Biomedical Research Journal

REVIEW ARTICLE
Year
: 2019  |  Volume : 6  |  Issue : 1  |  Page : 1--6

Klotho: An emerging factor in neurodegenerative diseases


Gauri V Pathare, Kavita K Shalia 
 Department of Biochemistry, Sir H. N. Medical Research Society, Mumbai, Maharashtra, India

Correspondence Address:
Ms. Gauri V Pathare
Sir H. N. Medical Research Society, Court House, L. T. Road, Mumbai - 400 002, Maharashtra
India

Abstract

Soluble Klotho protein is present in blood, urine, and cerebrospinal fluid and works as a humoral factor exerting different biological effects. Several animal studies have demonstrated the association of age-related neurodegeneration with Klotho deficiency. Lower Klotho levels have been reported in patients suffering from cognitive impairment, dementia, Alzheimer's disease, Parkinson's disease, multiple sclerosis, and other neurodegenerative diseases. Due to its antiaging properties, Klotho is the obvious choice to be studied as a protective/therapeutic agent in neurobiology. In this review, we have attempted to shed light on the different neurodegenerative diseases affected by deficiency of Klotho and its neuroprotective role against pathogenicity of the disease.



How to cite this article:
Pathare GV, Shalia KK. Klotho: An emerging factor in neurodegenerative diseases.Biomed Res J 2019;6:1-6


How to cite this URL:
Pathare GV, Shalia KK. Klotho: An emerging factor in neurodegenerative diseases. Biomed Res J [serial online] 2019 [cited 2024 Mar 29 ];6:1-6
Available from: https://www.brjnmims.org/text.asp?2019/6/1/1/257035


Full Text



 Introduction



The Klotho protein discovered over two decades ago is still a sought after molecule. Although its protective effects on physiology can be observed and have been demonstrated in several animal and human studies, its mechanism remains an enigma. Hence, even today, it can be referred to as “novel” as described by its discoverer in 1997.[1] Studies on Klotho have been commenced in almost all human pathophysiological disorders. Due to its antiaging properties, it is the obvious choice to be studied as a protective/therapeutic agent in neurobiology. In this review, we have attempted to shed light on the different neurodegenerative diseases affected by deficiency of Klotho. The role of Klotho in these diseases is briefly described in [Figure 1].{Figure 1}

 Klotho Gene and Protein



Kuro-o et al. have established a novel mouse autosomal recessive mutant, deficient in the Klotho gene, which exhibits multiple phenotypes very similar to those observed during human aging, which can be regarded as a model of human premature aging syndromes.[1] The human Klotho gene (KL), more precisely α-Klotho, located on chromosome 13q12, spans over 50 kb in length, and comprises five exons and four introns.[2] It is expressed primarily in the kidney, the parathyroid gland, and the choroid plexus in the brain.[3],[4] The Klotho gene encodes two transcripts that arise from alternative RNA splicing. The full-length transcript encodes a single-pass membrane protein (KL) that consists of (1) an N-terminal signal sequence, (2) an extracellular domain with two internal repeats (KL1 and KL2), (3) a single-pass transmembrane protein, and (4) a short intracellular carboxy-terminal domain.[2] The membrane form acts as co-receptor for bone-derived fibroblast growth factor-23 (FGF-23), thereby regulating phosphate and Vitamin D metabolism.[5] The other transcript encodes a truncated KL protein lacking the second internal repeat of the extracellular domain (KL2), the transmembrane domain, and the intracellular domain. This soluble form is also produced by cleavage of extracellular domain of the transmembrane protein by proteases (ADAM10 and ADAM17).[6] This soluble form is found to predominate over the membrane form and was concluded to be the major gene product in humans by Matsumura et al.[2] The soluble Klotho is present in blood, urine, and cerebrospinal fluid (CSF)[7] and works as a humoral factor exerting different biological effects and works independently of FGF-23 signaling.[8] Circulating Klotho plays an important role in the regulation of endothelial maintenance through regulation of nitric oxide availability,[9],[10] calcium homeostasis in the kidney,[11],[12] and inhibition of intracellular insulin and insulin-like growth factor-1 signaling.[13]

 Klotho and Neuroprotection



Klotho expression is downregulated with age.[14],[15],[16],[17] Recent years have seen a surge in the studies exploring the effects of lack as well as potential therapeutic effect of Klotho in age-related changes in the brain. Shiozaki et al. have proposed that Klotho knockout mouse is an apt model to study the central nervous system (CNS) aging.[18] Studies have reported CNS alterations such as hypomyelination, overexpression/phosphorylation of neurofilaments, synaptic loss, and behavioral impairments in Klotho-deficient mice similar to those observed in aged animals.[18],[19],[20] Moreover, selective reduction of Klotho levels in the choroid plexus in the brain promoted neuroinflammation.[17] So, how does Klotho protect the brain?

The common link between aging and neuropathological changes including cognitive deficits is “oxidative stress.” Vascular risk factors such as atrial fibrillation, hypertension, atherosclerosis, angina, and diabetes – whose pathogenesis has been attributed to oxidative stress – have also been reported to contribute in dementia and Alzheimer's disease (AD).[21] Therefore, the answer to how Klotho, as an antiaging protein, ameliorates such pathologies, could be by its ability to resist oxidative stress. In fact, its ability to induce expression of antioxidative enzymes via the inhibition of insulin/insulin-like growth factor 1 (IGF-1) signaling is the main pathway through which Klotho functions is an antiaging factor. Suppression of insulin/IGF-1 signaling is an evolutionarily conserved mechanism for longevity. However, there are no reports yet on the actual mechanism by which Klotho inhibits the activity of insulin/IGF-1 receptors in the brain.

Another way Klotho influences aging process in the brain is via regulation of Vitamin D synthesis. Although global Vitamin D3 deficiency is a risk factor for age-related cognitive decline,[22] hypervitaminosis D is a risk factor for cognitive impairment.[23],[24] Klotho here controls hypervitaminosis D by inhibiting the enzyme 1-α-hydroxylase that catalyzes the formation of 1,25-dihydroxyvitamin D3, the bioactive form of Vitamin D.[25] Thus, Klotho plays an important role in proper functioning of the CNS.

 Klotho Deficiency and Cognitive Impairment/decline



The reduced levels of Klotho as well as IGF-1 in the hippocampus of rat model of dementia suggests a significant role of these molecules in the pathogenesis of dementia.[26] Dubal et al. observed that mice with increased Klotho levels showed improved long-term potentiation (strengthening of synapses), as well as increased synaptic GluN2B (an N-methyl-D-aspartate receptor subunit, with vital learning and memory functions). They further reported that Klotho elevated this essential synaptic GluN2B through post-transcriptional mechanisms in a subunit-specific manner. The Klotho-mediated positive effects were, however, lost on blocking GluN2B. A comparison between Klotho overexpressing and control mice in their study showed that increased Klotho enhanced learning and memory in various tasks without any behavioral alteration. In fact, this suggested that the effect of elevated Klotho on survival, longevity, and improved cognition in mice is independent of age.[27]

A comparison study by Nagai et al. between Klotho mutant mice and wild-type mice for mnemonic function associated with aging, measured by an object recognition memory task requiring the hippocampus, showed that cognitive impairment was evident in the Klotho mutant mice. Increased lipid and DNA peroxide levels in the brain of Klotho mutant mice were found prior to the development of evident cognition deficits, suggesting a role for oxidative stress in the etiology of aging-associated cognitive impairment. Supporting this hypothesis was the fact that lowering oxidative stress in Klotho mutant mice with α-tocopherol, an antioxidant that reduces lipid peroxidation, was accompanied by prevention of apoptotic cell death in the hippocampus, and mitigation of memory impairment.[20] Klotho expression has been reported in adult hippocampus and has been associated with hippocampal cognitive performance in mammals. Adult hippocampal neurogenesis (AHN), an evolutionarily conserved mechanism in humans, is vital for several hippocampal cognitive processes including pattern separation, contextual fear conditioning, and spatial memory. Salech et al. have reported Klotho expression in the granular layer of the adult dentate gyrus (DG) of the hippocampus. Downregulating Klotho in the hippocampus decreased AHN and led to poorer hippocampal-dependent memory. On the other hand, providing recombinant Klotho accelerated the proliferation of neural progenitors. This study proposed Klotho's direct involvement in regulating AHN, thereby making it indispensable for cognitive functions.[28]

Laszczyk et al. suggested that Klotho is required for neurogenesis during the transition to adult brain functionality. Although they found that the highest expression of Klotho was observed in choroid plexus, mature hippocampal neuronal layers, including the DG, also expressed the protein. They further showed that the deficiency of Klotho in the hippocampus led to reduced numbers of neural stem cells, decreased proliferation, and impaired neuronal maturation, which collectively led to premature aging. Moreover, Klotho deficiency led to a smaller size and reduced proliferation in neurospheres, which were rectified by providing recombinant Klotho. Thus, the study suggested a regulatory role of Klotho in postnatal neurogenesis, including neural stem cell proliferation and maturation. Supporting this conclusion, was the evident influence of Klotho on hippocampal-dependent spatial memory function.[29]

To test the validity of Klotho as a neuroprotective agent, Zhou et al. injected a lentiviral vector, encoding the Klotho gene, into the bilateral lateral ventricles of 7-month-old senescence-accelerated mouse prone 8 (SAMP8) animals. The mice were observed for a period of 3 months, at the end of which the Y-maze alternation task and passive avoidance task were used to assess their memory deficits. They reported a significant increase in the Klotho expression in the brain of SAMP8 mice at 10 months, along with reduced memory deficits, neuronal loss, and synaptic damage. Further, increased expression of antioxidative enzymes (manganese superoxide dismutase and catalase), attributed to decreased Akt and forkhead box class O1 (FoxO1) phosphorylation, was caused by the upregulation of Klotho expression. Thus, Zhou et al. demonstrated that direct upregulation of Klotho in the brain may ameliorate age-related memory impairments possibly by curtailing oxidative stress.[30]

Further, the protective effects of Klotho were established through an experiment by Long et al. Ligustilide, a phthalide derivative, found in several medicinal plants minimizes the cognitive dysfunction and brain damage induced by cerebral ischemia.[31] It provides neuroprotection against cerebral ischemia in mice by regulating anti-inflammatory and antioxidant signaling pathways. Long et al. downregulated Klotho in the choroid plexus tissues by injecting lentiviruses into a specific location in the bilateral common carotid artery occlusion (BCCAO) mouse model of cerebral ischemia. They found ligustilide to upregulate the expression of Klotho in the choroid plexus. Moreover, downregulation of Klotho attenuated the protective effects of ligustilide on neurons in the hippocampus and caudate-putamen regions of mice with BCCAO-induced neural injury. Furthermore, silencing of Klotho gene prevented the inhibitory effects of ligustilide on the RIG-1/nuclear factor kappa-B p65 and Akt/FoxO1 pathways. Thus, they concluded that Klotho upregulation might contribute to the ligustilide-induced neuroprotection after cerebral ischemic injury in mice.[32]

The protective effects of Klotho have also been reported in dementia. In dementia model of rats, Wang et al. observed that Klotho levels were reduced at the hippocampus and suggested a relationship between Klotho and pathogenesis of dementia.[26] Participants from the population-based “Invecchiare in Chianti” or “Aging in Chianti” (InCHIANTI) study, a prospective cohort study comprising Italian adults over 55 years of age and without dementia, were recruited by Shardell et al. Their study demonstrated an independent association between higher plasma Klotho concentrations and lesser cognitive decline, as measured by the Mini-Mental State Examination.[33] Similar results were reported by Zou and Zhou in a case–control study which indicated lower Klotho protein levels to be associated with mild cognitive impairment (MCI) in aged individuals.[34] Further genetic studies by the same group have identified the contribution of promoter methylation of the Klotho gene to MCI in Xinjiang Han population in China. Modifications in promoter methylation are often found to affect gene expression and have been associated with neurodegenerative diseases.[35] They have further reported the difference in Klotho protein levels, and Klotho promoter hypermethylation-related risk varies according to different ethnic groups. Thus, further investigation pertinent to the ethnicity-related risks is required.

 Klotho in Neurodegenerative Diseases



Multiple sclerosis

Multiple sclerosis (MS) is the most common neurodegenerative disease that results from inflammatory demyelinating events. MS is characterized by a loss of oligodendrocytes (with partial protection of axons), astroglial scars, and multiple demyelinization areas. Chen et al. found a direct correlation between Klotho protein and oligodendrocyte maturation and myelination of the CNS in murine models.[36] Moreover, their Klotho knockout mice showed a significant reduction in major myelin protein and gene expression levels as well as significantly lower mature oligodendrocytes. Thus, Klotho evidently plays a key role in myelin biology and maybe a key therapeutic target involved in protecting myelin against age-dependent changes in the brain.

In humans, Emami Aleagha et al. found decreased Klotho concentration in the CSF of patients with relapsing-remitting MS (RRMS). Furthermore, they reported that Klotho levels showed a significant negative correlation with the Expanded Disability Status Scale. It has been established through several studies that the redox system plays an important role in the pathophysiology of neurodegenerative diseases. The authors also found evidence supporting this wherein they found total antioxidant capacity in the CSF to be low in patients suffering from RRMS as compared to controls as well as positively correlated to Klotho levels.[37]

Contrarily, increased serum Klotho levels were found in MS patients as compared to controls by two independent studies. As part of the FGF-23/Klotho axis, Klotho is involved in regulation of Vitamin D synthesis, which has been reported to play a role in the progression and prevalence of MS. A lower risk of MS in individuals exposed to sunlight in early life supports this theory. Vitamin D has been shown to have immunomodulatory effects on an experimental model of MS by Muthian et al.[38] Ellidag et al. observed lowered Vitamin D levels and elevated serum Klotho and FGF-23 levels in MS patients as compared to controls. On this basis, they concluded that the Klotho/FGF-23/Vitamin D axis is altered in MS and has a significant role in the pathogenesis and progression of the disease.[39] Similar elevated serum Klotho levels in MS have been reported by Ahmadi et al. However, they have attributed this to the use of immunomodulatory drugs in the treatment of MS or a compensatory response to increase CNS regeneration via Vitamin D biosynthesis.[40]

Alzheimer's disease

The formation of senile plaques consisting of the β-amyloid (Aβ) peptides is a hallmark feature of AD pathogenesis. It is generated via amyloidogenic processing of the amyloid precursor protein (APP), which also generates secreted ectodomain fragments (APPsα and APPsβ), and an APP intracellular domain.[41] Although the precise physiological function of APP is not known, APP overexpression shows a neurotrophic effect on neural cell health and growth.[42] Klotho is a physiological target of APP whose expression is mediated by APPsβ. Li et al. proposed that extracellular APP processing produces APP ectodomain derivatives, which promote Klotho expression and protect against Aβ neurotoxicity during aging.[41] Moreover, L-glutamate (major excitatory neurotransmitter) and Aβ are widely used as oxidative stressors in in vitro models of neurodegeneration for studying neuronal damage mechanisms. The neurotoxic effect of Aβ is also associated with cellular injury resulting from reactive oxygen species exposure. Zeldich et al. showed that Klotho protects hippocampal neurons which have the highest vulnerability to oxidative stress, from glutamate and oligomeric Aβ-induced toxicity. This protection could be achieved by providing exogenous Klotho or increasing endogenous Klotho levels as found in Klotho-overexpressing mice.[43]

In a study conducted in aged individuals, Semba et al. found significantly lower CSF Klotho concentrations in those with AD as compared to those with normal cognition. They also found CSF Klotho concentrations to be lower in older as compared to younger individuals. In addition, they reported higher CSF Klotho concentrations in men than women, an interesting finding considering the higher risk and incidence of the disease associated with women.[44] Similar favorable results for Klotho were observed by Yokoyama et al., where they found higher serum Klotho levels to be associated with greater intrinsic connectivity in key functional networks (such as frontoparietal and default-mode networks) of the brain vulnerable to age-dependent neurodegeneration as well as AD. Their findings suggested the role of Klotho in maintaining and promoting brain resilience to neurodegeneration, which was possibly facilitated by boosting network connectivity of the prefrontal and temporal cortex.[45]

Parkinson's disease

As Klotho has been shown to protect neurons against oxidative stress, it could be applied to the prevalent movement disorder in the elderly, Parkinson's disease (PD). Clinically, PD is characterized by deteriorating motor functions such as resting tremor, difficulty with coordination, muscle rigidity, stooping posture, and bradykinesia.[46] Increased oxidative stress, reduced antioxidant status, and inflammation mediated by protein kinase A (PKA)-dependent cascade play critical roles in PD pathogenesis.[47]

Loss of dopamine neurons in the substantia nigra pars compacta (SNC) is one of the main pathological features of PD.[48] The nigrostriatal pathway is a dopaminergic pathway that connects the SNC with the dorsal striatum. Baluchnejadmojarad et al. carried out an experiment to determine the ability of exogenous Klotho to mitigate the damage to nigrostriatal dopaminergic pathway in 6-hydroxydopamine (6-OHDA) rat model of PD. They demonstrated that pretreating rats with Klotho alleviated drug-induced rotational behavior of 6-OHDA-lesioned rats and improved their performance in narrow beam task. At the cellular level, Klotho prevented degeneration of tyrosine hydroxylase-positive neurons in the SNC. In addition, Klotho significantly ameliorated α-synuclein, glial fibrillary acidic protein, phosphorylated-cAMP-response element binding protein (CREB), DNA fragmentation, and oxidative stress. Moreover, the beneficial effect of Klotho was reportedly attenuated by administering a PKA inhibitor and Ca2+/calmodulin-dependent protein kinase II (CamKII) inhibitor. Thus, the neuroprotective effect of Klotho in 6-OHDA rat model of PD appears to be dependent on PKA/CaMKII/CREB signaling cascade.[49]

Synuclein is an important membrane protein in the pathogenesis of PD, which is responsible for regulating the exocytosis mechanisms. Overexpression of human α-synuclein (hSYN) in transgenic mice leads to accumulation of α-synuclein in neurons and synapses and manifests as motor and cognitive deficits in the mice, thereby simulating key aspects of neurodegenerative diseases. Leon et al. demonstrated that soluble Klotho could counteract cognitive deficits in this disease model. Moreover, peripheral Klotho treatment enhanced synaptic functions and brain resilience in these mice.[50] Thus, studying the protective effects of Klotho in mice can lead to potentially promising therapeutic strategies for humans in aging and neurodegenerative diseases.

 Conclusion



Neuronal cell death, particularly resulting from increased oxidative stress, has been implicated as a major risk factor for neurodegenerative diseases. The essential role of Klotho to counteract the same in healthy brain aging, its neuroprotective properties, and its ability to ameliorate neuronal damage associated with neurodegeneration have been evidenced in several studies. Klotho may even have immune-regulatory roles in the choroid plexus of the brain. Klotho-dependent pathways are impaired with normal aging but more so with neurodegenerative disease development. Thus, Klotho is instrumental in counteracting cognitive decline and age-related neurological disorders. The need of the hour is to investigate the therapeutic potential of Klotho for neurological disorders.

Financial support and sponsorship

The authors would like to acknowledge Sir H. N. Medical Research Society, Mumbai, India, for financial support.

Conflicts of interest

There are no conflicts of interest.

References

1Kuro-o M, Matsumura Y, Aizawa H, Kawaguchi H, Suga T, Utsugi T, et al. Mutation of the mouse Klotho gene leads to a syndrome resembling ageing. Nature 1997;390:45-51.
2Matsumura Y, Aizawa H, Shiraki-Iida T, Nagai R, Kuro-o M, Nabeshima Y. Identification of the human Klotho gene and its two transcripts encoding membrane and secreted Klotho protein. Biochem Biophys Res Commun 1998;242:626-30.
3Krajisnik T, Olauson H, Mirza MA, Hellman P, Akerström G, Westin G, et al. Parathyroid Klotho and FGF-receptor 1 expression decline with renal function in hyperparathyroid patients with chronic kidney disease and kidney transplant recipients. Kidney Int 2010;78:1024-32.
4Li SA, Watanabe M, Yamada H, Nagai A, Kinuta M, Takei K. Immunohistochemical localization of Klotho protein in brain, kidney, and reproductive organs of mice. Cell Struct Funct 2004;29:91-9.
5Kurosu H, Ogawa Y, Miyoshi M, Yamamoto M, Nandi A, Rosenblatt KP, et al. Regulation of fibroblast growth factor-23 signaling by Klotho. J Biol Chem 2006;281:6120-3.
6Bloch L, Sineshchekova O, Reichenbach D, Reiss K, Saftig P, Kuro-o M, et al. Klotho is a substrate for alpha-, beta- and gamma-secretase. FEBS Lett 2009;583:3221-4.
7Imura A, Iwano A, Tohyama O, Tsuji Y, Nozaki K, Hashimoto N, et al. Secreted Klotho protein in sera and CSF: Implication for post-translational cleavage in release of Klotho protein from cell membrane. FEBS Lett 2004;565:143-7.
8Gigante M, Lucarelli G, Divella C, Netti GS, Pontrelli P, Cafiero C, et al. Soluble serum αKlotho is a potential predictive marker of disease progression in clear cell renal cell carcinoma. Medicine (Baltimore) 2015;94:e1917.
9Saito Y, Nakamura T, Ohyama Y, Suzuki T, Iida A, Shiraki-Iida T, et al. In vivo Klotho gene delivery protects against endothelial dysfunction in multiple risk factor syndrome. Biochem Biophys Res Commun 2000;276:767-72.
10Semba RD, Cappola AR, Sun K, Bandinelli S, Dalal M, Crasto C, et al. Plasma Klotho and cardiovascular disease in adults. J Am Geriatr Soc 2011;59:1596-601.
11Nabeshima Y. Discovery of alpha-Klotho unveiled new insights into calcium and phosphate homeostasis. Proc Jpn Acad Ser B Phys Biol Sci 2009;85:125-41.
12Chang Q, Hoefs S, van der Kemp AW, Topala CN, Bindels RJ, Hoenderop JG. The beta-glucuronidase Klotho hydrolyzes and activates the TRPV5 channel. Science 2005;310:490-3.
13Kurosu H, Yamamoto M, Clark JD, Pastor JV, Nandi A, Gurnani P, et al. Suppression of aging in mice by the hormone Klotho. Science 2005;309:1829-33.
14Yamazaki Y, Imura A, Urakawa I, Shimada T, Murakami J, Aono Y, et al. Establishment of sandwich ELISA for soluble alpha-Klotho measurement: Age-dependent change of soluble alpha-Klotho levels in healthy subjects. Biochem Biophys Res Commun 2010;398:513-8.
15Duce JA, Podvin S, Hollander W, Kipling D, Rosene DL, Abraham CR. Gene profile analysis implicates Klotho as an important contributor to aging changes in brain white matter of the rhesus monkey. Glia 2008;56:106-17.
16King GD, Rosene DL, Abraham CR. Promoter methylation and age-related downregulation of Klotho in rhesus monkey. Age (Dordr) 2012;34:1405-19.
17Zhu L, Stein LR, Kim D, Ho K, Yu GQ, Zhan L, et al. Klotho controls the brain-immune system interface in the choroid plexus. Proc Natl Acad Sci U S A 2018;115:E11388-96.
18Shiozaki M, Yoshimura K, Shibata M, Koike M, Matsuura N, Uchiyama Y, et al. Morphological and biochemical signs of age-related neurodegenerative changes in Klotho mutant mice. Neuroscience 2008;152:924-41.
19Kim HK, Jeong BH. Lack of functional KL-VS polymorphism of the Klotho gene in the Korean population. Genet Mol Biol 2016;39:370-3.
20Nagai T, Yamada K, Kim HC, Kim YS, Noda Y, Imura A, et al. Cognition impairment in the genetic model of aging KLOTHO gene mutant mice: A role of oxidative stress. FASEB J 2003;17:50-2.
21Viswanathan A, Rocca WA, Tzourio C. Vascular risk factors and dementia: How to move forward? Neurology 2009;72:368-74.
22Schlögl M, Holick MF. Vitamin D and neurocognitive function. Clin Interv Aging 2014;9:559-68.
23Maji D. Vitamin D toxicity. Indian J Endocrinol Metab 2012;16:295-6.
24Ellis S, Tsiopanis G, Lad T. Risks of the 'sunshine pill' – A case of hypervitaminosis D. Clin Med (Lond) 2018;18:311-3.
25Yoshida T, Fujimori T, Nabeshima Y. Mediation of unusually high concentrations of 1,25-dihydroxy Vitamin D in homozygous Klotho mutant mice by increased expression of renal 1alpha-hydroxylase gene. Endocrinology 2002;143:683-9.
26Wang H, Yue J, Luo J, Tian P, Deng J. Expression of hippocampus Klotho protein and insulin-like growth factor-1 in rats with dementia. Sheng Wu Yi Xue Gong Cheng Xue Za Zhi 2013;30:808-11.
27Dubal DB, Yokoyama JS, Zhu L, Broestl L, Worden K, Wang D, et al. Life extension factor Klotho enhances cognition. Cell Rep 2014;7:1065-76.
28Salech F, Varela-Nallar L, Arredondo SB, Bustamante DB, Andaur GA, Cisneros R, et al. Local Klotho enhances neuronal progenitor proliferation in the adult hippocampus. J Gerontol A Biol Sci Med Sci 2017. doi: 10.1093/gerona/glx248. [Epub ahead of print].
29Laszczyk AM, Fox-Quick S, Vo HT, Nettles D, Pugh PC, Overstreet-Wadiche L, et al. Klotho regulates postnatal neurogenesis and protects against age-related spatial memory loss. Neurobiol Aging 2017;59:41-54.
30Zhou HJ, Zeng CY, Yang TT, Long FY, Kuang X, Du JR. Lentivirus-mediated Klotho up-regulation improves aging-related memory deficits and oxidative stress in senescence-accelerated mouse prone-8 mice. Life Sci 2018;200:56-62.
31Kuang X, Yao Y, Du JR, Liu YX, Wang CY, Qian ZM. Neuroprotective role of Z-ligustilide against forebrain ischemic injury in ICR mice. Brain Res 2006;1102:145-53.
32Long FY, Shi MQ, Zhou HJ, Liu DL, Sang N, Du JR. Klotho upregulation contributes to the neuroprotection of ligustilide against cerebral ischemic injury in mice. Eur J Pharmacol 2018;820:198-205.
33Shardell M, Semba RD, Rosano C, Kalyani RR, Bandinelli S, Chia CW, et al. Plasma Klotho and cognitive decline in older adults: Findings from the InCHIANTI study. J Gerontol A Biol Sci Med Sci 2016;71:677-82.
34Zou T, Zhou X. The relationship of serum Klotho protein and mild cognitive impairment. Alzheimers Dement J Alzheimers Assoc 2014;11:P639-40.
35Luo M, Zhou X, Ji H, Ma W, Liu G, Dai D, et al. Population difference in the associations of KLOTH promoter methylation with mild cognitive impairment in Xinjiang Uygur and Han populations. PLoS One 2015;10:e0132156.
36Chen CD, Sloane JA, Li H, Aytan N, Giannaris EL, Zeldich E, et al. The antiaging protein Klotho enhances oligodendrocyte maturation and myelination of the CNS. J Neurosci 2013;33:1927-39.
37Emami Aleagha MS, Siroos B, Ahmadi M, Balood M, Palangi A, Haghighi AN, et al. Decreased concentration of Klotho in the cerebrospinal fluid of patients with relapsing-remitting multiple sclerosis. J Neuroimmunol 2015;281:5-8.
38Muthian G, Raikwar HP, Rajasingh J, Bright JJ. 1,25 dihydroxyvitamin-D3 modulates JAK-STAT pathway in IL-12/IFNgamma axis leading to th1 response in experimental allergic encephalomyelitis. J Neurosci Res 2006;83:1299-309.
39Ellidag HY, Yilmaz N, Kurtulus F, Aydin O, Eren E, Inci A, et al. The three sisters of fate in multiple sclerosis: Klotho (Clotho), fibroblast growth factor-23 (Lachesis), and Vitamin D (Atropos). Ann Neurosci 2016;23:155-61.
40Ahmadi M, Emami Aleagha MS, Harirchian MH, Yarani R, Tavakoli F, Siroos B. Multiple sclerosis influences on the augmentation of serum Klotho concentration. J Neurol Sci 2016;362:69-72.
41Li H, Wang B, Wang Z, Guo Q, Tabuchi K, Hammer RE, et al. Soluble amyloid precursor protein (APP) regulates transthyretin and Klotho gene expression without rescuing the essential function of APP. Proc Natl Acad Sci U S A 2010;107:17362-7.
42Oh ES, Savonenko AV, King JF, Fangmark Tucker SM, Rudow GL, Xu G, et al. Amyloid precursor protein increases cortical neuron size in transgenic mice. Neurobiol Aging 2009;30:1238-44.
43Zeldich E, Chen CD, Colvin TA, Bove-Fenderson EA, Liang J, Tucker Zhou TB, et al. The neuroprotective effect of Klotho is mediated via regulation of members of the redox system. J Biol Chem 2014;289:24700-15.
44Semba RD, Moghekar AR, Hu J, Sun K, Turner R, Ferrucci L, et al. Klotho in the cerebrospinal fluid of adults with and without Alzheimer's disease. Neurosci Lett 2014;558:37-40.
45Yokoyama JS, Marx G, Brown JA, Bonham LW, Wang D, Coppola G, et al. Systemic Klotho is associated with Klotho variation and predicts intrinsic cortical connectivity in healthy human aging. Brain Imaging Behav 2017;11:391-400.
46Sveinbjornsdottir S. The clinical symptoms of Parkinson's disease. J Neurochem 2016;139 Suppl 1:318-24.
47Mosley RL, Benner EJ, Kadiu I, Thomas M, Boska MD, Hasan K, et al. Neuroinflammation, oxidative stress and the pathogenesis of Parkinson's disease. Clin Neurosci Res 2006;6:261-81.
48Dauer W, Przedborski S. Parkinson's disease: Mechanisms and models. Neuron 2003;39:889-909.
49Baluchnejadmojarad T, Eftekhari SM, Jamali-Raeufy N, Haghani S, Zeinali H, Roghani M, et al. The anti-aging protein Klotho alleviates injury of nigrostriatal dopaminergic pathway in 6-hydroxydopamine rat model of Parkinson's disease: Involvement of PKA/CaMKII/CREB signaling. Exp Gerontol 2017;100:70-6.
50Leon J, Moreno AJ, Garay BI, Chalkley RJ, Burlingame AL, Wang D, et al. Peripheral elevation of a Klotho fragment enhances brain function and resilience in young, aging, and α-synuclein transgenic mice. Cell Rep 2017;20:1360-71.