetic material has been reached, and this magnifies a signal to DDR (Gonzalo et al., 2017[127]; Imran et al., 2021[158]; Jones et al., 2015[168]). Activation of DDR pathways causes cell-cycle arrest, mostly directed towards cellular senescence via p53/p21 and p16 INK4a/Rb pathways (Turner et al., 2019[334]; Zhu et al., 2019[398]). This is due to the secretion of proinflammatory cytokines, growth factors, and proteases from senescent cells, where declaring the SASP can potentially promote tissue dysfunction and inflammation (Aubert, 2014[19]; Rossiello et al., 2022[288]). This is particularly important when considering aging kidneys, which accelerate age-related renal function deterioration, predisposing an individual to CKD (Prasad et al., 2017[275]). Thus, uncovering the mechanisms regarding the effect of telomere shortening on kidney aging is important for designing therapies to maintain the telomere length and to protect the cells against senescence in elderly kidneys (Mikhelson and Gamaley, 2012[237]; Morgan et al., 2018[244]; Opresko and Shay, 2017[263]).

Oxidative stress-induced senescence

Oxidative stress-induced senescence is a key step in cellular aging and tissue dysfunction (Li et al., 2022[203]). ROS are produced as by-products of metabolic reactions or by exposure to external stimuli, and they can damage DNA, proteins, and lipids (Chandra and Rajawat, 2021[64]; Lin and Epel, 2022[210]). The activation of DDR pathways is one of the principle phenotypes whereby age-related DNA damage triggers cell cycle arrest via p53/p21 and p16INK4a/Rb signaling (Liu et al., 2023[217]; Rellmann et al., 2021[283]). In more chronic conditions, oxidative stress in kidney cells has been shown to increase senescence, leading to inflammation, fibrosis and reduced regenerative capacity (Pintó-Marijuan and Munné-Bosch, 2014[272]). This is detrimental to age-related renal decline and induces CKD susceptibility (Kaarniranta et al., 2018[169]). The aging kidney requires therapeutic strategies capable of preventing or at least reducing the progression of kidney disease, and the elucidation of the pathways involved in oxidative stress-induced premature senescence is essential for the development and success of such interventions (Nousis et al., 2023[259]; Terao et al., 2022[321]).

Role of senescent cells in kidney aging and disease

Cells that have become senescent contribute to kidney aging and renal disease development (Wiggins, 2011[364]). Such accumulation in the kidneys over time is partly the result of aging factors - most notably oxidative stress, DNA damage and telomere shortening - and are cells that have undergone an irreversible cell cycle arrest (Kłoda et al., 2015[190]; Verbalis, 2014[343]). The differentiated cells enter a senescent state, and some acquire SASP, overproducing pro-inflammatory interleukins, chemokines, growth factors, and proteases (Denecke et al., 2015[89]; Smykiewicz et al., 2018[310]). The SASP drives transcriptomes for age-associated inflammation and promotes tissue remodeling, fibrosis, and widespread chronic inflammation, all dialing in a decreasing trajectory for kidney function with age (Fusco et al., 2016[118]). Senescent cells also limit kidney regenerative capacity, thus impeding normal kidney repair following injury (Fougeray and Pallet, 2015[114]). This further deteriorates age-induced renal insufficiency and then promotes vulnerability to CKD, such as glomerulosclerosis and tubulointerstitial fibrosis (Lamb et al., 2003[198]; Weide and Huber, 2011[360]). Therapeutic strategies targeting senescent cells and their deleterious effects may offer hope in maintaining kidney function and outcomes in the aging population (Bennett, 1990[36]; Musso et al., 2015[248]).

Senescence-associated secretory phenotype (SASP)

Senescent cells exhibit a three-part phenotype called the SASP, comprising hundreds of pro-inflammatory cytokines and chemokines, growth factors, and proteases (Guebre-Egziabher et al., 2013[131]. This secretory phenotype has a major effect on the tissue microenvironment and a role in various age-related pathologies (Charloux et al., 2008[66]). The SASP is a double-edged singer in this narrative on kidney aging. First, it maintains the growth arrest of senescent cells, which prevents cancer (Singh and Krishan, 2019[308]; Ungar et al., 2000[338]). The SASP, a mixture of pro-inflammatory and tissue remodeling factors released by senescent cells, amplifies chronic inflammatory responses and may cause fibrosis, contributing to kidney structural and functional decay (Abrass, 1990[2]). These secretions can alter the homeostasis in tissues by remodeling the ECM, inducing fibrosis deposition, and promoting chemotaxis of the immune cells to the inflammatory site where the inflammatory response is maintained, and further tissue damage continues to occur (Andrade et al., 2018[14]; Lindeman and Goldman, 1986[213]). Chronic exposure to SASP factors exacerbates the progression of CKD and other renal pathologies linked to aging (Glassock et al., 2017[125]). In this context, inhibition against SASP components or their signaling pathways has recently emerged as a potential therapeutic strategy to alleviate the adverse consequences of cellular senescence that are expected to rescue declining kidney function and restore the health span of elderly populations (Shiels et al., 2017[305]; Shu et al., 2015[306]; Yin et al., 2023[378]) (Figure 2(Fig. 2)).

SIRT1: A Key Regulator of Oxidative Stress and Senescence

Overview of SIRT1 and its functions

SIRT1 is a novel member of the sirtuin family of NAD+-dependent class III deacetylases that has been identified as a critical regulator of mammalian cellular metabolism and longevity (Cui et al., 2022[82]; Jalgaonkar et al., 2022[160]). SIRT1 is an evolutionarily conserved mammalian protein with a wide range of cellular functions related to metabolism, stress response, and aging processes (Liu et al., 2022[221]; Shen et al., 2021[301]). It mainly deacetylates multiple protein targets and controls their activity, stability and association with other molecules (Chen et al., 2021[74]; Shen et al., 2024[303]). SIRT1 has roles in the complete scheme of metabolic regulation (Tang, 2016[316]). It also deacetylates and activates metabolic regulators like PGC-1α, increasing mitochondrial biogenesis and function and FOXO transcription factors involved in stress resistance and longevity (D'Onofrio et al., 2018[97]; You and Liang, 2023[380]). SIRT1 regulates these pathways to increase cellular energy utilization efficiency and protect cells from stress-induced death (Chang and Guarente, 2014[65]). SIRT1 also maintains genomic stability (Chen et al., 2021[70]). By deacetylating histones and other proteins involved in DNA repair, SIRT6 helps the cell to repair DNA double-strand breaks and maintain chromosomal stability (Chen et al., 2020[68]; Kim et al., 2022[185]). This function is most critical in aging cells as DNA damage is not just associated with nuclear blebbing and a decrease in PTR but also leads to cellular dysfunction and senescence (Alves-Fernandes and Jasiulionis, 2019[12]; Wang et al., 2021[352]). SIRT1 has roles in metabolism, including effects on the genome, but it is also a potent modulator of inflammation (Singh and Ubaid, 2020[309]). It deacetylates the subunit p65 of the NF-κB, a fundamental transcription factor in inflammatory responses, impairing the expression of pro-inflammatory cytokines (DiNicolantonio et al., 2022[93]; Yang et al., 2022[373]). This intervention helps avoid the chronic inflammation purported to be central to aging and age-related diseases (Lu et al., 2023[224]). In the kidneys, SIRT1 inhibits oxidative stress and fibrosis (Kauppinen et al., 2013[175]). It controls the activity of fibrotic factors, including TGF-β and SMAD3, contributing to a protective mechanism against renal aging and disease (Garcia-Peterson and Li, 2021[120]; Patra et al., 2023[267]). Thus, defining the role of SIRT1 presents an essential axis for both targeting and understanding the therapeutic potential against age-induced kidney disease and possibly healthy aging (Meng et al., 2017[233]).

One pathway to SIRT1 expression involves progressing from ROS to cellular senescence and aging (Byrnes et al., 2022[58]). Elevated levels of ROS are known to induce oxidative stress, which can result in cellular damage and trigger a senescence response (Feng et al., 2024[109]). This cellular senescence is characterized by a stable cell cycle arrest and the secretion of pro-inflammatory cytokines, collectively known as the SASP. As cells age and accumulate damage, the expression of SIRT1 is upregulated as a protective mechanism to counteract ROS's deleterious effects and promote cellular repair and longevity (Xu et al., 2019[370]; Yu et al., 2018[381]). In addition to the direct progression from ROS to aging, alternative pathways such as ROS-induced epigenetic modifications can also lead to the expression of SIRT1. ROS can influence various epigenetic marks, including DNA methylation, histone modifications, and non-coding RNA expression (Wang et al., 2014[353]). These epigenetic changes can alter the transcriptional landscape of the cell, leading to the activation of genes involved in stress response and survival, including SIRT1 (Tang et al., 2021[318]; Thirupathi and de Souza, 2017[329]). ROS-induced histone acetylation changes can create a more accessible chromatin state, facilitating the binding of transcription factors that promote SIRT1 expression (Suntar et al., 2020[315]). Furthermore, non-coding RNAs such as miRNAs and lncRNAs can modulate the stability and translation of SIRT1 mRNA, thereby influencing its protein levels in response to oxidative stress (Schottlender et al., 2021[298]; Singh and Ubaid, 2020[309]).

SIRT1 and oxidative stress regulation

SIRT1 is associated with regulating oxidative stress, a central mechanism of cellular aging and a requirement of many diseases like the kidneys (Beyfuss and Hood, 2018[40]; Cui et al., 2022[82]). SIRT1 modulates the activity of several transcription factors and enzymes implicated in the cellular antioxidant defense system (Wu et al., 2022[367]). SIRT1 is one of those main factors affecting the balance of response to oxidative stress primarily to the transcription factor FOXO3a (Jalgaonkar et al., 2022[160]; Shen et al., 2021[301]). Therefore, SIRT1-deacetylated-FOXO3a in the cell nucleus can be involved in the expression of antioxidant enzymes, including SOD and catalase (Zhang et al., 2017[389]). They are key antioxidant enzymes detoxifying ROS and protecting cells from oxidative stress (Meng et al., 2020[232]). More interestingly, the PGC-1α a coactivator that promotes the biogenesis and function of mitochondria, is one of the SIRT1 substrates (D'Onofrio et al., 2018[97]) since improved mitochondria efficacy results in less ROS creation. A large part of these ROS results in the lowering of the standard of pro-oxidants, which manifest themselves in this phenomenon (Singh and Ubaid, 2020[309]; Xu et al., 2019[370]). SIRT1 binding at chromatin represses key pro-inflammatory NF-κB transcription factors via deacetylation of the p65 subunit, thereby decreasing transcription of pro-inflammatory genes that would otherwise compromise redox homeostasis and harm tissues (Chen et al., 2013[75]; Kong et al., 2017[191]; Shi et al., 2023[304]). This is significant in kidney health as the positive regulatory actions of SIRT1 (Jin et al., 2023[166]). In addition, by increasing antioxidant defenses and attenuating inflammation, SIRT1 performs an antioxidant function. It thereby diminishes oxidative stress, which is a major driving force of renal problems due to aging as well as kidney diseases (DiNicolantonio et al., 2022[93]; Fang et al., 2022[105]; Feng et al., 2021[108]). The function of SIRT1 in controlling oxidative stress provides novel perspectives on therapeutic intervention to safeguard the kidney and enhance healthy aging (Alam et al., 2021[7]; Winiarska et al., 2021[365]).

Modulation of antioxidant defenses

SIRT1 enhances antioxidant defenses, which is crucial in reducing oxidative stress and maintaining cellular health by activation of transcription factors which deacetylate them (Halliwell, 2024[136]; van der Pol et al., 2019[341]). Deacetylated form of FOXO3a induces the expression of antioxidant enzymes, such as SOD and catalase, which serve to reduce ROS concentrations (Forman and Zhang, 2021[113]). Activation of PGC-1α induces mitochondrial biogenesis and function, decreasing ROS production and improving mitochondrial efficiency (Bhatt et al., 2020[47]; Bjørklund et al., 2022[49]; Ma, 2013[227]). In parallel, SIRT1 blocks pro-inflammatory pathways, likely in part via deacetylation of the NF-κB p65 subunit, suppressing pro-inflammatory cytokine production that could promote oxidative damage (Arulselvan et al., 2016[17]; He et al., 2023[142]). SIRT1 also activates the cellular antioxidant defense system, which appears crucial for preventing oxidative stress-induced cellular damage and aging (Barnes, 2020[28]; Sies, 1997[307]).

Regulation of oxidative stress-related signaling pathways

SIRT1 affects oxidative stress by signaling pathways, mediating oxidative stress, and affecting cell survival (Liguori et al., 2018[208]). It deacetylates FOXO3a and PGC-1α, two pivotal transcription factors (do Nascimento et al., 2008[95]). The expression of antioxidant enzyme SOD, catalase, and other genes that neutralize ROS is enhanced by deacetylated FOXO3a (Pingitore et al., 2015[271]; Zeng and Lu, 2018[386]). PGC-1α activation improves mitochondrial function, decreasing ROS generation through ameliorating mitochondrial efficiency and biogenesis (Agarwal et al., 2012[4]; Katz et al., 2011[174]). SIRT1 also deacetylates the p65 subunit, modulating the NF-κB pathway and decreasing the expression of pro-inflammatory cytokines (Demirci-Çekiç et al., 2022[88]). This combats chronic inflammation and the toll on the system when oxidative stress increases (Chen et al., 2009[73]). These regulatory pathways maintain cellular redox homeostasis, which is required for the resistance to oxidant stress and the extension of cellular lifespan by SIRT1 (Stocker, 2016[313]).

SIRT1 and cellular senescence modulation

SIRT1 is a notable regulator of pathways linked to cellular senescence, an indispensable stress-response program that causes cells to exit the cell cycle permanently and which is thought to contribute to aging and a spectrum of age-associated diseases (Barnes et al., 2019[29]; Salminen and Kaarniranta, 2012[291]). Aging is most likely initiated by many stimulants, the most significant being oxidative stress, DNA impairment, and the thickening of telomeres, which leads to cellular senescence (Pyo et al., 2020[278]; Yao and Rahman, 2012[376]). Senescent cells that are kept in the body will increasingly become more inert and secrete a series of pro-inflammatory cytokines, chemokines and proteases known as the SASP, which ultimately might contribute to tissue function decline and chronic inflammation (Chung et al., 2010[79]; Hwang et al., 2013[156]). SIRT1 regulates cellular mechanisms of senescence through multiple pathways (Chen et al., 2020[68]). One of the major pathways consists of the deacetylation and, thereby, activation of the tumor suppressive protein p53 (Hekmatimoghaddam et al., 2017[143]; Yi and Luo, 2010[377]). Active p53 can then cause a cell to G2 arrest and undergo apoptosis in response to DNA damage (D'Onofrio et al., 2018[97]). In contrast, SIRT1 deacetylation of p53 leads to SIRT1 facilitating cell survival and DNA repair. Still, without the unwanted senescence characteristic of hyperacetylation-treated cells, it prevents the build-up of senescent cells and the preservation of tissue homeostasis (Lagunas-Rangel, 2022[197]; Lee et al., 2019[200]). SIRT1 also interacts with and deacetylates other components of the cellular senescence process, such as the FOXO family of transcription factors (Kida and Goligorsky, 2016[179]). The FOXO proteins are essential regulators of lifespan and stress resistance. SIRT1 enhances the transcriptional activity of FOXO through their physical interaction and deacetylation of FOXO, leading to up-regulating FOXO-dependent genes associated with the reduction in cellular senescence, such as DNA repair, antioxidant defense and cell cycle control (Cui et al., 2022[82]; Fukuda et al., 2020[117]).

SIRT1 also impacts the NF-κB signaling pathway, which plays important role in inflammation and senescence formation (Guo et al., 2020[133]). As a result of deacetylation of the p65 subunit of NF-κB, transcription of pro-inflammatory genes is lowered by SIRT1, reducing the inflammation that can induce senescence (Poulose and Raju, 2014[274]). In the context of renal aging, modulation of senescence is especially pronounced for SIRT1. Accumulation of senescent cells in the kidney contributes to fibrosis, reduced regenerative capacity, and loss of function (Ala and Ala, 2021[6]). SIRT1 is an important regulator of renal function with protective roles in delaying the pathogenesis and progression of age-associated kidney diseases through improved DNA repair, reduction in oxidative stress, and modulation of inflammatory responses (Wang et al., 2021[356]; Yacoub et al., 2014[371]). Therefore, the current review aims to provide insights into the outcomes of SIRT1 activity modulation on cellular senescence and its underlying molecular mechanism as the regulators of cellular senescence, which promise prospective potential therapeutic options to counteract against aging process and extend health span, especially in age-vulnerable organs such as the kidney (Famulski and Halloran, 2005[103]; Lamb et al., 2003[198]; Maertens and Van Den Noortgate, 2008[228]).

SIRT1 as a Potential Therapeutic Target in Kidney Aging

Experimental evidence for SIRT1 activation in kidney aging

Experimental evidence highlights a potential role of SIRT1 in kidney aging, recent studies have demonstrated protection against various kidney age-related changes by activating SIRT1 (Guo et al., 2020[133]; Kitai et al., 2021[189]). Studies on animal models have shown that overexpression of SIRT1 in the kidneys ameliorated age-related renal dysfunction (Kume et al., 2013[194]). For example, transgenic mice with SIRT1 overexpression show less oxidative stress, fibrosis, and improved renal function than their wild-type littermates (Tanriover et al., 2023[319]). Drugs that activate SIRT1, such as resveratrol, have also achieved positive outcomes (Wang et al., 2018[355]). Conversely, in aged mice, resveratrol treatment enhances SIRT1 enzymatic activity, promoting antioxidant enzyme expression, reducing inflammation and protecting against renal fibrosis (Kanbay et al., 2021[171]; Yacoub et al., 2014[371]). These effects together favour the maintenance of kidney integrity and function (Jin et al., 2024[165]). In addition, in vitro culture studies with renal cells have shown that SIRT1 activation improves cellular resistance to oxidative stress and delays the onset of cell senescence (Guan and Hao, 2016[130]). Together, these results suggest that it may be therapeutically advantageous to manipulate SIRT1 to mitigate age-related kidney decay and maintain aging kidney health. Further understanding these mechanisms could open new avenues for interventions in age-associated kidney diseases (Meng et al., 2017[233]; Qi et al., 2022[279]).

Animal studies

Experimental evidence in animal models shows that activation of SIRT1 attenuates aged-related kidney injury (Yan et al., 2022[372]). Activation of SIRT1 minimizes oxidative stress, inflammation, fibrosis, and renal function improvement (Kitada et al., 2013[187]). Such studies in aged mice reveal it improved kidney structure and function upon SIRT1 overexpression, suggesting its protecting role in renal aging and associated pathologies (Ogura et al., 2021[262]). D-galactose is a monosaccharide; a simple sugar. D-galactose has a history in cellular metabolism and aging analyses (Azman and Zakaria, 2019[21]). High intake induces oxidative stress and age-related damage in animal studies (Wang et al., 2022[354]). Fang et al. reported that empagliflozin attenuates renal senescence in D-galactose-induced aging mice via SIRT1 and oxidative stress reduction.

Conversely, treatment of empagliflozin reversed the changes of BTBD9 and alleviated age-related kidney injury as well as upregulating SIRT1, SOD1, and SOD2. Similarly, in vitro empagliflozin exerted protection effects at the cellular level, whereas a SIRT1 inhibitor decreased these effects. These findings demonstrate the ability of empagliflozin to alleviate renal aging by concentrating on SIRT1-related oxidative stress (Fang et al., 2023[106]). Nur77 is an orphan nuclear receptor that participates in multiple cellular functions, such as apoptosis, inflammation, and metabolism (Lith and de Vries, 2021[214]). It is closely related to immune regulation, cancer formation and metabolic diseases (Liu et al., 2021[218]; Zhang, 2007[390]). Yu et al. showed that oxidative stress inhibits the Nur77-SIRT1 axis to reduce SIRT1 expression levels, thereby accelerating aging. The overexpression of Nur77 stabilizes SIRT1, indicating that Nur77 regulates protein expression and renal insulin sensitivity in the aged kidney. Nur77 depletion exacerbated age-related nephropathy, demonstrating the importance of Nur77-SIRT1 signaling in kidney aging and functions. This work provides a novel understanding of oxidative stress and SIRT1 regulation at the molecular level (Yu et al., 2023[382]).

IUGR is a condition in which a fetus does not grow at the expected rate during pregnancy. This can then cause low birth weight, birth complications and lifelong health problems (Longo et al., 2014[223]). This may be due to placenta issues, mother complications, or genetics (Darendeliler, 2019[83]). Keshavjee et al. assessed whether suppression of SIRT1 expression and oxidative stress contribute to, a nutshell, improved NAFLD. We studied SIPS using an IUGR rat model. The study revealed pronounced oxidative stress and senescence markers in IUGR males, which may provide a potential pathway to metabolic disease. This work further highlights the repercussions of early life stress into adulthood kidney health (Keshavjee et al., 2022[177]). NAD+, an important coenzyme required for cellular metabolism, is an essential coenzyme required for DNA repair and energy production (Yoshino et al., 2018[379]). A central player in redox reactions involves moving electrons from one molecule to another. Decreasing cellular function and health as NAD+ levels drop with age (Covarrubias et al., 2021[81]; Imai and Guarente, 2014[157]). Braidy et al. demonstrated that NAD+ levels and SIRT1 activity declined as Wistar rats aged in all studied organs, related to an augmentation of oxidative stress and a decrease in mitochondrial function. Reduced SIRT1 activity was accompanied by heightened DNA damage and protein carbonylation. Our study illustrates the potential benefits of preserving NAD+ and SIRT1 levels in protecting against oxidative damage and suggests its therapeutic relevance to aging and aging-associated disorders (Braidy et al., 2011[54]) (Figure 3(Fig. 3)).

Glomerular endothelial cells are important in blood filtration, filtration barrier maintenance, and glomerular structure (Benzing and Salant, 2021[37]; Bose and Cattran, 2014[51]). Chuang et al. demonstrated that the podocyte-specific deletion of SIRT1 in aging mice exacerbated kidney injury, manifested by worsened glomerulosclerosis, albuminuria, and oxidative stress. The results from this study indicated that decreased SIRT1 expression indeed promoted the higher expression of cellular senescence markers and the weaker activation of major transcription factors, which consequently suggested that SIRT1 was essential to protect podocytes from age-related injury and oxidative stress, unveiling its therapeutic potential in this context (Chuang et al., 2017[78]). Cadmium is a toxic heavy metal present as a pollutant in the environment mainly due to industrial activities that include mining and smelting (Peana et al., 2022[268]). This builds up in an individual's organs, destroying them and leading to severe health problems such as kidney failure, bone brittleness and cancer (Genchi et al., 2020[123]; Satarug et al., 2010[293]). Dong et al. in vivo experiments were carried out to detect the potential role played by SIRT1 in CKD. The results showed that Cd accelerated the process of renal senescence, fibrosis, and dedifferentiation, which NAC effectively attenuated through the SIRT1-P53 pathway. NAC reduced oxidative stress and reversed senescence, attenuating kidney damage associated with CKD progression. The research indicates that the SIRT1-P53 pathway may offer a therapeutic target in cadmium-induced CKD (Dong et al., 2023[96]). The platinum-based chemotherapy drug is cisplatin, which helps treat testicular, ovarian, bladder, and lung cancers (Leal and García-Perdomo, 2019[199]). It does this by creating damage in the DNA of cancer cells, eventually leading to cancer cell death. It has multiple adverse effects, like nephrotoxicity, ototoxicity, and gastrointestinal disturbances (Rossi and Di Maio, 2016[287]; Xian et al., 2021[368]). Li et al., in a murine model, found that cisplatin caused kidney senescence and fibrosis prematurely. Administration of NAC significantly suppressed this adverse effect by promoting SIRT1 activation and the deacetylation of p53. The findings of this study suggest that SIRT1 serves a protective role against cisplatin-induced renal injury and that SIRT1 activation is a potential target for the prevention of renal dysfunction chronicity after AKI and p53 deacetylation (Li et al., 2019[201]). Hasegawa et al. overexpressed SIRT1 in mice, specifically in kidneys, and created transgenic mice, which protected SIRT1 from cisplatin-induced AKI through peroxisome quality maintenance. Renal tubular apoptosis was diminished by SIRT1 overexpression, which was associated with decreased reactive oxygen species, peroxisome number, and function preservation. This study demonstrates that SIRT1 might have a role in maintaining peroxisomes and protecting against AKI, a plausible mechanism that implicates SIRT1 as a therapeutic target for renal protection (Hasegawa et al., 2010[139]).

Similarly, Lim et al. explored the state and fate of the kidney in aged mice beyond what was known from similar experiments on a background of atherosclerosis. Accordingly, the study showed that the kidneys of older mice exhibit oxidative stress and lower SIRT1, ERR-1α, PGC-1α, PPARα expression, and reduced klotho, an antiaging factor. These changes were accompanied by upregulated albuminuria, mesangial volume, and tubulointerstitial fibrosis. These results indicate that approaches to modulate SIRT1 and its downstream signaling may be beneficial in reducing oxidative stress and pathophysiological changes in the aging kidney (Lim et al., 2012[209]).

Chronic rising blood pressure can cause hypertensive kidney injury, which arises from damaging blood vessels by the raised blood pressure for so long that it injures the nephrons and leads to the kidney functioning less efficiently (Messerer et al., 2021[234]; Radi, 2019[282]), which can easily lead to chronic kidney disease and can lead to proteinuria, glomerulosclerosis, renal failure. All the more reason for early intervention (Poston and Koyner, 2019[273]). Pushpakumar et al. studied hypertensive kidney injury in aging and reported that oxidative stress and hypermethylation of antioxidant enzymes amplified kidney damage. In aged kidneys, expression of SIRT1, SODs and catalase was decreased and further decreased in ang-II treatment. The results reveal that epigenetic regulation of oxidative stress is fundamentally important in this setting. They imply that it is targeted as a therapeutic approach to ameliorate hypertensive kidney injury with aging (Pushpakumar et al., 2020[277]).

Furthermore, Bai et al. proved that melatonin protected rats from AKI after severe burns through up-regulating SIRT1. Melatonin significantly attenuated oxidative stress, inflammation and apoptosis in renal tissues. Inhibition of SIRT1 abolished the protective effects, which correlated with enhanced SIRT1 expression. This study reinforces the protective ability of melatonin by activating SIRT1 and supports its clinical application in burn-related kidney damage (Bai et al., 2016[25]) (Figure 4(Fig. 4)).

In another study, Grosjean et al. observed the reversal of glomerulosclerosis in diabetic, sclerosis-prone mice by treatment with FDA-approved drugs (pyridoxamine, enalapril, and pentosan polysulfate) that lower levels of oxidative stress and inflammation. Treatment up-regulated Nrf2, SIRT1, ERα and AGER1 expression, reduced albuminuria and protected against sclerosis. The findings provide significant evidence for an oxidative stress and inflammatory therapeutic target for early diabetic kidney disease protection (Grosjean et al., 2018[129]). The AMPK/PPARα/ UCP2 pathway is important in cellular energy homeostasis and metabolism (Carling, 2017[61]). AMPK activation inhibits PPARα leading to fatty acid oxidation, and UCP2 decreases mitochondrial membrane potential, reducing reactive oxygen species production and increasing energy efficiency (Herzig and Shaw, 2018[144]; Trefts and Shaw, 2021[331]). Rubattu et al. spontaneous hypertensive rat strains demonstrated that the AMPK/PPARα/UCP2 were significantly down-regulated in stroke-prone rats, and this might contribute to the age-associated increase in oxidative stress and inflammation in the brain. These changes were associated with increased vulnerability to hypertension-induced target organ damage. The study demonstrates that this antioxidant pathway is downregulated early in this model, and the data would strongly suggest that this contributes to the vulnerability of the rat to the development of hypertension-induced damage (Rubattu et al., 2015[289]).

Furthermore, Uneda et al. showed that ATRAP-KO mice aged their kidneys faster, developed higher oxidative stress, and had shorter lifespans than their WT mice. The expression of SIRT1 was reduced, and these effects were accompanied by a decreased expression of sirtuin-1. These results collectively indicate that ATRAP may have protective effects against the aging of the kidney via SIRT1-dependent mechanisms, independently of AT1R-signaling, implicating ATRAP as an attractive therapeutic target for the prevention of senescence-driven renal decay (Uneda et al., 2017[337]) (Table 1(Tab. 1); References in Table 1: Bai et al., 2016[25]; Braidy et al., 2011[54]; Chuang et al., 2017[78]; Dong et al., 2023[96]; Fang et al., 2023[106]; Grosjean et al., 2018[129]; Hasegawa et al., 2010[139]; Keshavjee et al., 2022[177]; Li et al., 2019[201]; Lim et al., 2012[209]; Pushpakumar et al., 2020[277]; Rubattu et al., 2015[289]; Uneda et al., 2017[337]; Yu et al., 2023[382]).

Human clinical studies

It was previously demonstrated that human clinical studies reveal that SIRT1 activation has beneficial effects against age-induced renal manifestations through inhibition of oxidative stress and bettering mitochondrial function and autophagy on experimentally induced kidney aging (Afsar et al., 2021[3]; Liu et al., 2015[216]; Poulose and Raju, 2014[274]). Increased kidney SIRT1 is associated with superior renal outcomes and lower expression of senescence markers, supporting it as a potential treatment for aging-related renal pathology (Guo and Bechtel-Walz, 2023[132]). Zbroch et al. measured serum SIRT1 and αKlotho hemodialysis patients with end-stage renal disease. Results showed higher SIRT1 and lower αKlotho levels in hemodialysis patients than healthy controls. The relationship between SIRT1 and αKlotho was dependent on kidney function, considering that high SIRT1 values and low αKlotho levels were associated with reduced kidney function, as established, which suggests their as future novel biomarkers for oxidative stress and cardiovascular disease in renal patients (Zbroch et al., 2020[385]). In another investigation Vlassara et al. studies in diabetic CKD patients have demonstrated that sevelamer carbonate also significantly reduces HbA1c, serum AGEs, triglycerides and markers of inflammation and oxidative stress. This was associated with increased SIRT1 and AGE receptor 1 expression and PMNC TNF levels by this treatment. These results would indicate that the treatment of incipient nephropathy would be interfered with by senility of the gut-derived AGEs of metabolic and inflammatory anomalies and that sevelamer carbonate may be one of the end-stage renal disease-vulnerability factors to retard AGEs (Vlassara et al., 2012[346]).

Similarly, Serrano et al. found that sevelamer carbonate decreased AGEs circulating and cellular in those with type 2 diabetes and diabetic kidney disease, raised antioxidant defenses and reduced pro-oxidants. On the other hand, across the board, the treatment didn't significantly alter HbA1c or albumin/creatinine ratios but did yield benefits for specific subgroups. These findings indicate that sevelamer carbonate may ameliorate oxidative stress and inflammation of DKD, which is worth further confirming (Yubero-Serrano et al., 2015[383]).

Strategies for SIRT1 modulation

Small molecule activators

Small molecule activators of SIRT1, such as resveratrol and SRT2104, enhance SIRT1 activity, promoting longevity and metabolic health (Panickar and Jewell, 2015[264]). These activators bind to SIRT1, inducing conformational changes that increase deacetylase activity (Sturmlechner et al., 2017[314]). This modulation helps reduce oxidative stress, delay cellular senescence, and improve mitochondrial function, offering therapeutic potential for aging-related diseases (Ungar et al., 2000[338]). SLGT2 inhibitors are a class of drugs used to treat type 2 diabetes. They prevent glucose reabsorption in the kidneys, thereby increasing glucose excretion in urine and reducing blood sugar levels (Escobar et al., 2023[101]).

Additionally, and importantly, SGLT2 inhibitors are beneficial in terms of decreased cardiovascular and renal events beyond their effects on glycemia, rendering them among the preferred options for therapy (Ugusman et al., 2021[336]). Wicik et al. carried out a bioinformatic analysis. They thus identified the SGLT2 interaction network, which singled out SIRT1 as one of the most important interactors and indicated that SGLT2 inhibitors might exert systemic effects via SIRT1 regulation. This analysis uncovered central interactions of SGLT2 with oxidative stress and aging-related proteins, which may indicate the pleiotropic impact of SGLT2 inhibitors that extends beyond glucose lowering (Wicik et al., 2022[362]). Doxorubicin is an anthracycline chemotherapy agent used to treat a variety of cancers, including breast cancer, bladder cancer and lymphoma (Carvalho et al., 2009[62]), by blocking the growth and spread of cancer cells. As an injectable form, it intercalates DNA and inhibits topoisomerase II, thereby generating a free radical which causes DNA double-strand breaks that cannot be repaired. Adverse effects include alopecia, emetesia and cardiomyopathy (Rivankar, 2014[284]; Wu et al., 2022[366]). Xiang et al. showed that fasudil reduced oxidative stress, apoptosis, and senescence in the mouse model of doxorubicin-induced nephrotoxicity with potential involvement of SIRT1 activation. Pathway-directed treatment with Fasudil restored kidney function, DNA damage, and oxidative stress markers. The study implies that fasudil protects against cisplatin nephrotoxicity through oxidative stress and cell senescence (Xiang et al., 2021[369]).

PPARα agonists are substances activating the PPARα receptor, increasing lipid metabolism, decreasing inflammation, and increasing fatty acid oxidation (Bougarne et al., 2018[53]). They are most commonly employed in treating a range of conditions associated with hyperlipidemia and metabolic syndrome (Janani and Ranjitha Kumari, 2015[161]; Montaigne et al., 2021[243]). Kim et al. showed that the PPARα agonist fenofibrate prevents aging-related decline in renal function, proteinuria, inflammation, and fibrosis in mice. Results of the study revealed that compounds could activate AMPK and SIRT1 pathways, which could improve oxidative stress and mitochondrial dysfunction. PPARα is activated during aging, which is renal protective, due to the activation of AMPK-SIRT1 signaling, which can delay the aging process (Kim et al., 2016[184]).

Tropisetron is an antiemetic medicine used to prevent nausea and vomiting caused by cancer chemotherapy, radiation therapy, as well as after surgery. It acts on the serotonin receptors of the brain and the gut (de Bruijn, 1992[86]; Yang and Zhang, 2020[374]). Mirshafa et al. examined the impact of tropisetron on aging-mediated renal injury in the D-galactose-induced mouse model. All these beneficial effects of tropisetron treatment were associated with decreased oxidative stress, reduced mitochondrial dysfunction and inflammation, and induction of SIRT1. Moreover, these effects on tropisetron improved renal histopathology and function with the possibility of SIRT1-modulated renoprotective for preventing renal aging (Mirshafa et al., 2024[240]). Resveratrol is a natural polyphenol found abundantly in grapes, berries, and peanuts (Zhou et al., 2021[393]). They are rich in antioxidants and have been reported to have many health benefits, including anti-aging and its positive impact on cardiovascular health and actions against inflammation (Breuss et al., 2019[55]; Galiniak et al., 2019[119]). Wang et al. studied RSV improved mitochondria function and oxidative damage in rat kidneys following hemorrhage shock. SIRT1 and PGC1-α expression was also enhanced by RSV treatment in parallel with the elevation of antioxidant defenses. RSV supplementation during resuscitation ameliorated mitochondrial respiratory capacity impairment. It reduced ROS and lipid peroxidation, which may play key roles in its AKI protection, in conjunction with the improvement of mitochondrial functions and decrease of oxidative stress, as evidenced by the present study (Wang et al., 2015[351]). Pyridoxamine is a form of vitamin B6 having potential protective therapeutic profiles (Giannoukakis, 2005[124]). It serves mainly as an antioxidant, fighting against harmful free radicals and preventing the formation of AGEs (Voziyan and Hudson, 2005[347]). It is currently being studied for diabetes complications and kidney diseases (Voziyan and Hudson, 2005[348]). Simon et al. observed that pyridoxamine treatment in aged female mice increased glomerular SIRT1, ERα, and AGER1 expression and reduced TGFβ expression and collagen deposition. This study demonstrates that pyridoxamine can attenuate age-related oxidant stress and not only age-related kidney damage but perhaps AGE accumulation and, therefore, may be a beneficial therapeutic agent in preventing renal aging (Pereira-Simon et al., 2016[269]).

Natural compounds and nutraceuticals

Modulation of SIRT1 activity with natural compounds and nutraceuticals, such as resveratrol, is a powerful strategy, similar to other compounds such as quercetin, curcumin, and pterostilbene which also activates SIRT1, further promoting lifespan extension and anti-age-related diseases (Du et al., 2022[98]; Zhu et al., 2020[397]). They are adrenal cortex hormones, more commonly known as corticosteroids in the meal, used in diets to take advantage of their anti-inflammatory properties (Liu et al., 2023[219]; Tu et al., 2016[332]). Trehalose is sugar that consists of two glucose molecules and is limited to two glucose molecules. It acts as a power pack and a stress shield in different life forms (Vanaporn and Titball, 2020[342]). Trehalose: A sugar well known to stabilize proteins and cells in a dry state or under heat stress (Chen et al., 2022[67]). Bahri et al. demonstrate that aging rats supplemented with trehalose have lowered SIRT1 levels, mitigated oxidative stress inflammation, and improved kidney histopathology, indicating that trehalose could effectively serve as a dietary intervention for renal aging. Trehalose increases antioxidant ability and reduces malondialdehyde. The data suggest that modulation of SIRT1 by trehalose could attenuate at least some features of age-induced kidney injury (Bahri et al., 2021[24]).

Nrf2 is a transcription factor that, through a basic leucine zipper motif, regulates the genes that help in the protection of redox homeostasis and assists organisms in the prevention of tissue damage initiated by ROS through the upregulation of antioxidant proteins (Baird and Yamamoto, 2020[26]; He et al., 2020[141]). It is key in cellular defense, decreasing inflammation and detoxification mechanisms (Bellezza et al., 2018[35]). Kim et al. identified resveratrol, an Nrf2 activator, suppressed SIRT1/Nrf2 signaling, alleviated oxidative stress and mitochondrial dysfunction, and attenuated aging-related renal injury in mice. Resveratrol treatment improved renal function, reduced proteinuria, and attenuated age-associated pathophysiologic changes. The current study highlighted the role of resveratrol as an inducer of SIRT1 in attenuating oxidative stress and mitochondrial dysfunction in aging kidneys (Kim et al., 2018[183]). Ellagic acid is a natural plant polyphenol shown to scavenge oxygen-derived free radicals with a potential protective effect on mitochondria (Zhu et al., 2022[395]). Almost as focused on its antioxidant energy, it helps to fight free radicals - thereby decreasing oxidative force and infection (Shakeri et al., 2018[299]). This chemical can also be an anti-cancer, anti-inflammatory, and cardioprotective agent, thus becoming an important dietary constituent concerning human health (Deepika and Maurya, 2022[87]). Naghibi et al. showed that ellagic acid increased SIRT1 and NRF2 in elderly ventral kidneys, decreased oxidative stress, and improved renal function and histopathology. In conclusion, ellagic acid caused increases in antioxidative enzymes and decreases in malondialdehyde, an index of lipid peroxidation, were observed. As conclusion, these results indicate that ellagic acid reduces the susceptibility of the aging kidneys to damage by attenuating local oxidative stress to activate SIRT1 and NRF2 signaling, thus affirming the potential therapeutic agent for renal aging (Naghibi et al., 2023[251]).

Astragalus membranes contain numerous bioactive components, and APS is one of the foremost effective ingredients (Li et al., 2022[202]). Due to their immunomodulatory antioxidant, antioxidant and anti-inflammatory properties, ticks APS can be used in therapeutic aspects of supporting immune function, reducing oxidative stress and anti-inflammatory diseases (Li et al., 2023[205]; Zhang et al., 2019[391]). Miao et al. demonstrated that Astragalus polysaccharides could alleviate aortic endothelial senescence in aged rats by regulating SIRT1 and downstream p21, p53, and P16. APS treatment improved vascular function and decreased oxidative stress, possibly reflecting its anti-aging effects. The results indicated that APS may alleviate endothelial senescence and oxidative stress via the SIRT1/p53 signaling pathway (Bhat et al., 2024[45]; Miao et al., 2024[236]; Thapa et al., 2024[322]). Similarly, Tousian et al. used Alpha-mangostin to address high glucose-induced memory aging in human umbilical vein endothelial cells. Alpha-mangostin administration had similar beneficial effects to metformin regarding its impact on cell viability, oxidative stress, and accruing senescence markers, as shown in the study. These effects were mediated by the SIRT1 pathway, which indicated that alpha-mangostin may protect against vascular dysfunction and cellular senescence in the hyperglycemia state (Tousian et al., 2020[330]). Ginseng, a formidable traditional medicinal plant, has ginsenoside as a bioactive component (Kiefer and Pantuso, 2003[180]). Because of its health characteristics, ginsenoside functions as an anti-inflammatory, antioxidant, and anti-cancer, which improves the immune system, enhances energy, and brings wellness (Chen et al., 2019[72]; Mancuso and Santangelo, 2017[229]). Kim et al. showed the targeting of Akt/FoxO1 pathways and oxidative counterparts as potential issues of ginsenoside Rc in human renal cells. They show that treatment with the Rc increased catalase expression and scavenged reactive species by interacting with CBP and SIRT1. These results indicate that ginsenoside Rc may partially prevent oxidative stress-induced renal injury by regulating FoxO1 and the SIRT1 pathway (Kim et al., 2014[182]). Chlorogenic acid is one of the many natural compounds in coffee, fruits, and vegetables (Naveed et al., 2018[253]). These properties, which include antioxidant and anti-inflammatory properties, can be partly responsible for their potential health benefits, including improved glucose metabolism, weight management, and cardiovascular protection (Miao and Xiang, 2020[235]; Yun et al., 2022[384]). Hada et al. demonstrated that CGA attenuated vascular senescence by activating the Nrf2/HO-1 signaling pathway. Treatment of CGA increased the expression of SIRT1 and eNOS but decreased the protein levels of oxidative stress markers and senescence-associated proteins. This study confirmed that CGA could alleviate H2O2-induced endothelial cell senescence and suggested that it is a potential exogenous antioxidant against vascular aging. These results imply CGA as a possible medicine against vascular nutrients (Hada et al., 2020[135]).

Baicalin is a flavonoid isolated from the roots of Scutellaria baicalensis (Hu et al., 2021[150]). It has several pharmacological properties, such as anti-inflammatory, antioxidant, and neuroprotective effects (Bajek-Bil et al., 2023[27]). Baicalin has been widely studied for its potential therapeutic effects, particularly for cancer, cardiovascular disease, and neurodegenerative diseases (Wen et al., 2023[361]). Kim et al. stated that the flavonoid baicalin modulated FoxO1 phosphorylation and acetylation, thus promoting antioxidant defenses in aged rat kidneys. The therapeutic effect of Baicalin may inhibit the production of reactive species and increase the catalase expression, which was accomplished by SIRT1 and the PI3K/Akt pathway. Their results indicate that baicalin may inhibit oxidative stress and aging-related damage, and the underlying mechanism may be related to the modulation of the multiple signaling pathways, providing baicalin as a potential anti-aging agent (Kim et al., 2012[181]). Furthermore Zhang et al. showed that resveratrol prevents high-fat diet-mediated renal injury and cellular senescence in mice through regulation of SIRT1. These results suggested the capacity of resveratrol to protect the kidney from obesity-related renal disease by reducing oxidative stress and ameliorating renal histopathology. This study demonstrates the renoprotective effect of SIRT1 activation by resveratrol, suggesting its value as a therapeutic agent for the renal protection (Zhang et al., 2016[388]). Hydrogen sulfide is a colorless poisonous gas that smells like rotten eggs (Aroca and Gotor, 2022[15]; Bhat et al., 2024[42]; Thapa et al., 2023[324]). It is found naturally in volcanic sources, hot springs, and mineral waters. It is generated naturally by the bacterial decomposition of organic residues without oxygen (Dilek et al., 2020[92]; Huang and Xie, 2023[151]). Hou et al. studied the role of H2S in aging kidneys and demonstrated that H2S levels were lowered, the expression of H2S-producing enzymes CSE and CBS were downregulated in the kidneys of old mice H2S donor treatment also alleviated oxidative stress, collagen deposition, and induced antioxidant protein expressions. The amelioration of age-related kidney dysfunction by H2S was associated with the activation of Nrf2, suggesting that H2S supplementation is a potential therapeutic approach to attenuate age-related kidney dysfunction via decreasing oxidative stress and reinforcing the antioxidant defenses (Hou et al., 2016[147]). In another study Niu et al. showed that the consumption of long-term administration of EGCG expanded the life span of healthy rats by ameliorating liver and kidney functions, arresting age-associated inflammation and oxidative stress. The frequency of death and expression of NF-κB decreased significantly, while the expression of SIRT1 and FOXO3a significantly increased. Accordingly, EGCG treatment could postpone death. Based on these results, EGCG may mitigate hepatic and renal injury and potentially mediate inflammatory and oxidative cascades indicative of increase life span (Niu et al., 2013[258]).

Lifestyle interventions

Several strategies affect SIRT1 lifestyle interventions, such as exercise, caloric restriction, and an antioxidant-rich diet (Kitada et al., 2017[188]). Exercise increases the SIRT1 function, restores metabolic health, and reduces inflammation. And this thing is that caloric restriction upregulates SIRT1, promoting longevity and cellular repair (Saldanha et al., 2013[290]; Wang et al., 2021[356]). Those polyphenol-rich diets in berries, green tea, and red wine can also increase SIRT1 levels, which helps increase stress resistance and metabolic function (Qiu et al., 2021[281]). Ning et al. showed that short-term CR spared renal intrinsic aging in aged rats, decreasing oxidative damage and autophagic activity levels. CR increased SIRT1 and AMPK, reduced mTOR, increased autophagy, and lowered oxidative stress markers (Bhat et al., 2023[43]; Thapa et al., 2023[326]). Taken together, this study indicates that short-term CR, via the regulations of the main metabolic modulators and through induction of autophagy, is an efficient intervention to delay the progression of renal aging (Ning et al., 2013[257]). In another study, Wang et al. found that SIRT2 could deacetylate FOXO3a upon oxidative stress and caloric restriction. In the kidney and white adipose tissue of these animals, high expression of SIRT2 leads to increased FOXO3a DNA binding and gene expression of the targets SOD2 and catalase, accompanied by a decrease in reactive oxygen species levels. Its result indicates that SIRT2 is a critical player in the oxidative stress resistance and the survival of the cells during caloric restriction (Wang et al., 2007[350]) (Table 2(Tab. 2); References in Table 2: Bahri et al., 2021[24]; Fang et al., 2023[106]; Hada et al., 2020[135]; Kim et al., 2012[181], 2014[182], 2016[184], 2018[183]; Mirshafa et al., 2024[240]; Naghibi et al., 2023[251]; Pereira-Simon et al., 2016[269]; Wang et al., 2015[351]; Xiang et al., 2021[369]).

Challenges and limitations of SIRT1-based interventions

Although promising, interventions based on SIRT1 have several challenges and limitations that should be overcome to develop an effective therapy against age-related kidney dysfunction (Casalena et al., 2012[63]; Hao and Haase, 2010[138]). Most importantly, the delivery and bioavailability of SIRT1 activators, including resveratrol, are the major roadblocks to putting SIRT1 activation into clinical use, although preclinical studies suggest their benefit (Clark, 2000[80]). Resveratrol ingestion is characterized by poor bioavailability, associated with its rapid metabolism and elimination, which largely preclude significant clinical impact (Grinyó, 2000[128]; Ungar et al., 2000[339]). Improving the stability and delivery modalities of SIRT1 activators is important for developing effective drugs so that these compounds can reach sufficient therapeutic concentrations in target tissues (Moritz et al., 1997[246]). While this can lead to several side effects, as many of these activators also interact with other enzymes in the sirtuin family, some other compounds show SIRT1 specificity (Thapa et al., 2023[326], 2024[328]). Such non-specific activation may lead to collateral damage, minimizing the therapeutic effect (Bar-Shai et al., 2008[30]; Bertram, 2013[38]). However, developing selective sirtuin activators for SIRT1 that do not bind undesirable off-target interactions is needed for this safety to be adequate (Luckey and Parsa, 2003[225]). In addition, the chronic consequences of pro-longevity pathways, including SIRT1 activation, are incompletely characterized (Krzesinski and Delanaye, 2014[193]). Although short-term studies have shown beneficial effects on renal aging by SIRT1 activation, the longer-term consequences of chronic SIRT1 activation over a long period remain uncertain (Diz, 2008[94]; Weide and Huber, 2011[360]). Although a role for SIRT1 in repressing cellular senescence might be acknowledged, the possibility that sustained SIRT1 activation may be deleterious in the context of cancer progression and other age-related diseases necessitate further long-term studies to be conducted (Denecke et al., 2015[89]; Maruyama et al., 2010[230]; Vernier et al., 1971[344]). Additionally, differing individual responses to SIRT1 activators represents a further obstacle to clinical utility (Passmore et al., 2005[266]). Many factors, including genetic background, concurrent disease and the environment's influence, can impact the efficacy of SIRT1-based therapies (Davison, 1998[85]; Lindeman, 1993[212]). Given the genuine variation in these individual considerations, personalized strategies will be crucial to ensuring that the effectiveness of therapeutic intervention is optimized and risks minimized (Wang et al., 2021[356]). Lastly, translating conclusions from animal models to human studies is not without its symptoms (Campbell et al., 2021[60]). Most of the preclinical research showing the advantages of SIRT1 activation is in animal models (Nguyen and Corvera, 2024[255]). Human physiology and heterogeneity of human diseases together contribute to different consequences than can be seen in pre-clinical models and require robust clinical trials to evaluate the potential efficacy of SIRT1-based interventions in humans (Betjes, 2020[39]; Kitada et al., 2017[188]; Kuro, 2021[196]). While targeting Klotho, SIRT1, or the SIRT1/Klotho axis is very promising for the pharmacotherapy for kidney aging, inevitable issues, including delivery, specificity, long-term affordability, individual variability, and transgenic and translatable approach from rodents to humans will be essential to be overcome (Das et al., 2014[84]; Kashihara et al., 2012[173]; Little, 2011[215]). Extensive further research and development is required to address these restrictions and realize the full therapeutic capabilities of SIRT1 in age-related kidney dysfunction (Bagnasco, 2005[22]; Morrissey and Yango, 2006[247]).

Conclusion and Future Perspective

The interplay between oxidative stress and cellular senescence is significant in renal aging, driving functional decline and heightened disease susceptibility in these organs. Among those is the NAD+-dependent deacetylase SIRT1, which appears to be a major player in dampening these harmful processes. SIRT1 regulates the cellular ROS level by upregulating the transcription factors such as FOXO3a, PGC-1α and the antioxidant enzyme, including SOD, catalase, which in turn neutralize the ROS and keep the cellular redox homeostasis. SIRT1 also regulates senescence by deacetylating proteins like p53 and FOXO, activating DNA repair enzymes, lowering inflammation, and boosting stress resistance. SIRT1, a NAD-dependent protein deacetylase, protects against aging-induced renal injury, as in vitro and animal experimental evidence shows.

Further research is also needed to improve the bioavailability and stability of SIRT1 activators through developing more effective delivery systems. Such delivery could be fundamentally revolutionized using advanced drug delivery platforms, including nanotechnology, to target tissues at sustained concentrations that are pharmacologically active. Therefore, discovering selective SIRT1 activators without off-target effects may contribute to their safety and efficacy. Targeting SIRT1 for protective intervention into kidney aging is of great promise. Thus, tackling challenges related to mechanisms of action, delivery, specificity, long-term toxicity, intersubject variability, and translational studies is essential. Further research and development are necessary to fully exploit the benefits of SIRT1 in the context of improved renal health and extend kidney and health span in the aging population.

Declaration

Consent for publication

All authors gave their consent for publication.

Competing interests

No authors have any conflict of interest or competing interests to declare.

Funding

No funding was received to perform this study.

Author contributions

WHA researched the data and wrote the first draft of the manuscript. SSA edited the manuscript. All authors reviewed and approved the final version of the manuscript. Waleed Hassan Almalki is the guarantor of this work.

 

References

1. Aaseth J, Alexander J, Alehagen U, Tinkov A, Skalny A, Larsson A, et al. The aging kidney-as influenced by heavy metal exposure and selenium supplementation. Biomolecules. 2021;11(8):1078
2. Abrass IB. The biology and physiology of aging. West J Med. 1990;153:641-5
3. Afsar B, Afsar RE, Copur S, Sag AA, Ortiz A, Kanbay M. The effect of energy restriction on development and progression of chronic kidney disease: review of the current evidence. Br J Nutr. 2021;125:1201-14
4. Agarwal A, Aponte-Mellado A, Premkumar BJ, Shaman A, Gupta S. The effects of oxidative stress on female reproduction: a review. Reprod Biol Endocrinol. 2012;10:49
5. Akasaka-Manya K, Manya H, Endo T. Function and change with aging of α-klotho in the kidney. Vitam Horm. 2016;101:239-56
6. Ala M, Ala M. Metformin for cardiovascular protection, inflammatory bowel disease, osteoporosis, periodontitis, polycystic ovarian syndrome, neurodegeneration, cancer, inflammation and senescence: what is next? ACS Pharmacol Transl Sci. 2021;4:1747-70
7. Alam F, Syed H, Amjad S, Baig M, Khan TA, Rehman R. Interplay between oxidative stress, SIRT1, reproductive and metabolic functions. Curr Res Physiol. 2021;4:119-24
8. Alharbi KS, Almalki WH, Albratty M, Meraya AM, Najmi A, Vyas G, et al. The therapeutic role of nutraceuticals targeting the Nrf2/HO-1 signaling pathway in liver cancer. J Food Biochem. 2022;46(10):e14357
9. Alharbi KS, Almalki WH, Makeen HA, Albratty M, Meraya AM, Nagraik R, et al. Role of medicinal plant-derived nutraceuticals as a potential target for the treatment of breast cancer. J Food Biochem. 2022;46(12):e14387
10. Alharbi KS, Fuloria NK, Fuloria S, Rahman SB, Al-Malki WH, Javed Shaikh MA, et al. Nuclear factor-kappa B and its role in inflammatory lung disease. Chem Biol Interact. 2021;345:109568
11. Alharbi KS, Javed Shaikh MA, Afzal O, Alfawaz Altamimi AS, Almalki WH, Alzarea SI, et al. An overview of epithelial growth factor receptor (EGFR) inhibitors in cancer therapy. Chem Biol Interact. 2022;366:110108
12. Alves-Fernandes DK, Jasiulionis MG. The role of SIRT1 on DNA damage response and epigenetic alterations in cancer. Int J Mol Sci. 2019;20(13):3153
13. Alvis BD, Hughes CG. Physiology considerations in geriatric patients. Anesthesiol Clin. 2015;33:447-56
14. Andrade L, Rodrigues CE, Gomes SA, Noronha IL. Acute kidney injury as a condition of renal senescence. Cell Transplant. 2018;27:739-53
15. Aroca A, Gotor C. Hydrogen sulfide action in the regulation of plant autophagy. FEBS Lett. 2022;596:2186-97
16. Artioli GG, Sale C, Jones RL. Carnosine in health and disease. Eur J Sport Sci. 2019;19(1):30-9
17. Arulselvan P, Fard MT, Tan WS, Gothai S, Fakurazi S, Norhaizan ME, et al. Role of antioxidants and natural products in inflammation. Oxid Med Cell Longev. 2016;2016:5276130
18. Ashapkin VV, Kutueva LI, Vanyushin BF. Aging as an epigenetic phenomenon. Curr Genomics. 2017;18:385-407
19. Aubert G. Telomere dynamics and aging. Prog Mol Biol Transl Sci. 2014;125:89-111
20. Ayissi VB, Ebrahimi A, Schluesenner H. Epigenetic effects of natural polyphenols: a focus on SIRT1-mediated mechanisms. Mol Nutr Food Res. 2014;58(1):22-32
21. Azman KF, Zakaria R. D-Galactose-induced accelerated aging model: an overview. Biogerontology. 2019;20:763-82
22. Bagnasco SM. Role and regulation of urea transporters. Pflugers Arch. 2005;450:217-26
23. Bahl G, Pathak Y, Hussain MS, Gupta Y, Saraswat N. Navigating Sheehan syndrome's silent onset: A case report. J Clin Transl Endocrinol Case Rep. 2024;32:100168
24. Bahri F, Khaksari M, Movahedinia S, Shafiei B, Rajizadeh MA, Nazari-Robati M. Improving SIRT1 by trehalose supplementation reduces oxidative stress, inflammation, and histopathological scores in the kidney of aged rats. J Food Biochem. 2021;45(10):e13931
25. Bai X-Z, He T, Gao J-X, Liu Y, Liu J-Q, Han S-C, et al. Melatonin prevents acute kidney injury in severely burned rats via the activation of SIRT1. Sci Rep. 2016;6(1):32199
26. Baird L, Yamamoto M. The molecular mechanisms regulating the KEAP1-NRF2 pathway. Mol Cell Biol. 2020;40(13):e00099-20
27. Bajek-Bil A, Chmiel M, Włoch A, Stompor-Gorący M. Baicalin - current trends in detection methods and health-promoting properties. Pharmaceuticals (Basel). 2023;16(4):570
28. Barnes PJ. Oxidative stress-based therapeutics in COPD. Redox Biol. 2020;33:101544
29. Barnes PJ, Baker J, Donnelly LE. Cellular senescence as a mechanism and target in chronic lung diseases. Am J Respir Crit Care Med. 2019;200:556-64
30. Bar-Shai M, Carmeli E, Ljubuncic P, Reznick AZ. Exercise and immobilization in aging animals: the involvement of oxidative stress and NF-kappaB activation. Free Radic Biol Med. 2008;44:202-14
31. Bauer JH, Reams GP, Wu Z. The aging hypertensive kidney: pathophysiology and therapeutic options. Am J Med. 1991;90(4B):21s-7s
32. Baylis C. Changes in renal hemodynamics and structure in the aging kidney;sexual dimorphism and the nitric oxide system. Exp Gerontol. 2005;40:271-8
33. Baylis C. Sexual dimorphism: the aging kidney, involvement of nitric oxide deficiency, and angiotensin II overactivity. J Gerontol A Biol Sci Med Sci. 2012;67:1365-72
34. Baylis C, Corman B. The aging kidney: insights from experimental studies. J Am Soc Nephrol. 1998;9:699-709
35. Bellezza I, Giambanco I, Minelli A, Donato R. Nrf2-Keap1 signaling in oxidative and reductive stress. Biochim Biophys Acta Mol Cell Res. 2018;1865:721-33
36. Bennett WM. Geriatric pharmacokinetics and the kidney. Am J Kidney Dis. 1990;16:283-8
37. Benzing T, Salant D. Insights into glomerular filtration and albuminuria. N Engl J Med. 2021;384:1437-46
38. Bertram JF. Estimating glomerular number: why we do it and how. Clin Exp Pharmacol Physiol. 2013;40:785-8
39. Betjes MG. Uremia-associated ageing of the thymus and adaptive immune responses. Toxins (Basel). 2020;12(4):224
40. Beyfuss K, Hood DA. A systematic review of p53 regulation of oxidative stress in skeletal muscle. Redox Rep. 2018;23(1):100-17
41. Bhat AA, Afzal O, Afzal M, Gupta G, Thapa R, Ali H, et al. MALAT1: A key regulator in lung cancer pathogenesis and therapeutic targeting. Pathol Res Pract. 2024;253:154991
42. Bhat AA, Gilhotra RM, Thapa R, Subramaniyan V, Singh SK, Yadav HK, et al. Synergistic welfare of synbiotic nutraceuticals on neurological function. In: Dua K. (ed): Synbiotics in human health: biology to drug delivery (pp 571-80). Singapore: Springer, 2024
43. Bhat AA, Goyal A, Thapa R, Kazmi I, Alzarea SI, Singh M, et al. Uncovering the complex role of interferon-gamma in suppressing type 2 immunity to cancer. Cytokine. 2023;171:156376
44. Bhat AA, Gupta G, Alharbi KS, Afzal O, Altamimi ASA, Almalki WH, et al. Polysaccharide-based nanomedicines targeting lung cancer. Pharmaceutics. 2022;14(12):2788
45. Bhat AA, Kukreti N, Afzal M, Goyal A, Thapa R, Ali H, et al. Ferroptosis and circular RNAs: new horizons in cancer therapy. EXCLI J. 2024;23:570-99
46. Bhat AA, Thapa R, Afzal O, Agrawal N, Almalki WH, Kazmi I, et al. The pyroptotic role of Caspase-3/GSDME signalling pathway among various cancer: A Review. Int J Biol Macromol. 2023;242(Pt 2):124832
47. Bhatt S, Nagappa AN, Patil CR. Role of oxidative stress in depression. Drug Discov Today. 2020;25:1270-6
48. Bitzer M, Wiggins J. Aging biology in the kidney. Adv Chronic Kidney Dis. 2016;23(1):12-8
49. Bjørklund G, Shanaida M, Lysiuk R, Antonyak H, Klishch I, Shanaida V, et al. Selenium: an antioxidant with a critical role in anti-aging. Molecules. 2022;27(19):6613
50. Bolignano D, Mattace-Raso F, Sijbrands EJ, Zoccali C. The aging kidney revisited: a systematic review. Ageing Res Rev. 2014;14:65-80
51. Bose B, Cattran D. Glomerular diseases: FSGS. Clin J Am Soc Nephrol. 2014;9:626-32
52. Bouarich H, Chávez Guillén A, Rodríguez Puyol D. Kidney and hypertension in older adults. Med Clin (Barc). 2021;157:178-84
53. Bougarne N, Weyers B, Desmet SJ, Deckers J, Ray DW, Staels B, et al. Molecular actions of PPARα in lipid metabolism and inflammation. Endocr Rev. 2018;39:760-802
54. Braidy N, Guillemin GJ, Mansour H, Chan-Ling T, Poljak A, Grant R. Age related changes in NAD+ metabolism oxidative stress and Sirt1 activity in wistar rats. PLoS One. 2011;6(4):e19194
55. Breuss JM, Atanasov AG, Uhrin P. Resveratrol and its effects on the vascular system. Int J Mol Sci. 2019;20(7):1523
56. Bridges CC, Zalups RK. The aging kidney and the nephrotoxic effects of mercury. J Toxicol Environ Health B Crit Rev. 2017;20:55-80
57. Buemi M, Nostro L, Aloisi C, Cosentini V, Criseo M, Frisina N. Kidney aging: from phenotype to genetics. Rejuvenation Res. 2005;8:101-9
58. Byrnes K, Blessinger S, Bailey NT, Scaife R, Liu G, Khambu B. Therapeutic regulation of autophagy in hepatic metabolism. Acta Pharm Sin B. 2022;12(1):33-49
59. Cailleaux PE, Cohen-Solal M. Managing Musculoskeletal and kidney aging: a call for holistic insights. Clin Interv Aging. 2022;17:717-32
60. Campbell RA, Docherty MH, Ferenbach DA, Mylonas KJ. The role of ageing and parenchymal senescence on macrophage function and fibrosis. Front Immunol. 2021;12:700790
61. Carling D. AMPK signalling in health and disease. Curr Opin Cell Biol. 2017;45:31-7
62. Carvalho C, Santos RX, Cardoso S, Correia S, Oliveira PJ, Santos MS, et al. Doxorubicin: the good, the bad and the ugly effect. Curr Med Chem. 2009;16:3267-85
63. Casalena G, Daehn I, Bottinger E. Transforming growth factor-β, bioenergetics, and mitochondria in renal disease. Semin Nephrol. 2012;32:295-303
64. Chandra A, Rajawat J. Skeletal aging and osteoporosis: mechanisms and therapeutics. Int J Mol Sci. 2021;22 (7):3553
65. Chang HC, Guarente L. SIRT1 and other sirtuins in metabolism. Trends Endocrinol Metab. 2014;25:138-45
66. Charloux A, Brandenberger G, Piquard F, Geny B. Dysregulation of pulsatility in aging IV. Pulsatile signaling and cardiovascular aging: functions and regulation of natriuretic peptide signaling. Ageing Res Rev. 2008;7:151-63
67. Chen A, Tapia H, Goddard JM, Gibney PA. Trehalose and its applications in the food industry. Compr Rev Food Sci Food Saf. 2022;21:5004-37
68. Chen C, Zhou M, Ge Y, Wang X. SIRT1 and aging related signaling pathways. Mech Ageing Dev. 2020;187:111215
69. Chen D, Xie R, Shu B, Landay AL, Wei C, Reiser J, et al. Wnt signaling in bone, kidney, intestine, and adipose tissue and interorgan interaction in aging. Ann N Y Acad Sci. 2019;1442(1):48-60
70. Chen J, Chen H, Pan L. SIRT1 and gynecological malignancies (Review). Oncol Rep. 2021;45(4):43
71. Chen L, Ma Y, Ma X, Liu L, Jv X, Li A, et al. TFEB regulates cellular labile iron and prevents ferroptosis in a TfR1-dependent manner. Free Radic Biol Med. 2023;208:445-57
72. Chen W, Balan P, Popovich DG. Review of Ginseng anti-diabetic studies. Molecules. 2019;24(24):4501
73. Chen Y, Mehta G, Vasiliou V. Antioxidant defenses in the ocular surface. Ocul Surf. 2009;7:176-85
74. Chen Y, Zhou F, Liu H, Li J, Che H, Shen J, et al. SIRT1, a promising regulator of bone homeostasis. Life Sci. 2021;269:119041
75. Chen Z, Shentu TP, Wen L, Johnson DA, Shyy JY. Regulation of SIRT1 by oxidative stress-responsive miRNAs and a systematic approach to identify its role in the endothelium. Antioxid Redox Signal. 2013;19 :1522-38
76. Cheng S, Huang M, Liu S, Yang M. Bisphenol F and bisphenol S induce metabolic perturbations in human ovarian granulosa cells. Arab J Chem. 2024;17(9):105904
77. Choudhury D, Levi M. Kidney aging - inevitable or preventable? Nat Rev Nephrol. 2011;7:706-17
78. Chuang PY, Cai W, Li X, Fang L, Xu J, Yacoub R, et al. Reduction in podocyte SIRT1 accelerates kidney injury in aging mice. Am J Physiol Renal Physiol. 2017;313:F621-8
79. Chung S, Yao H, Caito S, Hwang JW, Arunachalam G, Rahman I. Regulation of SIRT1 in cellular functions: role of polyphenols. Arch Biochem Biophys. 2010;501(1):79-90
80. Clark B. Biology of renal aging in humans. Adv Ren Replace Ther. 2000;7(1):11-21
81. Covarrubias AJ, Perrone R, Grozio A, Verdin E. NAD(+) metabolism and its roles in cellular processes during ageing. Nat Rev Mol Cell Biol. 2021;22:119-41
82. Cui Z, Zhao X, Amevor FK, Du X, Wang Y, Li D, et al. Therapeutic application of quercetin in aging-related diseases: SIRT1 as a potential mechanism. Front Immunol. 2022;13:943321
83. Darendeliler F. IUGR: Genetic influences, metabolic problems, environmental associations/triggers, current and future management. Best Pract Res Clin Endocrinol Metab. 2019;33(3):101260
84. Das BC, Thapa P, Karki R, Das S, Mahapatra S, Liu TC, et al. Retinoic acid signaling pathways in development and diseases. Bioorg Med Chem. 2014;22:673-83
85. Davison AM. Renal disease in the elderly. Nephron. 1998;80(1):6-16
86. de Bruijn KM. Tropisetron. A review of the clinical experience. Drugs. 1992;43(Suppl 3):11-22
87. Deepika, Maurya PK. Ellagic acid: insight into its protective effects in age-associated disorders. 3 Biotech. 2022;12(12):340
88. Demirci-Çekiç S, Özkan G, Avan AN, Uzunboy S, Çapanoğlu E, Apak R. Biomarkers of oxidative stress and antioxidant defense. J Pharm Biomed Anal. 2022;209:114477
89. Denecke C, Biebl M, Pratschke J. Optimizing clinical utilization and allocation of older kidneys. Curr Opin Organ Transplant. 2015;20:431-7
90. Denic A, Glassock RJ, Rule AD. Structural and functional changes with the aging kidney. Adv Chronic Kidney Dis. 2016;23(1):19-28
91. Denic A, Rule AD, Glassock RJ. Healthy and unhealthy aging on kidney structure and function: human studies. Curr Opin Nephrol Hypertens. 2022;31:228-34
92. Dilek N, Papapetropoulos A, Toliver-Kinsky T, Szabo C. Hydrogen sulfide: An endogenous regulator of the immune system. Pharmacol Res. 2020;161:105119
93. DiNicolantonio JJ, McCarty MF, O'Keefe JH. Nutraceutical activation of Sirt1: a review. Open Heart. 2022;9(2):e002171
94. Diz DI. Lewis K. Dahl memorial lecture: the renin-angiotensin system and aging. Hypertension. 2008;52 (1):37-43
95. do Nascimento JL, Oliveira KR, Crespo-Lopez ME, Macchi BM, Maués LA, Pinheiro Mda C, et al. Methylmercury neurotoxicity & antioxidant defenses. Indian J Med Res. 2008;128:373-82
96. Dong W, Zhang K, Gong Z, Luo T, Li J, Wang X, et al. N-acetylcysteine delayed cadmium-induced chronic kidney injury by activating the sirtuin 1-P53 signaling pathway. Chem Biol Interact. 2023;369:110299
97. D'Onofrio N, Servillo L, Balestrieri ML. SIRT1 and SIRT6 signaling pathways in cardiovascular disease protection. Antioxid Redox Signal. 2018;28:711-32
98. Du Y, Zhu YJ, Zhou YX, Ding J, Liu JY. Metformin in therapeutic applications in human diseases: its mechanism of action and clinical study. Mol Biomed. 2022;3(1):41
99. Dybiec J, Szlagor M, Młynarska E, Rysz J, Franczyk B. Structural and functional changes in aging kidneys. Int J Mol Sci. 2022;23(23):15435
100. Epstein M. Aging and the kidney. J Am Soc Nephrol. 1996;7:1106-22
101. Escobar C, Pascual-Figal D, Manzano L, Nuñez J, Camafort M. Current tole of SLGT2 inhibitors in the management of the whole spectrum of heart failure: focus on dapagliflozin. J Clin Med. 2023;12(21):6798
102. Esposito C, Dal Canton A. Functional changes in the aging kidney. J Nephrol. 2010;23(Suppl 15):S41-5
103. Famulski KS, Halloran PF. Molecular events in kidney ageing. Curr Opin Nephrol Hypertens. 2005;14:243-8
104. Fan J, Lei W, Wang L, Ge W. A nomogram for predicting the risk of treatment failure of roxadustat in peritoneal dialysis with renal anemia. Sci Rep. 2024;14(1):7622
105. Fang C, Xu H, Yuan L, Zhu Z, Wang X, Liu Y, et al. Natural compounds for SIRT1-mediated oxidative stress and neuroinflammation in stroke: a potential therapeutic target in the future. Oxid Med Cell Longev. 2022;2022:1949718
106. Fang R, Chen J, Long J, Zhang B, Huang Q, Li S, et al. Empagliflozin improves kidney senescence induced by D-galactose by reducing sirt1-mediated oxidative stress. Biogerontology. 2023;24:771-82
107. Fang Y, Gong AY, Haller ST, Dworkin LD, Liu Z, Gong R. The ageing kidney: Molecular mechanisms and clinical implications. Ageing Res Rev. 2020;63:101151
108. Feng X, Chen W, Ni X, Little PJ, Xu S, Tang L, et al. Metformin, macrophage dysfunction and atherosclerosis. Front Immunol. 2021;12:682853
109. Feng Y, Chen Y, Wu X, Chen J, Zhou Q, Liu B, et al. Interplay of energy metabolism and autophagy. Autophagy. 2024;20(1):4-14
110. Ferenbach DA, Bonventre JV. Mechanisms of maladaptive repair after AKI leading to accelerated kidney ageing and CKD. Nat Rev Nephrol. 2015;11:264-76
111. Fernandes GFS, Silva GDB, Pavan AR, Chiba DE, Chin CM, Dos Santos JL. Epigenetic regulatory mechanisms induced by resveratrol. Nutrients. 2017;9(11):1201
112. Fledderus J, Vanchin B, Rots MG, Krenning G. The endothelium as a target for anti-atherogenic therapy: a focus on the epigenetic enzymes EZH2 and SIRT1. J Pers Med. 2021;11(2):103
113. Forman HJ, Zhang H. Targeting oxidative stress in disease: promise and limitations of antioxidant therapy. Nat Rev Drug Discov. 2021;20:689-709
114. Fougeray S, Pallet N. Mechanisms and biological functions of autophagy in diseased and ageing kidneys. Nat Rev Nephrol. 2015;11(1):34-45
115. Franzin R, Stasi A, Fiorentino M, Stallone G, Cantaluppi V, Gesualdo L, et al. Inflammaging and complement system: a link between acute kidney injury and chronic graft damage. Front Immunol. 2020;11:734
116. Frassetto LA, Sebastian A, DuBose TD Jr. How metabolic acidosis and kidney disease may accelerate the aging process. Eur J Clin Nutr. 2020;74(Suppl 1):27-32
117. Fukuda Y, Akimoto K, Homma T, Baker JR, Ito K, Barnes PJ, et al. Virus-induced asthma exacerbations: SIRT1 targeted approach. J Clin Med. 2020;9(8):2623
118. Fusco S, Garasto S, Corsonello A, Vena S, Mari V, Gareri P, et al. Medication-induced nephrotoxicity in older patients. Curr Drug Metab. 2016;17:608-25
119. Galiniak S, Aebisher D, Bartusik-Aebisher D. Health benefits of resveratrol administration. Acta Biochim Pol. 2019;66(1):13-21
120. Garcia-Peterson LM, Li X. Trending topics of SIRT1 in tumorigenicity. Biochim Biophys Acta Gen Subj. 2021;1865(9):129952
121. Gava AL, Freitas FP, Meyrelles SS, Silva IV, Graceli JB. Gender-dependent effects of aging on the kidney. Braz J Med Biol Res. 2011;44:905-13
122. Gekle M. Kidney and aging - A narrative review. Exp Gerontol. 2017;87(Pt B):153-5
123. Genchi G, Sinicropi MS, Lauria G, Carocci A, Catalano A. The effects of cadmium toxicity. Int J Environ Res Public Health. 2020;17(11):3782
124. Giannoukakis N. Pyridoxamine (BioStratum). Curr Opin Investig Drugs. 2005;6:410-8
125. Glassock R, Denic A, Rule AD. When kidneys get old: an essay on nephro-geriatrics. J Bras Nefrol. 2017;39 (1):59-64
126. Glassock RJ, Rule AD. Aging and the kidneys: anatomy, physiology and consequences for defining chronic kidney disease. Nephron. 2016;134(1):25-9
127. Gonzalo S, Kreienkamp R, Askjaer P. Hutchinson-Gilford Progeria Syndrome: A premature aging disease caused by LMNA gene mutations. Ageing Res Rev. 2017;33:18-29
128. Grinyó JM. Borderline kidney graft donors--what are the problems? Nephrol Dial Transplant. 2000;15:950-2
129. Grosjean F, Yubero-Serrano EM, Zheng F, Esposito V, Swamy S, Elliot SJ, et al. Pharmacologic control of oxidative stress and inflammation determines whether diabetic glomerulosclerosis progresses or decreases: A pilot study in sclerosis-prone mice. PLoS One. 2018;13(9):e0204366
130. Guan Y, Hao CM. SIRT1 and kidney function. Kidney Dis (Basel). 2016;1(4):258-65
131. Guebre-Egziabher F, Alix PM, Koppe L, Pelletier CC, Kalbacher E, Fouque D, et al. Ectopic lipid accumulation: A potential cause for metabolic disturbances and a contributor to the alteration of kidney function. Biochimie. 2013;95:1971-9
132. Guo H, Bechtel-Walz W. The interplay of autophagy and oxidative stress in the kidney: what do we know? Nephron. 2023;147:627-42
133. Guo J, Zheng HJ, Zhang W, Lou W, Xia C, Han XT, et al. Accelerated kidney aging in diabetes mellitus. Oxid Med Cell Longev. 2020;2020:1234059
134. Gupta G, Al-Malki WH, Kazmi I, Thangavelu L, Gupta PK, Jha NK, et al. The role of HGF/MET in liver cancer. Future Med Chem. 2021;13:1829-32
135. Hada Y, Uchida HA, Otaka N, Onishi Y, Okamoto S, Nishiwaki M, et al. The protective effect of chlorogenic acid on vascular senescence via the Nrf2/HO-1 pathway. Int J Mol Sci. 2020;21(12):4527
136. Halliwell B. Understanding mechanisms of antioxidant action in health and disease. Nat Rev Mol Cell Biol. 2024;25(1):13-33
137. Han X, Ding C, Sang X, Peng M, Yang Q, Ning Y, et al. Targeting Sirtuin1 to treat aging-related tissue fibrosis: From prevention to therapy. Pharmacol Ther. 2022;229:107983
138. Hao CM, Haase VH. Sirtuins and their relevance to the kidney. J Am Soc Nephrol. 2010;21:1620-7
139. Hasegawa K, Wakino S, Yoshioka K, Tatematsu S, Hara Y, Minakuchi H, et al. Kidney-specific overexpression of Sirt1 protects against acute kidney injury by retaining peroxisome function. J Biol Chem. 2010;285:13045-56
140. Hayashi K, Hishikawa A, Itoh H. DNA damage repair and DNA methylation in the kidney. Am J Nephrol. 2019;50:81-91
141. He F, Ru X, Wen T. NRF2, a transcription factor for stress response and beyond. Int J Mol Sci. 2020;21(13):4777
142. He X, Wan F, Su W, Xie W. Research progress on skin aging and active ingredients. Molecules. 2023;28(14):5556
143. Hekmatimoghaddam S, Dehghani Firoozabadi A, Zare-Khormizi MR, Pourrajab F. Sirt1 and Parp1 as epigenome safeguards and microRNAs as SASP-associated signals, in cellular senescence and aging. Ageing Res Rev. 2017;40:120-41
144. Herzig S, Shaw RJ. AMPK: guardian of metabolism and mitochondrial homeostasis. Nat Rev Mol Cell Biol. 2018;19:121-35
145. Hirakawa Y, Jao TM, Inagi R. Pathophysiology and therapeutics of premature ageing in chronic kidney disease, with a focus on glycative stress. Clin Exp Pharmacol Physiol. 2017;44(Suppl 1):70-7
146. Hommos MS, Glassock RJ, Rule AD. Structural and functional changes in human kidneys with healthy aging. J Am Soc Nephrol. 2017;28:2838-44
147. Hou CL, Wang MJ, Sun C, Huang Y, Jin S, Mu XP, et al. Protective effects of hydrogen sulfide in the ageing kidney. Oxid Med Cell Longev. 2016;2016:7570489
148. Hu HH, Cao G, Wu XQ, Vaziri ND, Zhao YY. Wnt signaling pathway in aging-related tissue fibrosis and therapies. Ageing Res Rev. 2020;60:101063
149. Hu J, Xu T, Shen H, Song Y, Yang J, Zhang A, et al. Accuracy of gallium-68 pentixafor positron emission tomography–computed tomography for subtyping diagnosis of primary aldosteronism. JAMA Network Open. 2023;6(2):e2255609-e
150. Hu Q, Zhang W, Wu Z, Tian X, Xiang J, Li L, et al. Baicalin and the liver-gut system: Pharmacological bases explaining its therapeutic effects. Pharmacol Res. 2021;165:105444
151. Huang J, Xie Y. Hydrogen sulfide signaling in plants. Antioxid Redox Signal. 2023;39(1-3):40-58
152. Huber TB, Edelstein CL, Hartleben B, Inoki K, Jiang M, Koya D, et al. Emerging role of autophagy in kidney function, diseases and aging. Autophagy. 2012;8:1009-31
153. Hussain MS, Afzal O, Gupta G, Altamimi ASA, Almalki WH, Alzarea SI, et al. Long non-coding RNAs in lung cancer: Unraveling the molecular modulators of MAPK signaling. Pathol Res Pract. 2023;249:154738
154. Hussain MS, Gupta G, Afzal M, Alqahtani SM, Samuel VP, Hassan Almalki W, et al. Exploring the role of lncrna neat1 knockdown in regulating apoptosis across multiple cancer types: A review. Pathol Res Pract. 2023;252:154908
155. Hussain MS, Maqbool M, Shaikh NK, Agrawal M, Sultana A. Therapeutic landscape of diabetic nephropathy: insights from long noncoding RNAs. Clin Diabetol. 2024;13(4):1-12
156. Hwang JW, Yao H, Caito S, Sundar IK, Rahman I. Redox regulation of SIRT1 in inflammation and cellular senescence. Free Radic Biol Med. 2013;61:95-110
157. Imai S, Guarente L. NAD+ and sirtuins in aging and disease. Trends Cell Biol. 2014;24:464-71
158. Imran SAM, Yazid MD, Idrus RBH, Maarof M, Nordin A, Razali RA, et al. Is there an interconnection between Epithelial-Mesenchymal Transition (EMT) and telomere shortening in aging? Int J Mol Sci. 2021;22 (8):3888
159. Islam MR, Rauf A, Alash S, Fakir MNH, Thufa GK, Sowa MS, et al. A comprehensive review of phytoconstituents in liver cancer prevention and treatment: targeting insights into molecular signaling pathways. Med Oncol. 2024;41(6):134
160. Jalgaonkar MP, Parmar UM, Kulkarni YA, Oza MJ. SIRT1-FOXOs activity regulates diabetic complications. Pharmacol Res. 2022;175:106014
161. Janani C, Ranjitha Kumari BD. PPAR gamma gene - a review. Diabetes Metab Syndr. 2015;9(1):46-50
162. Jassal SV, Oreopoulos DG. The aging kidney. Geriatr Nephrol Urol. 1998;8:141-7
163. Jia G, Aroor AR, Jia C, Sowers JR. Endothelial cell senescence in aging-related vascular dysfunction. Biochim Biophys Acta Mol Basis Dis. 2019;1865:1802-9
164. Jiang S, Li T, Yang Z, Yi W, Di S, Sun Y, et al. AMPK orchestrates an elaborate cascade protecting tissue from fibrosis and aging. Ageing Res Rev. 2017;38:18-27
165. Jin Q, Liu T, Ma F, Fu T, Yang L, Mao H, et al. Roles of Sirt1 and its modulators in diabetic microangiopathy: A review. Int J Biol Macromol. 2024;264(Pt 2):130761
166. Jin Q, Liu T, Qiao Y, Liu D, Yang L, Mao H, et al. Oxidative stress and inflammation in diabetic nephropathy: role of polyphenols. Front Immunol. 2023;14:1185317
167. Jo MJ, Lee JK, Kim JE, Ko GJ. Molecular mechanisms associated with aging kidneys and future perspectives. Int J Mol Sci. 2023;24(23):16912
168. Jones MJ, Goodman SJ, Kobor MS. DNA methylation and healthy human aging. Aging Cell. 2015;14:924-32
169. Kaarniranta K, Kajdanek J, Morawiec J, Pawlowska E, Blasiak J. PGC-1α protects RPE cells of the aging retina against oxidative stress-induced degeneration through the regulation of senescence and mitochondrial quality control. the significance for AMD pathogenesis. Int J Mol Sci. 2018;19(8):2317
170. Kanasaki K, Kitada M, Koya D. Pathophysiology of the aging kidney and therapeutic interventions. Hypertens Res. 2012;35:1121-8
171. Kanbay M, Demiray A, Afsar B, Covic A, Tapoi L, Ureche C, et al. Role of Klotho in the Development of essential hypertension. Hypertension. 2021;77:740-50
172. Karam Z, Tuazon J. Anatomic and physiologic changes of the aging kidney. Clin Geriatr Med. 2013;29:555-64
173. Kashihara N, Satoh M, Sasaki T. [Chronic kidney disease (CKD)--recent progress. Topics: VI. Chronic kidney disease (CKD) and associated disorders: 9. Kidney and senescence]. Nihon Naika Gakkai Zasshi. 2012;101:1332-9
174. Katz DL, Doughty K, Ali A. Cocoa and chocolate in human health and disease. Antioxid Redox Signal. 2011;15:2779-811
175. Kauppinen A, Suuronen T, Ojala J, Kaarniranta K, Salminen A. Antagonistic crosstalk between NF-κB and SIRT1 in the regulation of inflammation and metabolic disorders. Cell Signal. 2013;25:1939-48
176. Kaysen GA, Myers BD. The aging kidney. Clin Geriatr Med. 1985;1(1):207-22
177. Keshavjee B, Lambelet V, Coppola H, Viertl D, Prior JO, Kappeler L, et al. Stress-induced premature senescence related to oxidative stress in the developmental programming of nonalcoholic fatty liver disease in a rat model of intrauterine growth restriction. Antioxidants (Basel). 2022;11(9):1695
178. Khan S, Loi V, Rosner MH. Drug-induced kidney injury in the elderly. Drugs Aging. 2017;34:729-41
179. Kida Y, Goligorsky MS. Sirtuins, cell senescence, and vascular aging. Can J Cardiol. 2016;32:634-41
180. Kiefer D, Pantuso T. Panax ginseng. Am Fam Physician. 2003;68:1539-42
181. Kim DH, Kim JM, Lee EK, Choi YJ, Kim CH, Choi JS, et al. Modulation of FoxO1 phosphorylation/acetylation by baicalin during aging. J Nutr Biochem. 2012;23:1277-84
182. Kim DH, Park CH, Park D, Choi YJ, Park MH, Chung KW, et al. Ginsenoside Rc modulates Akt/FoxO1 pathways and suppresses oxidative stress. Arch Pharm Res. 2014;37:813-20
183. Kim EN, Lim JH, Kim MY, Ban TH, Jang IA, Yoon HE, et al. Resveratrol, an Nrf2 activator, ameliorates aging-related progressive renal injury. Aging (Albany NY). 2018;10(1):83-99
184. Kim EN, Lim JH, Kim MY, Kim HW, Park CW, Chang YS, et al. PPARα agonist, fenofibrate, ameliorates age-related renal injury. Exp Gerontol. 2016;81:42-50
185. Kim JY, Mondaca-Ruff D, Singh S, Wang Y. SIRT1 and autophagy: implications in endocrine disorders. Front Endocrinol (Lausanne). 2022;13:930919
186. Kimura T, Isaka Y, Yoshimori T. Autophagy and kidney inflammation. Autophagy. 2017;13:997-1003
187. Kitada M, Kume S, Takeda-Watanabe A, Kanasaki K, Koya D. Sirtuins and renal diseases: relationship with aging and diabetic nephropathy. Clin Sci (Lond). 2013;124:153-64
188. Kitada M, Ogura Y, Monno I, Koya D. Regulating autophagy as a therapeutic target for diabetic nephropathy. Curr Diab Rep. 2017;17(7):53
189. Kitai Y, Nangaku M, Yanagita M. Aging-related kidney diseases. Contrib Nephrol. 2021;199:266-73
190. Kłoda K, Drozd A, Borowiecka E, Domański L. [Telomere length and transrenal DNA isolated from transplanted kidney recipients' urine]. Postepy Hig Med Dosw (Online). 2015;69:649-53
191. Kong X, Guan J, Li J, Wei J, Wang R. P66(Shc)-SIRT1 regulation of oxidative stress protects against cardio-cerebral vascular disease. Mol Neurobiol. 2017;54:5277-85
192. Kooman JP, Shiels PG, Stenvinkel P. Premature aging in chronic kidney disease and chronic obstructive pulmonary disease: similarities and differences. Curr Opin Clin Nutr Metab Care. 2015;18:528-34
193. Krzesinski JM, Delanaye P. [How to manage chronic kidney disease in the elderly?]. Rev Med Liege. 2014;69:287-93
194. Kume S, Kitada M, Kanasaki K, Maegawa H, Koya D. Anti-aging molecule, Sirt1: a novel therapeutic target for diabetic nephropathy. Arch Pharm Res. 2013;36:230-6
195. Kuro-O M. A potential link between phosphate and aging--lessons from Klotho-deficient mice. Mech Ageing Dev. 2010;131:270-5
196. Kuro-O M. Klotho and calciprotein particles as therapeutic targets against accelerated ageing. Clin Sci (Lond). 2021;135:1915-27
197. Lagunas-Rangel FA. SIRT7 in the aging process. Cell Mol Life Sci. 2022;79(6):297
198. Lamb EJ, O'Riordan SE, Delaney MP. Kidney function in older people: pathology, assessment and management. Clin Chim Acta. 2003;334(1-2):25-40
199. Leal F, García-Perdomo HA. Effectiveness of platinum-based chemotherapy in patients with metastatic prostate cancer: systematic review and meta-analysis. Clin Genitourin Cancer. 2019;17(3):e627-e44
200. Lee SH, Lee JH, Lee HY, Min KJ. Sirtuin signaling in cellular senescence and aging. BMB Rep. 2019;52(1):24-34
201. Li C, Xie N, Li Y, Liu C, Hou FF, Wang J. N-acetylcysteine ameliorates cisplatin-induced renal senescence and renal interstitial fibrosis through sirtuin1 activation and p53 deacetylation. Free Radic Biol Med. 2019;130:512-27
202. Li CX, Liu Y, Zhang YZ, Li JC, Lai J. Astragalus polysaccharide: a review of its immunomodulatory effect. Arch Pharm Res. 2022;45:367-89
203. Li J, Jia B, Cheng Y, Song Y, Li Q, Luo C. Targeting molecular mediators of ferroptosis and oxidative stress for neurological disorders. Oxid Med Cell Longev. 2022;2022:3999083
204. Li T, Yang K, Gao W, Peng F, Zou X. Cellular senescence in acute kidney injury: Target and opportunity. Biochem Biophys Res Commun. 2024;706:149744
205. Li Z, Qi J, Guo T, Li J. Research progress of Astragalus membranaceus in treating peritoneal metastatic cancer. J Ethnopharmacol. 2023;305:116086
206. Li Z, Wang Z. Aging kidney and aging-related disease. Adv Exp Med Biol. 2018;1086:169-87
207. Li Z, Zhang Z, Ren Y, Wang Y, Fang J, Yue H, et al. Aging and age-related diseases: from mechanisms to therapeutic strategies. Biogerontology. 2021;22:165-87
208. Liguori I, Russo G, Curcio F, Bulli G, Aran L, Della-Morte D, et al. Oxidative stress, aging, and diseases. Clin Interv Aging. 2018;13:757-72
209. Lim JH, Kim EN, Kim MY, Chung S, Shin SJ, Kim HW, et al. Age-associated molecular changes in the kidney in aged mice. Oxid Med Cell Longev. 2012;2012:171383
210. Lin J, Epel E. Stress and telomere shortening: Insights from cellular mechanisms. Ageing Res Rev. 2022;73:101507
211. Lin X, Jin H, Chai Y, Shou S. Cellular senescence and acute kidney injury. Pediatr Nephrol. 2022;37:3009-18
212. Lindeman RD. Renal physiology and pathophysiology of aging. Contrib Nephrol. 1993;105:1-12
213. Lindeman RD, Goldman R. Anatomic and physiologic age changes in the kidney. Exp Gerontol. 1986;21:379-406
214. Lith SC, de Vries CJM. Nuclear receptor Nur77: its role in chronic inflammatory diseases. Essays Biochem. 2021;65:927-39
215. Little MH. Renal organogenesis: what can it tell us about renal repair and regeneration? Organogenesis. 2011;7:229-41
216. Liu FC, Tsai YF, Tsai HI, Yu HP. Anti-inflammatory and organ-protective effects of resveratrol in trauma-hemorrhagic injury. Mediators Inflamm. 2015;2015:643763
217. Liu HM, Cheng MY, Xun MH, Zhao ZW, Zhang Y, Tang W, et al. Possible mechanisms of oxidative stress-induced skin cellular senescence, inflammation, and cancer and the therapeutic potential of plant polyphenols. Int J Mol Sci. 2023;24(4):3755
218. Liu L, Ma D, Zhuo L, Pang X, You J, Feng J. Progress and promise of Nur77-based therapeutics for central nervous system disorders. Curr Neuropharmacol. 2021;19:486-97
219. Liu Q, Chen J, Zeng A, Song L. Pharmacological functions of salidroside in renal diseases: facts and perspectives. Front Pharmacol. 2023;14:1309598
220. Liu RM. Aging, cellular senescence, and Alzheimer's disease. Int J Mol Sci. 2022;23(4):1989
221. Liu Z, Wang C, Pei J, Li M, Gu W. SIRT1: A novel protective molecule in pre-eclampsia. Int J Med Sci. 2022;19:993-1002
222. Lonergan ET. Aging and the kidney: adjusting treatment to physiologic change. Geriatrics. 1988;43(3):27-30, 32-3
223. Longo S, Borghesi A, Tzialla C, Stronati M. IUGR and infections. Early Hum Dev. 2014;90(Suppl 1):S42-4
224. Lu C, Zhao H, Liu Y, Yang Z, Yao H, Liu T, et al. Novel role of the SIRT1 in endocrine and metabolic diseases. Int J Biol Sci. 2023;19:484-501
225. Luckey AE, Parsa CJ. Fluid and electrolytes in the aged. Arch Surg. 2003;138:1055-60
226. Lyulcheva-Bennett K, Williams S, Howse M, McCann E. Genomic testing in patients with renal disease. Br J Hosp Med. 2023;84(7):1-11
227. Ma Q. Role of Nrf2 in oxidative stress and toxicity. Annu Rev Pharmacol Toxicol. 2013;53:401-26
228. Maertens S, Van Den Noortgate NJ. Kidney in old age. Acta Clin Belg. 2008;63(1):8-15
229. Mancuso C, Santangelo R. Panax ginseng and Panax quinquefolius: From pharmacology to toxicology. Food Chem Toxicol. 2017;107(Pt A):362-72
230. Maruyama N, Ishigami A, Kondo Y. Pathophysiological significance of senescence marker protein-30. Geriatr Gerontol Int. 2010;10(Suppl 1):S88-98
231. Meng P, Zhu M, Ling X, Zhou L. Wnt signaling in kidney: the initiator or terminator? J Mol Med (Berl). 2020;98:1511-23
232. Meng T, Qin W, Liu B. SIRT1 Antagonizes Oxidative stress in diabetic vascular complication. Front Endocrinol (Lausanne). 2020;11:568861
233. Meng X, Tan J, Li M, Song S, Miao Y, Zhang Q. Sirt1: Role under the condition of ischemia/hypoxia. Cell Mol Neurobiol. 2017;37(1):17-28
234. Messerer DAC, Halbgebauer R, Nilsson B, Pavenstädt H, Radermacher P, Huber-Lang M. Immunopathophysiology of trauma-related acute kidney injury. Nat Rev Nephrol. 2021;17:91-111
235. Miao M, Xiang L. Pharmacological action and potential targets of chlorogenic acid. Adv Pharmacol. 2020;87:71-88
236. Miao X, Rong L, Fu B, Cui S, Gu Z, Hu F, et al. Astragalus polysaccharides attenuate rat aortic endothelial senescence via regulation of the SIRT-1/p53 signaling pathway. BMC Complement Med Ther. 2024;24(1):80
237. Mikhelson VM, Gamaley IA. Telomere shortening is a sole mechanism of aging in mammals. Curr Aging Sci. 2012;5:203-8
238. Minami S, Yamamoto T, Yamamoto-Imoto H, Isaka Y, Hamasaki M. Autophagy and kidney aging. Prog Biophys Mol Biol. 2023;179:10-5
239. Mir RH, Maqbool M, Mir PA, Hussain MS, Wani SUD, Pottoo FH, et al. Green synthesis of silver nanoparticles and their potential applications in mitigating cancer. Curr Pharm Des. 2024;30:2445-67
240. Mirshafa A, Shokati Sayyad M, Mohammadi E, Talebpour Amiri F, Shaki F. 5-HT3 antagonist, tropisetron, ameliorates age-related renal injury induced by D-galactose in male mice: Up-regulation of sirtuin 1. Iran J Basic Med Sci. 2024;27:577-87
241. Modi D, Hussain MS, Ainampudi S, Prajapati BG. Long acting injectables for the treatment of prostate cancer. J Drug Deliv Sci Technol. 2024;100:105996
242. Mohamad Kamal NS, Safuan S, Shamsuddin S, Foroozandeh P. Aging of the cells: Insight into cellular senescence and detection Methods. Eur J Cell Biol. 2020;99(6):151108
243. Montaigne D, Butruille L, Staels B. PPAR control of metabolism and cardiovascular functions. Nat Rev Cardiol. 2021;18:809-23
244. Morgan RG, Donato AJ, Walker AE. Telomere uncapping and vascular aging. Am J Physiol Heart Circ Physiol. 2018;315(1):H1-5
245. Morigi M, Perico L, Benigni A. Sirtuins in renal health and disease. J Am Soc Nephrol. 2018;29:1799-809
246. Moritz KM, Lim GB, Wintour EM. Developmental regulation of erythropoietin and erythropoiesis. Am J Physiol. 1997;273:R1829-44
247. Morrissey PE, Yango AF. Renal transplantation: older recipients and donors. Clin Geriatr Med. 2006;22:687-707
248. Musso CG, Belloso WH, Scibona P, Bellizzi V, Macías Núñez JF. Impact of renal aging on drug therapy. Postgrad Med. 2015;127:623-9
249. Musso CG, Giordani MC, Imperiali N. Aging kidney transplantation. Rev Invest Clin. 2016;68:68-74
250. Musso CG, Oreopoulos DG. Aging and physiological changes of the kidneys including changes in glomerular filtration rate. Nephron Physiol. 2011;119(Suppl 1):1-5
251. Naghibi N, Sadeghi A, Movahedinia S, Rahimi Naiini M, Rajizadeh MA, Bahri F, et al. Ellagic acid ameliorates aging-induced renal oxidative damage through upregulating SIRT1 and NRF2. BMC Complement Med Ther. 2023;23(1):77
252. Nakatani Y, Inagi R. Epigenetic regulation through SIRT1 in podocytes. Curr Hypertens Rev. 2016;12:89-94
253. Naveed M, Hejazi V, Abbas M, Kamboh AA, Khan GJ, Shumzaid M, et al. Chlorogenic acid (CGA): A pharmacological review and call for further research. Biomed Pharmacother. 2018;97:67-74
254. Nguyen T, Foster Y, Cekaj S. Older adult kidney function assessment and rounding creatinine led to medication dosing error. Am J Ther. 2018;25:e439-e46
255. Nguyen TT, Corvera S. Adipose tissue as a linchpin of organismal ageing. Nat Metab. 2024;6:793-807
256. Nguyen TV, Goldfarb DS. The older adult patient and kidney function. Consult Pharm. 2012;27:431-44
257. Ning YC, Cai GY, Zhuo L, Gao JJ, Dong D, Cui S, et al. Short-term calorie restriction protects against renal senescence of aged rats by increasing autophagic activity and reducing oxidative damage. Mech Ageing Dev. 2013;134:570-9
258. Niu Y, Na L, Feng R, Gong L, Zhao Y, Li Q, et al. The phytochemical, EGCG, extends lifespan by reducing liver and kidney function damage and improving age-associated inflammation and oxidative stress in healthy rats. Aging Cell. 2013;12:1041-9
259. Nousis L, Kanavaros P, Barbouti A. Oxidative stress-induced cellular senescence: is labile iron the connecting link? Antioxidants (Basel). 2023;12(6):1250
260. Obas V, Vasan RS. The aging heart. Clin Sci (Lond). 2018;132:1367-82
261. Ogrodnik M. Cellular aging beyond cellular senescence: Markers of senescence prior to cell cycle arrest in vitro and in vivo. Aging Cell. 2021;20(4):e13338
262. Ogura Y, Kitada M, Koya D. Sirtuins and renal oxidative stress. Antioxidants (Basel). 2021;10(8):1198
263. Opresko PL, Shay JW. Telomere-associated aging disorders. Ageing Res Rev. 2017;33:52-66
264. Panickar KS, Jewell DE. The beneficial role of anti-inflammatory dietary ingredients in attenuating markers of chronic low-grade inflammation in aging. Horm Mol Biol Clin Investig. 2015;23(2):59-70
265. Pannarale G, Carbone R, Del Mastro G, Gallo C, Gattullo V, Natalicchio L, et al. The aging kidney: structural changes. J Nephrol. 2010;23(Suppl 15):S37-40
266. Passmore JC, Joshua IG, Rowell PP, Tyagi SC, Falcone JC. Reduced alpha adrenergic mediated contraction of renal preglomerular blood vessels as a function of gender and aging. J Cell Biochem. 2005;96:672-81
267. Patra S, Praharaj PP, Singh A, Bhutia SK. Targeting SIRT1-regulated autophagic cell death as a novel therapeutic avenue for cancer prevention. Drug Discov Today. 2023;28(9):103692
268. Peana M, Pelucelli A, Chasapis CT, Perlepes SP, Bekiari V, Medici S, et al. Biological effects of human exposure to environmental cadmium. Biomolecules. 2022;13(1):36
269. Pereira-Simon S, Rubio GA, Xia X, Cai W, Choi R, Striker GE, et al. Inhibition of advanced glycation end products (AGEs) accumulation by pyridoxamine modulates glomerular and mesangial cell estrogen receptor α expression in aged female mice. PLoS One. 2016;11(7):e0159666
270. Perico L, Remuzzi G, Benigni A. Sirtuins in kidney health and disease. Nat Rev Nephrol. 2024;20:313-29
271. Pingitore A, Lima GP, Mastorci F, Quinones A, Iervasi G, Vassalle C. Exercise and oxidative stress: potential effects of antioxidant dietary strategies in sports. Nutrition. 2015;31:916-22
272. Pintó-Marijuan M, Munné-Bosch S. Photo-oxidative stress markers as a measure of abiotic stress-induced leaf senescence: advantages and limitations. J Exp Bot. 2014;65:3845-57
273. Poston JT, Koyner JL. Sepsis associated acute kidney injury. BMJ. 2019;364:k4891
274. Poulose N, Raju R. Aging and injury: alterations in cellular energetics and organ function. Aging Dis. 2014;5:101-8
275. Prasad KN, Wu M, Bondy SC. Telomere shortening during aging: Attenuation by antioxidants and anti-inflammatory agents. Mech Ageing Dev. 2017;164:61-6
276. Presta P, Lucisano G, Fuiano L, Fuiano G. The kidney and the elderly: why does the risk increase? Int Urol Nephrol. 2012;44:625-32
277. Pushpakumar S, Ren L, Juin SK, Majumder S, Kulkarni R, Sen U. Methylation-dependent antioxidant-redox imbalance regulates hypertensive kidney injury in aging. Redox Biol. 2020;37:101754
278. Pyo IS, Yun S, Yoon YE, Choi JW, Lee SJ. Mechanisms of aging and the preventive effects of resveratrol on age-related diseases. Molecules. 2020;25(20):4649
279. Qi W, Hu C, Zhao D, Li X. SIRT1-SIRT7 in diabetic kidney disease: biological functions and molecular mechanisms. Front Endocrinol (Lausanne). 2022;13:801303
280. Qiu H, Chen X, Luo Z, Zhao L, Zhang T, Yang N, et al. Inhibition of endogenous hydrogen sulfide production exacerbates the inflammatory response during urine‑derived sepsis‑induced kidney injury. Exp Therap Med. 2018;16:2851-8
281. Qiu Y, Zhou X, Liu Y, Tan S, Li Y. The role of sirtuin-1 in immune response and systemic lupus erythematosus. Front Immunol. 2021;12:632383
282. Radi ZA. Kidney pathophysiology, toxicology, and drug-induced injury in drug development. Int J Toxicol. 2019;38:215-27
283. Rellmann Y, Eidhof E, Dreier R. Review: ER stress-induced cell death in osteoarthritic cartilage. Cell Signal. 2021;78:109880
284. Rivankar S. An overview of doxorubicin formulations in cancer therapy. J Cancer Res Ther. 2014;10:853-8
285. Rodríguez-Castro EM, Córdova HR. Aging and the kidney. Bol Asoc Med P R. 2011;103(3):57-62
286. Rohilla S, Singh M, Priya S, Almalki WH, Haniffa SM, Subramaniyan V, et al. Exploring the mechanical perspective of a new anti-tumor agent: melatonin. J Environ Pathol Toxicol Oncol. 2023;42(1):1-16
287. Rossi A, Di Maio M. Platinum-based chemotherapy in advanced non-small-cell lung cancer: optimal number of treatment cycles. Expert Rev Anticancer Ther. 2016;16:653-60
288. Rossiello F, Jurk D, Passos JF, d'Adda di Fagagna F. Telomere dysfunction in ageing and age-related diseases. Nat Cell Biol. 2022;24:135-47
289. Rubattu S, Bianchi F, Busceti CL, Cotugno M, Stanzione R, Marchitti S, et al. Differential modulation of AMPK/PPARα/UCP2 axis in relation to hypertension and aging in the brain, kidneys and heart of two closely related spontaneously hypertensive rat strains. Oncotarget. 2015;6:18800-18
290. Saldanha JF, Leal Vde O, Stenvinkel P, Carraro-Eduardo JC, Mafra D. Resveratrol: why is it a promising therapy for chronic kidney disease patients? Oxid Med Cell Longev. 2013;2013:963217
291. Salminen A, Kaarniranta K. AMP-activated protein kinase (AMPK) controls the aging process via an integrated signaling network. Ageing Res Rev. 2012;11:230-41
292. Sands JM. Urine concentrating and diluting ability during aging. J Gerontol A Biol Sci Med Sci. 2012;67:1352-7
293. Satarug S, Garrett SH, Sens MA, Sens DA. Cadmium, environmental exposure, and health outcomes. Environ Health Perspect. 2010;118:182-90
294. Sato Y, Yanagita M. Immune cells and inflammation in AKI to CKD progression. Am J Physiol Renal Physiol. 2018;315:F1501-12
295. Sato Y, Yanagita M. Immunology of the ageing kidney. Nat Rev Nephrol. 2019;15:625-40
296. Satturwar S, Parwani AV. Cytomorphology of papillary renal neoplasm with reverse polarity. Cytojournal. 2023;20:43
297. Schmitt R, Susnik N, Melk A. Molecular aspects of renal senescence. Curr Opin Organ Transplant. 2015;20:412-6
298. Schottlender N, Gottfried I, Ashery U. Hyperbaric oxygen treatment: effects on mitochondrial function and oxidative stress. Biomolecules. 2021;11(12):1827
299. Shakeri A, Zirak MR, Sahebkar A. Ellagic acid: a logical lead for drug development? Curr Pharm Des. 2018;24:106-22
300. Shankland SJ, Wang Y, Shaw AS, Vaughan JC, Pippin JW, Wessely O. Podocyte aging: why and how getting old matters. J Am Soc Nephrol. 2021;32:2697-713
301. Shen P, Deng X, Chen Z, Ba X, Qin K, Huang Y, et al. SIRT1: A potential therapeutic target in autoimmune diseases. Front Immunol. 2021;12:779177
302. Shen Q-q, Jv X-h, Ma X-z, Li C, Liu L, Jia W-t, et al. Cell senescence induced by toxic interaction between α-synuclein and iron precedes nigral dopaminergic neuron loss in a mouse model of Parkinson’s disease. Acta Pharmacol Sin. 2024;45:268-81
303. Shen S, Shen M, Kuang L, Yang K, Wu S, Liu X, et al. SIRT1/SREBPs-mediated regulation of lipid metabolism. Pharmacol Res. 2024;199:107037
304. Shi S, Chen Y, Luo Z, Nie G, Dai Y. Role of oxidative stress and inflammation-related signaling pathways in doxorubicin-induced cardiomyopathy. Cell Commun Signal. 2023;21(1):61
305. Shiels PG, McGuinness D, Eriksson M, Kooman JP, Stenvinkel P. The role of epigenetics in renal ageing. Nat Rev Nephrol. 2017;13:471-82
306. Shu B, Shi Q, Wang YJ. Shen (Kidney)-tonifying principle for primary osteoporosis: to treat both the disease and the Chinese medicine syndrome. Chin J Integr Med. 2015;21:656-61
307. Sies H. Oxidative stress: oxidants and antioxidants. Exp Physiol. 1997;82:291-5
308. Singh G, Krishan P. Dietary restriction regimens for fighting kidney disease: Insights from rodent studies. Exp Gerontol. 2019;128:110738
309. Singh V, Ubaid S. Role of silent information regulator 1 (SIRT1) in regulating oxidative stress and inflammation. Inflammation. 2020;43:1589-98
310. Smykiewicz P, Segiet A, Keag M, Żera T. Proinflammatory cytokines and ageing of the cardiovascular-renal system. Mech Ageing Dev. 2018;175:35-45
311. Sobamowo H, Prabhakar SS. The kidney in aging: physiological changes and pathological implications. Prog Mol Biol Transl Sci. 2017;146:303-40
312. Speeckaert MM, Vanfraechem C, Speeckaert R, Delanghe JR. Peroxisome proliferator-activated receptor agonists in a battle against the aging kidney. Ageing Res Rev. 2014;14:1-18
313. Stocker R. Antioxidant defenses in human blood plasma and extra-cellular fluids. Arch Biochem Biophys. 2016;595:136-9
314. Sturmlechner I, Durik M, Sieben CJ, Baker DJ, van Deursen JM. Cellular senescence in renal ageing and disease. Nat Rev Nephrol. 2017;13(2):77-89
315. Suntar I, Sureda A, Belwal T, Sanches Silva A, Vacca RA, Tewari D, et al. Natural products, PGC-1 α, and Duchenne muscular dystrophy. Acta Pharm Sin B. 2020;10:734-45
316. Tang BL. Sirt1 and the mitochondria. Mol Cells. 2016;39(2):87-95
317. Tang C, Livingston MJ, Liu Z, Dong Z. Autophagy in kidney homeostasis and disease. Nat Rev Nephrol. 2020;16:489-508
318. Tang G, Li S, Zhang C, Chen H, Wang N, Feng Y. Clinical efficacies, underlying mechanisms and molecular targets of Chinese medicines for diabetic nephropathy treatment and management. Acta Pharm Sin B. 2021;11:2749-67
319. Tanriover C, Copur S, Mutlu A, Peltek IB, Galassi A, Ciceri P, et al. Early aging and premature vascular aging in chronic kidney disease. Clin Kidney J. 2023;16:1751-65
320. Tao Z, Jin Z, Wu J, Cai G, Yu X. Sirtuin family in autoimmune diseases. Front Immunol. 2023;14:1186231
321. Terao R, Ahmed T, Suzumura A, Terasaki H. Oxidative stress-induced cellular senescence in aging retina and age-related macular degeneration. Antioxidants (Basel). 2022;11(11):2189
322. Thapa R, Afzal M, Goyal A, Gupta G, Bhat AA, Almalki WH, et al. Exploring ncRNA-mediated regulation of EGFR signalling in glioblastoma: From mechanisms to therapeutics. Life Sci. 2024;345:122613
323. Thapa R, Afzal O, Afzal M, Gupta G, Bhat AA, Hassan Almalki W, et al. From LncRNA to metastasis: The MALAT1-EMT axis in cancer progression. Pathol Res Pract. 2024;253:154959
324. Thapa R, Afzal O, Altamimi ASA, Goyal A, Almalki WH, Alzarea SI, et al. Galangin as an inflammatory response modulator: An updated overview and therapeutic potential. Chem Biol Interact. 2023;378:110482
325. Thapa R, Afzal O, Bhat AA, Goyal A, Alfawaz Altamimi AS, Almalki WH, et al. New horizons in lung cancer management through ATR/CHK1 pathway modulation. Future Med Chem. 2023;15:1807-18
326. Thapa R, Ali H, Afzal O, Bhat AA, Almalki WH, Alzarea SI, et al. Unlocking the potential of mesoporous silica nanoparticles in breast cancer treatment. J Nanopart Res. 2023;25(8):169
327. Thapa R, Gupta G, Bhat AA, Almalki WH, Alzarea SI, Kazmi I, et al. A review of Glycogen Synthase Kinase-3 (GSK3) inhibitors for cancers therapies. Int J Biol Macromol. 2023;253(Pt 7):127375
328. Thapa R, Moglad E, Afzal M, Gupta G, Bhat AA, Kazmi I, et al. ncRNAs and their impact on dopaminergic neurons: autophagy pathways in Parkinson's disease. Ageing Res Rev. 2024;98:102327
329. Thirupathi A, de Souza CT. Multi-regulatory network of ROS: the interconnection of ROS, PGC-1 alpha, and AMPK-SIRT1 during exercise. J Physiol Biochem. 2017;73:487-94
330. Tousian H, Razavi BM, Hosseinzadeh H. Effects of alpha-mangostin on memory senescence induced by high glucose in human umbilical vein endothelial cells. Iran J Basic Med Sci. 2020;23:1261-7
331. Trefts E, Shaw RJ. AMPK: restoring metabolic homeostasis over space and time. Mol Cell. 2021;81:3677-90
332. Tu Y, Sun W, Chen DP, Wan YG, Wu W, Yao J. [Molecular mechanisms of autophagy in regulating renal aging and interventional effects of Chinese herbal medicine]. Zhongguo Zhong Yao Za Zhi. 2016;41:3914-8
333. Tufail M, Wu C, Hussain MS. Dietary, addictive and habitual factors, and risk of colorectal cancer. Nutrition. 2024;120:112334
334. Turner KJ, Vasu V, Griffin DK. Telomere biology and human phenotype. Cells. 2019;8(1):73
335. Uddin MJ, Farjana M, Moni A, Hossain KS, Hannan MA, Ha H. Prospective pharmacological potential of resveratrol in delaying kidney aging. Int J Mol Sci. 2021;22(15):8258
336. Ugusman A, Kumar J, Aminuddin A. Endothelial function and dysfunction: Impact of sodium-glucose cotransporter 2 inhibitors. Pharmacol Ther. 2021;224:107832
337. Uneda K, Wakui H, Maeda A, Azushima K, Kobayashi R, Haku S, et al. Angiotensin II Type 1 receptor-associated protein regulates kidney aging and lifespan independent of angiotensin. J Am Heart Assoc. 2017;6(8):e006120
338. Ungar A, Castellani S, Di Serio C, Cantini C, Cristofari C, Vallotti B, et al. Changes in renal autacoids and hemodynamics associated with aging and isolated systolic hypertension. Prostaglandins Other Lipid Mediat. 2000;62:117-33
339. Ungar A, Cristofari C, Torrini M, Di Serio C, Cantini C, Vallotti B, et al. Changes in renal autacoids in aged human hypertensives. J Physiol Pharmacol. 2000;51 (4 Pt 1):619-30
340. van den Anker J, Reed MD, Allegaert K, Kearns GL. Developmental changes in pharmacokinetics and pharmacodynamics. J Clin Pharmacol. 2018;58(Suppl 10): S10-25
341. van der Pol A, van Gilst WH, Voors AA, van der Meer P. Treating oxidative stress in heart failure: past, present and future. Eur J Heart Fail. 2019;21:425-35
342. Vanaporn M, Titball RW. Trehalose and bacterial virulence. Virulence. 2020;11(1):1192-202
343. Verbalis JG. Renal physiology of nocturia. Neurourol Urodyn. 2014;33(Suppl 1):S6-9
344. Vernier RL, Mauer SM, Fish AJ, Michael AF. The mesangial cell in glomerulonephritis. Adv Nephrol Necker Hosp. 1971;1:31-46
345. Vlassara H, Torreggiani M, Post JB, Zheng F, Uribarri J, Striker GE. Role of oxidants/inflammation in declining renal function in chronic kidney disease and normal aging. Kidney Int Suppl. 2009;114:S3-11
346. Vlassara H, Uribarri J, Cai W, Goodman S, Pyzik R, Post J, et al. Effects of sevelamer on HbA1c, inflammation, and advanced glycation end products in diabetic kidney disease. Clin J Am Soc Nephrol. 2012;7:934-42
347. Voziyan PA, Hudson BG. Pyridoxamine as a multifunctional pharmaceutical: targeting pathogenic glycation and oxidative damage. Cell Mol Life Sci. 2005;62:1671-81
348. Voziyan PA, Hudson BG. Pyridoxamine: the many virtues of a maillard reaction inhibitor. Ann N Y Acad Sci. 2005;1043:807-16
349. Wakino S, Itoh H. [Aging and kidney disease]. Nihon Rinsho. 2016;74:1531-40
350. Wang F, Nguyen M, Qin FX, Tong Q. SIRT2 deacetylates FOXO3a in response to oxidative stress and caloric restriction. Aging Cell. 2007;6:505-14
351. Wang H, Guan Y, Karamercan MA, Ye L, Bhatti T, Becker LB, et al. Resveratrol rescues kidney mitochondrial function following hemorrhagic shock. Shock. 2015;44:173-80
352. Wang L, Xu C, Johansen T, Berger SL, Dou Z. SIRT1 - a new mammalian substrate of nuclear autophagy. Autophagy. 2021;17:593-5
353. Wang S, Moustaid-Moussa N, Chen L, Mo H, Shastri A, Su R, et al. Novel insights of dietary polyphenols and obesity. J Nutr Biochem. 2014;25(1):1-18
354. Wang SS, Zhang X, Ke ZZ, Wen XY, Li WD, Liu WB, et al. D-galactose-induced cardiac ageing: A review of model establishment and potential interventions. J Cell Mol Med. 2022;26:5335-59
355. Wang SY, Cai GY, Chen XM. Energy restriction in renal protection. Br J Nutr. 2018;120:1149-58
356. Wang W, Li J, Cai L. Research progress of sirtuins in renal and cardiovascular diseases. Curr Opin Nephrol Hypertens. 2021;30(1):108-14
357. Wang X, Bonventre JV, Parrish AR. The aging kidney: increased susceptibility to nephrotoxicity. Int J Mol Sci. 2014;15:15358-76
358. Wang Y, Wang Y, Yang M, Ma X. Implication of cellular senescence in the progression of chronic kidney disease and the treatment potencies. Biomed Pharmacother. 2021;135:111191
359. Wei SY, Pan SY, Li B, Chen YM, Lin SL. Rejuvenation: Turning back the clock of aging kidney. J Formos Med Assoc. 2020;119:898-906
360. Weide T, Huber TB. Implications of autophagy for glomerular aging and disease. Cell Tissue Res. 2011;343:467-73
361. Wen Y, Wang Y, Zhao C, Zhao B, Wang J. The pharmacological efficacy of baicalin in inflammatory diseases. Int J Mol Sci. 2023;24(11):9317
362. Wicik Z, Nowak A, Jarosz-Popek J, Wolska M, Eyileten C, Siller-Matula JM, et al. Characterization of the SGLT2 interaction network and its regulation by SGLT2 inhibitors: a bioinformatic analysis. Front Pharmacol. 2022;13:901340
363. Wiggins J. Podocytes and glomerular function with aging. Semin Nephrol. 2009;29:587-93
364. Wiggins J. Why do our kidneys get old? Nephron Exp Nephrol. 2011;119(Suppl 1):e1-5
365. Winiarska A, Knysak M, Nabrdalik K, Gumprecht J, Stompór T. Inflammation and oxidative stress in diabetic kidney disease: the targets for SGLT2 inhibitors and GLP-1 receptor agonists. Int J Mol Sci. 2021;22(19):10822
366. Wu BB, Leung KT, Poon EN. Mitochondrial-targeted therapy for doxorubicin-induced cardiotoxicity. Int J Mol Sci. 2022;23(3):1912
367. Wu QJ, Zhang TN, Chen HH, Yu XF, Lv JL, Liu YY, et al. The sirtuin family in health and disease. Signal Transduct Target Ther. 2022;7(1):402
368. Xian C, Chen H, Xiong F, Fang Y, Huang H, Wu J. Platinum-based chemotherapy via nanocarriers and co-delivery of multiple drugs. Biomater Sci. 2021;9:6023-36
369. Xiang C, Yan Y, Zhang D. Alleviation of the doxorubicin-induced nephrotoxicity by fasudil in vivo and in vitro. J Pharmacol Sci. 2021;145(1):6-15
370. Xu F, Xu J, Xiong X, Deng Y. Salidroside inhibits MAPK, NF-κB, and STAT3 pathways in psoriasis-associated oxidative stress via SIRT1 activation. Redox Rep. 2019;24(1):70-4
371. Yacoub R, Lee K, He JC. The role of SIRT1 in diabetic kidney disease. Front Endocrinol (Lausanne). 2014;5:166
372. Yan J, Wang J, He JC, Zhong Y. Sirtuin 1 in chronic kidney disease and therapeutic potential of targeting sirtuin 1. Front Endocrinol (Lausanne). 2022;13:917773
373. Yang Y, Liu Y, Wang Y, Chao Y, Zhang J, Jia Y, et al. Regulation of SIRT1 and its roles in inflammation. Front Immunol. 2022;13:831168
374. Yang Y, Zhang L. A narrative review of tropisetron and palonosetron for the control of chemotherapy-induced nausea and vomiting. Chin Clin Oncol. 2020;9 (2):17
375. Yang YY, Shi LX, Li JH, Yao LY, Xiang DX. Piperazine ferulate ameliorates the development of diabetic nephropathy by regulating endothelial nitric oxide synthase. Mol Med Rep. 2019;19:2245-53
376. Yao H, Rahman I. Perspectives on translational and therapeutic aspects of SIRT1 in inflammaging and senescence. Biochem Pharmacol. 2012;84:1332-9
377. Yi J, Luo J. SIRT1 and p53, effect on cancer, senescence and beyond. Biochim Biophys Acta. 2010;1804:1684-9
378. Yin S, Zhou Z, Fu P, Jin C, Wu P, Ji C, et al. Roles of extracellular vesicles in ageing-related chronic kidney disease: Demon or angel. Pharmacol Res. 2023;193:106795
379. Yoshino J, Baur JA, Imai SI. NAD(+) intermediates: the biology and therapeutic potential of NMN and NR. Cell Metab. 2018;27:513-28
380. You Y, Liang W. SIRT1 and SIRT6: The role in aging-related diseases. Biochim Biophys Acta Mol Basis Dis. 2023;1869(7):166815
381. Yu Q, Dong L, Li Y, Liu G. SIRT1 and HIF1α signaling in metabolism and immune responses. Cancer Lett. 2018;418:20-6
382. Yu Y, Song X, Wang X, Zheng L, Ma G, Liu W, et al. Oxidative stress impairs the Nur77-Sirt1 axis resulting in a decline in organism homeostasis during aging. Aging Cell. 2023;22(5):e13812
383. Yubero-Serrano EM, Woodward M, Poretsky L, Vlassara H, Striker GE. Effects of sevelamer carbonate on advanced glycation end products and antioxidant/pro-oxidant status in patients with diabetic kidney disease. Clin J Am Soc Nephrol. 2015;10:759-66
384. Yun Z, Zou Z, Sun S, Che H. Chlorogenic acid improves food allergy through the AMPK/ACC/CPT-1 pathway. J Food Biochem. 2022;46(12):e14505
385. Zbroch E, Bazyluk A, Malyszko J, Koc-Zorawska E, Rydzewska-Rosolowska A, Kakareko K, et al. The serum concentration of anti-aging proteins, sirtuin1 and αklotho in patients with end-stage kidney disease on maintenance hemodialysis. Clin Interv Aging. 2020;15:387-93
386. Zeng J, Lu J. Mechanisms of action involved in ozone-therapy in skin diseases. Int Immunopharmacol. 2018;56:235-41
387. Zhang JQ, Li YY, Zhang XY, Tian ZH, Liu C, Wang ST, et al. Cellular senescence of renal tubular epithelial cells in renal fibrosis. Front Endocrinol (Lausanne). 2023;14:1085605
388. Zhang N, Li Z, Xu K, Wang Y, Wang Z. Resveratrol protects against high-fat diet induced renal pathological damage and cell senescence by activating SIRT1. Biol Pharm Bull. 2016;39:1448-54
389. Zhang W, Huang Q, Zeng Z, Wu J, Zhang Y, Chen Z. Sirt1 inhibits oxidative stress in vascular endothelial cells. Oxid Med Cell Longev. 2017;2017:7543973
390. Zhang XK. Targeting Nur77 translocation. Expert Opin Ther Targets. 2007;11(1):69-79
391. Zhang Z, Zhang L, Xu H. Effect of Astragalus polysaccharide in treatment of diabetes mellitus: a narrative review. J Tradit Chin Med. 2019;39(1):133-8
392. Zheng J, Yue R, Yang R, Wu Q, Wu Y, Huang M, et al. Visualization of zika virus infection via a light-initiated bio-orthogonal cycloaddition labeling strategy. Front Bioeng Biotechnol. 2022;10:940511
393. Zhou DD, Luo M, Huang SY, Saimaiti A, Shang A, Gan RY, et al. Effects and mechanisms of resveratrol on aging and age-related diseases. Oxid Med Cell Longev. 2021;2021:9932218
394. Zhou XJ, Rakheja D, Yu X, Saxena R, Vaziri ND, Silva FG. The aging kidney. Kidney Int. 2008;74:710-20
395. Zhu H, Yan Y, Jiang Y, Meng X. Ellagic acid and its anti-aging effects on central nervous system. Int J Mol Sci. 2022;23(18):10937
396. Zhu J, Pan S, Chai H, Zhao P, Feng Y, Cheng Z, et al. Microfluidic impedance cytometry enabled one‐step sample preparation for efficient single‐cell mass spectrometry. Small. 2024;20(26):2310700
397. Zhu R, Chen B, Bai Y, Miao T, Rui L, Zhang H, et al. Lycopene in protection against obesity and diabetes: A mechanistic review. Pharmacol Res. 2020;159:104966
398. Zhu Y, Liu X, Ding X, Wang F, Geng X. Telomere and its role in the aging pathways: telomere shortening, cell senescence and mitochondria dysfunction. Biogerontology. 2019;20(1):1-16
 
 

Figure 1: Graphical abstract

Figure 2: The image illustrates the process of kidney aging, highlighting key factors such as telomere aging, inflammation and oxidative stress, mitochondrial dysfunction, ER stress, and fibrosis. Each factor contributes to the overall decline in kidney function associated with aging.

Figure 3: The image illustrates the pathway of cell apoptosis in aging and aging-associated kidney disorders, involving DNA damage and PARP activation, leading to NAD+ and ATP depletion. SIRT deacetylates P53, regulating apoptotic mediators, while DNA repair mechanisms attempt to restore cell health, preventing apoptosis.

Figure 4: The image illustrates the protective role of SIRT1 in preventing kidney injury from severe burns. Melatonin activates SIRT1, which deacetylates p53 and p65, reducing apoptosis and inflammation. SIRT1 also enhances FOXO1 activity, mitigating oxidative stress and kidney damage.

 

Table 1: This table summarizes various studies on the role of SIRT1 in renal health, highlighting the models used, mechanisms involved, effects on oxidative stress and SIRT1 activation, and potential therapeutic implications. It provides a concise overview of how targeting SIRT1-related pathways may mitigate kidney aging and disease

Table 2: This table summarizes various compounds and agents studied for their effects on renal aging, detailing their mechanisms, key findings, and therapeutic implications. Each entry includes SIRT1 modulation and oxidative stress reduction.

[*] Corresponding Author:

Waleed Hassan Almalki, Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia, eMail: whmalki21@gmail.com or Whmalki@uqu.edu.sa