ABSTRACT
Klotho, a multifunctional protein, acts as a co-receptor in fibroblast growth factor 23 and exerts its impact through various molecular pathways, including Wnt, hypoxia-inducible factor and insulin-like growth factor 1 pathways. The physiological significance of Klotho is the regulation of vitamin D and phosphate metabolism as well as serving as a vital component in aging and neurodegeneration. The role of Klotho in aging and neurodegeneration in particular has gained considerable attention. In this narrative review we highlight several key insights into the molecular basis and physiological function of Klotho and synthesize current research on the role of Klotho in neurodegeneration and aging. Klotho deficiency was associated with cognitive impairment, reduced growth, diminished longevity and the development of age-related diseases in vivo. Serum Klotho levels showed a decline in individuals with advanced age and those affected by chronic kidney disease, establishing its potential diagnostic significance. Additionally, multiple medications have been demonstrated to influence Klotho levels. Therefore, this comprehensive review suggests that Klotho could open the door to novel interventions aimed at addressing the challenges of aging and neurodegenerative disorders.
Keywords: aging, chronic kidney disease, Klotho, neurodegeneration
INTRODUCTION
Aging and neurodegenerative disorders are complex medical conditions with poorly understood underlying pathophysiology that may affect virtually all tissues and organs. A limited number of genes and their transcripts have been associated with either premature aging, as seen in progeria, or with longevity [1, 2]. The α-Klotho protein, first identified in mice studies in 1997 [3], primarily functions as a co-receptor for fibroblast growth factor 23 (FGF23) in the kidneys and parathyroid gland and therefore has a crucial role in phosphate homeostasis, vitamin D metabolism and vascular calcification [4]. In addition, α-Klotho has been identified in a wide variety of tissues, consistent with its role in the aging process, including endocrine organs, arteries and reproductive, epithelial and neuronal tissue [5, 6]. β-Klotho is primarily expressed in the liver, adipose tissue and kidneys and plays a role in lipid and energy metabolism by acting as co-receptor for FGF15 and FGF21 [7, 8], while the γ-Klotho isoform is expressed in the brown adipose tissue, skin and kidneys with as yet poorly defined roles and by acting as co-receptor for FGF receptor 1b (FGFR1b), FGFR1c, FGFR2c and FGFR4 [4, 9, 10] and are not discussed in the current review. Nevertheless, preclinical studies conducted on Klotho knockout mice have revealed that Klotho deficiency is associated with impaired cognition, shorter lifespan, cardiac hypertrophy, vascular calcification, multi-organ atrophy and fibrosis and growth retardation [3, 11]. Moreover, overexpression of the Klotho gene has been shown to lengthen lifespan in mice [12], which raises the potential question of whether Klotho may be utilized as a target to control or reverse aging and/or neurodegeneration. One drawback of such models is the lack of distinction between soluble and transmembrane Klotho molecules, which may potentially have distinct physiological roles in the human body. Whether such physiological and pathological outcomes of Klotho deficiency and/or overexpression may simply be attributable to the role of Klotho on phosphate metabolism is unclear and should be evaluated with caution with the discovery of multiple phosphate metabolism–independent functions of Klotho protein. In this narrative review, our aim is to evaluate the potential pathophysiological and therapeutic role of Klotho protein in aging and neurodegenerative conditions.
The molecular basis
Klotho is a single-pass 130-kD transmembrane protein consisting of a short cytoplasmic tail, a transmembrane domain and two extracellular domains referred to as Kl1 and Kl2 and primarily expressed in kidneys, parathyroid glands and choroid plexus [13]. Membrane-bound Klotho functions as a co-receptor for FGF23 and FGFRs, while cleavage of the extracellular domains of Klotho protein leads to the formation of soluble Klotho [14]. The extracellular domain of Klotho is released into the blood circulation through cleavage by a disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) and ADAM17, which is stimulated by insulin and inhibited by metalloproteinase inhibitors [15]. It has been demonstrated that the kidney has a dual role in Klotho homeostasis, as it has been shown to produce and release Klotho into the circulation while also clearing Klotho from the blood into the urinary lumen [13]. Soluble Klotho is the primary form of Klotho present in urine via tubular transcytosis, not glomerular filtration [13], and acts as a paracrine and endocrine signal in many organ systems [16].
Klotho gene
The Klotho gene is located at chromosome 5 in humans between the PDS5B and STARD13 genes, which is homologous to the rat and mouse genomes [17]. The Klotho gene includes five exons and four introns [18]. The transcription of Klotho is downregulated in response to pro-inflammatory conditions, angiotensin II, nuclear factor κ light chain enhancer of activated B cells (NF-κB) signalling, matrix metalloproteinases and certain conditions such as dehydrated states and diabetic nephropathy [19–21]. On the other hand, vitamin D, the peroxisome proliferator-activated receptor γ (PPAR-γ) signalling pathway and multiple medications such as losartan, statins, fosinopril, erythropoietin and rapamycin upregulate Klotho gene expression [17, 22–24].
Physiological functions
The Klotho protein has multiple physiological functions in various organs systems through multiple signalling pathways.
Phosphate homeostasis
FGF23, a bone-derived hormone, is a phosphaturic hormone with gain-of-function mutations leading to resistance towards degradation by PHEX (phosphate regulating endopeptidase homolog, X-linked), leading to autosomal dominant hypophosphataemic rickets. FGF23 has weak affinity towards FGFRs in proximal tubules, while Klotho protein is crucial in the maintenance of such a link. The N-terminal region of FGF23 binds to FGFRs and the C-terminal region interacts with Klotho protein, while completion of such a complex leads to inhibition of phosphate reabsorption through proximal tubules due to downregulation of NaPi-2a and NaPi-2c and inhibition of active vitamin D generation due to downregulation of enzyme 1-alpha hydroxylase [25, 26]. Other physiological actions of FGF23 on phosphate metabolism include the upregulation of enzyme 24-hydroxylase, which is involved in the inactivation of active vitamin D and attenuation of parathyroid hormone release [27, 28].
Wnt signalling pathway
The Wnt/β-catenin signalling pathway is involved in cell cycle arrest at the G2/M phase and upregulation of pro-fibrotic cytokines and molecules [29]. Also, the Wnt/β-catenin signalling pathway is a universal pathway involved in embryogenesis via axis development, cell proliferation and cell migration [29]. Klotho has been shown to inhibit the Wnt signalling pathway via binding to multiple natural Wnt ligands [30]. Klotho is also involved in the regulation of intracellular calcium homeostasis [31], which may lead to inducement of μ-calpain, a calcium-dependent protease, thus leading to degradation of β-catenin [32]. Moreover, a decrease in the levels of Klotho in chronic kidney disease (CKD) patients may contribute to the development of kidney injury and fibrosis due to loss of suppression on the Wnt/β-catenin signalling pathway [32]. Additionally, augmented Wnt signalling has been linked to stem and progenitor cell dysfunction and cellular senescence. Therefore, Klotho-mediated downregulation of the Wnt signalling pathway may reverse such a condition and improve tissue regeneration [30].
Insulin-like growth factor 1 (IGF-1) signalling pathway
Multiple studies have demonstrated that Klotho suppresses the downstream signalling pathways of IGF or insulin receptor substrate (IRS) without direct interaction [33, 34]. One possible explanation is through the interactions over forkhead box proteins (FOXOs) in which activation of the IGF-1 signalling pathway results in upregulation of the PI3K/Akt pathway and phosphorylation of FOXO1/3a/4 proteins. Such phosphorylation prevents the nuclear translocation of FOXOs and thereby interferes with their transcription activity, including glucose-6-phosphatase activity involved in gluconeogenesis in the liver [35]. Also, FOXO1 is involved in the downregulation of adipogenesis via binding to the promoter region of PPARγ while Klotho protein leads to contradictory action via binding to the same region [36]. Another anti-aging property of Klotho protein is the inhibition of the IGF-1 signalling-mediated increase in reactive oxygen species, leading to downregulation of oxidative stress [34].
Hypoxia-inducible factor (HIF) pathway
HIF [37] is a protein complex essential for controlling vascularization and homeostasis. It promotes vascularization in hypoxic areas and acts as a transcription factor for many target genes in response to low oxygen levels [38]. It has been indicated that the transcription level of HIF1α protein is negatively regulated by Klotho [39]. Moreover, exogenous supplementation of Klotho can reduce the HIF level [40]. Klotho deficiency leads to an increase in erythropoietin (EPO) production through HIF activation. However, this increase in EPO production does not lead to an improvement in haematopoiesis because it is observed that haematopoietic stem cells in bone marrow of klotho-deficient mice are decreased [41, 42]. Shedding of soluble Klotho by the kidneys is induced by hypoxia [43], thus indicating a negative feedback loop as part of the complex interaction between tissue oxygenation, HIF1α, EPO, FGF23 and Klotho.
Enzymatic role
The two internal repeats of the extracellular domain of Klotho protein, namely KL1 and KL2, have structural and amino acid sequence homology to family 1 glycosidase enzymes that hydrolyse β-glycosidic bonds in glycoproteins and glycolipids [44]. Nevertheless, such β-glycosidic activity has not been detected in in vitro studies [45]. On the other hand, Klotho protein exerts β-glucuronidase and sialidase activity in in vitro models [31]. Such activity is involved in the regulation of transcellular transport of phosphate in renal proximal tubules via proteolytic degradation of NaPi-2a transporter via β-glucuronidase activity of Klotho, leading to phosphaturia [44, 46]. Nevertheless, there is a clear need for future studies for a better understanding of the enzymatic role of Klotho protein in human physiology.
Others
α-Klotho and soluble Klotho proteins are involved in multiple other physiological events, including the upregulation of antioxidant defence mechanisms, synthesis of proteins involved in the cell cycle such as p15 and p21, regulation of senescence and apoptosis via p53 or transforming growth factor β (TGF-β) signalling pathway [47, 48].
Klotho as an anti-aging target
The serum levels of Klotho have been shown to decrease with certain diseases, including CKD, neurodegenerative conditions, diabetes mellitus and aging. A recent cross-sectional cohort study including 346 healthy adult participants 18–85 years of age demonstrated that senior adults (ages 55–85 years) have the lowest serum α-Klotho levels (P < .01) and serum Klotho levels have negative correlation with age (P < .001) [49]. Moreover, Klotho-deficient mice have illustrated higher rates of vascular calcification, cardiac hypertrophy, cognitive impairment, hypercalcaemia, hyperphosphataemia, multi-organ atrophy and osteopenia [3, 50]. A national cohort study in the USA involving 10 069 adult participants ages 40–79 years with serum Klotho levels measured via enzyme-linked immunosorbent assay revealed that participants with low serum levels of Klotho (<666 pg/ml) have 31% higher risk of all-cause mortality compared with participants with higher serum levels of Klotho (>985 pg/ml) {hazard ratio 1.31 [95% confidence interval (CI) 1.00–1.71], P = .05} during a median follow-up period of 58 months. Such a statistically significant association persists for cardiovascular disease–related mortality and malignancy-related mortality [51]. Such a negative association between serum Klotho levels and all-cause mortality has also been established in another observational cohort study conducted in Italy, involving >804 adult participants ≥65 years of age [52]. Moreover, a meta-analysis of six cohort studies, including adult patients with CKD, revealed similar outcomes of higher risk for all-cause mortality among patients with lower serum Klotho levels [relative risk 1.88 (95% CI 1.29–2.74), P < .05] [53]. Furthermore, overexpression of the Klotho gene in transgenic mice has been shown to reverse or delay aging in preclinical studies, however, the therapeutic applicability of recombinant Klotho in human subjects is unknown [12].
Increasing Klotho levels emerges as a promising strategy in CKD. Experiments in rodents via viral delivery of Klotho cDNA or stimulating endogenous production through various means have shown promising results [54]. These methods demonstrate improvements in kidney function and protection from related complications [54]. Existing drugs such as paricalcitol and sevelamer carbonate also show potential in elevating Klotho levels [16]. However, clinical application faces challenges, with safety concerns and a lack of knowledge regarding upregulation and degradation mechanisms. Administering exogenous Klotho protein has demonstrated effective results in animal models [55], offering both restoration of levels and promotion of endogenous production, which is crucial in kidney failure scenarios. Despite success in animal trials, implementing Klotho replacement therapy in humans encounters significant obstacles [16]. A study conducted by Zhong et al. [56] investigated soluble Klotho (s-Klotho) derived from two distinct mammalian expression systems and found notable differences in their in vitro activities, in vivo pharmacokinetic properties and post-translational modifications (PTMs). A mutant s-Klotho with enhanced glycosidase functions exhibited reduced FGF23 co-receptor activity, suggesting structural divergence between the two well-established functions of s-Klotho. These findings offer valuable insights into the structure–function relationship of s-Klotho and its PTM biology, providing guidance for future therapeutic protein engineering of s-Klotho [56]. Additionally, fragment-based drug discovery methods could potentially overcome obstacles such as screening problems, early degradation resulting in a short half-life, low potency and challenges in crossing barriers. This approach may be particularly beneficial for developing small peptides of Klotho or other molecules with similar efficacy.
Multiple currently available drugs have been shown to upregulate serum levels of Klotho in human subjects.
Sodium–glucose co-transporter 2 (SGLT2) inhibitors
SGLT2 inhibitors constitute a new class of oral antidiabetic agents that improve glycaemic control by increasing renal sodium and glucose excretion. In addition to, and independent of, the effect on blood glucose, SGLT2 inhibitors have cardio- and renoprotective effects and can slow CKD progression [57]. Experimental studies have demonstrated that SGLT2 inhibitors prevent the downregulation of Klotho by inflammatory cytokines and high glucose in renal tubular cells, while they improved serum and urine Klotho levels and decreased urinary inflammatory markers and albuminuria in patients with early diabetic nephropathy [58]. In an animal model of unilateral ureteric obstruction, the SGLT2 inhibitor empagliflozin partially inhibited kidney function decline and the activation of inflammatory and profibrotic pathways [59]. Despite these promising clinical and experimental results, further studies are required to elucidate the role of Klotho in the cardio- and renoprotective effects of SGLT2 inhibitors.
Renin–angiotensin–aldosterone system (RAAS) inhibitors
RAAS blockers are commonly prescribed medications used in the management of hypertension, congestive heart failure, diabetic nephropathy, CKD and hyperaldosteronism. Losartan and valsartan have been shown to lead to an upregulation in serum Klotho levels in preclinical and clinical studies, however, the clinical importance of such an observation is less well understood [60–63]. The efficiency of valsartan and fluvastatin therapy separately or in combination in terms of anti-aging properties has been evaluated in a study including 130 healthy adult male participants. Participants received either fluvastatin 10 mg/day alone, valsartan 20 mg/day alone, fluvastatin 10 mg/day and valsartan 20 mg/day in combination or placebo for 30 days. Blood samples were obtained prior to therapy, after 30 days of therapy and 5 months after the discontinuation of therapy, while expression levels of six genes (SIRT1, PRKAA, KLOTHO, NFE2L2, mTOR and NF-κB) were analysed. The combination therapy leads to a 1.8-fold increase in SIRT1 (P < .0001), a 1.5-fold increase in PRKAA (P = .262) and a 1.7-fold increase in KLOTHO (P < .0001) levels while either therapy alone only leads to a slight increase in SIRT1 expression. Such a pattern of gene expression disappeared following the discontinuation of therapy [64]. This study is significant by demonstrating alterations in gene expression levels with commonly utilized medications, however, whether such alterations have clinical significance in terms of mortality or morbidity is unclear. Such an association has been validated in a randomized controlled clinical trial conducted in 76 patients with type 2 diabetes mellitus and diabetic kidney disease (DKD) in which participants were randomized to receive either valsartan/hydrochlorothiazide (n = 37) or amlodipine therapy (n = 39). Participants receiving valsartan/hydrochlorothiazide therapy had a statistically significant increase in serum Klotho levels (from 432.7 ± 179 to 506.4 ± 226.8 pg/ml; P = .01) even after comparison with the amlodipine group (P = .03) [60]. Furthermore, a similar pattern of elevation at serum Klotho levels in patients with DKD has been demonstrated with losartan therapy as well [62].
On the other hand, there is no clinical trial, to the best of our knowledge, conducted on human subjects regarding the effects of spironolactone or epleronone, aldosterone receptor antagonists, on serum Klotho levels. A preclinical trial conducted on mice and human embryonic kidney cells demonstrated that spironolactone therapy leads to upregulation of Klotho gene expression [65]. Nevertheless, there is a clear need for confirmatory studies in human participants for a better understanding of this issue.
Statins
Statins, 3-hydroxy-3-methyl-glutaryl-coenzyme A (HMG-CoA) reductase inhibitors, are widely used medications in the management of atherosclerosis, hyperlipidaemia, ischaemic cardiovascular or cerebrovascular diseases and peripheral artery diseases. Atorvastatin, pitavastatin and simvastatin have been shown to upregulate Klotho expression in preclinical studies [24, 66, 67]. An animal model of cognitive decline induced via intracerebroventricular streptozotocin administration has been utilized to evaluate the response to statin therapy in terms of cognitive functions. Klotho expression decreased significantly following the administration of streptozotocin compared with control models. Spatial performance was improved in animal models treated with simvastatin for 3 weeks, which may be attributable to the upregulation of Klotho and manganese superoxide dismutase [68].
Mammalian target of rapamycin (mTOR) inhibitors
The mTOR inhibitors are widely used immunosuppressive medications utilized in autoimmune disorders and transplantation. Serum Klotho levels were evaluated in an observational study including 36 kidney transplant recipients either taking everolimus or not in which serum Klotho levels were measured before and 1 year after kidney transplantation. Serum Klotho levels were higher 1 year after kidney transplantation as expected with the improvement in renal function (369.3 versus 211.8 pg/ml). Moreover, serum Klotho levels have been found to be higher in patients receiving everolimus therapy compared with others, in a statistically significant manner (536.7 versus 332.4 pg/ml) [69]. Another study conducted on 100 kidney transplant recipients, 50 treated with rapamycin and 50 treated with calcineurin inhibitors, showed similar findings, while elevated Klotho expression has been linked to the activation of mTORC2 via the phosphorylation of AKT [70].
Others
PPARγ agonists such as pioglitazone and rosiglitazone and vitamin D supplementations have been shown to upregulate Klotho expression in experimental animal models. Nevertheless, their applicability to human subjects is doubtful [71–75]. Multiple preclinical therapeutic modalities primarily targeting Klotho expression or functions are present in the literature.
γ-Aminobutyric acid receptor (GABA) agonists
The use of GABA agonists is a novel therapeutic approach currently in preclinical studies. Administration of GABA has been shown to increase the levels of circulating Klotho and Klotho levels at pancreatic islet of Langerhans among streptozotocin-induced diabetic mice subjects, which may be diminished in Klotho-knockdown subjects. However, infusion of soluble Klotho may reverse such conditions, thus indicating the pathophysiological importance of Klotho in the underlying mechanism of both type 1 diabetes mellitus and GABA-induced improvement [76]. In another study conducted on immunodeficient streptozotocin-induced diabetic mice models with human pancreatic β cell transplantation, the superiority of GABA + dipeptidyl peptidase (DPP)-IV inhibitor, namely sitagliptin, was revealed [77]. A preclinical study conducted on human islet cells incubated under cytokine mixture for 24 hours demonstrated that cytokine-induced apoptosis is significantly reduced by either GABA or glucagon-like peptide-1 analogue, namely exendin-4, therapy. However, the combination therapy has led to superior outcomes via upregulation of Klotho and sirtuin 1 expression and activation of the AKT signalling pathway [78].
Recombinant protein and gene therapy
Gene therapy and the use of recombinant proteins in the management of certain conditions are novel areas of medical research, while considerable efforts have been made regarding such therapeutic alternatives linked to Klotho over the last decade. Supplementation with recombinant Klotho has been shown to protect against kidney fibrosis caused by hypoxia or other insults via inhibiting TGF-β signalling in cell lines and animal models [79–81]. Also, recombinant Klotho therapy has been shown to exert protective actions on myocardial ischaemia–reperfusion injury, insulitis in diabetic subjects and hypertension in various models [40, 82, 83]. Moreover, Klotho-derived peptide 6 (KP6) mimics the function of the anti-aging protein Klotho and shows promise in treating DKD. KP6 reversed proteinuria, reduced kidney damage and inhibited β-catenin activation, suggesting a potential therapeutic strategy for DKD treatment [84]. Furthermore, gene therapies focusing on Klotho are being evaluated for therapeutic alternatives for conditions such as diabetes mellitus and neurodegenerative diseases such as epilepsy and Alzheimer's disease [85–87]. Nevertheless, such therapeutic alternatives are far from any clinical use and require multiple large-scale studies evaluating their efficiency and safety in human subjects.
Diet and nutrition
The serum level of Klotho is also affected by food and diet types. To decipher the role of diet in the serum level of Klotho is crucial to its relationship with aging and related diseases. A cross-sectional study based on 2007–2016 data with 7906 participants in the USA indicated that the serum Klotho level significantly increases [β coefficient 9.41 (95% CI 6.08–12.74), P < .001; 9.41 pg/ml increase per 1 point in the Mediterranean Diet Score) in people who adhere to a Mediterranean diet, probably due to a synergistic effect of food combinations, while in people who prefer low-fat and low-carbohydrate diets, non-significant associations were seen [88]. Additionally, a cross-sectional study with 8456 participants ages 40–79 years in the USA demonstrated a positive correlation between a Healthy Eating Index 2015 score and the serum Klotho level [β 0.74 (95% CI 0.21–1.27), P = .0067] [89]. Moreover, another study showed that Klotho is a significant mediator in the enhancement of kidney function by a healthy diet (Healthy Eating Index 2015 score) [β 0.05 (95% CI 0.02–0.08), P < .001] [90]. According to a cross-sectional study based on data from 2015–2016 with 2637 participants, 40–79 years old, in the USA, a high-fibre diet in men increases serum Klotho level [91], while a similar study with 11 282 participants (2007–2016 data from participants 40–79 years old in the USA) showed that a high-fibre diet in overweight, obese and elderly participants has a more robust association with serum Klotho levels [92]. Although the benefit of a healthy and high-fibre diet on Klotho levels has been shown in multiple studies, it is crucial to note that the connection between serum Klotho and diet is not entirely understood. More studies are required to demonstrate a definite causal relationship between eating habits and Klotho levels. Furthermore, genetics, lifestyle variables and other environmental influences all have a role in klotho levels and overall health. Fig. 1 demonstrates the summary of negative and positive factors affecting Klotho production as well as the signalling pathways with which Klotho interacts to exert its physiological impacts.
Figure 1:
Summary of the molecular basis and physiological impacts of Klotho. Factors that negatively affect Klotho production are shown in the upper left part of the figure and those that have positive impacts are shown in the upper right. The molecular pathways affected by Klotho, including IGF-1, Wnt, HIF [36] and phosphate homeostasis, are shown. The respective physiological alterations are depicted. Blunt and conventional arrows indicate inhibition and activation, respectively.
Klotho in aging
Aging can be defined as an irreversible process that leads to the gradual deterioration of tissue and organ function, eventually resulting in death. It is characterized by molecular and cellular changes such as genomic instability, oxidative stress, mitochondrial dysfunction and telomere shortening [93]. The role of Klotho protein in aging has been evaluated in multiple preclinical and clinical studies. Overexpression of Klotho mediated via the control of human elongation factor 1α promoter has been linked to an increase in the lifespan of mice [12]. A meta-analysis regarding the genetic variation of Klotho and its relation to longevity indicated a significant association between healthy aging and the KL-VS variant of the Klotho gene [94]. On the other hand, a case–control study showed no significant relation between the KL-VS variant of Klotho and longevity [95]. A prospective cohort study conducted in 804 adult participants ≥65 years of age with a 6-year follow-up period demonstrated that low serum Klotho levels (<575 pg/ml) are linked to a higher risk for mortality compared with higher serum Klotho levels (<763 pg/ml) after multivariate adjustments [52]. Another prospective study conducted in 1023 adult participants demonstrated that plasma Klotho levels are inversely correlated with age (P < .001) and serum C-reactive protein levels (P < .001). Moreover, age-adjusted serum Klotho levels are inversely correlated with the risk of cardiovascular disease (P < .001). Additionally, after adjustment for multiple cardiovascular disease–related variables (age, gender, smoking, lipid profile, diabetes mellitus, systolic blood pressure), log plasma Klotho levels were associated with cardiovascular disease risk [odds ratio per 1 standard deviation increase = 0.85 (95% CI 0.72–0.99)] [96]. Other cohort studies have illustrated a statistically significant inverse correlation between serum Klotho levels and cognitive decline or physical performance [97, 98].
Furthermore, several studies have shown that serum Klotho levels are inversely correlated with age. In other words, younger people tend to have higher plasma Klotho levels [99, 100]. Also, deficiency of Klotho contributes to hypertension, arterial stiffening, endothelial dysfunction and many other age-associated disorders [4]. It has been shown that expression impairments of the Klotho gene in mice produce a bundle of symptoms similar to aging in humans, such as decreased lifetime, skeletal and dermal problems, infertility and arterial stiffening [3].
Nevertheless, one significant concern and confounding factor in such research is the clear link between CKD and aging irrespective of serum Klotho levels [101, 102]. Multiple pathophysiological mechanisms, including uraemic toxin–mediated alterations, traditional cardiovascular disease–related neurodegeneration, polypharmacy, anaemia and dialysis-related factors are involved in this complex pathway [102]. Therefore it is important not to overlook such confounding factors in the discussion and evaluation of the role of Klotho in aging and neurodegenerative conditions.
Klotho in neurodegenerative disorders
The highest level of Klotho production in the brain occurs at the choroid plexus, while lower production occurs in the hippocampus, substantia nigra, medulla and cerebral/cerebellar cortex [103]. Klotho expression is seen in both neurons and oligodendrocytes, while the production starts in utero and diminishes with age. Mean cerebrospinal fluid Klotho levels are significantly lower in older adults compared with younger individuals (P = .005) and in patients with Alzheimer's disease (P = .02) [104]. An observational study conducted in 103 elderly patients in Spain revealed that lower serum Klotho levels in adults are associated with higher frailty and more likely to experience falls in the following 6 months [105]. The physiological functions of Klotho in the central nervous system have initially been illustrated via studies conducted on Klotho-knockout animal subjects and include diminished axonal transport, formation of a myelin sheath and maturation of oligodendrocytes, a decrease in the proliferation of hippocampal neural progenitor cells and cognitive impairment [106, 107]. Additional physiological functions have been identified via studies including administration of recombinant Klotho or epigenetic studies inducing the overexpression of Klotho protein. Incubation of hippocampal neuronal cell cultures with soluble Klotho leads to a decrease in oxidative stress via upregulation of superoxide dismutase activity and pro-inflammatory cytokines such as interleukin-1, tumour necrosis factor-α and NF-κB and causes protection against amyloid-β or glutamate-induced oxidative toxicity [108, 109]. Administration of soluble Klotho peripherally to mice subjects has led to enhancement of synaptic plasticity and glutamate receptor signalling, while similar findings have been recorded when secreted Klotho is overexpressed in mice models [110, 111]. Furthermore, overexpression of the full-length Klotho gene results in a decrease in oxidative and endoplasmic reticulum stress, inhibition of NLRP3 inflammasomes, improvement in synaptic plasticity and amyloid-β clearance mechanisms, protection of dopaminergic and hippocampal neurons and improvement in memory and cognition (Fig. 2) [112–115].
Figure 2:
Infographic summary of the promising aspects, concerns and research recommendations of Klotho.
On the other hand, Klotho does not cross the blood–brain barrier (BBB). However, Leon et al. [111] demonstrated that administering the alpha Klotho fragment (α-KL-F) peripherally results in an immediate enhancement of cognitive function and heightened neural resilience and plasticity in mice at various life stages. Interestingly, peripherally administered α-KL-F shows its effect by cleavage of the N-methyl-D-aspartate receptor (NMDAR) subunit GluN2B and results in increased NMDAR-dependent synaptic plasticity [111]. Additionally, it has been shown that platelet factor 4 induced by Klotho can cross the BBB and can increase hippocampal synaptic plasticity [116]. Despite the promising discoveries regarding the action and mechanism of Klotho, there are still crucial gaps in the literature, especially how Klotho exerts its effects on memory and neurocognition.
CONCLUSION
Knowledge regarding the physiological functions of Klotho protein, first identified in 1997, has expanded over the years, despite initial studies primarily focusing on its role in vitamin D metabolism, vascular calcification and phosphate homeostasis, especially among patients with CKD. Preclinical studies have demonstrated potential links between serum Klotho levels and multiple conditions including CKD progression, cardiovascular fibrosis, aging and neurodegenerative disorders. Emerging clinical and experimental insights suggest Klotho deficiency not only as a risk factor, but also a modifiable therapeutic target. Even though there is a clear need for future large-scale human studies in order to develop clinical and therapeutic strategies involving Klotho proteins in humans, this field appears to be promising.
ACKNOWLEDGEMENTS
This paper was written on behalf of the ERA CKD-MBD working group, which is an official body of the European Renal Association (ERA).
Contributor Information
Mehmet Kanbay, Department of Medicine, Nephrology, Koc University School of Medicine, Istanbul, Turkey.
Sidar Copur, Department of Medicine, Koc University School of Medicine, Istanbul, Turkey.
Lasin Ozbek, Department of Medicine, Koc University School of Medicine, Istanbul, Turkey.
Ali Mutlu, Department of Medicine, Koc University School of Medicine, Istanbul, Turkey.
Daniel Cejka, Department of Medicine III – Nephrology, Hypertension, Transplantation, Rheumatology, Geriatrics, Ordensklinikum Linz – Elisabethinen Hospital, Linz, Austria.
Paola Ciceri, Department of Health Sciences, Renal Division, University of Milan, Milan, Italy.
Mario Cozzolino, Department of Health Sciences, Renal Division, University of Milan, Milan, Italy.
Mathias Loberg Haarhaus, Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden.
CONFLICT OF INTEREST STATEMENT
M.K. and M.C. are members of the CKJ editorial board.
REFERENCES
- 1. Coppede F. Mutations involved in premature-ageing syndromes. Appl Clin Genet 2021;14:279–95. 10.2147/TACG.S273525 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Javidnia S, Cranwell S, Mueller SH et al. Mendelian randomization analyses implicate biogenesis of translation machinery in human aging. Genome Res 2022;32:258–65. 10.1101/gr.275636.121 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Kuro-o M, Matsumura Y, Aizawa H et al. Mutation of the mouse klotho gene leads to a syndrome resembling ageing. Nature 1997;390:45–51. 10.1038/36285 [DOI] [PubMed] [Google Scholar]
- 4. Buchanan S, Combet E, Stenvinkel P et al. Klotho, aging, and the failing kidney. Front Endocrinol (Lausanne) 2020;11:560. 10.3389/fendo.2020.00560 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Kanbay M, Demiray A, Afsar B et al. Role of Klotho in the development of essential hypertension. Hypertension 2021;77:740–50. 10.1161/HYPERTENSIONAHA.120.16635 [DOI] [PubMed] [Google Scholar]
- 6. Lim K, Groen A, Molostvov G et al. α-klotho expression in human tissues. J Clin Endocrinol Metab 2015;100:E1308–18. 10.1210/jc.2015-1800 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Kuro-o M. The Klotho proteins in health and disease. Nat Rev Nephrol 2019;15:27–44. 10.1038/s41581-018-0078-3 [DOI] [PubMed] [Google Scholar]
- 8. Ogawa Y, Kurosu H, Yamamoto M et al. βKlotho is required for metabolic activity of fibroblast growth factor 21. Proc Natl Acad Sci USA 2007;104:7432–7. 10.1073/pnas.0701600104 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Kim JH, Hwang KH, Park KS et al. Biological role of anti-aging protein klotho. J Lifestyle Med 2015;5:1–6. 10.15280/jlm.2015.5.1.1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Fon Tacer K, Bookout AL, Ding X et al. Research resource: comprehensive expression atlas of the fibroblast growth factor system in adult mouse. Mol Endocrinol 2010;24:2050–64. 10.1210/me.2010-0142 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Kinoshita S, Kawai M. The FGF23/KLOTHO regulatory network and its roles in human disorders. Vitam Horm 2016;101:151–74. 10.1016/bs.vh.2016.02.001 [DOI] [PubMed] [Google Scholar]
- 12. Kurosu H, Yamamoto M, Clark JD et al. Suppression of aging in mice by the hormone Klotho. Science 2005;309:1829–33. 10.1126/science.1112766 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Hu MC, Shi M, Zhang J et al. Renal production, uptake, and handling of circulating αKlotho. J Am Soc Nephrol 2016;27:79–90. 10.1681/ASN.2014101030 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Saar-Kovrov V, Donners M, van der Vorst EPC. Shedding of Klotho: functional implications in chronic kidney disease and associated vascular disease. Front Cardiovasc Med 2020;7:617842. 10.3389/fcvm.2020.617842 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Chen CD, Podvin S, Gillespie E et al. Insulin stimulates the cleavage and release of the extracellular domain of Klotho by ADAM10 and ADAM17. Proc Natl Acad Sci USA 2007;104:19796–801. 10.1073/pnas.0709805104 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Neyra JA, Hu MC, Moe OW. Klotho in clinical nephrology: diagnostic and therapeutic implications. Clin J Am Soc Nephrol 2020;16:162–76. 10.2215/CJN.02840320 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Xu Y, Sun Z. Molecular basis of Klotho: from gene to function in aging. Endocr Rev 2015;36:174–93. 10.1210/er.2013-1079 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Yahata K, Mori K, Arai H et al. Molecular cloning and expression of a novel klotho-related protein. J Mol Med (Berl) 2000;78:389–94. 10.1007/s001090000131 [DOI] [PubMed] [Google Scholar]
- 19. Saito K, Ishizaka N, Mitani H et al. Iron chelation and a free radical scavenger suppress angiotensin II-induced downregulation of klotho, an anti-aging gene, in rat. FEBS Lett 2003;551:58–62. 10.1016/S0014-5793(03)00894-9 [DOI] [PubMed] [Google Scholar]
- 20. Moreno JA, Izquierdo MC, Sanchez-Niño MD et al. The inflammatory cytokines TWEAK and TNFα reduce renal klotho expression through NFκB. J Am Soc Nephrol 2011;22:1315–25. 10.1681/ASN.2010101073 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Asai O, Nakatani K, Tanaka T et al. Decreased renal α-klotho expression in early diabetic nephropathy in humans and mice and its possible role in urinary calcium excretion. Kidney Int 2012;81:539–47. 10.1038/ki.2011.423 [DOI] [PubMed] [Google Scholar]
- 22. Zhang R, Zheng F. PPAR-gamma and aging: one link through klotho? Kidney Int 2008;74:702–4. 10.1038/ki.2008.382 [DOI] [PubMed] [Google Scholar]
- 23. Forster RE, Jurutka PW, Hsieh JC et al. Vitamin D receptor controls expression of the anti-aging klotho gene in mouse and human renal cells. Biochem Biophys Res Commun 2011;414:557–62. 10.1016/j.bbrc.2011.09.117 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Narumiya H, Sasaki S, Kuwahara N et al. HMG-CoA reductase inhibitors up-regulate anti-aging klotho mRNA via RhoA inactivation in IMCD3 cells. Cardiovasc Res 2004;64:331–6. 10.1016/j.cardiores.2004.07.011 [DOI] [PubMed] [Google Scholar]
- 25. Liu S, Gupta A, Quarles LD. Emerging role of fibroblast growth factor 23 in a bone-kidney axis regulating systemic phosphate homeostasis and extracellular matrix mineralization. Curr Opin Nephrol Hypertens 2007;16:329–35. 10.1097/MNH.0b013e3281ca6ffd [DOI] [PubMed] [Google Scholar]
- 26. Kuro-o M. Klotho as a regulator of fibroblast growth factor signaling and phophate/calcium metabolism. Curr Opin Nephrol Hypertens 2006;15:437–41. 10.1097/01.mnh.0000232885.81142.83 [DOI] [PubMed] [Google Scholar]
- 27. Shimada T, Yamazaki Y, Takahashi M et al. Vitamin D receptor-independent FGF23 actions in regulating phosphate and vitamin D metabolism. Am J Physiol Renal Physiol 2005;289:F1088–95. 10.1152/ajprenal.00474.2004 [DOI] [PubMed] [Google Scholar]
- 28. Ben-Dov IZ, Galitzer H, Lavi-Moshayoff V et al. The parathyroid is a target organ for FGF23 in rats. J Clin Invest 2007;117:4003–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Komiya Y, Habas R. Wnt signal transduction pathways. Organogenesis 2008;4:68–75. 10.4161/org.4.2.5851 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Liu H, Fergusson MM, Castilho RM et al. Augmented Wnt signaling in a mammalian model of accelerated aging. Science 2007;317:803–6. 10.1126/science.1143578 [DOI] [PubMed] [Google Scholar]
- 31. Chang Q, Hoefs S, van der Kemp AW et al. The beta-glucuronidase klotho hydrolyzes and activates the TRPV5 channel. Science 2005;310:490–3. 10.1126/science.1114245 [DOI] [PubMed] [Google Scholar]
- 32. Zhou L, Li Y, Zhou D et al. Loss of Klotho contributes to kidney injury by derepression of Wnt/β-catenin signaling. J Am Soc Nephrol 2013;24:771–85. 10.1681/ASN.2012080865 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Mitobe M, Yoshida T, Sugiura H et al. Oxidative stress decreases klotho expression in a mouse kidney cell line. Nephron Exp Nephrol 2005;101:e67–74. 10.1159/000086500 [DOI] [PubMed] [Google Scholar]
- 34. Yamamoto M, Clark JD, Pastor JV et al. Regulation of oxidative stress by the anti-aging hormone klotho. J Biol Chem 2005;280:38029–34. 10.1074/jbc.M509039200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Armoni M, Harel C, Karni S et al. FOXO1 represses peroxisome proliferator-activated receptor-gamma1 and -gamma2 gene promoters in primary adipocytes. A novel paradigm to increase insulin sensitivitiy. J Biol Chem 2006;281:19881–91. [DOI] [PubMed] [Google Scholar]
- 36. Chihara Y, Rakugi H, Ishikawa K et al. Klotho protein promotes adipocyte differentiation. Endocrinology 2006;147:3835–42. 10.1210/en.2005-1529 [DOI] [PubMed] [Google Scholar]
- 37. Briet M, Barhoumi T, Mian MO et al. Effects of recombinant human erythropoietin on resistance artery endothelial function in stage 4 chronic kidney disease. J Am Heart Assoc 2013;2:e000128. 10.1161/JAHA.113.000128 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Semenza GL. HIF-1: mediator of physiological and pathophysiological responses to hypoxia. J Apply Physiol (1985) 2000;88:1474–80. 10.1152/jappl.2000.88.4.1474 [DOI] [PubMed] [Google Scholar]
- 39. Li Q, Li Y, Liang L et al. Klotho negatively regulated aerobic glycolysis in colorectal cancer via ERK/HIF1α axis. Cell Commun Signal 2018;16:26. 10.1186/s12964-018-0241-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Takenaka T, Kobori H, Miyazaki T et al. Klotho protein supplementation reduces blood pressure and renal hypertrophy in db/db mice, a model of type 2 diabetes. Acta Physiol (Oxf) 2019;225:e13190. 10.1111/apha.13190 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Vadakke Madathil S, Coe LM, Casu C et al. Klotho deficiency disrupts hematopoietic stem cell development and erythropoiesis. Am J Pathol 2014;184:827–41. 10.1016/j.ajpath.2013.11.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Afsar B, Kanbay M, Afsar RE. Interconnections of fibroblast growth factor 23 and klotho with erythropoietin and hypoxia-inducible factor. Mol Cell Biochem 2022;477:1973–85. 10.1007/s11010-022-04422-3 [DOI] [PubMed] [Google Scholar]
- 43. Picciotto D, Murugavel A, Ansaldo F et al. The organ handling of soluble klotho in humans. Kidney Blood Press Res 2019;44:715–26. 10.1159/000501316 [DOI] [PubMed] [Google Scholar]
- 44. Tohyama O, Imura A, Iwano A et al. Klotho is a novel β-glucuronidase capable of hydrolyzing steroid β-glucuronides. J Biol Chem 2004;279:9777–84. 10.1074/jbc.M312392200 [DOI] [PubMed] [Google Scholar]
- 45. Mian IS. Sequence, structural, functional, and phylogenetic analyses of three glycosidase families. Blood Cells Mol Dis 1998;24:83–100. 10.1006/bcmd.1998.9998 [DOI] [PubMed] [Google Scholar]
- 46. Hu MC, Shi M, Zhang J et al. Klotho: a novel phosphaturic substance acting as an autocrine enzyme in the renal proximal tubule. FASEB J 2010;24:3438–50. 10.1096/fj.10-154765 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. de Oliveira RM. Klotho RNAi induces premature senescence of human cells via a p53/p21 dependent pathway. FEBS Lett 2006;580:5753–8. 10.1016/j.febslet.2006.09.036 [DOI] [PubMed] [Google Scholar]
- 48. Doi S, Zou Y, Togao O et al. Klotho inhibits transforming growth factor-β1 (TGF-β1) signaling and suppresses renal fibrosis and cancer metastasis in mice. J Biol Chem 2011;286:8655–65. 10.1074/jbc.M110.174037 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Espuch-Oliver A, Vázquez-Lorente H, Jurado-Fasoli L et al. References values of soluble α-klotho serum levels using an enzyme-linked immunosorbent assay in healthy adults aged 18–85 years. J Clin Med 2022;11:2415. 10.3390/jcm11092415 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Kuro-o M. A potential link between phosphate and aging—lessons from Klotho-deficient mice. Mech Ageing Dev 2010;131:270–5. 10.1016/j.mad.2010.02.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Kresovich JK, Bulka CM. Low serum klotho associated with all-cause mortality among a nationally representative sample of American adults. J Gerontol A Biol Sci Med Sci 2022;77:452–6. 10.1093/gerona/glab308 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Semba RD, Cappola AR, Sun K et al. Plasma klotho and mortality risk in older community-dwelling adults. J Gerontol A Biol Sci Med Sci 2011;66:794–800. 10.1093/gerona/glr058 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Charoenngam N, Ponvilawan B, Ungprasert P. Lower circulating soluble Klotho level is associated with increased risk of all-cause mortality in chronic kidney disease patients: a systematic review and meta-analysis. Int Urol Nephrol 2020;52:1543–50. 10.1007/s11255-020-02510-1 [DOI] [PubMed] [Google Scholar]
- 54. Wang Y, Sun Z. Klotho gene delivery prevents the progression of spontaneous hypertension and renal damage. Hypertension 2009;54:810–7. 10.1161/HYPERTENSIONAHA.109.134320 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Shi M, Flores B, Gillings N et al. αKlotho mitigates progression of AKI to CKD through activation of autophagy. J Am Soc Nephrol 2016;27:2331–45. 10.1681/ASN.2015060613 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Zhong X, Jagarlapudi S, Weng Y et al. Structure-function relationships of the soluble form of the antiaging protein Klotho have therapeutic implications for managing kidney disease. J Biol Chem 2020;295:3115–33. 10.1074/jbc.RA119.012144 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Packer M. Role of impaired nutrient and oxygen deprivation signaling and deficient autophagic flux in diabetic CKD development: implications for understanding the effects of sodium-glucose cotransporter 2-inhibitors. J Am Soc Nephrol 2020;31:907–19. 10.1681/ASN.2020010010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Mora-Fernández C, Sánchez-Niño MD, Donate-Correa J et al. Sodium-glucose co-transporter-2 inhibitors increase Klotho in patients with diabetic kidney disease: a clinical and experimental study. Biomed Pharmacother 2022;154:113677. 10.1016/j.biopha.2022.113677 [DOI] [PubMed] [Google Scholar]
- 59. Abbas NAT, El Salem A, Awad MM. Empagliflozin, SGLT2 inhibitor, attenuates renal fibrosis in rats exposed to unilateral ureteric obstruction: potential role of klotho expression. Naunyn Schmiedebergs Arch Pharmacol 2018;391:1347–60. 10.1007/s00210-018-1544-y [DOI] [PubMed] [Google Scholar]
- 60. Karalliedde J, Maltese G, Hill B et al. Effect of renin-angiotensin system blockade on soluble Klotho in patients with type 2 diabetes, systolic hypertension, and albuminuria. Clin J Am Soc Nephrol 2013;8:1899–905. 10.2215/CJN.02700313 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Li X, Zhou Q. [Effect of valsartan on ultrastructure in the brain tissue and expression of Klotho gene in spontaneously hypertensive rats]. Zhong Nan Da Xue Xue Bao Yi Xue Ban 2010;35:254–61. [DOI] [PubMed] [Google Scholar]
- 62. Lim SC, Liu JJ, Subramaniam T et al. Elevated circulating alpha-klotho by angiotensin II receptor blocker losartan is associated with reduction of albuminuria in type 2 diabetic patients. J Renin Angiotensin Aldosterone Syst 2014;15:487–90. 10.1177/1470320313475905 [DOI] [PubMed] [Google Scholar]
- 63. Yoon HE, Ghee JY, Piao S et al. Angiotensin II blockade upregulates the expression of Klotho, the anti-ageing gene, in an experimental model of chronic cyclosporine nephropathy. Nephrol Dial Transplant 2011;26:800–13. 10.1093/ndt/gfq537 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Janić M, Lunder M, Novaković S et al. Expression of longevity genes induced by a low-dose fluvastatin and valsartan combination with the potential to prevent/treat “aging-related disorders”. Int J Mol Sci 2019;20:1844. 10.3390/ijms20081844 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Alesutan I, Feger M, Pakladok T et al. 25-Hydroxyvitamin D3 1-α-hydroxylase-dependent stimulation of renal klotho expression by spironolactone. Kidney Blood Press Res 2013;37:475–87. 10.1159/000355728 [DOI] [PubMed] [Google Scholar]
- 66. Kuwahara N, Sasaki S, Kobara M et al. HMG-CoA reductase inhibition improves anti-aging klotho protein expression and arteriosclerosis in rats with chronic inhibition of nitric oxide synthesis. Int J Cardiol 2008;123:84–90. 10.1016/j.ijcard.2007.02.029 [DOI] [PubMed] [Google Scholar]
- 67. Yoon HE, Lim SW, Piao SG et al. Statin upregulates the expression of klotho, an anti-aging gene, in experimental cyclosporine nephropathy. Nephron Exp Nephrol 2012;120:e123–33. 10.1159/000342117 [DOI] [PubMed] [Google Scholar]
- 68. Adeli S, Zahmatkesh M, Tavoosidana G et al. Simvastatin enhances the hippocampal klotho in a rat model of streptozotocin-induced cognitive decline. Prog Neuropsychopharmacol Biol Psychiatry 2017;72:87–94. 10.1016/j.pnpbp.2016.09.009 [DOI] [PubMed] [Google Scholar]
- 69. Mizusaki K, Hasuike Y, Kimura T et al. Inhibition of the mammalian target of rapamycin may augment the increase in soluble klotho levels in renal transplantation recipients. Blood Purif 2019;47(Suppl 2):12–8. 10.1159/000496630 [DOI] [PubMed] [Google Scholar]
- 70. Tataranni T, Biondi G, Cariello M et al. Rapamycin-induced hypophosphatemia and insulin resistance are associated with mTORC2 activation and Klotho expression. Am J Transplant 2011;11:1656–64. 10.1111/j.1600-6143.2011.03590.x [DOI] [PubMed] [Google Scholar]
- 71. Shen D, Li H, Zhou R et al. Pioglitazone attenuates aging-related disorders in aged apolipoprotein E deficient mice. Exp Gerontol 2018;102:101–8. 10.1016/j.exger.2017.12.002 [DOI] [PubMed] [Google Scholar]
- 72. Huang KC, Cherng YG, Chen LJ et al. Rosiglitazone is effective to improve renal damage in type-1-like diabetic rats. Horm Metab Res 2014;46:240–4. [DOI] [PubMed] [Google Scholar]
- 73. Cheng L, Zhang L, Yang J et al. Activation of peroxisome proliferator-activated receptor γ inhibits vascular calcification by upregulating Klotho. Exp Ther Med 2017;13:467–74. 10.3892/etm.2016.3996 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Chen LJ, Cheng MF, Ku PM et al. Rosiglitazone increases cerebral klotho expression to reverse baroreflex in type 1-like diabetic rats. Biomed Res Int 2014;2014:309151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Haussler MR, Livingston S, Sabir ZL et al. Vitamin D receptor mediates a myriad of biological actions dependent on its 1,25-dihydroxyvitamin D ligand: distinct regulatory themes revealed by induction of klotho and fibroblast growth factor-23. JBMR Plus 2021;5:e10432. 10.1002/jbm4.10432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Prud'homme GJ, Glinka Y, Kurt M et al. The anti-aging protein Klotho is induced by GABA therapy and exerts protective and stimulatory effects on pancreatic beta cells. Biochem Biophys Res Commun 2017;493:1542–7. 10.1016/j.bbrc.2017.10.029 [DOI] [PubMed] [Google Scholar]
- 77. Liu W, Lau HK, Son DO et al. Combined use of GABA and sitagliptin promotes human β-cell proliferation and reduces apoptosis. J Endocrinol 2021;248:133–43. 10.1530/JOE-20-0315 [DOI] [PubMed] [Google Scholar]
- 78. Son DO, Liu W, Li X et al. Combined effect of GABA and glucagon-like peptide-1 receptor agonist on cytokine-induced apoptosis in pancreatic β-cell line and isolated human islets. J Diabetes 2019;11:563–72. 10.1111/1753-0407.12881 [DOI] [PubMed] [Google Scholar]
- 79. Yuan Q, Ren Q, Li L et al. A Klotho-derived peptide protects against kidney fibrosis by targeting TGF-β signaling. Nat Commun 2022;13:438. 10.1038/s41467-022-28096-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Ali MF, Venkatarayappa SKB, Benny M et al. Effects of Klotho supplementation on hyperoxia-induced renal injury in a rodent model of postnatal nephrogenesis. Pediatr Res 2020;88:565–70. 10.1038/s41390-020-0803-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Chen TH, Kuro OM, Chen CH et al. The secreted Klotho protein restores phosphate retention and suppresses accelerated aging in Klotho mutant mice. Eur J Pharmacol 2013;698:67–73. 10.1016/j.ejphar.2012.09.032 [DOI] [PubMed] [Google Scholar]
- 82. Prud'homme GJ, Glinka Y, Kurt M et al. Systemic Klotho therapy protects against insulitis and enhances beta-cell mass in NOD mice. Biochem Biophys Res Commun 2020;525:693–8. 10.1016/j.bbrc.2020.02.123 [DOI] [PubMed] [Google Scholar]
- 83. Myung J, Beom JH, Kim JH et al. Recombinant Klotho protein ameliorates myocardial ischemia/reperfusion injury by attenuating sterile inflammation. Biomedicines 2022;10:894. 10.3390/biomedicines10040894 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Sacks D, Baxter B, Campbell BCV et al. Multisociety consensus quality improvement revised consensus statement for endovascular therapy of acute ischemic stroke. Int J Stroke 2018;13:612–32. [DOI] [PubMed] [Google Scholar]
- 85. Xiang T, Luo X, Zeng C et al. Klotho ameliorated cognitive deficits in a temporal lobe epilepsy rat model by inhibiting ferroptosis. Brain Res 2021;1772:147668. 10.1016/j.brainres.2021.147668 [DOI] [PubMed] [Google Scholar]
- 86. Lin Y, Sun Z. In vivo pancreatic β-cell-specific expression of antiaging gene Klotho: a novel approach for preserving β-cells in type 2 diabetes. Diabetes 2015;64:1444–58. 10.2337/db14-0632 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Zeng CY, Yang TT, Zhou HJ et al. Lentiviral vector-mediated overexpression of Klotho in the brain improves Alzheimer's disease-like pathology and cognitive deficits in mice. Neurobiol Aging 2019;78:18–28. 10.1016/j.neurobiolaging.2019.02.003 [DOI] [PubMed] [Google Scholar]
- 88. Wu SE, Chen YJ, Chen WL. Adherence to Mediterranean diet and soluble klotho level: the value of food synergy in aging. Nutrients 2022;14:3910. 10.3390/nu14193910 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Ma TC, Zhou J, Wang CX et al. Associations between the Healthy Eating Index-2015 and S-Klotho plasma levels: a cross-sectional analysis in middle-to-older aged adults. Front Nutr 2022;9:904745. 10.3389/fnut.2022.904745 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Cai Q, Hu S, Qi C et al. Serum anti-aging protein α-klotho mediates the association between diet quality and kidney function. Nutrients 2023;15:2744. 10.3390/nu15122744 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Ostojic SM, Hillesund ER, Øverby NC et al. Individual nutrients and serum klotho levels in adults aged 40–79 years. Food Sci Nutr 2023;11:3279–86. 10.1002/fsn3.3310 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Liu S, Wu M, Wang Y et al. The association between dietary fiber intake and serum klotho levels in Americans: a cross-sectional study from the National Health and Nutrition Examination Survey. Nutrients 2023;15:3147. 10.3390/nu15143147 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Cheikhi A, Barchowsky A, Sahu A et al. Klotho: an elephant in aging research. J Gerontol A Biol Sci Med Sci 2019;74:1031–42. 10.1093/gerona/glz061 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Di Bona D, Accardi G, Virruso C et al. Association of Klotho polymorphisms with healthy aging: a systematic review and meta-analysis. Rejuvenation Res 2014;17:212–6. 10.1089/rej.2013.1523 [DOI] [PubMed] [Google Scholar]
- 95. Amin HA, Cordell HJ, Martin-Ruiz C et al. No evidence that genetic variation at the Klotho locus is associated with longevity in Caucasians from the Newcastle 85+ Study and the UK Biobank. J Gerontol A Biol Sci Med Sci 2022;77:457–61. 10.1093/gerona/glab361 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Semba RD, Cappola AR, Sun K et al. Plasma klotho and cardiovascular disease in adults. J Am Geriatr Soc 2011;59:1596–601. 10.1111/j.1532-5415.2011.03558.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Shardell M, Semba RD, Rosano C et al. Plasma klotho and cognitive decline in older adults: findings from the InCHIANTI study. J Gerontol A Biol Sci Med Sci 2016;71:677–82. 10.1093/gerona/glv140 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Semba RD, Cappola AR, Sun K et al. Relationship of low plasma klotho with poor grip strength in older community-dwelling adults: the InCHIANTI study. Eur J Appl Physiol 2012;112:1215–20. 10.1007/s00421-011-2072-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Xiao NM, Zhang YM, Zheng Q et al. Klotho is a serum factor related to human aging. Chin Med J (Engl) 2004;117:742–7. [PubMed] [Google Scholar]
- 100. Yamazaki Y, Imura A, Urakawa I et al. Establishment of sandwich ELISA for soluble alpha-klotho measurement: age-dependent change of soluble alpha-klotho levels in healthy subjects. Biochem Biophys Res Commun 2010;398:513–8. 10.1016/j.bbrc.2010.06.110 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Liu X, Ou YN, Ma YH et al. Renal function and neurodegenerative diseases: a two-sample Mendelian randomization study. Neurol Res 2023;45:456–64. 10.1080/01616412.2022.2158640 [DOI] [PubMed] [Google Scholar]
- 102. Drew DA, Weiner DE, Sarnak MJ. Cognitive impairment in CKD: pathophysiology, management, and prevention. Am J Kidney Dis 2019;74:782–90. 10.1053/j.ajkd.2019.05.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Hanson K, Fisher K, Hooper NM. Exploiting the neuroprotective effects of α-klotho to tackle ageing- and neurodegeneration-related cognitive dysfunction. Neuronal Signal 2021;5:NS20200101. 10.1042/NS20200101 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Semba RD, Moghekar AR, Hu J et al. Klotho in the cerebrospinal fluid of adults with and without Alzheimer's disease. Neurosci Lett 2014;558:37–40. 10.1016/j.neulet.2013.10.058 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Sanz B, Arrieta H, Rezola-Pardo C et al. Low serum klotho concentration is associated with worse cognition, psychological components of frailty, dependence, and falls in nursing home residents. Sci Rep 2021;11:9098. 10.1038/s41598-021-88455-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Chen CD, Sloane JA, Li H et al. The antiaging protein Klotho enhances oligodendrocyte maturation and myelination of the CNS. J Neurosci 2013;33:1927–39. 10.1523/JNEUROSCI.2080-12.2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Uchida A, Komiya Y, Tashiro T et al. Neurofilaments of Klotho, the mutant mouse prematurely displaying symptoms resembling human aging. J Neurosci Res 2001;64:364–70. 10.1002/jnr.1087 [DOI] [PubMed] [Google Scholar]
- 108. Sedighi M, Baluchnejadmojarad T, Afshin-Majd S et al. Anti-aging Klotho protects SH-SY5Y cells against amyloid β1-42 neurotoxicity: involvement of Wnt1/pCREB/Nrf2/HO-1 signaling. J Mol Neurosci 2021;71:19–27. 10.1007/s12031-020-01621-9 [DOI] [PubMed] [Google Scholar]
- 109. Chen CD, Li H, Liang J et al. The anti-aging and tumor suppressor protein Klotho enhances differentiation of a human oligodendrocytic hybrid cell line. J Mol Neurosci 2015;55:76–90. 10.1007/s12031-014-0336-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Li D, Jing D, Liu Z et al. Enhanced expression of secreted α-klotho in the hippocampus alters nesting behavior and memory formation in mice. Front Cell Neurosci 2019;13:133. 10.3389/fncel.2019.00133 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Leon J, Moreno AJ, Garay BI et al. Peripheral elevation of a Klotho fragment enhances brain function and resilience in young, aging, and α-synuclein transgenic mice. Cell Rep 2017;20:1360–71. 10.1016/j.celrep.2017.07.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Banerjee S, Zhao Y, Sarkar PS et al. Klotho ameliorates chemically induced endoplasmic reticulum (ER) stress signaling. Cell Physiol Biochem 2013;31:659–72. 10.1159/000350085 [DOI] [PubMed] [Google Scholar]
- 113. Zhou HJ, Zeng CY, Yang TT et al. Lentivirus-mediated klotho up-regulation improves aging-related memory deficits and oxidative stress in senescence-accelerated mouse prone-8 mice. Life Sci 2018;200:56–62. 10.1016/j.lfs.2018.03.027 [DOI] [PubMed] [Google Scholar]
- 114. Zhao Y, Zeng CY, Li XH et al. Klotho overexpression improves amyloid-β clearance and cognition in the APP/PS1 mouse model of Alzheimer's disease. Aging Cell 2020;19:e13239. 10.1111/acel.13239 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Dubal DB, Zhu L, Sanchez PE et al. Life extension factor klotho prevents mortality and enhances cognition in hAPP transgenic mice. J Neurosci 2015;35:2358–71. 10.1523/JNEUROSCI.5791-12.2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Park C, Hahn O, Gupta S et al. Platelet factors are induced by longevity factor klotho and enhance cognition in young and aging mice. Nat Aging 2023;3:1067–78. 10.1038/s43587-023-00468-0 [DOI] [PMC free article] [PubMed] [Google Scholar]