History and POIs

The Discovery of Klotho

The Discovery of the Anti-Aging Molecule

The world of anti-aging research is both a thrilling and daunting challenge. It has rapidly become a multibillion dollar industry in pursuit of a trillion dollar miracle molecule. This journey has seen some remarkable discoveries, one of which occurred more than 25 years ago at the hands of Makoto Kuro-O, M.D., Ph.D.

The initial aim of Kuro-o and his team was to understand the mechanisms of vascular calcification and atherosclerosis in mice. Their research involved the creation of a genetically modified mouse model. The focus was the Na+-K+-ATPase ion transporter, an enzyme located in the plasma membrane of all animal cells necessary for calcium/phosphate control and cell functioning.

Unexpectedly, the genetically modified mice started showing symptoms similar to human aging, such as motor disturbance, arteriosclerosis, infertility, skin atrophy, osteoporosis, emphysema, and many other features that closely resembled human aging. This led to further investigations, revealing that a neighboring gene had unintentionally been disrupted resulting in its deletion and an accelerated aging syndrome. It took two additional years of rigorous research to identify this gene and understand its biological function. This work resulted in the identification of a protein that's now ubiquitously referred to by scientist as the anti-aging protein: Klotho.

The paper, titled "Mutation of the mouse klotho gene leads to a syndrome resembling ageing, was published in the scientific journal Nature 26 years ago.

Makoto, K.O., Yutaka, M., Hiroki, A., Hiroshi, K., Tatsuo, S., Toshihiro, U., Yoshio, O., Masahiko, K., Tadashi, K., Eisuke, K. and Hitoshi, I., 1997. Mutation of the mouse klotho gene leads to a syndrome resembling ageing. Nature, 390(6655), pp.45-51.

Following the discovery of FGF23 deficient mice displaying an identical aging phenotype as Klotho deficient mice, the Klotho protein was functionally identified as a required co-receptor for Fibroblast Growth Factor-23 (FGF23) signaling.

Shimada T, Kakitani M, Yamazaki Y, Hasegawa H, Takeuchi Y, Fujita T, et al. Targeted ablation of Fgf23 demonstrates an essential physiological role of FGF23 in phosphate and vitamin D metabolism. J Clin Invest. (2004) 113:561–8. doi: 10.1172/JCI200419081

The Legacy of Klotho

They named the gene after the Greek mythological goddess Clotho. The head of Makoto Kuro-o’s previous lab (Dr. Yoichi Nabeshima) actually came up with the name, because it sounded like Kuro-o’s name and because of the mythology of aging it represented. Clotho is one of three sisters born to Zeus and Themis who collectively determined human fate. Clotho, the youngest of the sisters, spun the thread of human life and in so doing, determined an individual’s birth, death, and lifespan.  It is highly ironic that from 1995-1997, when this research first occurred, aging as a therapeutic, reversible target was still considered a completely unrealistic concept. This perception is in large part why developing Klotho as an anti-aging, therapeutic intervention has taken 26 years. 

The Klotho protein is a pivotal regulator of multiple physiological pathways, largely leveraging the network effect through reciprocal regulation and feedback mechanisms that influence the aging process. One key pathway involves its action as a co-receptor for Fibroblast Growth Factor 23 (FGF23), modulating phosphate and vitamin D metabolism. This bidirectional relationship has a vital role in maintaining mineral homeostasis in the body. Furthermore, Klotho inhibits Insulin and Insulin-like Growth Factor-1 (IGF-1) pathways, contributing to enhanced longevity by modulating glucose metabolism. In the renin-angiotensin system (RAS), Klotho suppresses angiotensin II actions, reducing oxidative stress, inflammation, and fibrosis. Conversely, RAS activation leads to Klotho downregulation, once again forming a feedback loop. It also regulates the Wnt signaling pathway, which controls cell proliferation and differentiation; in the absence of Klotho, Wnt signaling becomes overactive, leading to premature aging symptoms. Moreover, Klotho down regulates Transforming Growth Factor-beta (TGF-beta) signaling pathway, affecting tissue fibrosis and inflammation.

Klotho has also recientely demonstrated its influence on other factors such as the Telomerase, mTOR, and sirtuins, all of which play significant roles in aging.

In the context of hierarchical dominance in the aging process, Klotho can be seen as a master regulator. Its impact on multiple crucial pathways, its regulatory interactions in feedback loops, and its effects on critical biological processes such as oxidative stress, inflammation, and cellular senescence place Klotho in a dominant position in aging control.

Klotho operates as a critical signaling factor across numerous pathways, contributing to its powerful influence on aging and immunity:

[AMPK] [cAMP] [Epigenetic modulation] [FGF19] [FGF21] [FGF23] [HGH] [IGF1] [INF-ƴ] [Insulin] [MPAK] [mTOR] [NF-κß] [Nrf2] [p53/p21] [PGC-1α] [PKC] [PPAR-ƴ] [SIRT] [TGF-β] [VDRE] [Vit D] [Wnt]

This section taken from XO Re-GEN 10.1A

The anti-ageing protein Klotho acts as a hormone, and it is secreted into the blood from the kidneys, but its presence decreases with age leading to the vascular and arterial system to stiffen. In this study, salt-sensitive was confirmed to increase in aged mice and revealed that the cause was that the blood concentration of Klotho protein decreasing with age. To add to this the researchers clarified the molecular mechanism Wnt5a-RhoA pathway for the first time. Findings indicate that Klotho supplementation may prevent the development of hypertension, and levels of Klotho may be a predictive marker for the development of hypertension.

 

Factors That Increase Klotho Levels

Klotho is an antiaging protein that modulates oxidative stress, angiogenesis and fibrosis.[[i]] Klotho interacts with various membrane proteins such as receptors for transforming growth factor-β (TGF-β) and insulin-like growth factor (IGF). Klotho protein supplementation enhanced glomerular filtration rate, renal expression of superoxide dismutase, and klotho itself (P < 0.05). Klotho supplementation attenuated renal expressions of TGF-β and collagen type I and diminished renal abundance of Twist, phosphorylated Akt, and mammalian target of rapamycin (P < 0.05). Pathological examination revealed that klotho decreased the fibrosis index and nuclear staining of Smad in PKD kidneys (P < 0.05). Our data indicate that klotho protein supplementation ameliorates the renin-angiotensin system, reducing blood pressure in PKD mice. Furthermore, the present results implicate klotho supplementation in the suppression of Akt/mammalian target of rapamycin signaling, slowing cystic expansion. Finally, our findings suggest that klotho protein supplementation attenuated fibrosis at least partly by inhibiting epithelial mesenchymal transition in PKD. [[ii]]

 

  • Exercise – in both humans and mice. Klotho only increases post-exercise in people who completed a 16-week training program, with younger people having a bigger increase than older people. Based on mouse studies, it’s believed that muscle injury and new muscle cells secrete Klotho [7].

  • Vitamin D (Calcitriol) increases Klotho. However, Vitamin D supplements apparently don’t work in humans to increase Klotho, even when calcitriol goes up. In dialysis patients, vitamin D supplements actually decrease Klotho [23, 24, 25, 26].

  • Insulin increases klotho. This is one downside to a really low carb diet [27].

  • PPARgamma activators [12, 28].

  • Activated charcoal by binding to a toxin that decreases klotho (Indoxyl sulfate) [29].

  • Probiotics such as Acidophilus + L Lactis in aging mice [30].

  • Cordyceps reverses the Angiotensin II decrease in klotho [31].

  • Gentian root extract – might simply stabilize some part of the process after its production [32].

  • ACE inhibitors

  • Statins (HMG -CoA reductase inhibitors) – might simply stabilize some part of the process after its production [33, 34, 32].

  • [[iii] [iv] ]

Forward Signaling (Klotho Activation of Molecular Pathways):

  • (A) (Klotho) activates (B) (FGF23), which downregulates (C) (PI3K/Akt), affecting (D) (Insulin/IGF-1).

  • (A) (Klotho)  upregulates (E) Epigenetic mechanisms) that lead to the increase of (F) (Nrf2) and (G) (PPAR-γ).

  • (A) (Klotho) also upregulates  (H) (SIRT1) and (I) (AMPK) and modifies (J) (Ion Channels), downregulating (K) (MAPK/ERK).

  • (A) (Klotho)  inhibits (L) (mTOR), (M) (NF-Kb), and (N) (Wnt).

  • (A) (Klotho) , can increase the activity of (O) Antioxidant pathways, particularly through (F) (Nrf2), which can upregulate cellular (O) Antioxidant responses and decrease oxidative stress.

Klotho regulation is a complex interplay of numerous molecular pathways. First, Klotho (A) acts as a co-receptor for Fibroblast Growth Factor 23 (FGF23, B), enhancing its ability to bind to its receptor, FGFR. Activation of the FGF23 pathway leads to downregulation of Phosphatidylinositide 3-kinases (PI3K/Akt, C), a crucial component in the insulin and Insulin-like Growth Factor 1 (IGF-1, D) signaling pathways. This, in turn, leads to decreased insulin and IGF-1 activity, which can affect aging and longevity.

Furthermore, Klotho indirectly upregulates Epigenetic mechanisms (E), which then trigger an increase in Nuclear factor erythroid 2-related factor 2 (Nrf2, F), a key controller of the antioxidant response. Through Epigenetic regulation, Klotho also upregulates Peroxisome proliferator-activated receptor gamma (PPAR-γ, G), which further promotes antioxidant pathways and modulates energy metabolism.

The sirtuin 1 (SIRT1, H) and AMP-activated protein kinase (AMPK, I) pathways, known for their role in cellular energy metabolism and longevity, are also upregulated by Klotho. The interaction of Klotho with Ion Channels (J) modulates intracellular calcium levels, influencing several signaling pathways, including the Mitogen-Activated Protein Kinase/Extracellular Signal-Regulated Kinase (MAPK/ERK, K) pathway, which is consequently downregulated.

The activation of Klotho inhibits the mechanistic target of rapamycin (mTOR, L), a key nutrient-sensing pathway. This feeds into the PI3K/Akt pathway, forming a negative feedback loop. On the other hand, Klotho can inhibit the Nuclear Factor kappa B (NF-Kb, M) pathway, which plays a crucial role in inflammatory responses and aging.

Lastly, the Klotho protein also plays a role in Wnt signaling (N). In the presence of Klotho, the Wnt pathway is inhibited, contributing to the regulation of cellular growth and differentiation.

Thus, in summary, A (Klotho) activates B (FGF23), which downregulates C (PI3K/Akt), affecting D (Insulin/IGF-1). Simultaneously, A upregulates E (Epigenetic mechanisms) that lead to the increase of F (Nrf2) and G (PPAR-γ). A also upregulates H (SIRT1) and I (AMPK) and modifies J (Ion Channels), downregulating K (MAPK/ERK). Finally, A inhibits L (mTOR), M (NF-Kb), and N (Wnt).

Backward Signaling (Molecular Pathway Activation of Klotho): 

  • ••(A) (KLOTHO) 

  • ••(B) (FGF23) stimulates the production of (A) (Klotho).

  • Upregulation of ••(C) (PI3K/Akt), often via stimulation by (D) (Insulin/IGF-1), is associated with enhanced Klotho expression.

  • ••(E) (Epigenetic mechanisms) such as DNA methylation and histone modifications can alter the expression of the Klotho gene, leading to increased production of (A).

  • Activation of ••(F) (Nrf2) can increase Klotho gene transcription.

  • ••(G) (PPAR-γ) activation is associated with upregulation of Klotho.

  • Stimulation of ••(H) (SIRT1) or (I) (AMPK) can lead to an increase in Klotho production.

  • Alterations in ••(J) (Ion Channels) can also stimulate Klotho expression.

  • Downregulation of ••(K) (MAPK/ERK) can lead to upregulation of Klotho.

  • Inhibition of ••(L) (mTOR) or ••(M) (NF-Kb) has been linked with increased Klotho expression.

  • Inhibition of the ••(N (Wnt) signaling pathway can upregulate Klotho.

  • Activation of (O) Antioxidant pathways, often through ••(F) (Nrf2), particularly under conditions of oxidative stress, can lead to an increase in the expression and activity of (A) Klotho.

B (FGF23) is known to stimulate the production of A (Klotho), creating a positive feedback loop. Upregulation of C (PI3K/Akt) signaling, often via stimulation by D (Insulin/IGF-1), has been associated with enhanced Klotho expression, especially in the context of calorie restriction or exercise.

E (Epigenetic mechanisms) such as DNA methylation and histone modifications, can alter the expression of the Klotho gene, leading to increased production of A. Activation of F (Nrf2), particularly under oxidative stress conditions, can increase Klotho gene transcription. G (PPAR-γ) activation, often via anti-diabetic drugs, is associated with upregulation of Klotho.

Stimulation of H (SIRT1) or I (AMPK), particularly under conditions of cellular stress or energy deprivation, can lead to an increase in Klotho production. Alterations in J (Ion Channels), particularly those controlling calcium and potassium levels, can also stimulate Klotho expression, potentially via effects on cellular stress responses.

Downregulation of K (MAPK/ERK) can lead to upregulation of Klotho, potentially via effects on cellular growth and differentiation processes. Inhibition of L (mTOR) or M (NF-Kb), particularly under conditions of cellular stress, has been linked with increased Klotho expression, potentially via effects on cellular aging and inflammation.

Lastly, inhibition of the N (Wnt) signaling pathway can upregulate Klotho, as the Wnt pathway negatively regulates Klotho expression. Inhibition of Wnt signaling thereby leads to enhanced Klotho production.

Thus, to summarize, B (FGF23) and C (PI3K/Akt, stimulated by D [Insulin/IGF-1]) can enhance A (Klotho) production. E (Epigenetic mechanisms), F (Nrf2), and G (PPAR-γ) can increase A transcription. Activation of H (SIRT1) or I (AMPK), and modifications in J (Ion Channels) can lead to increased A expression. Downregulation of K (MAPK/ERK), or inhibition of L (mTOR) or M (NF-Kb), can upregulate A. Finally, inhibition of N (Wnt) can enhance A production.

As with the previous explanation, please note that this is a simplified overview of complex molecular interactions. Each of these systems is subject to various levels of regulation, feedback, and cross-talk with other signaling pathways.

Interventional Rational

These molecular pathway networks and  signaling cascades provide a grounded  rationale for effective augmentation of the systemic levels of Klotho supported by the literature. 

The nature of the Klotho/Molecular Pathways and Molecular Pathways/Klotho feedback loops provide a large number of targets of opportunity to increase Klotho gene expression and/or soluble Klotho levels utilizing readily available drugs, supplements and nutraceuticals. These molecular pathway networks and signaling cascades provide a grounded  rationale for effective augmentation of the systemic levels of Klotho. Each individual Target-Agent is supported by the literature for its ability to upregulate, up or downstream molecular pathways resulting in the direct or indirect up-regulation of the Klotho gene resulting in increased systemic levels of soluble or alpha-Klotho.

[i] Soluble Klotho, a biomarker and therapeutic strategy to reduce bronchopulmonary dysplasia and pulmonary hypertension in preterm infants

[ii] Klotho supplementation ameliorates blood pressure and renal function in DBA/2-pcy mice, a model of polycystic kidney disease

[iii] Klotho, the elusive kidney-derived anti-ageing factor 

[iv] Towards Age-Related Anti-Inflammatory Therapy: Klotho Suppresses Activation of ER and Golgi Stress Response in Senescent Monocytes

 Core KAP Agents



Identified Core Active-Agents

1) []  Curcumin [15, 102]: Curcumin is a natural compound found in turmeric, it has been shown to up-regulate Klotho Expression Directly. Curcumin demonstrates a secondary Klotho up-regulation component, potent Antioxidant and anti-inflammatory properties. Curcumin can also elicit DNA demethylation. Curcumin binds the VDR element resulting in an increase in Klotho levels. Curcumin has been shown to activate the Nrf2 pathway, leading to the expression of various antioxidant and cytoprotective genes.  Curcumin also down regulates TNF-α, resulting in an increase in Klotho levels. Oral bioavailability is low and can be increased by the addition of peperine and by lyphoization and/or micronization.  Curcumin is generally regarded as safe (GRAS). 

[2017] A Novel Highly Bioavailable Curcumin Formulation Improves Symptoms and Diagnostic Indicators in Rheumatoid Arthritis Patients:
A Randomized, Double-Blind, Placebo-Controlled, Two-Dose, Three-Arm, and Parallel-Group Study

[2018] Upregulation of klotho and erythropoietin contributes to the neuroprotection induced by curcumin-loaded nanoparticles in experimental model of chronic epilepsy

[2016] Curcumin attenuates cyclosporine A‑induced renal fibrosis by inhibiting hypermethylation of the klotho promoter

[2021] Epigenetic and non-epigenetic regulation of Klotho in kidney disease

Click [√] to Enlarge

2) []  Resveratrol [14, 102] : This compound, found in red wine and grapes, can upregulate Klotho Expression Directly. Resveratrol and Curcumin produce a synergistic result because both compounds are known to activate the SIRT1, Nrf2, and PPARγ pathways, and both also have DNA demethylating activity that directly influence Klotho expression. Resveratrol is a potent Antioxidant. Each of these pathways are known to be signaling activators of Klotho.

[] [2019] Resveratrol and 1,25-dihydroxyvitamin D co-administration protects the heart against d-galactose-induced aging in rats- evaluation of serum and cardiac levels of klotho

[2014] Resveratrol increases anti-aging Klotho gene expression via the activating transcription factor 3/c-Jun complex-mediated signaling pathway

[2021] Resveratrol (RV)- A pharmacological review and call for further research. (Image Source)

3) Quercetin [15, 14] : This flavonoid has been shown to have DNA demethylating effets.  This flavonoid, found in a variety of fruits and vegetables, has been shown to activate the Nrf2 pathway. Quercetin is a potent Direct Scavenging Antioxidant.


4) []  EGCG [13, 9] : (Epigallocatechin-3-gallate), a polyphenol found in green tea, can also be beneficial due to its activation of the Nrf2 pathway and DNA demethylating properties, which can lead to a synergistic increase in Klotho expression. This is the main polyphenol present in green tea, and it's known to activate the Nrf2 pathway, leading to enhanced antioxidant defenses.  EGCG is a Direct Scavenging Antioxidant.


5) Berberine [13, 0] : An alkaloid found in several plants, berberine is traditionally used in Chinese medicine. It has been reported to activate AMPK, leading to upregulation of SIRT1. This plant alkaloid, often used for its antimicrobial, anti-inflammatory, and blood-glucose-lowering effects, also stimulates Nrf2 activity.  Berberine is a Direct Scavenging Antioxidant.


Genistein [13, 5] Genistein is a naturally occurring isoflavone found predominantly in soy-based products. As a phytoestrogen, it has the ability to bind to estrogen receptors, and it has been implicated in various cellular pathways. One pathway of significant interest is the PI3K/Akt signaling pathway, which Genistein can modulate, leading to effects on cell growth, apoptosis, and angiogenesis.

Regarding Klotho, an anti-aging protein, Genistein has been reported to upregulate Klotho expression. The mechanism by which this occurs may involve the suppression of oxidative stress, thus enhancing Klotho gene expression. Additionally, through its effects on the Wnt signaling pathway, Genistein can suppress Wnt activity, which in turn may lead to increased Klotho expression.


6) []  Vitamin D [14, 21] : is known to directly up-regulate the Klotho gene and increase systemic levels of soluble or α-Klotho.  The active form of Vitamin D (calcitriol) has been shown to upregulate Klotho expression. Vitamin D forms a complex with Klotho and FGF23 to regulate phosphate metabolism. Vitamin D binds to the vitamin D receptor complex (VDR), and this complex interacts with specific DNA sequences known as vitamin D response elements (VDREs) present in the promoter region of the Klotho gene, stimulating its transcription and increasing Klotho production. Vitamin D enhances the expression of FGF23, which in turn forms a complex with membrane Klotho in the kidneys. In our study, vitamin D supplementation caused higher levels of klotho compared to control group which possibly meant that vitamin D increased mRNA levels due to transcriptional activation of klotho. The FGF23/Klotho complex activates downstream signaling pathways that also enhance the expression of Klotho.

[2020] Effect of vitamin D supplementation on klotho protein, antioxidant status and nitric oxide in the elderly- A randomized, double-blinded, placebocontrolled clinical trial


7) Alpha-lipoic acid (ALA) [15, 0] : ALA, a fatty acid found in every cell of the body, is a potent antioxidant that can also stimulate Nrf2 activity.


8) Oleanolic Acid (OA): A molecule found in olive oil, OA is reported to have potent Nrf2-activating properties.


8) Vitamin C [9, 23] can be integral components of this regimen. Vitamin C, a potent antioxidant, can enhance the activity of the Nrf2 pathway and contribute to overall cellular health and longevity, supporting Klotho expression. Vitamin C contributes to the active demethylation of DNA by acting as a cofactor for enzymes that belong to the family of Ten-Eleven Translocation (TET) dioxygenases. TET enzymes catalyze the conversion of 5-methylcytosine (5mC), a methylated form of the DNA base cytosine, to 5-hydroxymethylcytosine (5hmC), effectively "demethylating" the DNA. This conversion is a key step in active DNA demethylation.


9) Omega-3 [11, 34] : fatty acids and 5) Berberine [15, 0] are included for their demonstrated ability to stimulate both the Nrf2 and PPARγ pathways. Nrf2 plays a vital role in defending against oxidative stress, a key factor in aging and age-related diseases, while PPARγ has been linked to improved Klotho expression. Berberine is also known to activate the AMPK pathway which in turn can activate SIRT1 and PPARγ increasing Klotho expression. 


10) Magnesium [7, 7] : Magnesium is essential for many biological processes and has been associated with positive effects on longevity and the aging process. It plays a vital role in DNA replication, repair, and RNA synthesis, affecting rate limiting reactions that effect Klotho expression. 


11) Zinc [8, 0] : Zinc is known for its antioxidant properties and involvement in the immune response, DNA synthesis, and cell division, which can contribute to healthy aging and affect Klotho expression. Zinc plays an important part in the protein-protein interactions that are crucial in agent-target interactions. Zinc plays an important role in the binding kinetics and multiple conformational changes required for effective signals transduction.

Vitamin B12


Manganise


Each of these compounds and agents act on specific, but cross talking pathways. Some are directing or redirecting feedback loops. Combined they provide a comprehensive and synergistic approach to upregulating Klotho expression. Collectively these agent reduce oxidative stress, activate key cellular signaling pathways, and influence the epigenetic landscape, and when combined contribute to an optimal biological environment that should facilitate increased Klotho levels. 

Provided below are the four main pathways targeted by this strategy. Each is followed by the most potent agents that are able to upregulate each of the respective pathways.

Click [√] to Enlarge

Composite dietary antioxidant index (CDAI) is a measure of individual antioxidant profile based on a combination of 6 dietary antioxidants, including manganese, selenium, zinc, and vitamins A, C, and E. Individually only one of these (Vitamin A) antioxidants demonstrated a stistcically significant increase in Klotho levels. CDAI collectively achieves significance as a Klotho increasing interventional opportunity. See table above right.

[2023] Composite Dietary Antioxidant Index and Plasma Levels of Soluble Klotho- Insights from NHANES

Vitamin A (CDAI):

Curcumin: As a sirtuin activator, it may indirectly support mechanisms associated with klotho.  Potential synergies: Quercetin, Omega-3 fatty acids.

Resveratrol: As a sirtuin activator, it may indirectly support mechanisms associated with klotho. Potential synergies: Quercetin, Omega-3 fatty acids and Vitamin E.

Omega-3 fatty acids: EPA and DHA have anti-inflammatory properties and could work in conjunction with other compounds to enhance klotho-related effects. Potential synergies: Resveratrol, Vitamin E.

Vitamin D: Klotho is co-expressed with the vitamin D receptor in the kidney, and they have a reciprocal relationship. Potential synergies: Vitamin E, Omega-3 fatty acids.

Quercetin: It has shown potential in increasing klotho expression in certain studies.  Potential synergies: Resveratrol, Curcumin.

Green Tea Polyphenols (especially EGCG): EGCG has demonstrated potential in promoting klotho expression. Potential synergies: Curcumin, Resveratrol.

Alpha-lipoic acid: This antioxidant supports mitochondrial function, which can indirectly support klotho-related pathways. Potential synergies: Omega-3 fatty acids, Vitamin E.

Astaxanthin: A potent antioxidant that could help with cellular protection in conjunction with klotho enhancement. Potential synergies: Omega-3 (especially since astaxanthin is often sourced from algae, similarly to some forms of omega-3), Vitamin E.

Sulforaphane: Known to activate the Nrf2 pathway, which plays a role in antioxidant defenses. This might complement klotho-associated benefits. Potential synergies: Curcumin, Resveratrol.

Vitamin E (CDAI) : As a lipid-soluble antioxidant, it can protect cellular membranes, potentially complementing klotho's protective effects. Potential synergies: Resveratrol, Omega-3 fatty acids, Vitamin D. Vitamin E is also a TNF-α inhibitor. VitE enhanced the expression of klotho and subsequently impaired the protein expression of LPS-induced IL12p70,

Vitamin C (CDAI) :

Manganese (CDAI) :

Selenium (CDAI) :

Zinc (CDAI) :

SIRT1

Resveratrol: Perhaps the most well-known activator of SIRT1, resveratrol is a naturally occurring polyphenol found in grapes and red wine. It has been shown to activate SIRT1, leading to various beneficial effects like improved mitochondrial function and longevity in various model organisms.

Metformin: This is a first-line medication for the treatment of type 2 diabetes. It has been shown to activate the AMPK pathway, which can lead to the activation of SIRT1.

NAD+ Boosters (Nicotinamide Riboside and Nicotinamide Mononucleotide): SIRT1 is a NAD+-dependent enzyme, so increasing the cellular levels of NAD+ can enhance SIRT1 activity. Nicotinamide riboside (NR) and nicotinamide mononucleotide (NMN) are two supplements that can increase cellular NAD+ levels.

Quercetin: This flavonoid, found in a variety of fruits and vegetables, has been shown to upregulate SIRT1.

Berberine: An alkaloid found in several plants, berberine is traditionally used in Chinese medicine. It has been reported to activate AMPK, leading to upregulation of SIRT1.

Curcumin: Curcumin, found in the spice turmeric, is reported to upregulate SIRT1. It's known for its antioxidant and anti-inflammatory properties.

Fasting and Caloric Restriction: Fasting and caloric restriction have been shown to increase the expression of SIRT1, leading to various beneficial effects on health and longevity.

Nfr2

Sulforaphane: Found in cruciferous vegetables like broccoli, sulforaphane is one of the most potent activators of the Nrf2 pathway.

Curcumin: This polyphenol, found in turmeric, has been shown to activate the Nrf2 pathway, leading to the expression of various antioxidant and cytoprotective genes.

Resveratrol: Apart from activating SIRT1, resveratrol also activates the Nrf2 pathway, adding to its multitude of health benefits.

Quercetin: This flavonoid, found in a variety of fruits and vegetables, has been shown to activate the Nrf2 pathway.

Epigallocatechin-3-gallate (EGCG): This is the main polyphenol present in green tea, and it's known to activate the Nrf2 pathway, leading to enhanced antioxidant defenses.

Alpha-lipoic acid (ALA): ALA, a fatty acid found in every cell of the body, is a potent antioxidant that can also stimulate Nrf2 activity.

Oleanolic Acid (OA): A molecule found in olive oil, OA is reported to have potent Nrf2-activating properties.

Berberine: This plant alkaloid, often used for its antimicrobial, anti-inflammatory, and blood-glucose-lowering effects, also stimulates Nrf2 activity.

Spirulina: This blue-green algae has been shown to activate Nrf2, leading to the induction of antioxidant enzymes.

Epigenetics

Decitabine (5-Aza-2'-deoxycytidine) and Azacitidine (5-Azacytidine): These are FDA-approved DNA methyltransferase inhibitors that cause demethylation and reactivation of silenced genes.

Epigallocatechin-3-gallate (EGCG): This compound found in green tea can inhibit DNA methyltransferase activity and thus lead to DNA demethylation.

Genistein: This isoflavone, abundant in soy and other legumes, is known for its weak DNA methyltransferase inhibitory effect leading to demethylation and gene activation.

Resveratrol: Apart from its antioxidant properties, resveratrol also possesses DNA demethylating effects.

Curcumin: Known for its anti-inflammatory and antioxidant properties, curcumin can also cause DNA demethylation.

Quercetin: This flavonoid has been shown to have DNA demethylating effects.

Parthenolide: This sesquiterpene lactone, found in the medicinal plant feverfew, can cause DNA demethylation.

Vitamin C (Ascorbic Acid): Vitamin C enhances the activity of enzymes involved in active DNA demethylation, such as the Ten-eleven Translocation (TET) enzymes.

[2013] Ascorbate Induces Ten-Eleven Translocation (Tet) Methylcytosine Dioxygenase-mediated Generation of 5-Hydroxymethylcytosine

[2019] Reprogramming the Epigenome With Vitamin C

[2021] Ascorbic Acid in Epigenetic Reprogramming

[2023] Vitamin C- From nutrition to oxygen sensing and epigenetics

Synergistic

Resveratrol: This compound is known to activate SIRT1, Nrf2, and PPARγ pathways and has a DNA demethylating effect, which might impact Klotho expression. By acting on multiple pathways, resveratrol could potentially exert a synergistic effect on Klotho upregulation.

Curcumin: Curcumin is known to activate Nrf2 and PPARγ and has a DNA demethylating effect. This combination of actions might lead to a synergistic effect on Klotho expression.

Omega-3 fatty acids: These fatty acids have been shown to activate Nrf2 and PPARγ pathways, potentially leading to synergistic effects on Klotho expression.

Berberine: This compound activates PPARγ and has been reported to upregulate SIRT1, possibly contributing to a synergistic effect on Klotho upregulation.

EGCG (Epigallocatechin-3-gallate): EGCG can activate Nrf2 and has DNA demethylating properties, which might result in a synergistic upregulation of Klotho.

Indirect Clinical or Laboratory Markers Indicating an Increase in Klotho:

1 Reduction in phosphate levels

2 Increase in soluble Klotho levels in urine

[2013] Klotho, phosphate and FGF‑23 in ageing and disturbed mineral metabolism

The tissue-specific expression of Klotho also explains why the kidney and parathyroid gland are FGF-23 target organs despite the fact that many other tissues express FGFRs (Figure 1).

  1. Reduced FGF23 Levels: Lower levels may indicate increased Klotho activity since Klotho serves as a co-receptor for FGF23. This would be especially pertinent in the context of mineral metabolism.

  2. Improved Renal Function: Markers such as decreased serum creatinine and increased glomerular filtration rate (GFR) could indicate enhanced Klotho activity.

  3. Calcium/Phosphorus Homeostasis: Decreased serum phosphorus and increased serum calcium could indicate Klotho-mediated regulation of mineral ion homeostasis.

  4. Reduced Markers of Oxidative Stress: Lower levels of reactive oxygen species (ROS) or increases in antioxidant enzymes like superoxide dismutase (SOD) and glutathione peroxidase.

  5. Insulin Sensitivity: Improved glucose tolerance and lower fasting insulin levels could indirectly indicate increased Klotho levels.

  6. Anti-Inflammatory Markers: Reduced levels of pro-inflammatory cytokines like TNF-α and IL-6.

  7. Improved Lipid Profile: Lower levels of LDL cholesterol and increased HDL cholesterol could be indicative.

  8. Reduced Blood Pressure: Klotho has been shown to have vasculoprotective effects; thus, a decrease in blood pressure could be an indirect marker.

  9. Improvement in Cognitive Tests: Given the neuroprotective role of Klotho, cognitive performance could serve as an indirect but functionally relevant marker.

  10. Decrease in Cellular Senescence Markers: Reduction in markers like p16, p21, and p53 could be indicative of increased Klotho activity.

  11. Improved Bone Density: Considering Klotho's role in phosphate metabolism, improvements in bone density scans could also serve as an indirect marker.

  12. Hormone Levels: Alterations in hormones like IGF-1, which have inverse relationships with Klotho, could also be looked at as indirect markers.

  13. Grip strength