PreprintPDF Available

Metadichol ; An agonist that expresses anti-aging gene Klotho in various cell lines

Authors:

Abstract

Klotho is a well-known tumor suppressor hormone that exhibits anti-cancer and anti-aging properties. Klotho levels are low or non-existent in cancer patients. Klotho protein levels decrease with aging; maintaining consistent levels may prevent disease and promote healthier aging. Metadichol is a nano emulsion of long-chain alcohols C26, C28, and C30, of which C-28 constitutes over 85%. Any small molecule that can elevate Klotho can, in principle, help reverse many diseases in which Klotho levels are low. Previously, we showed that treatment of the pancreatic cancer cell lines PANC1, MIA-PACA, and COLO-205, combined with Metadichol, a lipid emulsion consisting of long-chain alcohols at 1-100 pg/mL concentrations, resulted in a 4- to 10-fold increase in Klotho expression as determined by qRT-PCR, This study aimed to demonstrate that Metadichol promotes Klotho expression in a wide variety of cell lines, such as primary cancer, stem, and somatic cell lines. Cells were treated with various concentrations of Metadichol ranging from 1 pg to 1 µg. Three to fifteen fold increase in Klotho expression was observed compared with baseline, as measured by qRT-PCR and qualified by western blot analysis. Metadichol is a natural agonist of Klotho expression and is non-toxic at levels up to 5000 mg/kg in rats. and has a potential therapeutic role in cancer and reversing aging.
Page 1/12
Metadichol ; An agonist that expresses anti-aging
gene Klotho in various cell lines
Palayakotai R Raghavan ( raghavan@nanorxinc.com )
nanorx inc
Research Article
Keywords: Klotho, Metadichol, anti-aging, anti-tumor, primary cancer cells, broblasts
Posted Date: February 28th, 2023
DOI: https://doi.org/10.21203/rs.3.rs-2635049/v1
License: This work is licensed under a Creative Commons Attribution 4.0 International License. 
Read Full License
Metadichol promotes the expression of the anti-aging gene Klotho in various cell lines
Palayakotai R. Raghavan
Nanorx Inc., PO Box 131, Chappaqua, NY 10514, USA
Email: raghavan@nanorxinc.com
Abstract
Klotho is a well-known tumor suppressor hormone that exhibits anti-cancer and anti-aging
properties. Klotho levels are low or non-existent in cancer patients. Klotho protein levels
decrease with aging; maintaining consistent levels may prevent disease and promote healthier
aging. Metadichol is a nano emulsion of long-chain alcohols C26, C28, and C30, of which C-28
constitutes over 85%. Any small molecule that can elevate Klotho can, in principle, help reverse
many diseases in which Klotho levels are low. . Previously, we showed that treatment of the
pancreatic cancer cell lines PANC1, MIA-PACA, and COLO-205, combined with Metadichol, a
lipid emulsion consisting of long-chain alcohols at 1-100 pg/mL concentrations, resulted in a 4-
to 10-fold increase in Klotho expression as determined by qRT-PCR, This study aimed to
demonstrate that Metadichol promotes Klotho expression in a wide variety of cell lines, such as
primary cancer, stem, and somatic cell lines. Cells were treated with various concentrations of
Metadichol ranging from 1 pg to 1 µg. Three to fifteen fold increase in Klotho expression was
observed compared with baseline, as measured by qRT-PCR and qualified by western blot
analysis. Metadichol is a natural agonist of Klotho expression and is non-toxic at levels up to
5000 mg/kg in rats. and has a potential therapeutic role in cancer and reversing aging.
Keywords: Klotho, Metadichol, anti-aging, anti-tumor, primary cancer cells, fibroblasts
Introduction
In Greek methodology, Klotho was known as the goddess of lifespan regulation [1]. Following
its initial discovery in the kidneys in 1997 [2], Klotho has been shown to block tumor growth and
metastasis, modulate chemotherapeutic drug resistance and improve overall survival [3]. The
injection of Klotho into animals decreased breast cancer growth regarding tumor size, weight,
and visual appearance and resulted in no significant side effects. Low levels of Klotho are
associated with cancer and many other diseases [4-6], as shown in Table 1. An increasing number
of studies [4] have focused on the use of Klotho in extending longevity and counteracting the
effects of aging on physical function. Recently, significant progress has been made in identifying
factors (7) contributing to the aging process, which has contributed to developing therapeutic
strategies to prevent, delay, or reverse the age-related decline. Many studies have attempted to
induce Klotho gene expression, emphasizing its potential benefit when injected into animals.
Clinical and preclinical studies have demonstrated that Klotho is an essential anti-aging molecule
[8] that affects lifespan, health, and cognitive function [9]. Laboratory animals that do not
express Klotho exhibit a shorter lifespan and cognitive impairment. In contrast, mice that
overexpress Klotho have a longer lifespan and enhanced cognition and memory [10]. Klotho
significantly inhibits the growth of lung cancer [11], pancreatic carcinoma [12], colorectal
carcinoma [13], breast cancer [14], hepatocellular carcinoma [15], ovarian carcinoma [16],
melanoma [17], diffuse large B cell lymphoma [18].
Several molecules, such as PPARγ agonists [19], testosterone [20], and resveratrol [21], either
directly promote Klotho over expression in vitro or inhibit Klotho down regulation in vivo. Jung
et al reported a novel molecular mechanism by which a small molecule [N-(2-chlorophenyl)-1H-
indole-3-caboxamid] induces Klotho expression [22]. Other studies [23] have used CRISPR
methodology to upregulate Klotho transcription and production in two different cell lines, one of
which was a neuron-like cell line.
Pharmaceutical companies have been developing Klotho agonists that up regulate Klotho
expression are of significant interest for treating diseases. For example, Klotho Therapeutics has
developed a patent-pending treatment based on Klotho that affects aging [24]. Klogenix has been
working on a two-pronged approach to target endogenous Klotho's natural production and
regulation and deliver Klotho genetic material directly to patient cells, thus enabling them to
produce the protein [25].
Previously, we showed that treatment of the pancreatic cancer cell lines PANC1, MIA-PACA,
and COLO-205, combined with Metadichol, a lipid emulsion consisting of long-chain alcohols at
1-100 pg/mL concentrations, resulted in a 4- to 15-fold increase in Klotho expression as
determined by qRT-PCR [26] . In the present study, we have extended our original work to
determine the effects of Metadichol in many other cell lines, including primary cancer cell lines,
stem cell lines, and fibroblasts. (Figure1)
Methods
Experimental procedures
This work was carried out by a service provider: Skanda Life Sciences, Bangalore, India.
Chemicals and reagents
The A549, Colo-205, PANC1, MDAMB31, HeLA, HepG2, and human cardiac fibroblast cells
were purchased from the ATCC (USA). Primary breast cancer cells were obtained from BIOIVT
(Detroit, Michigan, USA). Primary antibodies were purchased from ABclonal (Woburn,
Massachusetts, USA) and E-lab Science (Maryland, USA). Primers were obtained from
SahaGene, Hyderabad, India (Table 2). All other molecular biology reagents were purchased
from Sigma Aldrich.
Cell line maintenance and seeding
The cells were cultured in a suitable medium with or without supplements in the presence of 1%
antibiotics in a humidified atmosphere of 5% CO2 at 37°C. The medium was changed
periodically until the cells reached confluency. Cell survival was determined using a
hemocytometer. When the cells reached 70–80% confluency, single-cell suspensions were
seeded into 6-well plates at a density of 106 cells per well and incubated for 24 h at 37°C in 5%
CO2. Afterward, we rinsed the cell monolayer with a serum-free medium and added Metadichol
at predefined concentrations.
Cell treatment
Metadichol was prepared at various concentrations (1 pg/mL, 100 pg/mL, one ng/mL, and 100
ng/mL) in serum-free media, and the mixture was added to predesignated wells. The control cells
received media without drugs. The cells were incubated, then gently rinsed with sterile
phosphate-buffered saline (PBS) solution. Quantitative RT-PCR (q RT-PCR) and western blot
analysis were done as described below.
RNA isolation
RNA was isolated from each treatment group using TRIzol reagent (Invitrogen). Cells (106) were
collected into 1.5-mL microcentrifuge tubes and centrifuged at 5,000 rpm for 5 min at 4°C. Then,
650 µL of TRIzol was added to the pellet, mixed, and incubated on ice for 20 min. Subsequently,
we added 300 µL of chloroform, mixed the samples with gentle inversion for 1–2 min, and set
them on ice for 10 min. The samples were centrifuged at 12,000 rpm for 15 min at 4°C. The
upper aqueous layer was transferred to a new sterile 1.5 mL centrifuge tube, and an equal amount
of pre-chilled isopropanol was added. The samples were incubated at 20°C for 60 min, then
centrifuged at 12,000 rpm for 15 min at 4°C. The supernatant was carefully removed, and the
RNA pellet was washed with 1.0 mL of 100% ethanol and 700 µL of 70% ethanol with
centrifugation as previously described. The RNA pellet was air-dried at room temperature for
approximately 15–20 min and resuspended in 30 µL of DEPC-treated water. RNA concentration
was measured using a Spectradrop (Spectramax i3x, USA) spectrophotometer (Molecular
Devices).
cDNA synthesis
Complementary DNA (cDNA) was synthesized from 2 µg of total RNA using the Prime Script
cDNA synthesis kit (Takara) with oligo dT primers following the manufacturer’s instructions.
The reaction volume was 20 µL, and cDNA synthesis was performed at 50°C for 30 min, then
incubating at 85°C for 5 min using an Applied Biosystems instrument (Veritii). The resulting
cDNA was used as a template for qPCR.
Primers and qPCR
A final reaction volume of 20 µL of PCR mixture was prepared, consisting of 1 µL of cDNA, 10
µL of SYBR green Master Mix, and one µM complementary forward and reverse gene-specific
primers. The samples were run under the following conditions: initial denaturation at 95°C for 5
min followed by 30 cycles of secondary denaturation at 95°C for 30 seconds, annealing at the
optimized temperature for 30 seconds, and extension at 72°C for 1 min. We defined the number
of cycles that allowed amplification in the exponential range without reaching a plateau as the
optimal number of cycles. The results were obtained using CFX Maestro software. We calculated
fold-change using the comparative CT method and determined the relative expression of each
target gene relative to a housekeeping gene (β-actin) and untreated control cells. ΔCT was
calculated for each treatment using the following formulas: ΔCt = Ct (target gene)–Ct (reference
gene), ΔΔ Ct = ΔCt (treatment group)–ΔCt (control group). The fold-change was calculated for
target gene expression for each treatment using the formula: Fold change = 2^ (−ΔΔCt).
Protein isolation and western blot analysis
Total protein was isolated from 106 cells using radio immuno precipitation assay buffer
supplemented with the protease inhibitor phenylmethyl sulfonyl fluoride. The cells were lysed
for 30 min at four °C with gentle inversion, centrifuged at 10,000 rpm for 15 min, and the
supernatant was transferred to a new sterile tube. The Bradford method ( BIO-RAD USA) was
used to measure the protein concentration. Protein (25 µg) was mixed with 1X sample loading
dye containing SDS and loaded onto a polyacrylamide gel. The proteins were separated under
denaturing conditions using a Tris-glycine running buffer. The proteins were transferred to
PVDF membranes (Invitrogen) using a Turbo transblot system (Bio-Rad, USA), blocked with
5% BSA for one h, incubated with the respective primary antibody overnight at four °C,
followed by a species-specific secondary antibody for one h at RT. After washing, the
membranes were incubated with ECL substrate (Merck) for 1 min in the dark. The images at
suitable exposure settings were captured using the ChemiDoc XRS system (Bio–Rad, USA).
Results and Discussion
The highest Klotho expression was observed in MDAMB-231 ( Triple-Negative Breast Cancer
cells developed at MD Anderson Institute Houston) , followed by stem cells, then cardiac
fibroblasts (NHCFs) ( Figure 1) . Although the expression of Klotho induced by small molecules
is elevated in kidney cells [27], we found that Metadichol increased Klotho expression in many
different cell types and thus may represent a universal Klotho agonist. Because Klotho exhibits
anticancer activity, its level of expression in cancer cells is significant. Down regulation of
Klotho has been observed in several cancers, such as pancreatic cancer, HCC, and others
(28-29).. Epigenetic modulation, such as promoter methylation and histone deacetylation, also
contributes to the dysregulation of Klotho in cancer.
Forester et al. [30-31] suggested that the liganded vitamin D receptor (VDR) upregulates Klotho
via vitamin D response elements (VDRE). Metadichol is a VDR inverse/protein agonist [32-33].
Among the consequences of the enhanced expression of Klotho is an increase in telomerase
activity. Similarly, Metadichol can up regulate telomerase [34] and thus potentially prevent stem
cell aging [35].
Consequently, the down regulation of Klotho enhances proliferation and reduces apoptosis in
cancer cells. Conversely, the over expression of Klotho results in cancer cell inhibition [36-38].
Similarly, our results suggest that Metadichol could be useful in inducing apoptosis in cancer
cells by increasing Klotho expression and this also has benefits in other diseases. The current
study and previously published results (39-42) suggest that the observed results could have been
been due to increased Klotho expression (43). The potential therapeutic utility of Metadichol in
elevating Klotho levels warrants further study in vitro and in vivo.
Abbreviated Cell Lines Description
PANC1: human pancreatic cancer cell line
isolated from a pancreatic carcinoma of
ductal cell origin
HCAF-PPCC: (Human Cancer associated
Fibroblast primary prostate cancer cell.
MIA-PACA: Human pancreatic cancer cell
line
HCAF-TNBCC; Human Cancer associated
Fibroblasts-Triple Negative Breast Cancer
Panel
COLO-205: Cell line is made up of epithelial
cells isolated from ascitic fluid derived from a
70-year-old, male with colon cancer
A-549: Adenocarcinomic human alveolar
basal epithelial cell
HEPG2 : human liver cancer cell line
HELA; Immortalized cell line used in
scientific research. It is the oldest and most
commonly used human cell line. The line is
derived from cervical cancer cells
hESC BGO1V; cells are pluripotent and can
differentiate to representatives of the three
primary germ layers.
MDAMB 231; isolated at M D Anderson
Institute, Houston, USA from a pleural
effusion of a patient with invasive ductal
carcinoma) is commonly used to model late-
stage breast cancer
Author Contributions
All work was planned and supervised by the author (PPR), who is solely responsible for its
content.
Acknowledgments; We thank Dr Michelle Muller of Sferlap S.A Switzerland for helpful
discussion.
Conflicts of Interest; None
Funding; Nanorx, Inc. R&D Budget provided funding.
Competing Interests: None
Availability of data and material
All data are in the manuscript and the supplementary material provided.
Supplementary material : Western Blot data and Klotho Diseases Network
References
1. Michael Lichtenauer, et al. Uncoupling fate: Klotho—Goddess of fate and regulator of life and
ageing. 2020; Australasian Journal of aging. vol 39,2, 161-163. https://doi.org/10.1111/
ajag.12772
2. Kuro-o M, et al. Mutation of the mouse klotho gene leads to a syndrome resembling aging.
Nature. 1997; 390(6655):45–51.
3. Ligumsky H, ,et al 2022. The role of α-klotho in human cancer: molecular and clinical
aspects. Oncogene. 2022; 41(40):4487–97. https://doi.org/10.1038/s41388-022-02440-5 .
4. Kuro-o M. 2019. The Klotho proteins in health and disease. Nature Reviews Nephrology.
15(1):27-44.
5. Buchanan S, et al. 2020. Klotho, aging, and the failing kidney. Frontiers in endocrinology.
11:560.
6. Chen K, et al, 2021.Klotho deficiency causes heart aging via impairing the Nrf2-GR pathway.
Circulation research, 128(4):492-507.
7. Feder A, et al, 2019. The biology of human resilience: opportunities for enhancing resilience
across the life span. Biol psychiatry, 86(6):443-53.
8. Prud’homme GJ, et al, 2022. Pathobiology of the klotho anti-aging protein and therapeutic
considerations. Front Aging, 3:931331. https://doi.org/10.3389/fragi.2022.931331
9. Abraham CR, et al, 2016. Klotho is a neuroprotective and cognition-enhancing protein. Vitam
Horm. 2016; 101:215–38. https://doi.org/10.1016/bs.vh.2016.02.004.
10.Kurosu H,, Kuro-o M et al. 2005. Suppression of aging in mice by the hormone klotho.
Science. 2005; 309(5742):1829–33. https://doi.org/10.1126/science.1112766.
11.Doi S, Kuro-O M et al, 2011. Klotho inhibits transforming growth factor-β1 (TGF-1)
signaling and suppresses renal fibrosis and cancer metastasis in mice. J Biol Chem 2011; 286
(10):8655–65. https://doi.org/10.1074/jbc.M110.174037.
12. Abramovitz L, teal 2011. . KL1 internal repeat mediates klotho tumor suppressor activities
and inhibits bFGF and IGF-I signaling in pancreatic cancer. Clin Cancer Res. 17(13):4254–66.
https://doi.org/10.1074/jbc.M110.174037
13.Li XX, et al 2014. . Klotho suppresses the growth and invasion of colon cancer cells through
inhibition of the IGF1R-mediated PI3K/AKT pathway. Int J Oncol 45(2):611–8.
14. Ligumsky et al 2015. . Tumor suppressor activity of klotho in breast cancer is revealed by
structure-function analysis. Mol Cancer Res.13(10): 1398–1407. https://doi.org/
10.1158/1541-7786.MCR-15-0141
15.Tang X, et al 2016. Klotho: a tumor suppressor and modulator of the Wnt/ β-catenin pathway
in human hepatocellular carcinoma. Lab Invest. 96(2):197–205. https://doi.org/10.1038/
labinvest.2015.86.
16.Yan Y, et al 2017. . Reduced Klotho expression contributes to poor survival rates in human
patients with ovarian cancer, and over expression of Klotho inhibits the progression of ovarian
cancer partly via the inhibition of systemic inflammation in nude mice. Mol Med Rep.
15(4):1777–85. https://doi.org/10.3892/mmr.2017.6172.
17.Behera R, et al 2017. . Inhibition of age-related therapy resistance in melanoma by
rosiglitazone-mediated induction of klotho. Clin Cancer Res. 2017; 23(12):3181–90. https://
doi.org/10.1158/1078-0432.CCR-17-0201.
18.Zhou X, et al 2017. Klotho, an anti-aging gene, is a tumor suppressor and inhibitor of
IGF-1R signaling in diffuse large B cell lymphoma. J Hematol Oncol. 10(1):37. https://
doi.org/10.1186/s13045-017-0391-5.
19. Zhang H, et al 2008. . Klotho is a target gene of PPAR–γ. Kidney Int. 74(6):732–9. https://
doi.org/10.1038/ki.2008.244.
20. Hsu SC, et al 2014. Testosterone increases renal anti-aging klotho gene expression via the
androgen receptor-mediated pathway. Biochem J, 464(2):221–9. https://doi.org/10.1042/
BJ20140739.
21.Hsu SC, et al 2014. Resveratrol increases anti-aging klotho gene expression via the activating
transcription factor 3/c-Jun complex-mediated signaling pathway. Int J Biochem Cell Biol.
53:361–71. https://doi.org/10.1016/j.biocel.2014.06.002.
22. Jung D, et al 2018. Induction of anti-aging klotho with a small chemical compound that
demethylates CpG islands. Oncotarget, 8(29):46745–55.
23. Chen CD, et al 2018. . Activation of the anti-aging and cognition-enhancing gene klotho by
CRISPR-dCas9 transcriptional effector complex. J Mol Neurosci. 64(2):175–84. https://
doi.org/10.1007/s12031-017-1011-0.
24. www.klotho.com.
25. www.klogenix.com.
26.Raghavan PR. 2018. Metadichol® a novel agonist of the anti-aging klotho gene in cancer cell
lines. J Cancer Sci Ther.10(11):351–7. https://doi.org/10.4172/1948-5956.1000567.
27.Zhou X, and Wang X, 2015. Klotho: a novel biomarker for cancer. J Cancer Res Clin Oncol.
141(6):961–9. https://doi.org/10.1007/s00432-014-1788-y.
28.Li Y, et al 2020. Overexpression of klotho suppresses growth and pulmonary metastasis of
osteosarcoma in vivo. Genet Mol Biol. 43(2):e20190229. doi: 10.1590/1678-4685-
GMB-2019-0229
29.King GD, et al 2012. Identification of novel small molecules that elevate klotho expression.
Biochem J. 441(1):453–61. https://doi.org/10.1042/BJ20101909.
30.Forster RE, et al., 2011. . Vitamin D receptor controls the expression of the anti-aging klotho
gene in mouse and human renal cells. Biochem Biophys Res Commun. 414(3):557–62. https://
doi.org/10.1016/j.bbrc.2011.09.117.
31.Haussler MR, et al, 2016. 1,25-Dihydroxyvitamin D and klotho: a tale of two renal hormones
coming of age. Vitam Horm. 2016; 100:165–230. https://doi.org/10.1016/bs.vh.2015.11.005
32.Neubig RR. 2007. Missing links: mechanisms of protean agonism, Mol Pharmacol.
71(5):1200–2. https://doi.org/10.1124/mol.107.034926.
33.P.R. Raghavan 2015, Metadichol ® Liquid and gel nanopaarticle formulations. US patent
9,006.292.
34.Raghavan PR. 2019. The quest for immortality: introducing Metadichol®, a novel telomerase
activator. Stem Cell Res Ther. 2019; 9(446):2. https://doi.org/10.4172/2157- 7633.1000446
35..Ullah M and Sun Z. Klotho deficiency accelerates stem cell aging by impairing telomerase
activity. J Gerontol A Biol Sci Med Sci.74(9):1396–407. https://doi.org/10.1093/gerona/
gly261.
36.Zhou X, Wang X. 2015. Klotho: a novel biomarker for cancer. J Cancer Res Clin Oncol.
141(6):961-9.
37.Bo Chen, et al (2012). Inhibition of lung cancer cells growth, motility and induction of
apoptosis by Klotho, a novel secreted Wnt antagonist, in a dose-dependent manner, Cancer
Biology & Therapy, 13:12, 1221-1228, DOI: 10.4161/cbt.21420
38.Ligumsky H, et al 2022. The role of α-klotho in human cancer: Molecular and clinical
aspects. Oncogene, 41(40):4487–97. https://doi.org/10.1038/s41388-022-02440-5.
39.Raghavan PR. 2017. Metadichol, a novel ROR gamma inverse agonist, and its applications in
psoriasis. J Clin Exp Dermatol Res, 8:433. https://doi.org/10.4172/2155-9554.1000433.
40.Raghavan PR. 2017. Metadichol® and vitamin C increase in vivo, an open-label study. Vitam
Miner, 6(16):3. http://dx.doi.org/10.4172/2376-1318.1000169
41.Raghavan PR. 2017. Rheumatoid arthritis and osteoporosis: a case study. J Arthritis,
6(3):1000240. https://doi.org/10.4172/2167-7921.1000240.
42.Raghavan PR. 2017. Systolic and diastolic BP control in metabolic syndrome patients with
Metadichol®, a novel nanoemulsion lipid. J Cardiol Cardiovasc Ther. 5(2): 555660. https://
doi.org/10.19080/JOCCT.2017.05.555660.
43.Kuro-O M. 2019. The Klotho proteins in health and disease. Nat Rev Nephrol.
Jan;15(1):27-44. doi: 10.1038/s41581-018-0078-3.
Tables
Table 2. Primers used for qPCR
Table 1. List of diseases associated with low levels of Klotho
Acetylcholine and Nitric Oxide Dysregulation
Aging (highly accelerated)
All-Cause Mortality
Anemia
Anorexia
Atherosclerosis (as well as calcification of the
arteries)
Bone Loss (such as osteoporosis and low bone mass)
Cancers
Bone
Brain
Breast
Colon
Stomach
Kidney
Liver
Gene
Primer pair
Sequence
Tm
P ro d u c t s i z e
(base pairs)
βActin
FP
TCCTCCTGAGCGCAAGTACTCT
62.1
153
RP
GCTCAGTAACAGTCCGCCTAGAA
62.4
KLOTHO
FP
GGGAGGTCAGGTGTCCATTG
55.88
152
RP
TGCTCTCGGGATAGTCACCA
53.83
Figure 1. Klotho fold expression changes in various cell lines
KLOTHO-WESTERN BLOT DATA IN HESC BG01V AND
PANC1,MIAPACA AND COLO205 CELL LINES
hESC BG01V cell line culture and treatment
!Cell line:
!hESC BG01V (ATCC® SCRC-2002™), Homo sapiens, embryonic stem cells
!Cell Culture:
!hESC BG01V cells were cultured in 1:1 Mixture of Dulbecco's Modified Eagles Medium and Ham's F-12 medium containing 1.2g/L sodium bicarbonate,
2.5mM L-glutamine, 15mM HEPES and 0.5mM sodium pyruvate, supplemented with 2mM L-Alanyl-L-Glutamine, 0.1mM Non-essential amino acids,
0.1mM 2-mercaptoethanol; 4ng/ml bFGF (80%), Knockout serum replacement (5%), fetal bovine serum (15%), 100units/ml penicillin G, and 100µg/ml
streptomycin at 37°C, 5% CO2 incubator. hESC BG01V cells were treated with various concentrations of test sample Metadichol (1pg/ml, 100pg/ml, 1ng/
ml, and 100ng/ml incubated for 24hrs. Post incubation, the cells were harvested for isolation of protein using RIPA buffer (Sigma; R0-278)
Isolation of protein using RIPA buffer
1. The cells, post harvesting, were washed twice using 1XPBS.
2. The cell pellets were gently suspended in 500µl of RIPA buffer with 1X Protease Inhibitor (Sigma; P-8340)
3. The cells were incubated for 30mins by gentle mixing every 5mins.
4. Post incubation, the cells were centrifuged at 10,000rpm for 12-15 minutes.
5. The protein lysates in the supernatant was transferred to fresh sterile tubes and stored in -20˚C until further use.
SDS-PAGE and Western Blot procedure
1. 100ug protein sample from each cell lysate was mixed with 5X loading dye and heated for 6 min at 980C.
2. Protein samples were loaded and separated on 12% SDS-PAGE gel using Mini protean Tetra cell (Bio-Rad).
3. Methanol activated 0.45uM PVDF membrane was pre-wet in transfer buffer for 10 min @ RT.
4. Protein transfer was done for 10 min in Turbo Transblot (Bio-Rad) apparatus.
5. Blot was blocked in 5% BSA + TBST for 1 hr @RT.
6. Blot was incubated with 10 Ab @ dilution: 1: 1000 for overnight @ 40C on shaker.
7. Washed 3 times with TBST for 5min @ RT
8. Blot was incubated with 20 Ab (Anti-Rabbit HRP- IgG) @ dilution 1: 1000 for 1hr @ RT.
9. Washed 3 times with TBST for 5min @ RT
10. Blot was rinsed with ECL reagent (two component system) for 1 min in dark and image was captured with 40 sec
exposure in Chemidoc MP imaging system (Bio-Rad).
Weste rn Blo t f or the ex pre ssi on of G APD H, KLO THO in hE SC BG 01V
cells.
Metadicho
l
Band!Intensity!Of!
Protein
Normalised!
Protein
expression
Relative!Protein!!
Expression
GAPDH
KLOTHO
Control
32916.56
15318.803
0.47
1.00
1pg!
28754.782
16261.953
0.57
1.22
100pg!
27463.368
14343.711
0.52
1.12
1ng
22421.296
6319.468
0.28
0.61
100ng!
28068.631
4156.054
0.15
0.32
KLOTHO Protein expression in hESC BG01Vtreated with
sample
Fold regulation
0.00
0.33
0.65
0.98
1.30
Treament
Control
1pg
100pg
1ng
100ng
0.3182
0.6056
1.1223
1.2152
1
36KDa!
65!KDa
Western Blot of Klotho in Panc-1 cells
Panc 1
Klotho expression
Metadichol (Conc.)
Lane
Relative Gene
Expression
Control
1
1.00
1µg/ml
2
0.65
1ng/ml
3
0.58
1pg/ml
4
1.23
β-actin
Klotho
Western Blot of Klotho in Panc 1 cells
Expression of Klotho Expression of β-Actin
Weste rn blo t d ata on an aly sis of ex pre ssi on of Kl oth o p rot ei n
Panc 1
Metadichol
(Conc.)
Band Intensity Proteins
Normalised
Relative Gene
Expression
β-actin
Klotho
Control
18858.602
18953.894
1.01
1.00
1µg/ml
16428.522
10724.974
0.65
0.65
1ng/ml
16059.894
9369.045
0.58
0.58
1pg/ml
22344.338
27602.016
1.24
1.23
Klotho Protein Expression in Panc 1 cells treated with Metadichol
Western Blot of Klotho in Panc-1 cells
β-actin
Klotho
Colo 205
Klotho expression
Metadichol (Conc.)
Lane
Relative Gene
Expression
Control
1
1.00
1µg/ml
2
0.93
1ng/ml
3
0.87
1pg/ml
4
0.48
Western Blot of Klotho in Colo 205 cells
Expression of Klotho Expression of β-Actin
Weste rn blo t d ata on an aly sis of ex pre ssi on of Kl oth o p rot ei n
Colo 205
Metadichol
(Conc.)
Band Intensity Proteins
Normalised
Relative Gene
Expression
β-actin
Klotho
Control
25292.128
26710.68
1.06
1.00
1µg/ml
20753.811
20309.42
0.98
0.93
1ng/ml
18488.711
16999.69
0.92
0.87
1pg/ml
32729.957
16477.58
0.50
0.48
Klotho Protein Expression in Colo 205 cells treated with Metadichol
Western Blot of Klotho in Panc-1 cells
Panc 1
Klotho expression
Metadichol (Conc.)
Lane
Relative Gene
Expression
Control
1
1.00
1µg/ml
2
0.65
1ng/ml
3
0.58
1pg/ml
4
1.23
β-actin
Klotho
Western Blot of Klotho in Panc 1 cells
Expression of Klotho Expression of β-Actin
Weste rn blo t d ata on an aly sis of ex pre ssi on of Kl oth o p rot ei n
Panc 1
Metadichol
(Conc.)
Band Intensity Proteins
Normalised
Relative Gene
Expression
β-actin
Klotho
Control
18858.602
18953.894
1.01
1.00
1µg/ml
16428.522
10724.974
0.65
0.65
1ng/ml
16059.894
9369.045
0.58
0.58
1pg/ml
22344.338
27602.016
1.24
1.23
Klotho Protein Expression in Panc 1 cells treated with Metadichol
SEARCH TUTORIAL ABOUT CONTACT US DOWNLOAD
Disease-Connect.org
Gene KL
7
>
Gene View: The network shows the diseases associated with KL based on the GWAS/OMIM/DEG records (P < 1e-8).
Legend Label Layout P-value Edge Option Export Table View Hierarchical View
Neoplasms, Complex and Mixed
P-value < 1.0e-12 Node: 40
(<0
,- I••
+ J
Edge: 339
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
The α-Klotho protein (henceforth denoted Klotho) has antiaging properties, as first observed in mice homozygous for a hypomorphic Klotho gene (kl/kl). These mice have a shortened lifespan, stunted growth, renal disease, hyperphosphatemia, hypercalcemia, vascular calcification, cardiac hypertrophy, hypertension, pulmonary disease, cognitive impairment, multi-organ atrophy and fibrosis. Overexpression of Klotho has opposite effects, extending lifespan. In humans, Klotho levels decline with age, chronic kidney disease, diabetes, Alzheimer’s disease and other conditions. Low Klotho levels correlate with an increase in the death rate from all causes. Klotho acts either as an obligate coreceptor for fibroblast growth factor 23 (FGF23), or as a soluble pleiotropic endocrine hormone (s-Klotho). It is mainly produced in the kidneys, but also in the brain, pancreas and other tissues. On renal tubular-cell membranes, it associates with FGF receptors to bind FGF23. Produced in bones, FGF23 regulates renal excretion of phosphate (phosphaturic effect) and vitamin D metabolism. Lack of Klotho or FGF23 results in hyperphosphatemia and hypervitaminosis D. With age, human renal function often deteriorates, lowering Klotho levels. This appears to promote age-related pathology. Remarkably, Klotho inhibits four pathways that have been linked to aging in various ways: Transforming growth factor β (TGF-β), insulin-like growth factor 1 (IGF-1), Wnt and NF-κB. These can induce cellular senescence, apoptosis, inflammation, immune dysfunction, fibrosis and neoplasia. Furthermore, Klotho increases cell-protective antioxidant enzymes through Nrf2 and FoxO. In accord, preclinical Klotho therapy ameliorated renal, cardiovascular, diabetes-related and neurodegenerative diseases, as well as cancer. s-Klotho protein injection was effective, but requires further investigation. Several drugs enhance circulating Klotho levels, and some cross the blood-brain barrier to potentially act in the brain. In clinical trials, increased Klotho was noted with renin-angiotensin system inhibitors (losartan, valsartan), a statin (fluvastatin), mTOR inhibitors (rapamycin, everolimus), vitamin D and pentoxifylline. In preclinical work, antidiabetic drugs (metformin, GLP-1-based, GABA, PPAR-γ agonists) also enhanced Klotho. Several traditional medicines and/or nutraceuticals increased Klotho in rodents, including astaxanthin, curcumin, ginseng, ligustilide and resveratrol. Notably, exercise and sport activity increased Klotho. This review addresses molecular, physiological and therapeutic aspects of Klotho.
Article
Full-text available
Klotho has been recognized as a gene involved in the aging process in mammals for over 30 years, where it regulates phosphate homeostasis and the activity of members of the fibroblast growth factor (FGF) family. The α-Klotho protein is the receptor for Fibroblast Growth Factor-23 (FGF23), regulating phosphate homeostasis and vitamin D metabolism. Phosphate toxicity is a hallmark of mammalian aging and correlates with diminution of Klotho levels with increasing age. As such, modulation of Klotho activity is an attractive target for therapeutic intervention in the diseasome of aging; in particular for chronic kidney disease (CKD), where Klotho has been implicated directly in the pathophysiology. A range of senotherapeutic strategies have been developed to directly or indirectly influence Klotho expression, with varying degrees of success. These include administration of exogenous Klotho, synthetic and natural Klotho agonists and indirect approaches, via modulation of the foodome and the gut microbiota. All these approaches have significant potential to mitigate loss of physiological function and resilience accompanying old age and to improve outcomes within the diseasome of aging.
Article
Full-text available
Klotho is originally discovered as an anti-aging gene and knock-out of klotho accelerates aging in mice. Subsequent studies support the anti-carcinogenesis role of klotho in a variety of human malignancies. The present study investigated the role of klotho on growth and metastasis of osteosarcoma cells. The osteosarcoma cells were transduced with lentivirus particles encoding klotho or scramble control. The reconstructed osteosarcoma cells were injected into the femoral medullary cavity of nude mice to establish a xenograft animal model. The anti-tumor properties of klotho were evaluated in terms of tumor growth, apoptosis, glycogen production, and pulmonary metastasis. Lentivirus-mediated overexpression of klotho significantly decreased tumor volume and weight in osteosarcoma mice. Determination of PCNA and Ki67 expression revealed that overexpression of klotho inhibited cell proliferation in tumor tissues obtained from osteosarcoma xenografts. PAS staining also showed that overexpression of klotho significantly decreased the production of glycogen in osteosarcoma. Moreover, TUNEL positive cells were significantly increased after lentivirus-mediated overexpression of klotho. Furthermore, lentivirus-mediated upregulation of klotho reduced the number of pulmonary metastatic lesions in mice compared to control mice. These findings demonstrated that elevated klotho could inhibit osteosarcoma cell growth and pulmonary metastasis in vivo, suggesting that klotho may be a valuable therapeutic target for osteosarcoma.
Article
Full-text available
Recent scientific and technological advances have brought us closer to being able to apply a true biopsychosocial approach to the study of resilience in humans. Decades of research have identified a range of psychosocial protective factors in the face of stress and trauma. Progress in resilience research is now advancing our understanding of the biology underlying these protective factors at multiple phenotypic levels, including stress response systems, neural circuitry function, and immune responses, in interaction with genetic factors. It is becoming clear that resilience involves active and unique biological processes that buffer the organism against the impact of stress, not simply involve a reversal of pathological mechanisms. Here, we provide an overview of recent progress in the field, highlighting key psychosocial milestones and accompanying biological changes during development, and into adulthood and old age. Continued advances in our understanding of psychological, social, and biological determinants of resilience will contribute to the development of novel interventions and help optimize the type and timing of intervention for those most at risk, resulting in a possible new framework for enhancing resilience across the life span.
Article
Full-text available
Klotho is an anti-aging protein that is mostly secreted by the kidneys, the brain, and the thyroid. It plays a significant role in regulating kidney function and vascular health. Klotho gene is named after "the Spinner" (Clotho from Greek mythology), the goddess who spins the thread of life. Klotho is a transmembrane protein known to be a co-receptor for Fibroblast Growth Factor-23. Klotho gene is expressed in a variety of tissues changes in the levels are associated with many diseases. Klotho is a tumor suppressor in breast cancer and its expression is reduced in human pancreatic adenocarcinoma, and treatment with klotho inhibits the growth of pancreatic cancer cells in vitro and in vivo. Growing evidence suggests that an increase in KL expression may be beneficial for age-related diseases such as arteriosclerosis and diabetes. It remains a challenge today to induce Klotho expression. Herein we show that treating pancreatic cancer cells PANC1, MIAPACA and COLO-205 with Metadichol® a novel food based lipid emulsion of long chain alcohols at picogram/ml, concentration led to a 4-10 fold increase in Klotho expression as seen quantitative RT-PCR. These results suggest the use of Metadichol® given its constituents that are present in foods we consume every day is a novel therapeutic intervention for pancreatic cancer and other diseases.
Article
Full-text available
Multiple lines of evidence show that the anti-aging and cognition-enhancing protein Klotho fosters neuronal survival, increases the anti-oxidative stress defense, and promotes remyelination of demyelinated axons. Thus, upregulation of the Klotho gene can potentially alleviate the symptoms and/or prevent the progression of age-associated neurodegenerative diseases such as Alzheimer’s disease and demyelinating diseases such as multiple sclerosis. Here we used a CRISPR-dCas9 complex to investigate single-guide RNA (sgRNA) targeting the Klotho promoter region for efficient transcriptional activation of the Klotho gene. We tested the sgRNAs within the − 1 to − 300 bp of the Klotho promoter region and identified two sgRNAs that can effectively enhance Klotho gene transcription. We examined the transcriptional activation of the Klotho gene using three different systems: a Firefly luciferase (FLuc) and NanoLuc luciferase (NLuc) coincidence reporter system, a NLuc knock-in in Klotho 3′-UTR using CRISPR genomic editing, and two human cell lines: neuronal SY5Y cells and kidney HK-2 cells that express Klotho endogenously. The two sgRNAs enhanced Klotho expression at both the gene and protein levels. Our results show the feasibility of gene therapy for targeting Klotho using CRISPR technology. Enhancing Klotho levels has a therapeutic potential for increasing cognition and treating age-associated neurodegenerative, demyelinating and other diseases, such as chronic kidney disease and cancer.
Article
Klotho is a well-established longevity hormone. Its most prominent function is the regulation of phosphate homeostasis. However, klotho possesses multiple pleiotropic activities, including inhibition of major signaling pathways, reducing oxidative stress and suppressing inflammation. These activities are tightly associated with cancer, and klotho was discovered as a universal tumor suppressor. We review here novel molecular aspects of klotho activity in cancer, focusing on its structure–function relationships and clinical aspects regarding its expression, blood levels, clinical risk, and prognostic value in the clinical setting. In addition, the potential benefit of klotho treatment combined with chemotherapy, biological therapy, or immunotherapy, are discussed. Finally, as klotho was shown in preclinical models to inhibit cancer development and growth, we discuss various approaches to developing klotho-based therapies.
Article
Rationale: Cardiac aging is an important contributing factor for heart failure which affects a large population but remains poorly understood. Objective: The purpose of this study is to investigate whether Klotho plays a role in cardiac aging. Methods and Results: Heart function declined in old mice (24 months), as evidenced by decreases in fractional shortening, ejection fraction, and cardiac output. Heart size and weight, cardiomyocyte size and cardiac fibrosis were increased in old mice, indicating that aging causes cardiac hypertrophy and remodeling. Circulating Klotho levels were dramatically decreased in old mice, which prompted us to investigate whether the Klotho decline may cause heart aging. We found that Klotho gene mutation (KL-/-) largely decreased serum klotho levels and impaired heart function. Interestingly, supplement of exogenous secreted Klotho prevented heart failure, hypertrophy, and remodeling in both old mice and KL (-/-) mice. Secreted Klotho treatment inhibited excessive cardiac oxidative stress, senescence and apoptosis in old mice and KL (-/-) mice. Serum phosphate levels in KL (-/-) mice were kept in the normal range, suggesting that Klotho deficiency-induced heart aging is independent of phosphate metabolism. Mechanistically, Klotho deficiency suppressed glutathione reductase (GR) expression and activity in the heart via inhibition of transcription factor Nrf2. Furthermore, cardiac-specific overexpression of GR prevented excessive oxidative stress, apoptosis, and heart failure in both old and KL (-/-) mice. Conclusions: Klotho deficiency causes cardiac aging via impairing the Nrf2-GR pathway. Supplement of exogenous secreted Klotho represents a promising therapeutic strategy for aging-associated cardiomyopathy and heart failure.
Article
The Klotho proteins, αKlotho and βKlotho, are essential components of endocrine fibroblast growth factor (FGF) receptor complexes, as they are required for the high-affinity binding of FGF19, FGF21 and FGF23 to their cognate FGF receptors (FGFRs). Collectively, these proteins form a unique endocrine system that governs multiple metabolic processes in mammals. FGF19 is a satiety hormone that is secreted from the intestine on ingestion of food and binds the βKlotho–FGFR4 complex in hepatocytes to promote metabolic responses to feeding. By contrast, under fasting conditions, the liver secretes the starvation hormone FGF21, which induces metabolic responses to fasting and stress responses through the activation of the hypothalamus–pituitary–adrenal axis and the sympathetic nervous system following binding to the βKlotho–FGFR1c complex in adipocytes and the suprachiasmatic nucleus, respectively. Finally, FGF23 is secreted by osteocytes in response to phosphate intake and binds to αKlotho–FGFR complexes, which are expressed most abundantly in renal tubules, to regulate mineral metabolism. Growing evidence suggests that the FGF–Klotho endocrine system also has a crucial role in the pathophysiology of ageing-related disorders, including diabetes, cancer, arteriosclerosis and chronic kidney disease. Therefore, targeting the FGF–Klotho endocrine axes might have therapeutic benefit in multiple systems; investigation of the crystal structures of FGF–Klotho–FGFR complexes is paving the way for the development of drugs that can regulate these axes.