From Wikipedia, the free encyclopedia
KL
Identifiers
Aliases KL, entrez:9365, klotho, HFTC3, KLA
External IDs OMIM: 604824 MGI: 1101771 HomoloGene: 68415 GeneCards: KL
Orthologs
SpeciesHumanMouse
Entrez
Ensembl
UniProt
RefSeq (mRNA)

NM_004795
NM_153683

NM_013823

RefSeq (protein)

NP_004786

NP_038851

Location (UCSC) Chr 13: 33.02 – 33.07 Mb Chr 5: 150.88 – 150.92 Mb
PubMed search [3] [4]
Wikidata
View/Edit Human View/Edit Mouse

Klotho is an enzyme that in humans is encoded by the KL gene. [5] The three subfamilies of klotho are α-klotho, β-klotho, and γ-klotho. [6] α-klotho activates FGF23, and β-klotho activates FGF19 and FGF21. [7] When the subfamily is not specified, the word "klotho" typically refers to the α-klotho subfamily, because α-klotho was discovered before the other members. [8] [7]

α-klotho is highly expressed in the brain, liver and kidney. [9] β-klotho is predominantly expressed in the liver. [10] [9] γ-klotho is expressed in the skin. [9]

Klotho can exist in a membrane-bound form or a (hormonal) soluble, circulating form. [11] Proteases can convert the membrane-bound form into the circulating form. [12]

The KL gene encodes a type-I single-pass transmembrane protein [7] that is related to β-glucuronidases. Reduced production of this protein has been observed in patients with chronic kidney failure (CKF), and this may be one of the factors underlying degenerative processes (e.g., arteriosclerosis, osteoporosis, and skin atrophy) seen in CKF. Mutations within the family have been associated with ageing, bone loss and alcohol consumption. [13] [14] Transgenic mice that overexpress Klotho live longer than wild-type mice. [15]

Structure

The α-klotho gene is located on chromosome 13, and is translated into a single-pass integral membrane protein. [9] The intracellular portion of the α-klotho protein is short (11 amino acids), whereas the extracellular portion is long (980 amino acids). [9] The transmembrane portion is also comparatively short (21 amino acids). [9] The extracellular portion contains two repeat sequences, termed the KL1 (about 450 amino acids) and KL2 (about 430 amino acids) domains. [9] [7] In the kidney and the choroid plexus of the brain, the transmembrane protein can be proteolytically cleaved to produce a 130- Kilo- Dalton, soluble form of α-klotho protein, released into the circulation and cerebrospinal fluid, respectively. [9] In humans, the secreted form of klotho is more dominant than the membrane form. [16]

The β-Klotho gene is located on chromosome 4. The protein shares homology (43.1% identity and 60.1% similarity) with α-klotho. [17] It should not be confused with the alpha-cut and beta-cut of alpha-klotho, which releases KL1+KL2 and KL2 domain, respectively.

Function

Klotho is a transmembrane protein that, in addition to other effects, provides some control over the sensitivity of the organism to insulin and appears to be involved in ageing. Its discovery was documented in 1997 by Makoto Kuro-o et al. [18] The name of the gene comes from Klotho or Clotho, one of the Moirai, or Fates, in Greek mythology, who spins the thread of human life. [7]

The klotho protein is a novel β-glucuronidase ( EC number 3.2.1.31) capable of hydrolyzing steroid β-glucuronides. Genetic variants in KLOTHO have been associated with human aging, [19] and klotho protein has been shown to be a circulating factor detectable in serum that declines with age. [20]

The binding of the endocrine fibroblast growth factors (FGF's, viz., FGF19 and FGF21) to their fibroblast growth factor receptors, is promoted via their interactions as co-receptors with β-klotho. [21] [22] [16] [7] Loss of β-Klotho abolishes all effects of FGF21. [23]

α-klotho, which binds to the endocrine FGF FGF23 changes cellular calcium homeostasis, by both increasing the expression and activity of TRPV5 (decreasing phosphate reabsorption in the kidney) and decreasing that of TRPC6 (decreasing phosphate absorption from the intestine). [24] α-klotho increases kidney calcium reabsorption by stabilizing TPRV5. [25] About 95% to 98% of Ca2+ filtered from the blood by the kidney is normally reabsorbed by the kidney's renal tubule, which is mediated by TRPV5. [26]

Clinical significance

α-klotho can suppress oxidative stress and inflammation, thereby reducing endothelial dysfunction and atherosclerosis. [8] Blood plasma α-klotho is increased by aerobic exercise, thereby reducing endothelial dysfunction. [27]

β-klotho activation of FGF21 protein has a protective effect on heart muscle cells. [28] Obesity is characterized by FGF21 resistance, believed to be caused by the inhibition of β-klotho by the inflammatory cell signalling protein ( cytokine) tumor necrosis factor alpha, [28] but there is evidence against this mechanism. [16]

Klotho is required for oligodendrocyte maturation, myelin integrity, and can protect neurons from toxic effects. [29] Mice deficient in klotho have a reduced number of synapses and cognitive deficits, whereas mice overexpressing klotho have enhanced learning and memory. [30] Research with injections of klotho in primates demonstrates a positive effect on memory that lasts for as long as two weeks. [31]

Reduced klotho expression is seen in the lung macrophages of smokers. [32] An abnormal form of autophagy associated with reduced expression of klotho is linked to the pathogenesis of chronic obstructive pulmonary disease. [32] (Although normal autophagy helps maintain muscle, excessive autophagy causes loss of muscle mass. [32])

It has been found that the decreased klotho expression may be due to DNA hypermethylation, which may have been induced by the overexpression of DNMT3a. [33] Klotho may be a reliable gene for early detection of methylation changes in oral tissues, and can be used as a target for therapeutic modification in oral cancer during the early stages.

Klotho-deficient mice manifest a syndrome resembling accelerated human ageing and display extensive and accelerated arteriosclerosis. Additionally, they exhibit impaired endothelium dependent vasodilation and impaired angiogenesis, suggesting that klotho protein may protect the cardiovascular system through endothelium-derived nitric oxide production. [16]

Klotho could play a protective role in Alzheimer's disease patients. [34] [35]

Research with injections of α-klotho in primates suggests a positive effect on memory that could have implications for research with humans. [31] Interestingly the cognitive effects in rhesus monkeys were observed even with subcutaneous injection despite previous results showing that klotho protein fails to cross the blood–brain barrier. [36]

Effects on aging

Reduced α-klotho or FGF23 can result in impaired phosphate excretion from the kidney, leading to hyperphosphatemia. [7] In mice, this leads to a phenotype characteristic of premature aging, which can be mitigated by feeding the mice a low phosphate diet. [7]

The plasma (soluble) form of α-klotho is most easily measured, and has been shown to decrease after 40 years of age in humans. [37] Lower plasma levels of α-klotho in older adults is associated with increased frailty and all-cause mortality. [37] Physical activity has been shown to increase plasma α-klotho. [37]

Mice lacking either fibroblast growth factor 23 or the α-klotho enzyme display premature aging due to hyperphosphatemia. [24] Many of these symptoms can be alleviated by feeding the mice a low phosphate diet. [7]

Although the majority of research has explored klotho's absence, it was demonstrated that klotho over-expression in mice extended their average life span between 19% and 31% compared to normal mice. [15] In addition, variations in the Klotho gene (SNP Rs9536314) are associated with both life extension and increased cognition in human populations and mice, but only if the gene expression was heterozygous, not homozygous. [38] [9] The cognitive benefits of α-klotho are primarily seen late in life. [9]

Klotho increases membrane expression of the inward rectifier ATP-dependent potassium channel ROMK. [24] Klotho-deficient mice show increased production of vitamin D, and altered mineral-ion homeostasis is suggested to cause premature aging‑like phenotypes, because reduced vitamin D activity from dietary restriction reverses the premature aging‑like phenotypes and prolongs survival in these mutants. These results suggest that aging‑like phenotypes were due to klotho-associated vitamin D metabolic abnormalities (hypervitaminosis). [39] [40] [41] [42]

Klotho is an antagonist of the Wnt signaling pathway, and chronic Wnt stimulation can lead to stem cell depletion and aging. [43] Klotho inhibition of Wnt signaling can inhibit cancer. [32] The anti-aging effects of klotho are also a consequence of increased resistance to inflammation and oxidative stress. [16]

Extracellular vesicles (EV) in young mice carried more copies of klotho-producing mRNA than those from old mice. Transfusing young EVs into older mice helped rebuild their muscles. [44]

The presence of senescent cells decreases α-klotho levels. Senolytic drugs reduce the level of these cells, allowing α-klotho levels to increase. [45]

References

  1. ^ a b c GRCh38: Ensembl release 89: ENSG00000133116Ensembl, May 2017
  2. ^ a b c GRCm38: Ensembl release 89: ENSMUSG00000058488Ensembl, May 2017
  3. ^ "Human PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  4. ^ "Mouse PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  5. ^ Matsumura Y, Aizawa H, Shiraki-Iida T, Nagai R, Kuro-o M, Nabeshima Y (January 1998). "Identification of the human klotho gene and its two transcripts encoding membrane and secreted klotho protein". Biochemical and Biophysical Research Communications. 242 (3): 626–630. doi: 10.1006/bbrc.1997.8019. PMID  9464267.
  6. ^ Dolegowska K, Marchelek-Mysliwiec M, Nowosiad-Magda M, Slawinski M, Dolegowska B (June 2019). "FGF19 subfamily members: FGF19 and FGF21". Journal of Physiology and Biochemistry. 75 (2): 229–240. doi: 10.1007/s13105-019-00675-7. PMC  6611749. PMID  30927227.
  7. ^ a b c d e f g h i Kuro-O M (January 2019). "The Klotho proteins in health and disease". Nature Reviews. Nephrology. 15 (1): 27–44. doi: 10.1038/s41581-018-0078-3. PMID  30455427. S2CID  53872296.
  8. ^ a b Lim K, Halim A, Lu TS, Ashworth A, Chong I (September 2019). "Klotho: A Major Shareholder in Vascular Aging Enterprises". International Journal of Molecular Sciences. 20 (18): E4637. doi: 10.3390/ijms20184637. PMC  6770519. PMID  31546756.
  9. ^ a b c d e f g h i j Hanson K, Fisher K, Hooper NM (June 2021). "Exploiting the neuroprotective effects of α-klotho to tackle ageing- and neurodegeneration-related cognitive dysfunction". Neuronal Signaling. 5 (2): NS20200101. doi: 10.1042/NS20200101. PMC  8204227. PMID  34194816.
  10. ^ Kurt B, Kurtz A (March 2015). "Plasticity of renal endocrine function". American Journal of Physiology. Regulatory, Integrative and Comparative Physiology. 308 (6): R455–R466. doi: 10.1152/ajpregu.00568.2013. PMID  25608752. S2CID  37452911.
  11. ^ Buendía P, Ramírez R, Aljama P, Carracedo J (2016). "Klotho Prevents Translocation of NFκB". Klotho. Vitamins & Hormones. Vol. 101. pp. 119–150. doi: 10.1016/bs.vh.2016.02.005. ISBN  9780128048191. PMID  27125740.
  12. ^ Martín-González C, González-Reimers E, Quintero-Platt G, Martínez-Riera A, Santolaria-Fernández F (May 2019). "Soluble α-Klotho in Liver Cirrhosis and Alcoholism". Alcohol and Alcoholism. 54 (3): 204–208. doi: 10.1093/alcalc/agz019. PMC  6731336. PMID  30860544.
  13. ^ "Entrez Gene: klotho".
  14. ^ Schumann G, Liu C, O'Reilly P, Gao H, Song P, Xu B, et al. (December 2016). "KLB is associated with alcohol drinking, and its gene product β-Klotho is necessary for FGF21 regulation of alcohol preference". Proceedings of the National Academy of Sciences of the United States of America. 113 (50): 14372–14377. Bibcode: 2016PNAS..11314372S. doi: 10.1073/pnas.1611243113. PMC  5167198. PMID  27911795.
  15. ^ a b Kurosu H, Yamamoto M, Clark JD, Pastor JV, Nandi A, Gurnani P, et al. (September 2005). "Suppression of aging in mice by the hormone Klotho". Science. 309 (5742): 1829–1833. Bibcode: 2005Sci...309.1829K. doi: 10.1126/science.1112766. PMC  2536606. PMID  16123266.
  16. ^ a b c d e Baranowska B, Kochanowski J (September 2020). "The metabolic, neuroprotective cardioprotective and antitumor effects of the Klotho protein". Neuro Endocrinology Letters. 41 (2): 69–75. PMID  33185993.
  17. ^ "Klotho Beta". Retrieved 12 September 2023.
  18. ^ Kuro-o M, Matsumura Y, Aizawa H, Kawaguchi H, Suga T, Utsugi T, et al. (November 1997). "Mutation of the mouse klotho gene leads to a syndrome resembling ageing". Nature. 390 (6655): 45–51. Bibcode: 1997Natur.390...45K. doi: 10.1038/36285. PMID  9363890. S2CID  4428141.
  19. ^ Arking DE, Krebsova A, Macek M, Macek M, Arking A, Mian IS, et al. (January 2002). "Association of human aging with a functional variant of klotho". Proceedings of the National Academy of Sciences of the United States of America. 99 (2): 856–861. Bibcode: 2002PNAS...99..856A. doi: 10.1073/pnas.022484299. PMC  117395. PMID  11792841.
  20. ^ Xiao NM, Zhang YM, Zheng Q, Gu J (May 2004). "Klotho is a serum factor related to human aging". Chinese Medical Journal. 117 (5): 742–747. PMID  15161545.[ permanent dead link]
  21. ^ Helsten T, Schwaederle M, Kurzrock R (September 2015). "Fibroblast growth factor receptor signaling in hereditary and neoplastic disease: biologic and clinical implications". Cancer and Metastasis Reviews. 34 (3): 479–496. doi: 10.1007/s10555-015-9579-8. PMC  4573649. PMID  26224133.
  22. ^ Talukdar S, Owen BM, Song P, Hernandez G, Zhang Y, Zhou Y, et al. (February 2016). "FGF21 Regulates Sweet and Alcohol Preference". Cell Metabolism. 23 (2): 344–349. doi: 10.1016/j.cmet.2015.12.008. PMC  4749404. PMID  26724861.
  23. ^ Flippo KH, Potthoff MJ (March 2021). "Metabolic Messengers: FGF21". Nature Metabolism. 3 (3): 309–317. doi: 10.1038/s42255-021-00354-2. PMC  8620721. PMID  33758421.
  24. ^ a b c Huang CL (May 2010). "Regulation of ion channels by secreted Klotho: mechanisms and implications". Kidney International. 77 (10): 855–860. doi: 10.1038/ki.2010.73. PMID  20375979.
  25. ^ van Goor MK, Hoenderop JG, van der Wijst J (June 2017). "TRP channels in calcium homeostasis: from hormonal control to structure-function relationship of TRPV5 and TRPV6". Biochimica et Biophysica Acta (BBA) - Molecular Cell Research. 1864 (6): 883–893. doi: 10.1016/j.bbamcr.2016.11.027. PMID  27913205.
  26. ^ Wolf MT, An SW, Nie M, Bal MS, Huang CL (December 2014). "Klotho up-regulates renal calcium channel transient receptor potential vanilloid 5 (TRPV5) by intra- and extracellular N-glycosylation-dependent mechanisms". The Journal of Biological Chemistry. 289 (52): 35849–35857. doi: 10.1074/jbc.M114.616649. PMC  4276853. PMID  25378396.
  27. ^ Saghiv MS, Sira DB, Goldhammer E, Sagiv M (2017). "The effects of aerobic and anaerobic exercises on circulating soluble-Klotho and IGF-I in young and elderly adults and in CAD patients". Journal of Circulating Biomarkers. 6: 1849454417733388. doi: 10.1177/1849454417733388. PMC  5644364. PMID  29081845.
  28. ^ a b Olejnik A, Franczak A, Krzywonos-Zawadzka A, Kałużna-Oleksy M, Bil-Lula I (2018). "The Biological Role of Klotho Protein in the Development of Cardiovascular Diseases". BioMed Research International. 2018: 5171945. doi: 10.1155/2018/5171945. PMC  6323445. PMID  30671457.
  29. ^ Torbus-Paluszczak M, Bartman W, Adamczyk-Sowa M (October 2018). "Klotho protein in neurodegenerative disorders". Neurological Sciences. 39 (10): 1677–1682. doi: 10.1007/s10072-018-3496-x. PMC  6154120. PMID  30062646.
  30. ^ Vo HT, Laszczyk AM, King GD (August 2018). "Klotho, the Key to Healthy Brain Aging?". Brain Plasticity. 3 (2): 183–194. doi: 10.3233/BPL-170057. PMC  6091049. PMID  30151342.
  31. ^ a b Tozer L (4 July 2023). "Anti-ageing protein injection boosts monkeys' memories". Nature. 619 (7969): 234. Bibcode: 2023Natur.619..234T. doi: 10.1038/d41586-023-02214-3. PMID  37402904. S2CID  259334272.
  32. ^ a b c d Zhou H, Pu S, Zhou H, Guo Y (2021). "Klotho as Potential Autophagy Regulator and Therapeutic Target". Frontiers in Pharmacology. 12: 755366. doi: 10.3389/fphar.2021.755366. PMC  8560683. PMID  34737707.
  33. ^ Adhikari BR, Uehara O, Matsuoka H, Takai R, Harada F, Utsunomiya M, et al. (September 2017). "Immunohistochemical evaluation of Klotho and DNA methyltransferase 3a in oral squamous cell carcinomas". Medical Molecular Morphology. 50 (3): 155–160. doi: 10.1007/s00795-017-0156-9. PMID  28303350. S2CID  22810635.
  34. ^ Paroni G, Panza F, De Cosmo S, Greco A, Seripa D, Mazzoccoli G (March 2019). "Klotho at the Edge of Alzheimer's Disease and Senile Depression". Molecular Neurobiology. 56 (3): 1908–1920. doi: 10.1007/s12035-018-1200-z. PMID  29978424. S2CID  49567009.
  35. ^ Lehrer S, Rheinstein PH (2020). "Alignment of Alzheimer's disease amyloid β-peptide and klotho". World Academy of Sciences Journal. 2 (6): 1. doi: 10.3892/wasj.2020.68. PMC  7521834. PMID  32999998.
  36. ^ Castner SA, Gupta S, Wang D, Moreno AJ, Park C, Chen C, et al. (July 2023). "Longevity factor klotho enhances cognition in aged nonhuman primates". Nature Aging. 3 (8): 931–937. doi: 10.1038/s43587-023-00441-x. PMC  10432271. PMID  37400721. S2CID  259322607.
  37. ^ a b c Veronesi F, Borsari V, Cherubini A, Fini M (October 2021). "Association of Klotho with physical performance and frailty in middle-aged and older adults: A systematic review". Experimental Gerontology. 154: 111518. doi: 10.1016/j.exger.2021.111518. PMID  34407459. S2CID  237011996.
  38. ^ Dubal DB, Yokoyama JS, Zhu L, Broestl L, Worden K, Wang D, et al. (May 2014). "Life extension factor klotho enhances cognition". Cell Reports. 7 (4): 1065–1076. doi: 10.1016/j.celrep.2014.03.076. PMC  4176932. PMID  24813892.
  39. ^ Kuro-o M (October 2009). "Klotho and aging". Biochimica et Biophysica Acta (BBA) - General Subjects. 1790 (10): 1049–1058. doi: 10.1016/j.bbagen.2009.02.005. PMC  2743784. PMID  19230844.
  40. ^ Medici D, Razzaque MS, Deluca S, Rector TL, Hou B, Kang K, et al. (August 2008). "FGF-23-Klotho signaling stimulates proliferation and prevents vitamin D-induced apoptosis". The Journal of Cell Biology. 182 (3): 459–465. doi: 10.1083/jcb.200803024. PMC  2500132. PMID  18678710.
  41. ^ Tsujikawa H, Kurotaki Y, Fujimori T, Fukuda K, Nabeshima Y (December 2003). "Klotho, a gene related to a syndrome resembling human premature aging, functions in a negative regulatory circuit of vitamin D endocrine system". Molecular Endocrinology. 17 (12): 2393–2403. doi: 10.1210/me.2003-0048. hdl: 2433/145275. PMID  14528024.
  42. ^ Imura A, Tsuji Y, Murata M, Maeda R, Kubota K, Iwano A, et al. (June 2007). "alpha-Klotho as a regulator of calcium homeostasis". Science. 316 (5831): 1615–1618. Bibcode: 2007Sci...316.1615I. doi: 10.1126/science.1135901. PMID  17569864. S2CID  40529168.
  43. ^ Liu H, Fergusson MM, Castilho RM, Liu J, Cao L, Chen J, et al. (August 2007). "Augmented Wnt signaling in a mammalian model of accelerated aging". Science. 317 (5839): 803–806. doi: 10.1038/ki.2010.73. PMID  17690294.
  44. ^ Irving M (2021-12-10). ""Young blood" particles that help old mice fight aging identified". New Atlas. Retrieved 2021-12-11.
  45. ^ Irving M (2022-03-16). "Senolytic drugs boost protein that protects against effects of aging". New Atlas. Retrieved 2022-03-16.

Further reading

External links

This article incorporates text from the United States National Library of Medicine, which is in the public domain.

From Wikipedia, the free encyclopedia
KL
Identifiers
Aliases KL, entrez:9365, klotho, HFTC3, KLA
External IDs OMIM: 604824 MGI: 1101771 HomoloGene: 68415 GeneCards: KL
Orthologs
SpeciesHumanMouse
Entrez
Ensembl
UniProt
RefSeq (mRNA)

NM_004795
NM_153683

NM_013823

RefSeq (protein)

NP_004786

NP_038851

Location (UCSC) Chr 13: 33.02 – 33.07 Mb Chr 5: 150.88 – 150.92 Mb
PubMed search [3] [4]
Wikidata
View/Edit Human View/Edit Mouse

Klotho is an enzyme that in humans is encoded by the KL gene. [5] The three subfamilies of klotho are α-klotho, β-klotho, and γ-klotho. [6] α-klotho activates FGF23, and β-klotho activates FGF19 and FGF21. [7] When the subfamily is not specified, the word "klotho" typically refers to the α-klotho subfamily, because α-klotho was discovered before the other members. [8] [7]

α-klotho is highly expressed in the brain, liver and kidney. [9] β-klotho is predominantly expressed in the liver. [10] [9] γ-klotho is expressed in the skin. [9]

Klotho can exist in a membrane-bound form or a (hormonal) soluble, circulating form. [11] Proteases can convert the membrane-bound form into the circulating form. [12]

The KL gene encodes a type-I single-pass transmembrane protein [7] that is related to β-glucuronidases. Reduced production of this protein has been observed in patients with chronic kidney failure (CKF), and this may be one of the factors underlying degenerative processes (e.g., arteriosclerosis, osteoporosis, and skin atrophy) seen in CKF. Mutations within the family have been associated with ageing, bone loss and alcohol consumption. [13] [14] Transgenic mice that overexpress Klotho live longer than wild-type mice. [15]

Structure

The α-klotho gene is located on chromosome 13, and is translated into a single-pass integral membrane protein. [9] The intracellular portion of the α-klotho protein is short (11 amino acids), whereas the extracellular portion is long (980 amino acids). [9] The transmembrane portion is also comparatively short (21 amino acids). [9] The extracellular portion contains two repeat sequences, termed the KL1 (about 450 amino acids) and KL2 (about 430 amino acids) domains. [9] [7] In the kidney and the choroid plexus of the brain, the transmembrane protein can be proteolytically cleaved to produce a 130- Kilo- Dalton, soluble form of α-klotho protein, released into the circulation and cerebrospinal fluid, respectively. [9] In humans, the secreted form of klotho is more dominant than the membrane form. [16]

The β-Klotho gene is located on chromosome 4. The protein shares homology (43.1% identity and 60.1% similarity) with α-klotho. [17] It should not be confused with the alpha-cut and beta-cut of alpha-klotho, which releases KL1+KL2 and KL2 domain, respectively.

Function

Klotho is a transmembrane protein that, in addition to other effects, provides some control over the sensitivity of the organism to insulin and appears to be involved in ageing. Its discovery was documented in 1997 by Makoto Kuro-o et al. [18] The name of the gene comes from Klotho or Clotho, one of the Moirai, or Fates, in Greek mythology, who spins the thread of human life. [7]

The klotho protein is a novel β-glucuronidase ( EC number 3.2.1.31) capable of hydrolyzing steroid β-glucuronides. Genetic variants in KLOTHO have been associated with human aging, [19] and klotho protein has been shown to be a circulating factor detectable in serum that declines with age. [20]

The binding of the endocrine fibroblast growth factors (FGF's, viz., FGF19 and FGF21) to their fibroblast growth factor receptors, is promoted via their interactions as co-receptors with β-klotho. [21] [22] [16] [7] Loss of β-Klotho abolishes all effects of FGF21. [23]

α-klotho, which binds to the endocrine FGF FGF23 changes cellular calcium homeostasis, by both increasing the expression and activity of TRPV5 (decreasing phosphate reabsorption in the kidney) and decreasing that of TRPC6 (decreasing phosphate absorption from the intestine). [24] α-klotho increases kidney calcium reabsorption by stabilizing TPRV5. [25] About 95% to 98% of Ca2+ filtered from the blood by the kidney is normally reabsorbed by the kidney's renal tubule, which is mediated by TRPV5. [26]

Clinical significance

α-klotho can suppress oxidative stress and inflammation, thereby reducing endothelial dysfunction and atherosclerosis. [8] Blood plasma α-klotho is increased by aerobic exercise, thereby reducing endothelial dysfunction. [27]

β-klotho activation of FGF21 protein has a protective effect on heart muscle cells. [28] Obesity is characterized by FGF21 resistance, believed to be caused by the inhibition of β-klotho by the inflammatory cell signalling protein ( cytokine) tumor necrosis factor alpha, [28] but there is evidence against this mechanism. [16]

Klotho is required for oligodendrocyte maturation, myelin integrity, and can protect neurons from toxic effects. [29] Mice deficient in klotho have a reduced number of synapses and cognitive deficits, whereas mice overexpressing klotho have enhanced learning and memory. [30] Research with injections of klotho in primates demonstrates a positive effect on memory that lasts for as long as two weeks. [31]

Reduced klotho expression is seen in the lung macrophages of smokers. [32] An abnormal form of autophagy associated with reduced expression of klotho is linked to the pathogenesis of chronic obstructive pulmonary disease. [32] (Although normal autophagy helps maintain muscle, excessive autophagy causes loss of muscle mass. [32])

It has been found that the decreased klotho expression may be due to DNA hypermethylation, which may have been induced by the overexpression of DNMT3a. [33] Klotho may be a reliable gene for early detection of methylation changes in oral tissues, and can be used as a target for therapeutic modification in oral cancer during the early stages.

Klotho-deficient mice manifest a syndrome resembling accelerated human ageing and display extensive and accelerated arteriosclerosis. Additionally, they exhibit impaired endothelium dependent vasodilation and impaired angiogenesis, suggesting that klotho protein may protect the cardiovascular system through endothelium-derived nitric oxide production. [16]

Klotho could play a protective role in Alzheimer's disease patients. [34] [35]

Research with injections of α-klotho in primates suggests a positive effect on memory that could have implications for research with humans. [31] Interestingly the cognitive effects in rhesus monkeys were observed even with subcutaneous injection despite previous results showing that klotho protein fails to cross the blood–brain barrier. [36]

Effects on aging

Reduced α-klotho or FGF23 can result in impaired phosphate excretion from the kidney, leading to hyperphosphatemia. [7] In mice, this leads to a phenotype characteristic of premature aging, which can be mitigated by feeding the mice a low phosphate diet. [7]

The plasma (soluble) form of α-klotho is most easily measured, and has been shown to decrease after 40 years of age in humans. [37] Lower plasma levels of α-klotho in older adults is associated with increased frailty and all-cause mortality. [37] Physical activity has been shown to increase plasma α-klotho. [37]

Mice lacking either fibroblast growth factor 23 or the α-klotho enzyme display premature aging due to hyperphosphatemia. [24] Many of these symptoms can be alleviated by feeding the mice a low phosphate diet. [7]

Although the majority of research has explored klotho's absence, it was demonstrated that klotho over-expression in mice extended their average life span between 19% and 31% compared to normal mice. [15] In addition, variations in the Klotho gene (SNP Rs9536314) are associated with both life extension and increased cognition in human populations and mice, but only if the gene expression was heterozygous, not homozygous. [38] [9] The cognitive benefits of α-klotho are primarily seen late in life. [9]

Klotho increases membrane expression of the inward rectifier ATP-dependent potassium channel ROMK. [24] Klotho-deficient mice show increased production of vitamin D, and altered mineral-ion homeostasis is suggested to cause premature aging‑like phenotypes, because reduced vitamin D activity from dietary restriction reverses the premature aging‑like phenotypes and prolongs survival in these mutants. These results suggest that aging‑like phenotypes were due to klotho-associated vitamin D metabolic abnormalities (hypervitaminosis). [39] [40] [41] [42]

Klotho is an antagonist of the Wnt signaling pathway, and chronic Wnt stimulation can lead to stem cell depletion and aging. [43] Klotho inhibition of Wnt signaling can inhibit cancer. [32] The anti-aging effects of klotho are also a consequence of increased resistance to inflammation and oxidative stress. [16]

Extracellular vesicles (EV) in young mice carried more copies of klotho-producing mRNA than those from old mice. Transfusing young EVs into older mice helped rebuild their muscles. [44]

The presence of senescent cells decreases α-klotho levels. Senolytic drugs reduce the level of these cells, allowing α-klotho levels to increase. [45]

References

  1. ^ a b c GRCh38: Ensembl release 89: ENSG00000133116Ensembl, May 2017
  2. ^ a b c GRCm38: Ensembl release 89: ENSMUSG00000058488Ensembl, May 2017
  3. ^ "Human PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  4. ^ "Mouse PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  5. ^ Matsumura Y, Aizawa H, Shiraki-Iida T, Nagai R, Kuro-o M, Nabeshima Y (January 1998). "Identification of the human klotho gene and its two transcripts encoding membrane and secreted klotho protein". Biochemical and Biophysical Research Communications. 242 (3): 626–630. doi: 10.1006/bbrc.1997.8019. PMID  9464267.
  6. ^ Dolegowska K, Marchelek-Mysliwiec M, Nowosiad-Magda M, Slawinski M, Dolegowska B (June 2019). "FGF19 subfamily members: FGF19 and FGF21". Journal of Physiology and Biochemistry. 75 (2): 229–240. doi: 10.1007/s13105-019-00675-7. PMC  6611749. PMID  30927227.
  7. ^ a b c d e f g h i Kuro-O M (January 2019). "The Klotho proteins in health and disease". Nature Reviews. Nephrology. 15 (1): 27–44. doi: 10.1038/s41581-018-0078-3. PMID  30455427. S2CID  53872296.
  8. ^ a b Lim K, Halim A, Lu TS, Ashworth A, Chong I (September 2019). "Klotho: A Major Shareholder in Vascular Aging Enterprises". International Journal of Molecular Sciences. 20 (18): E4637. doi: 10.3390/ijms20184637. PMC  6770519. PMID  31546756.
  9. ^ a b c d e f g h i j Hanson K, Fisher K, Hooper NM (June 2021). "Exploiting the neuroprotective effects of α-klotho to tackle ageing- and neurodegeneration-related cognitive dysfunction". Neuronal Signaling. 5 (2): NS20200101. doi: 10.1042/NS20200101. PMC  8204227. PMID  34194816.
  10. ^ Kurt B, Kurtz A (March 2015). "Plasticity of renal endocrine function". American Journal of Physiology. Regulatory, Integrative and Comparative Physiology. 308 (6): R455–R466. doi: 10.1152/ajpregu.00568.2013. PMID  25608752. S2CID  37452911.
  11. ^ Buendía P, Ramírez R, Aljama P, Carracedo J (2016). "Klotho Prevents Translocation of NFκB". Klotho. Vitamins & Hormones. Vol. 101. pp. 119–150. doi: 10.1016/bs.vh.2016.02.005. ISBN  9780128048191. PMID  27125740.
  12. ^ Martín-González C, González-Reimers E, Quintero-Platt G, Martínez-Riera A, Santolaria-Fernández F (May 2019). "Soluble α-Klotho in Liver Cirrhosis and Alcoholism". Alcohol and Alcoholism. 54 (3): 204–208. doi: 10.1093/alcalc/agz019. PMC  6731336. PMID  30860544.
  13. ^ "Entrez Gene: klotho".
  14. ^ Schumann G, Liu C, O'Reilly P, Gao H, Song P, Xu B, et al. (December 2016). "KLB is associated with alcohol drinking, and its gene product β-Klotho is necessary for FGF21 regulation of alcohol preference". Proceedings of the National Academy of Sciences of the United States of America. 113 (50): 14372–14377. Bibcode: 2016PNAS..11314372S. doi: 10.1073/pnas.1611243113. PMC  5167198. PMID  27911795.
  15. ^ a b Kurosu H, Yamamoto M, Clark JD, Pastor JV, Nandi A, Gurnani P, et al. (September 2005). "Suppression of aging in mice by the hormone Klotho". Science. 309 (5742): 1829–1833. Bibcode: 2005Sci...309.1829K. doi: 10.1126/science.1112766. PMC  2536606. PMID  16123266.
  16. ^ a b c d e Baranowska B, Kochanowski J (September 2020). "The metabolic, neuroprotective cardioprotective and antitumor effects of the Klotho protein". Neuro Endocrinology Letters. 41 (2): 69–75. PMID  33185993.
  17. ^ "Klotho Beta". Retrieved 12 September 2023.
  18. ^ Kuro-o M, Matsumura Y, Aizawa H, Kawaguchi H, Suga T, Utsugi T, et al. (November 1997). "Mutation of the mouse klotho gene leads to a syndrome resembling ageing". Nature. 390 (6655): 45–51. Bibcode: 1997Natur.390...45K. doi: 10.1038/36285. PMID  9363890. S2CID  4428141.
  19. ^ Arking DE, Krebsova A, Macek M, Macek M, Arking A, Mian IS, et al. (January 2002). "Association of human aging with a functional variant of klotho". Proceedings of the National Academy of Sciences of the United States of America. 99 (2): 856–861. Bibcode: 2002PNAS...99..856A. doi: 10.1073/pnas.022484299. PMC  117395. PMID  11792841.
  20. ^ Xiao NM, Zhang YM, Zheng Q, Gu J (May 2004). "Klotho is a serum factor related to human aging". Chinese Medical Journal. 117 (5): 742–747. PMID  15161545.[ permanent dead link]
  21. ^ Helsten T, Schwaederle M, Kurzrock R (September 2015). "Fibroblast growth factor receptor signaling in hereditary and neoplastic disease: biologic and clinical implications". Cancer and Metastasis Reviews. 34 (3): 479–496. doi: 10.1007/s10555-015-9579-8. PMC  4573649. PMID  26224133.
  22. ^ Talukdar S, Owen BM, Song P, Hernandez G, Zhang Y, Zhou Y, et al. (February 2016). "FGF21 Regulates Sweet and Alcohol Preference". Cell Metabolism. 23 (2): 344–349. doi: 10.1016/j.cmet.2015.12.008. PMC  4749404. PMID  26724861.
  23. ^ Flippo KH, Potthoff MJ (March 2021). "Metabolic Messengers: FGF21". Nature Metabolism. 3 (3): 309–317. doi: 10.1038/s42255-021-00354-2. PMC  8620721. PMID  33758421.
  24. ^ a b c Huang CL (May 2010). "Regulation of ion channels by secreted Klotho: mechanisms and implications". Kidney International. 77 (10): 855–860. doi: 10.1038/ki.2010.73. PMID  20375979.
  25. ^ van Goor MK, Hoenderop JG, van der Wijst J (June 2017). "TRP channels in calcium homeostasis: from hormonal control to structure-function relationship of TRPV5 and TRPV6". Biochimica et Biophysica Acta (BBA) - Molecular Cell Research. 1864 (6): 883–893. doi: 10.1016/j.bbamcr.2016.11.027. PMID  27913205.
  26. ^ Wolf MT, An SW, Nie M, Bal MS, Huang CL (December 2014). "Klotho up-regulates renal calcium channel transient receptor potential vanilloid 5 (TRPV5) by intra- and extracellular N-glycosylation-dependent mechanisms". The Journal of Biological Chemistry. 289 (52): 35849–35857. doi: 10.1074/jbc.M114.616649. PMC  4276853. PMID  25378396.
  27. ^ Saghiv MS, Sira DB, Goldhammer E, Sagiv M (2017). "The effects of aerobic and anaerobic exercises on circulating soluble-Klotho and IGF-I in young and elderly adults and in CAD patients". Journal of Circulating Biomarkers. 6: 1849454417733388. doi: 10.1177/1849454417733388. PMC  5644364. PMID  29081845.
  28. ^ a b Olejnik A, Franczak A, Krzywonos-Zawadzka A, Kałużna-Oleksy M, Bil-Lula I (2018). "The Biological Role of Klotho Protein in the Development of Cardiovascular Diseases". BioMed Research International. 2018: 5171945. doi: 10.1155/2018/5171945. PMC  6323445. PMID  30671457.
  29. ^ Torbus-Paluszczak M, Bartman W, Adamczyk-Sowa M (October 2018). "Klotho protein in neurodegenerative disorders". Neurological Sciences. 39 (10): 1677–1682. doi: 10.1007/s10072-018-3496-x. PMC  6154120. PMID  30062646.
  30. ^ Vo HT, Laszczyk AM, King GD (August 2018). "Klotho, the Key to Healthy Brain Aging?". Brain Plasticity. 3 (2): 183–194. doi: 10.3233/BPL-170057. PMC  6091049. PMID  30151342.
  31. ^ a b Tozer L (4 July 2023). "Anti-ageing protein injection boosts monkeys' memories". Nature. 619 (7969): 234. Bibcode: 2023Natur.619..234T. doi: 10.1038/d41586-023-02214-3. PMID  37402904. S2CID  259334272.
  32. ^ a b c d Zhou H, Pu S, Zhou H, Guo Y (2021). "Klotho as Potential Autophagy Regulator and Therapeutic Target". Frontiers in Pharmacology. 12: 755366. doi: 10.3389/fphar.2021.755366. PMC  8560683. PMID  34737707.
  33. ^ Adhikari BR, Uehara O, Matsuoka H, Takai R, Harada F, Utsunomiya M, et al. (September 2017). "Immunohistochemical evaluation of Klotho and DNA methyltransferase 3a in oral squamous cell carcinomas". Medical Molecular Morphology. 50 (3): 155–160. doi: 10.1007/s00795-017-0156-9. PMID  28303350. S2CID  22810635.
  34. ^ Paroni G, Panza F, De Cosmo S, Greco A, Seripa D, Mazzoccoli G (March 2019). "Klotho at the Edge of Alzheimer's Disease and Senile Depression". Molecular Neurobiology. 56 (3): 1908–1920. doi: 10.1007/s12035-018-1200-z. PMID  29978424. S2CID  49567009.
  35. ^ Lehrer S, Rheinstein PH (2020). "Alignment of Alzheimer's disease amyloid β-peptide and klotho". World Academy of Sciences Journal. 2 (6): 1. doi: 10.3892/wasj.2020.68. PMC  7521834. PMID  32999998.
  36. ^ Castner SA, Gupta S, Wang D, Moreno AJ, Park C, Chen C, et al. (July 2023). "Longevity factor klotho enhances cognition in aged nonhuman primates". Nature Aging. 3 (8): 931–937. doi: 10.1038/s43587-023-00441-x. PMC  10432271. PMID  37400721. S2CID  259322607.
  37. ^ a b c Veronesi F, Borsari V, Cherubini A, Fini M (October 2021). "Association of Klotho with physical performance and frailty in middle-aged and older adults: A systematic review". Experimental Gerontology. 154: 111518. doi: 10.1016/j.exger.2021.111518. PMID  34407459. S2CID  237011996.
  38. ^ Dubal DB, Yokoyama JS, Zhu L, Broestl L, Worden K, Wang D, et al. (May 2014). "Life extension factor klotho enhances cognition". Cell Reports. 7 (4): 1065–1076. doi: 10.1016/j.celrep.2014.03.076. PMC  4176932. PMID  24813892.
  39. ^ Kuro-o M (October 2009). "Klotho and aging". Biochimica et Biophysica Acta (BBA) - General Subjects. 1790 (10): 1049–1058. doi: 10.1016/j.bbagen.2009.02.005. PMC  2743784. PMID  19230844.
  40. ^ Medici D, Razzaque MS, Deluca S, Rector TL, Hou B, Kang K, et al. (August 2008). "FGF-23-Klotho signaling stimulates proliferation and prevents vitamin D-induced apoptosis". The Journal of Cell Biology. 182 (3): 459–465. doi: 10.1083/jcb.200803024. PMC  2500132. PMID  18678710.
  41. ^ Tsujikawa H, Kurotaki Y, Fujimori T, Fukuda K, Nabeshima Y (December 2003). "Klotho, a gene related to a syndrome resembling human premature aging, functions in a negative regulatory circuit of vitamin D endocrine system". Molecular Endocrinology. 17 (12): 2393–2403. doi: 10.1210/me.2003-0048. hdl: 2433/145275. PMID  14528024.
  42. ^ Imura A, Tsuji Y, Murata M, Maeda R, Kubota K, Iwano A, et al. (June 2007). "alpha-Klotho as a regulator of calcium homeostasis". Science. 316 (5831): 1615–1618. Bibcode: 2007Sci...316.1615I. doi: 10.1126/science.1135901. PMID  17569864. S2CID  40529168.
  43. ^ Liu H, Fergusson MM, Castilho RM, Liu J, Cao L, Chen J, et al. (August 2007). "Augmented Wnt signaling in a mammalian model of accelerated aging". Science. 317 (5839): 803–806. doi: 10.1038/ki.2010.73. PMID  17690294.
  44. ^ Irving M (2021-12-10). ""Young blood" particles that help old mice fight aging identified". New Atlas. Retrieved 2021-12-11.
  45. ^ Irving M (2022-03-16). "Senolytic drugs boost protein that protects against effects of aging". New Atlas. Retrieved 2022-03-16.

Further reading

External links

This article incorporates text from the United States National Library of Medicine, which is in the public domain.


Videos

Youtube | Vimeo | Bing

Websites

Google | Yahoo | Bing

Encyclopedia

Google | Yahoo | Bing

Facebook