1
|
Gao X, Sun Z, Hu J, Li Y, Deng Q, Li R. Identification of the enzymatic cleavage relationship between anti-aging protein α-Klotho and Alzheimer's disease biomarker BACE1. J Alzheimers Dis 2025; 104:463-472. [PMID: 39994980 DOI: 10.1177/13872877251317730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
BackgroundThe α-Klotho is known to be involved in longevity and various age-related diseases, including cognitive impairment. BACE1, an important enzyme associated with the pathological process of Alzheimer's disease (AD), serves as a biomarker for predicting changes in cognitive function. Although both proteins are closely linked to age-related cognitive function, the mechanism of their interaction remains unclear.ObjectiveTo identify the enzymatic digestion relation between α-Klotho and BACE1 and the specific cleavage site.MethodsThirty elderly and forty-five young individuals were recruited. The cleavage product was identified by Coomassie blue staining, western blot, and MALDI-TOF mass spectrometry. The concentrations of plasma proteins were measured by ELISA.ResultsA new protein product was identified after the digestion reaction. BACE1 cleaved the α-Klotho peptide 951-981 at the F-T residues. When the F-T residues were replaced with K-K, BACE1 was unable to cleave the mutant peptide. The plasma levels of α-Klotho were significantly lower in elderly participants than in young participants (p < 0.0001). However, there was no significant difference in plasma BACE1 levels between elderly and young participants (p = 0.164). In elderly adults, there was a significant positive correlation between plasma BACE1 and α-Klotho protein levels (p = 0.009, r = 0.469), while this correlation was not observed in young adults (p = 0.170, r = -0.208).ConclusionsThe anti-aging protein α-Klotho is a substrate of BACE1 with a specific cleavage site at F-T. The BACE1/α-Klotho pathway may serve as a common axis for age-related cognitive decline.
Collapse
Affiliation(s)
- Xiang Gao
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
- Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - Zuoli Sun
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Jia Hu
- Central Laboratory, Capital Medical University, Beijing, China
| | - Yuhong Li
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Qi Deng
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Rena Li
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders & National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
2
|
Shibata K, Chen C, Tai XY, Manohar SG, Husain M. Impact of APOE, Klotho, and sex on cognitive decline with aging. Proc Natl Acad Sci U S A 2025; 122:e2416042122. [PMID: 39903109 PMCID: PMC11831164 DOI: 10.1073/pnas.2416042122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 12/28/2024] [Indexed: 02/06/2025] Open
Abstract
The effects of apolipoprotein E (APOE) and Klotho genes, both implicated in aging, on human cognition as a function of sex and age are yet to be definitively established. Here, we showed in the largest cohort studied to date (N = 320,861) that APOE homozygous ε4 carriers had a greater decline in cognition with aging compared to ε3 carriers (ε3/ε4 and ε3/ε3) as well as smaller hippocampi and amygdala (N = 29,510). Critically, sex and age differentially affected the decline in cognition. Younger (40 to 50 y) female homozygous ε4 carriers showed a cognitive advantage over female ε3 carriers, but this advantage was not present in males. By contrast, Klotho-VS heterozygosity did not affect cognition or brain volume, regardless of APOE genotype, sex, or age. These cognitive trajectories with aging demonstrate clear sex-dependent antagonistic pleiotropy effects of APOE ε4, but no effects of Klotho genotype on cognition and brain volume.
Collapse
Affiliation(s)
- Kengo Shibata
- Nuffield Department of Clinical Neurosciences, University of Oxford, OxfordOX3 9DU, United Kingdom
| | - Cheng Chen
- Department of Experimental Psychology, University of Oxford, OxfordOX2 6GG, United Kingdom
| | - Xin You Tai
- Nuffield Department of Clinical Neurosciences, University of Oxford, OxfordOX3 9DU, United Kingdom
- Division of Clinical Neurology, John Radcliffe Hospital, Oxford University Hospitals Trust, OxfordOX3 9DU, United Kingdom
| | - Sanjay G. Manohar
- Nuffield Department of Clinical Neurosciences, University of Oxford, OxfordOX3 9DU, United Kingdom
- Department of Experimental Psychology, University of Oxford, OxfordOX2 6GG, United Kingdom
- Division of Clinical Neurology, John Radcliffe Hospital, Oxford University Hospitals Trust, OxfordOX3 9DU, United Kingdom
| | - Masud Husain
- Nuffield Department of Clinical Neurosciences, University of Oxford, OxfordOX3 9DU, United Kingdom
- Department of Experimental Psychology, University of Oxford, OxfordOX2 6GG, United Kingdom
- Division of Clinical Neurology, John Radcliffe Hospital, Oxford University Hospitals Trust, OxfordOX3 9DU, United Kingdom
| |
Collapse
|
3
|
Errigo A, Dore MP, Mocci G, Pes GM. Lack of association between common polymorphisms associated with successful aging and longevity in the population of Sardinian Blue Zone. Sci Rep 2024; 14:30773. [PMID: 39730495 DOI: 10.1038/s41598-024-80497-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 11/19/2024] [Indexed: 12/29/2024] Open
Abstract
More than two decades ago, in the central-eastern region of the Mediterranean island of Sardinia, a mountain area was identified where the population displays exceptional longevity, especially among men (the Longevity Blue Zone, LBZ). This community was thoroughly investigated to understand the underlying causes of the phenomenon. The present study analyzed 11 genetic markers previously associated with increased survival in several long-lived populations. APOE (rs429358 and rs7412), APOE promoter (rs449647, rs769446, and rs405509), ACE1 (rs1799752), IL6 ‒174G/C (rs1800795), TNFα ‒308G/A (rs1800629), FOXO3A (rs2802292), KLOTHO (rs9536314) and G6PD (rs5030868) polymorphisms were investigated. PCR-based genotyping was performed following genomic DNA extraction from 150 nonagenarians living in the LBZ and 150 controls from a nearby area. No significant deviation in the frequency of the analyzed markers was detected between the two subgroups except for a weak association with the - 174G > C gene variant in the IL-6 gene (p = 0.040), which codes for a major modulator of the inflammatory response. Overall, the findings of this study do not support a significant association of known genetic variants on survival in the population of the Sardinian LBZ, suggesting that other genetic or epigenetic traits not yet identified might play a role.
Collapse
Affiliation(s)
- Alessandra Errigo
- Dipartimento di Medicina, Chirurgia e Farmacia, University of Sassari, Viale San Pietro 43, Sassari, 07100, Italy
| | - Maria Pina Dore
- Dipartimento di Medicina, Chirurgia e Farmacia, University of Sassari, Viale San Pietro 43, Sassari, 07100, Italy
- Baylor College of Medicine, One Baylor Plaza Blvd, Houston, TX, 77030, USA
| | - Giammarco Mocci
- Division of Gastroenterology, "Brotzu" Hospital, Cagliari, Italy
| | - Giovanni Mario Pes
- Dipartimento di Medicina, Chirurgia e Farmacia, University of Sassari, Viale San Pietro 43, Sassari, 07100, Italy.
- Sardinia Longevity Blue Zone Observatory, Ogliastra, 08040, Italy.
| |
Collapse
|
4
|
Cook N, Driscoll I, Gaitán JM, Glittenberg M, Betthauser TJ, Carlsson CM, Johnson SC, Asthana S, Zetterberg H, Blennow K, Kollmorgen G, Quijano-Rubio C, Dubal DB, Okonkwo OC. Amyloid-β positivity is less prevalent in cognitively unimpaired KLOTHO KL-VS heterozygotes. J Alzheimers Dis 2024; 102:480-490. [PMID: 39529379 PMCID: PMC12025201 DOI: 10.1177/13872877241289785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
BACKGROUND Klotho, encoded by the KLOTHO gene, is an anti-aging and neuroprotective protein. KLOTHO KL-VS heterozygosity (KL-VSHET) is hypothesized to be protective against the accumulation of Alzheimer's disease (AD) neuropathological hallmarks (amyloid-β (Aβ) and tau). OBJECTIVE We examine whether being positive for Aβ (A+) or tau (T+), or A/T joint status [positive for Aβ (A + T-), tau (A-T+), both (A + T+) or neither (A-T-)] vary by KL-VS and whether serum klotho protein levels vary based on A+, T+, or A/T status in a cohort enriched for AD risk. METHODS The sample consisted of 704 cognitively unimpaired, late middle-aged, and older adults; MeanAge(SD) = 64.9(8.3). Serum klotho was available for a sub-sample of 396 participants; MeanAge(SD) = 66.8(7.4). Covariate-adjusted logistic regression examined whether A + or T+, and multinomial regression examined whether A/T status, vary by KL-VS genotype. Covariate-adjusted linear regression examined whether serum klotho levels differ based on A+, T+, or A/T status. RESULTS A+ prevalence was lower in KL-VSHET (p = 0.05), with no differences in T + prevalence (p = 0.52). KL-VSHET also had marginally lower odds of being A + T- (p = 0.07). Serum klotho levels did not differ based on A+, T+, or A/T status (all ps ≥ 0.40). CONCLUSIONS KL-VSHET is associated with lower odds of being positive for Aβ, regardless of whether one is also positive for tau. Conversely, the likelihood of being tau positive did not differ based on KL-VS genotype. Our findings add to the growing KLOTHO literature and suggests the need for further research focused on understanding the mechanisms underlying KL-VS-related putative resilience to AD.
Collapse
Affiliation(s)
- Noah Cook
- Department of Medicine and Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Department of Population Health Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Ira Driscoll
- Department of Medicine and Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Wisconsin Alzheimer’s Institute, Madison, WI, USA
| | - Julian M. Gaitán
- Department of Medicine and Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Matthew Glittenberg
- Department of Medicine and Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Tobey J. Betthauser
- Department of Medicine and Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Cynthia M. Carlsson
- Department of Medicine and Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Wisconsin Alzheimer’s Institute, Madison, WI, USA
- Geriatric Research Education and Clinical Center, William S. Middleton VA Hospital, Madison, WI, USA
| | - Sterling C. Johnson
- Department of Medicine and Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Wisconsin Alzheimer’s Institute, Madison, WI, USA
| | - Sanjay Asthana
- Department of Medicine and Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Wisconsin Alzheimer’s Institute, Madison, WI, USA
- Geriatric Research Education and Clinical Center, William S. Middleton VA Hospital, Madison, WI, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | | | | | - Dena B. Dubal
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Ozioma C. Okonkwo
- Department of Medicine and Alzheimer’s Disease Research Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Wisconsin Alzheimer’s Institute, Madison, WI, USA
- Geriatric Research Education and Clinical Center, William S. Middleton VA Hospital, Madison, WI, USA
| |
Collapse
|
5
|
Sawada KM, de Moraes NS, Araújo LMQ, Gazoni FM, Lazaretti-Castro M, Cendoroglo MS, Bilezikian JP, Figueiredo MS, dos Santos FC. Blood concentrations of α-Klotho and FGF-23 exhibit no correlation with bone mineral density in elderly individuals. EINSTEIN-SAO PAULO 2024; 22:eAO0412. [PMID: 39319957 PMCID: PMC11461005 DOI: 10.31744/einstein_journal/2024ao0412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/21/2024] [Indexed: 09/26/2024] Open
Abstract
OBJECTIVE To investigating the relationship between α-Klotho and FGF-23 with bone biochemical markers and bone density findings in extremely aged individuals. METHODS A total of 55 individuals with a mean age of 85.6 years were subjected to clinical, biochemical, and bone mineral density analyses and the enzyme-linked immunosorbent assay-based detection of α-Klotho and FGF-23. The mean, standard deviation, median, and interquartile ranges of the sample values were determined, and Spearman's test for association assessments was used for statistical analysis. RESULTS The study participants expressed median FGF-23 and α-Klotho levels of 69.81 RU/mL (51.43 RU/mL) and 733.43 pg/mL (360.83 pg/mL), respectively. The majority of the participants possessed osteopenia (54.5%) and a vitamin D deficiency (57%). The 25-hydroxyvitamin D concentrations ranged between 7.1 and 47.5ng/mL, with a median of 18.1ng/mL. CONCLUSION No substantial associations were discovered between α-Klotho and FGF-23 levels and bone density in the study participants.
Collapse
Affiliation(s)
- Karina Moura Sawada
- Universidade Federal de São PauloEscola Paulista de MedicinaDepartment of MedicineSão PauloSPBrazilDiscipline of Geriatrics, Department of Medicine, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil.
| | - Niele Silva de Moraes
- Universidade Federal de São PauloEscola Paulista de MedicinaDepartment of MedicineSão PauloSPBrazilDiscipline of Geriatrics, Department of Medicine, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil.
| | - Lara Miguel Quirino Araújo
- Universidade Federal de São PauloEscola Paulista de MedicinaDepartment of MedicineSão PauloSPBrazilDiscipline of Geriatrics, Department of Medicine, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil.
| | - Fernanda Martins Gazoni
- Universidade Federal de São PauloEscola Paulista de MedicinaDepartment of MedicineSão PauloSPBrazilDiscipline of Geriatrics, Department of Medicine, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil.
| | - Marise Lazaretti-Castro
- Universidade Federal de São PauloEscola Paulista de MedicinaDepartment of MedicineSão PauloSPBrazilDiscipline of Endocrinology, Department of Medicine, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil.
| | - Maysa Seabra Cendoroglo
- Universidade Federal de São PauloEscola Paulista de MedicinaDepartment of MedicineSão PauloSPBrazilDiscipline of Geriatrics, Department of Medicine, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil.
| | - John P. Bilezikian
- Columbia UniversityCollege of Physicians and SurgeonsDepartment of MedicineNew YorkNew YorkUSDivision of Endocrinology, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York.
| | - Maria Stella Figueiredo
- Universidade Federal de São PauloEscola Paulista de MedicinaDepartment of Clinical and Experimental OncologySão PauloSPBrazilDiscipline of Hematology and Hemotherapy, Department of Clinical and Experimental Oncology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil.
| | - Fania Cristina dos Santos
- Universidade Federal de São PauloEscola Paulista de MedicinaDepartment of MedicineSão PauloSPBrazilDiscipline of Geriatrics, Department of Medicine, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
6
|
Münz S, Wolf L, Hoelzle LE, Chernyakov D, Edemir B, Föller M. Impact of cytotoxic agents or apoptosis stimulants on αklotho in MDCK, NRK-52E and HK2 kidney cells. Aging (Albany NY) 2022; 14:7282-7299. [PMID: 35997650 PMCID: PMC9550246 DOI: 10.18632/aging.204238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 08/09/2022] [Indexed: 11/25/2022]
Abstract
αKlotho is a transmembrane protein acting as a co-receptor for FGF23, a bone hormone regulating renal phosphate and vitamin D metabolism. αKlotho expression is controlled by PPARγ. Soluble αklotho (sKL) regulates cellular signaling impacting stress resistance and death. αKlotho deficiency causes early onset of aging-associated diseases while its overexpression markedly increases lifespan. Cellular stress due to cytotoxic therapeutics or apoptosis induction through caspase activation or serum deficiency may result in cell death. Owing to αklotho's role in cellular stress and aging, this study explored the effect of cytotoxic agents or apoptosis stimulants on cellular αklotho expression. Experiments were performed in renal MDCK, NRK-52E and HK-2 cells. Gene expression was determined by qRT-PCR, sKL by ELISA, apoptosis and necrosis by annexin V binding and a fluorescent DNA dye, and cell viability by MTT assay. Cytostatic drugs cisplatin, paclitaxel, and doxorubicin as well as apoptosis induction with caspase 3 activator PAC-1 and serum deprivation induced αklotho and PPARG gene expression while decreasing viability and proliferation and inducing apoptosis of MDCK and NRK-52E cells to a variable extent. PPARγ antagonism attenuated up-regulation of αklotho in MDCK cells. In HK-2 cells, αklotho gene expression and sKL protein were down-regulated by chemotherapeutics. SKL serum levels in patients following chemotherapy were not significantly changed. In summary, potentially fatal stress results in up-regulation of αKlotho gene expression in MDCK and NRK-52E cells and down-regulation in HK-2 cells. These results indicate that different renal cell lines may exhibit completely different regulation of αklotho.
Collapse
Affiliation(s)
- Sina Münz
- Department of Physiology, University of Hohenheim, Stuttgart 70599, Germany
| | - Lisa Wolf
- Department of Physiology, University of Hohenheim, Stuttgart 70599, Germany
| | - Ludwig E Hoelzle
- Institute of Animal Science, University of Hohenheim, Stuttgart 70599, Germany
| | - Dmitry Chernyakov
- Department of Oncology, Martin-Luther-University Halle-Wittenberg, Halle (Saale) 06120, Germany
| | - Bayram Edemir
- Department of Oncology, Martin-Luther-University Halle-Wittenberg, Halle (Saale) 06120, Germany
| | - Michael Föller
- Department of Physiology, University of Hohenheim, Stuttgart 70599, Germany
| |
Collapse
|
7
|
Ali M, Sung YJ, Wang F, Fernández MV, Morris JC, Fagan AM, Blennow K, Zetterberg H, Heslegrave A, Johansson PM, Svensson J, Nellgård B, Lleó A, Alcolea D, Clarimon J, Rami L, Molinuevo JL, Suárez-Calvet M, Morenas-Rodríguez E, Kleinberger G, Haass C, Ewers M, Levin J, Farlow MR, Perrin RJ, Cruchaga C. Leveraging large multi-center cohorts of Alzheimer disease endophenotypes to understand the role of Klotho heterozygosity on disease risk. PLoS One 2022; 17:e0267298. [PMID: 35617280 PMCID: PMC9135221 DOI: 10.1371/journal.pone.0267298] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/06/2022] [Indexed: 11/18/2022] Open
Abstract
Two genetic variants in strong linkage disequilibrium (rs9536314 and rs9527025) in the Klotho (KL) gene, encoding a transmembrane protein, implicated in longevity and associated with brain resilience during normal aging, were recently shown to be associated with Alzheimer disease (AD) risk in cognitively normal participants who are APOE ε4 carriers. Specifically, the participants heterozygous for this variant (KL-SVHET+) showed lower risk of developing AD. Furthermore, a neuroprotective effect of KL-VSHET+ has been suggested against amyloid burden for cognitively normal participants, potentially mediated via the regulation of redox pathways. However, inconsistent associations and a smaller sample size of existing studies pose significant hurdles in drawing definitive conclusions. Here, we performed a well-powered association analysis between KL-VSHET+ and five different AD endophenotypes; brain amyloidosis measured by positron emission tomography (PET) scans (n = 5,541) or cerebrospinal fluid Aβ42 levels (CSF; n = 5,093), as well as biomarkers associated with tau pathology: the CSF Tau (n = 5,127), phosphorylated Tau (pTau181; n = 4,778) and inflammation: CSF soluble triggering receptor expressed on myeloid cells 2 (sTREM2; n = 2,123) levels. Our results found nominally significant associations of KL-VSHET+ status with biomarkers for brain amyloidosis (e.g., CSF Aβ positivity; odds ratio [OR] = 0.67 [95% CI, 0.55-0.78], β = 0.72, p = 0.007) and tau pathology (e.g., biomarker positivity for CSF Tau; OR = 0.39 [95% CI, 0.19-0.77], β = -0.94, p = 0.007, and pTau; OR = 0.50 [95% CI, 0.27-0.96], β = -0.68, p = 0.04) in cognitively normal participants, 60-80 years old, who are APOE e4-carriers. Our work supports previous findings, suggesting that the KL-VSHET+ on an APOE ε4 genotype background may modulate Aβ and tau pathology, thereby lowering the intensity of neurodegeneration and incidence of cognitive decline in older controls susceptible to AD.
Collapse
Affiliation(s)
- Muhammad Ali
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Neurogenomics and Informatics Center, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Yun Ju Sung
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Neurogenomics and Informatics Center, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Fengxian Wang
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Neurogenomics and Informatics Center, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Maria V. Fernández
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Neurogenomics and Informatics Center, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - John C. Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Anne M. Fagan
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Department of Neuroscience and Physiology, University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Department of Neuroscience and Physiology, University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
| | - Amanda Heslegrave
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
| | - Per M. Johansson
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
- Department of Anesthesiology and Intensive Care Medicine, Sahlgrenska University Hospital, Mölndal, Sweden
- Institute of Clinical Sciences, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Johan Svensson
- Department of Internal Medicine, Institute of Medicine, The Sahlgrenska Academy at the University of Gothenburg, Göteborg, Sweden
| | - Bengt Nellgård
- Department of Internal Medicine, Institute of Medicine, The Sahlgrenska Academy at the University of Gothenburg, Göteborg, Sweden
| | - Alberto Lleó
- Neurology Department, Hospital de Sant Pau, Barcelona, Spain
| | - Daniel Alcolea
- Neurology Department, Hospital de Sant Pau, Barcelona, Spain
| | - Jordi Clarimon
- Neurology Department, Hospital de Sant Pau, Barcelona, Spain
| | - Lorena Rami
- IDIBAPS, Alzheimer´s Disease and Other Cognitive Disorders Unit, Neurology Service, ICN Hospital Clinic, Barcelona, Spain
| | - José Luis Molinuevo
- IDIBAPS, Alzheimer´s Disease and Other Cognitive Disorders Unit, Neurology Service, ICN Hospital Clinic, Barcelona, Spain
- Alzheimer´s Disease and Other Cognitive Disorders Unit, Neurology Service, ICN Hospital Clinic i Universitari, Barcelona, Spain
- BarcelonaBeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
| | - Marc Suárez-Calvet
- BarcelonaBeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
- Biomedical Center (BMC), Biochemistry, Ludwig‐Maximilians‐Universität München, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Estrella Morenas-Rodríguez
- Biomedical Center (BMC), Biochemistry, Ludwig‐Maximilians‐Universität München, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Gernot Kleinberger
- Biomedical Center (BMC), Biochemistry, Ludwig‐Maximilians‐Universität München, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Christian Haass
- Biomedical Center (BMC), Biochemistry, Ludwig‐Maximilians‐Universität München, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Michael Ewers
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Martin R. Farlow
- Indiana Alzheimer Disease Research Center, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Richard J. Perrin
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | | | | | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Neurogenomics and Informatics Center, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|
8
|
Gaitán JM, Asthana S, Carlsson CM, Engelman CD, Johnson SC, Sager MA, Wang D, Dubal DB, Okonkwo OC. Circulating Klotho Is Higher in Cerebrospinal Fluid than Serum and Elevated Among KLOTHO Heterozygotes in a Cohort with Risk for Alzheimer's Disease. J Alzheimers Dis 2022; 90:1557-1569. [PMID: 36314202 PMCID: PMC10139824 DOI: 10.3233/jad-220571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Klotho is a longevity and neuroprotective hormone encoded by the KLOTHO gene, and heterozygosity for the KL-VS variant confers a protective effect against neurodegenerative disease. OBJECTIVE Test whether klotho concentrations in serum or cerebrospinal fluid (CSF) vary as a function of KLOTHO KL-VS genotype, determine whether circulating klotho concentrations from serum and CSF differ from one another, and evaluate whether klotho levels are associated with Alzheimer's disease risk factors. METHODS Circulating klotho was measured in serum (n = 1,116) and CSF (n = 183) of cognitively intact participants (aged 62.4 ± 6.5 years; 69.5% female). KLOTHO KL-VS zygosity (non-carrier; heterozygote; homozygote) was also determined. Linear regression was used to test whether klotho hormone concentration varied as a function of KL-VS genotype, specimen source, and demographic and clinical characteristics. RESULTS Serum and CSF klotho were higher in KL-VS carriers than non-carriers. Klotho concentration was higher in CSF than in serum. Females had higher serum and CSF klotho, while younger age was associated with higher klotho in CSF. CONCLUSION In a cohort enriched for risk for Alzheimer's disease, heterozygotic and homozygotic carriers of the KL-VS allele, females, and younger individuals have higher circulating klotho. Fluid source, KL-VS genotype, age, and sex should be considered in analyses of circulating klotho on brain health.
Collapse
Affiliation(s)
- Julian M. Gaitán
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, 600 Highland Ave., Madison, WI 53792 USA
| | - Sanjay Asthana
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, 600 Highland Ave., Madison, WI 53792 USA
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, 610 Walnut St. Suite 957, Madison, WI 53726, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, 2500 Overlook Terrace, Madison, WI 53705, USA
| | - Cynthia M. Carlsson
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, 600 Highland Ave., Madison, WI 53792 USA
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, 610 Walnut St. Suite 957, Madison, WI 53726, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, 2500 Overlook Terrace, Madison, WI 53705, USA
| | - Corinne D. Engelman
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, 600 Highland Ave., Madison, WI 53792 USA
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, 610 Walnut St. Suite 957, Madison, WI 53726, USA
- Department of Population Health Sciences, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, 610 Walnut St. Suite 707, Madison, WI 53726, USA
| | - Sterling C. Johnson
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, 600 Highland Ave., Madison, WI 53792 USA
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, 610 Walnut St. Suite 957, Madison, WI 53726, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, 2500 Overlook Terrace, Madison, WI 53705, USA
| | - Mark A. Sager
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, 600 Highland Ave., Madison, WI 53792 USA
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, 610 Walnut St. Suite 957, Madison, WI 53726, USA
| | - Dan Wang
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA 94158, USA
| | - Dena B. Dubal
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA 94158, USA
| | - Ozioma C. Okonkwo
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, 600 Highland Ave., Madison, WI 53792 USA
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, 610 Walnut St. Suite 957, Madison, WI 53726, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, 2500 Overlook Terrace, Madison, WI 53705, USA
| |
Collapse
|
9
|
Amin HA, Cordell HJ, Martin-Ruiz C, Robinson L, Kirkwood T, Blakemore AI, Drenos F. No Evidence That Genetic Variation At The Klotho Locus Is Associated With Longevity In Caucasians From The Newcastle 85 Plus Study And The Uk Biobank. J Gerontol A Biol Sci Med Sci 2021; 77:457-461. [PMID: 34893828 PMCID: PMC8893196 DOI: 10.1093/gerona/glab361] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Indexed: 11/24/2022] Open
Abstract
The demographics of Western populations are changing, with an increase in the proportion of older adults. There is evidence to suggest that genetic factors may influence the aging process: studying these may lead to interventions to help individuals live a longer and healthier life. Evidence from several groups indicates that Klotho (KL), a gene encoding a single-pass transmembrane protein that acts as an FGF23 co-receptor, may be associated with longevity and healthy aging. We aimed to explore this area further by comparing the genotype counts in 642 long-lived individuals from the Newcastle 85+ Study with 18 295 middle-aged Newcastle-based controls from the UK Biobank to test whether variants at the KL gene locus are over- or under-represented in older individuals. If KL is associated with longevity, then we would expect the genotype counts to differ between the 2 cohorts. We found that the rs2283368 CC genotype and the rs9536338 C allele, but not the KL-VS haplotype, were associated with reaching very old age. However, these associations did not replicate in the remainder of the UK Biobank cohort. Thus, our results do not reliably support the role of KL as a longevity factor.
Collapse
Affiliation(s)
- Hasnat A Amin
- Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Heather J Cordell
- Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne, United Kingdom
| | - Carmen Martin-Ruiz
- BioScreening Core Facility, Biosciences Institute, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Louise Robinson
- Population Health Sciences Institute, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Tom Kirkwood
- Ageing Research Laboratories, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Alexandra I Blakemore
- Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom.,Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
| | | |
Collapse
|
10
|
Gagan JM, Cao K, Zhang YA, Zhang J, Davidson TL, Pastor JV, Moe OW, Hsia CCW. Constitutive transgenic alpha-Klotho overexpression enhances resilience to and recovery from murine acute lung injury. Am J Physiol Lung Cell Mol Physiol 2021; 321:L736-L749. [PMID: 34346778 DOI: 10.1152/ajplung.00629.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
AIMS Normal lungs do not express alpha-Klotho (Klotho) protein but derive cytoprotection from circulating soluble Klotho. It is unclear whether chronic supranormal Klotho levels confer additional benefit. To address this, we tested the age-related effects of Klotho overexpression on acute lung injury (ALI) and recovery. METHODS Transgenic Klotho-overexpressing (Tg-Kl) and wild-type (WT) mice (2 and 6 months old) were exposed to hyperoxia (95% O2; 72 h) then returned to normoxia (21% O2; 24 h) (Hx-R). Control mice were kept in normoxia. Renal and serum Klotho, lung histology, and bronchoalveolar lavage fluid oxidative damage markers were assessed. Effects of hyperoxia were tested in human embryonic kidney cells stably expressing Klotho. A549 lung epithelial cells transfected with Klotho cDNA or vector were exposed to cigarette smoke; lactate dehydrogenase and double-strand DNA breaks were measured. RESULTS Serum Klotho decreased with age. Hyperoxia suppressed renal Klotho at both ages and serum Klotho at 2-months of age. Tg-Kl mice at both ages and 2-months-old WT mice survived Hx-R; 6-months-old Tg-Kl mice showed lower lung damage than age-matched WT mice. Hyperoxia directly inhibited Klotho expression and release in vitro; Klotho transfection attenuated cigarette smoke-induced cytotoxicity and DNA double-strand breaks in lung epithelial cells. CONCLUSIONS Young animals with chronic high baseline Klotho expression are more resistant to ALI. Chronic constitutive Klotho overexpression in older Tg-Kl animals attenuates hyperoxia-induced lung damage and improves survival and short-term recovery despite an acute reduction in serum Klotho level during injury. We conclude that chronic enhancement of Klotho expression increases resilience to ALI.
Collapse
Affiliation(s)
- Joshuah M Gagan
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Khoa Cao
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Yu-An Zhang
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Jianning Zhang
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Taylor L Davidson
- Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Johanne V Pastor
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, United States.,Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Orson W Moe
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, United States.,Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, The University of Texas Southwestern Medical Center, Dallas, TX, United States.,Department of Physiology, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Connie C W Hsia
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
11
|
Klotho inhibits neuronal senescence in human brain organoids. NPJ Aging Mech Dis 2021; 7:18. [PMID: 34341344 PMCID: PMC8329278 DOI: 10.1038/s41514-021-00070-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 05/28/2021] [Indexed: 02/05/2023] Open
Abstract
Aging is a major risk factor for many neurodegenerative diseases. Klotho (KL) is a glycosylated transmembrane protein that is expressed in the choroid plexus and neurons of the brain. KL exerts potent anti-aging effects on multiple cell types in the body but its role in human brain cells remains largely unclear. Here we show that human cortical neurons, derived from human pluripotent stem cells in 2D cultures or in cortical organoids, develop the typical hallmarks of senescent cells when maintained in vitro for prolonged periods of time, and that moderate upregulation or repression of endogenous KL expression in cortical organoids inhibits and accelerates senescence, respectively. We further demonstrate that KL expression alters the expression of senescence-associated genes including, extracellular matrix genes, and proteoglycans, and can act in a paracrine fashion to inhibit neuronal senescence. In summary, our results establish an important role for KL in the regulation of human neuronal senescence and offer new mechanistic insight into its role in human brain aging.
Collapse
|
12
|
Belloy ME, Napolioni V, Han SS, Le Guen Y, Greicius MD. Association of Klotho-VS Heterozygosity With Risk of Alzheimer Disease in Individuals Who Carry APOE4. JAMA Neurol 2021; 77:849-862. [PMID: 32282020 DOI: 10.1001/jamaneurol.2020.0414] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Importance Identification of genetic factors that interact with the apolipoprotein e4 (APOE4) allele to reduce risk for Alzheimer disease (AD) would accelerate the search for new AD drug targets. Klotho-VS heterozygosity (KL-VSHET+ status) protects against aging-associated phenotypes and cognitive decline, but whether it protects individuals who carry APOE4 from AD remains unclear. Objectives To determine if KL-VSHET+ status is associated with reduced AD risk and β-amyloid (Aβ) pathology in individuals who carry APOE4. Design, Setting, and Participants This study combined 25 independent case-control, family-based, and longitudinal AD cohorts that recruited referred and volunteer participants and made data available through public repositories. Analyses were stratified by APOE4 status. Three cohorts were used to evaluate conversion risk, 1 provided longitudinal measures of Aβ CSF and PET, and 3 provided cross-sectional measures of Aβ CSF. Genetic data were available from high-density single-nucleotide variant microarrays. All data were collected between September 2015 and September 2019 and analyzed between April 2019 and December 2019. Main Outcomes and Measures The risk of AD was evaluated through logistic regression analyses under a case-control design. The risk of conversion to mild cognitive impairment (MCI) or AD was evaluated through competing risks regression. Associations with Aβ, measured from cerebrospinal fluid (CSF) or brain positron emission tomography (PET), were evaluated using linear regression and mixed-effects modeling. Results Of 36 530 eligible participants, 13 782 were excluded for analysis exclusion criteria or refusal to participate. Participants were men and women aged 60 years and older who were non-Hispanic and of Northwestern European ancestry and had been diagnosed as being cognitively normal or having MCI or AD. The sample included 20 928 participants in case-control studies, 3008 in conversion studies, 556 in Aβ CSF regression analyses, and 251 in PET regression analyses. The genotype KL-VSHET+ was associated with reduced risk for AD in individuals carrying APOE4 who were 60 years or older (odds ratio, 0.75 [95% CI, 0.67-0.84]; P = 7.4 × 10-7), and this was more prominent at ages 60 to 80 years (odds ratio, 0.69 [95% CI, 0.61-0.79]; P = 3.6 × 10-8). Additionally, control participants carrying APOE4 with KL-VS heterozygosity were at reduced risk of converting to MCI or AD (hazard ratio, 0.64 [95% CI, 0.44-0.94]; P = .02). Finally, in control participants who carried APOE4 and were aged 60 to 80 years, KL-VS heterozygosity was associated with higher Aβ in CSF (β, 0.06 [95% CI, 0.01-0.10]; P = .03) and lower Aβ on PET scans (β, -0.04 [95% CI, -0.07 to -0.00]; P = .04). Conclusions and Relevance The genotype KL-VSHET+ is associated with reduced AD risk and Aβ burden in individuals who are aged 60 to 80 years, cognitively normal, and carrying APOE4. Molecular pathways associated with KL merit exploration for novel AD drug targets. The KL-VS genotype should be considered in conjunction with the APOE genotype to refine AD prediction models used in clinical trial enrichment and personalized genetic counseling.
Collapse
Affiliation(s)
- Michael E Belloy
- Department of Neurology and Neurological Sciences, Functional Imaging in Neuropsychiatric Disorders (FIND) Lab, Stanford University, Stanford, California
| | - Valerio Napolioni
- Department of Neurology and Neurological Sciences, Functional Imaging in Neuropsychiatric Disorders (FIND) Lab, Stanford University, Stanford, California
| | - Summer S Han
- Department of Neurosurgery, Stanford University, Stanford, California.,Quantitative Sciences Unit, Stanford Medicine, Stanford, California
| | - Yann Le Guen
- Department of Neurology and Neurological Sciences, Functional Imaging in Neuropsychiatric Disorders (FIND) Lab, Stanford University, Stanford, California
| | - Michael D Greicius
- Department of Neurology and Neurological Sciences, Functional Imaging in Neuropsychiatric Disorders (FIND) Lab, Stanford University, Stanford, California
| | | |
Collapse
|
13
|
Gao X, Sun Z, Ma G, Li Y, Liu M, Zhang G, Xu H, Gao Y, Zhou J, Deng Q, Li R. Reduced Plasma Levels of α-Klotho and Their Correlation With Klotho Polymorphisms in Elderly Patients With Major Depressive Disorders. Front Psychiatry 2021; 12:682691. [PMID: 34721095 PMCID: PMC8548667 DOI: 10.3389/fpsyt.2021.682691] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 09/20/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Recent literature suggests that α-Klotho, a widely recognized anti-aging protein, is involved in longevity as well as in many diseases, including Alzheimer's disease, and depression. Although the Klotho gene encodes α-Klotho, a single transmembrane protein with intracellular and extracellular domains, the relationship between Klotho gene polymorphism and circulating α-Klotho levels in patients with major depressive disorder (MDD) is not clear. Methods: A total of 144 MDD patients and 112 age-matched healthy controls were included in this study. The Klotho genetic polymorphisms (rs9536314, rs9527025, and rs9315202) and plasma α-Klotho levels were measured by PCR and ELISA, respectively. The severity of depressive symptoms was estimated using the Hamilton Depression Scale (HAMD). Results: We found a significantly lower level of plasma α-Klotho in the MDD patients than in controls. Among them, only elderly MDD patients (first episode) showed significantly lower α-Klotho levels than the age-matched controls, while elderly recurrent and young MDD patients showed no difference in plasma α-Klotho levels from age-matched controls. The young MDD group showed a significantly earlier onset age, higher plasma α-Klotho levels, and lower HAMD scores than those in the elderly MDD group. While the plasma α-Klotho levels were higher in rs9315202 T alleles carrier regardless age or sex, the rs9315202 T allele was negatively correlated with disease severity only in the elderly MDD patients. Conclusion: The results of our study showed that only elderly MDD patients showed a decrease in plasma α-Klotho levels along with an increase in disease severity as well as an association with the number of rs9315202 T alleles, and not young MDD patients compared to age-matched controls. Our data suggest that circulating α-Klotho levels combined with Klotho genetic polymorphisms are important in elderly MDD patients, particularly carriers of the Klotho gene rs9315202 T allele.
Collapse
Affiliation(s)
- Xiang Gao
- Laboratory of Brain Disorders, Ministry of Science and Technology, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Zuoli Sun
- Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Guangwei Ma
- Laboratory of Brain Disorders, Ministry of Science and Technology, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Yuhong Li
- Laboratory of Brain Disorders, Ministry of Science and Technology, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Min Liu
- Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Guofu Zhang
- Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Hong Xu
- Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Yane Gao
- Laboratory of Brain Disorders, Ministry of Science and Technology, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Jixuan Zhou
- Laboratory of Brain Disorders, Ministry of Science and Technology, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Qi Deng
- Laboratory of Brain Disorders, Ministry of Science and Technology, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Rena Li
- Laboratory of Brain Disorders, Ministry of Science and Technology, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
14
|
Abstract
Variation in the klotho gene is linked to differences in health outcomes: klotho allele KL-VS heterozygosity is associated with longevity, better cognition and greater right frontal grey matter volume in late life. Contradicting reports, however, suggest that KL-VS’s effect on health might be age-dependent. Here we examine the relationship between KL-VS genotype, cognition and brain structure in childhood and adolescence. We hypothesized that KL-VS has early influences on cognitive and brain development. We investigated the associations of KL-VS carrier status with cognition and brain morphology in a cohort of 1387 children and adolescents aged 3–21 years, examining main effects and interactions between age, sex and socioeconomic circumstance. KL-VS had no main effect on either cognition or brain structure, though there was a significant KL-VS × age interaction for cognition (specifically executive function, attention, episodic memory, and general cognition), total grey matter and total brain volume. KL-VS heterozygotes had better cognition than non-carriers before age 11, but lower cognition after age 11. Heterozygotes had smaller brains than non-carriers did in early childhood. Sex moderated the association between KL-VS and white matter volume. Among girls, KL-VS heterozygotes had smaller white matter volumes than non-carriers. Among boys, heterozygotes had greater white matter volumes than non-carriers. However, a replication in a cohort of 2306 children aged 6–12 years showed no significant associations. In contrast to findings in late life, these results show that KL-VS does not have a main effect on cognition and brain structure. Furthermore, KL-VS’s influence may depend on age and sex.
Collapse
|
15
|
The association between Single Nucleotide Polymorphisms of Klotho Gene and Mortality in Elderly Men: The MrOS Sweden Study. Sci Rep 2020; 10:10243. [PMID: 32581247 PMCID: PMC7314825 DOI: 10.1038/s41598-020-66517-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 02/24/2020] [Indexed: 12/12/2022] Open
Abstract
The Klotho (KL) gene is involved in phosphate homeostasis. Polymorphisms in this gene have been reported to be associated with the risk of cardiovascular disease. Here we used computational tools to predict the damage-associated single nucleotide polymorphisms (SNPs) in the human KL gene. We further investigated the association of SNPs in the KL gene and mortality in the Swedish multicenter prospective Osteoporotic Fractures in Men (MrOS) cohort. This study included 2921 men (aged 69-81 years) with mean 4.49 ± 1.03 years follow-up. 18 SNPs in the KL gene were genotyped using Sequenom. These SNPs were identified by in silico tools for the coding and noncoding genome to predict the damaging SNPs. After quality analyses, SNPs were analyzed for mortality risk using two steps approach on logistic regression model screening and then Cox regression model confirmation. Two non-synonymous SNPs rs9536314 and rs9527025 were found to be potentially damaging SNPs that affect KL protein stability and expression. However, these two SNPs were not statistically significantly associated with all-cause mortality (crude Hazard ratio [HR] 1.72, 95% confidence interval [CI] 0.96-3.07 in rs9536314; crude HR 1.82, 95% CI 0.998-3.33 in rs9527025) or cardiovascular mortality (crude HR 1.52, 95% CI 0.56-4.14 in rs9536314; crude HR 1.54, 95% CI 0.55-4.33 in rs9527025) in additive model using Cox regression analysis. In conclusion, these two potentially damaging SNPs (rs9536314 and rs9527025) in the KL gene were not associated with all-cause mortality or cardiovascular mortality in MrOs cohort. Larger scales studies and meta-analysis are needed to confirm the correlation between polymorphisms of the KL gene and mortality.
Collapse
|
16
|
Sachdeva A, Gouge J, Kontovounisios C, Nikolaou S, Ashworth A, Lim K, Chong I. Klotho and the Treatment of Human Malignancies. Cancers (Basel) 2020; 12:cancers12061665. [PMID: 32585905 PMCID: PMC7352559 DOI: 10.3390/cancers12061665] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 06/16/2020] [Indexed: 12/24/2022] Open
Abstract
Klotho was first discovered as an anti-ageing protein linked to a number of age-related disease processes, including cardiovascular, renal, musculoskeletal, and neurodegenerative conditions. Emerging research has also demonstrated a potential therapeutic role for Klotho in cancer biology, which is perhaps unsurprising given that cancer and ageing share similar molecular hallmarks. In addition to functioning as a tumour suppressor in numerous solid tumours and haematological malignancies, Klotho represents a candidate therapeutic target for patients with these diseases, the majority of whom have limited treatment options. Here, we examine contemporary evidence evaluating the anti-neoplastic effects of Klotho and describe the modulation of downstream oncogenic signalling pathways, including Wnt/β-catenin, FGF, IGF1, PIK3K/AKT, TGFβ, and the Unfolded Protein Response. We also discuss possible approaches to developing therapeutic Klotho and consider technological advances that may facilitate the delivery of Klotho through gene therapy.
Collapse
Affiliation(s)
- Aishani Sachdeva
- The Royal Marsden NHS Foundation Trust, London SW6 6JJ, UK; (A.S.); (C.K.)
- Department of Surgery and Cancer, Chelsea and Westminster Hospital, London SW10 9NH, UK;
| | - Jerome Gouge
- Institute of Structural and Molecular Biology, Birkbeck College, London WC1E 7HX, UK;
| | - Christos Kontovounisios
- The Royal Marsden NHS Foundation Trust, London SW6 6JJ, UK; (A.S.); (C.K.)
- Department of Surgery and Cancer, Chelsea and Westminster Hospital, London SW10 9NH, UK;
| | - Stella Nikolaou
- Department of Surgery and Cancer, Chelsea and Westminster Hospital, London SW10 9NH, UK;
| | - Alan Ashworth
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94158, USA;
| | - Kenneth Lim
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202-5181, USA;
| | - Irene Chong
- The Royal Marsden NHS Foundation Trust, London SW6 6JJ, UK; (A.S.); (C.K.)
- The Institute of Cancer Research, London SW3 6JB, UK
- Correspondence:
| |
Collapse
|
17
|
Cheikhi A, Barchowsky A, Sahu A, Shinde SN, Pius A, Clemens ZJ, Li H, Kennedy CA, Hoeck JD, Franti M, Ambrosio F. Klotho: An Elephant in Aging Research. J Gerontol A Biol Sci Med Sci 2020; 74:1031-1042. [PMID: 30843026 DOI: 10.1093/gerona/glz061] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Indexed: 12/12/2022] Open
Abstract
The year 2017 marked the 20th anniversary of the first publication describing Klotho. This single protein was and is remarkable in that its absence in mice conferred an accelerated aging, or progeroid, phenotype with a dramatically shortened life span. On the other hand, genetic overexpression extended both health span and life span by an impressive 30%. Not only has Klotho deficiency been linked to a number of debilitating age-related illnesses but many subsequent reports have lent credence to the idea that Klotho can compress the period of morbidity and extend the life span of both model organisms and humans. This suggests that Klotho functions as an integrator of organ systems, making it both a promising tool for advancing our understanding of the biology of aging and an intriguing target for interventional studies. In this review, we highlight advances in our understanding of Klotho as well as key challenges that have somewhat limited our view, and thus translational potential, of this potent protein.
Collapse
Affiliation(s)
- Amin Cheikhi
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh.,Division of Geriatric Medicine, Department of Medicine, University of Pittsburgh
| | - Aaron Barchowsky
- Department of Environmental and Occupational Health, University of Pittsburgh.,Department of Pharmacology and Chemical Biology, University of Pittsburgh
| | - Amrita Sahu
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh.,Department of Environmental and Occupational Health, University of Pittsburgh
| | - Sunita N Shinde
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh
| | - Abish Pius
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh
| | - Zachary J Clemens
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh
| | - Hua Li
- Department of Biotherapeutics Discovery, Research Division, Boehringer Ingelheim Pharmaceuticals, Inc. Ridgefield, Connecticut
| | - Charles A Kennedy
- Department of Research Beyond Borders, Regenerative Medicine, Boehringer Ingelheim Pharmaceuticals, Inc. Ridgefield, Connecticut
| | - Joerg D Hoeck
- Department of Research Beyond Borders, Regenerative Medicine, Boehringer Ingelheim Pharmaceuticals, Inc. Ridgefield, Connecticut
| | - Michael Franti
- Department of Research Beyond Borders, Regenerative Medicine, Boehringer Ingelheim Pharmaceuticals, Inc. Ridgefield, Connecticut
| | - Fabrisia Ambrosio
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh.,Department of Environmental and Occupational Health, University of Pittsburgh.,Department of Bioengineering, University of Pittsburgh, Pennsylvania.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pennsylvania
| |
Collapse
|
18
|
Erickson CM, Schultz SA, Oh JM, Darst BF, Ma Y, Norton D, Betthauser T, Gallagher CL, Carlsson CM, Bendlin BB, Asthana S, Hermann BP, Sager MA, Blennow K, Zetterberg H, Engelman CD, Christian BT, Johnson SC, Dubal DB, Okonkwo OC. KLOTHO heterozygosity attenuates APOE4-related amyloid burden in preclinical AD. Neurology 2019; 92:e1878-e1889. [PMID: 30867273 PMCID: PMC6550504 DOI: 10.1212/wnl.0000000000007323] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 12/05/2018] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE To examine whether the KLOTHO gene variant KL-VS attenuates APOE4-associated β-amyloid (Aβ) accumulation in a late-middle-aged cohort enriched with Alzheimer disease (AD) risk factors. METHODS Three hundred nine late-middle-aged adults from the Wisconsin Registry for Alzheimer's Prevention and the Wisconsin Alzheimer's Disease Research Center were genotyped to determine KL-VS and APOE4 status and underwent CSF sampling (n = 238) and/or 11C-Pittsburgh compound B (PiB)-PET imaging (n = 183). Covariate-adjusted regression analyses were used to investigate whether APOE4 exerted expected effects on Aβ burden. Follow-up regression analyses stratified by KL-VS genotype (i.e., noncarrier vs heterozygous; there were no homozygous individuals) evaluated whether the influence of APOE4 on Aβ was different among KL-VS heterozygotes compared to noncarriers. RESULTS APOE4 carriers exhibited greater Aβ burden than APOE4-negative participants. This effect was stronger in CSF (t = -5.12, p < 0.001) compared with PiB-PET (t = 3.93, p < 0.001). In the stratified analyses, this APOE4 effect on Aβ load was recapitulated among KL-VS noncarriers (CSF: t = -5.09, p < 0.001; PiB-PET: t = 3.77, p < 0 .001). In contrast, among KL-VS heterozygotes, APOE4-positive individuals did not exhibit higher Aβ burden than APOE4-negative individuals (CSF: t = -1.03, p = 0.308; PiB-PET: t = 0.92, p = 0.363). These differential APOE4 effects remained after KL-VS heterozygotes and noncarriers were matched on age and sex. CONCLUSION In a cohort of at-risk late-middle-aged adults, KL-VS heterozygosity was associated with an abatement of APOE4-associated Aβ aggregation, suggesting KL-VS heterozygosity confers protections against APOE4-linked pathways to disease onset in AD.
Collapse
Affiliation(s)
- Claire M Erickson
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Stephanie A Schultz
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Jennifer M Oh
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Burcu F Darst
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Yue Ma
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Derek Norton
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Tobey Betthauser
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Catherine L Gallagher
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Cynthia M Carlsson
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Barbara B Bendlin
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Sanjay Asthana
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Bruce P Hermann
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Mark A Sager
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Kaj Blennow
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Henrik Zetterberg
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Corinne D Engelman
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Bradley T Christian
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Sterling C Johnson
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Dena B Dubal
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco
| | - Ozioma C Okonkwo
- From the Geriatric Research Education and Clinical Center (C.L.G., C.M.C., S.A., S.C.J., O.C.O.), William S. Middleton Memorial VA Hospital; Wisconsin Alzheimer's Disease Research Center (C.M.E., J.M.O., Y.M., C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., B.T.C., S.C.J., O.C.O.); Departments of Population Health Sciences (B.F.D., C.D.E.), Neurology (C.L.G., B.P.H.), Radiology (M.A.S.), Medical Physics (T.B., B.T.C.), and Biostatistics & Medical Informatics (D.N.), University of Wisconsin School of Medicine and Public Health, Madison; Division of Biology and Biomedical Sciences (S.A.S.), Washington University in St. Louis, MO; Department of Psychiatry and Neurochemistry (K.B., H.Z.), Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg; Clinical Neurochemistry Laboratory (K.B., H.Z.), Sahlgrenska University Hospital, Mölndal, Sweden; Institute of Neurology (C.L.G., H.Z.), University College London, Queen Square; UK Dementia Research Institute (H.Z.), London; Wisconsin Alzheimer's Institute (C.M.C., B.B.B., S.A., B.P.H., M.A.S., C.D.E., S.C.J., O.C.O.), Madison; and Department of Neurology and Weill Institute for Neurosciences (D.B.D.), University of California, San Francisco.
| |
Collapse
|
19
|
Li D, Jing D, Liu Z, Chen Y, Huang F, Behnisch T. Enhanced Expression of Secreted α-Klotho in the Hippocampus Alters Nesting Behavior and Memory Formation in Mice. Front Cell Neurosci 2019; 13:133. [PMID: 31001090 PMCID: PMC6454015 DOI: 10.3389/fncel.2019.00133] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 03/18/2019] [Indexed: 12/30/2022] Open
Abstract
The klotho gene family consists of α-, β-, and γ-Klotho, which encode type I single-pass transmembrane proteins with large extracellular domains. α-Klotho exists as a full-length membrane-bound and as a soluble form after cleavage of the extracellular domain. Due to gene splicing, a short extracellular Klotho form can be expressed and secreted. Inactivation of α-Klotho leads to a phenotype that resembles accelerated aging, as the expression level of the α-Klotho protein in the hippocampal formation of mice decreases with age. Here, we show that intrahippocampal viral expression of secreted human α-Klotho alters social behavior and memory formation. Interestingly, overexpression of secreted human α-Klotho in the CA1 changed the nest-building behavior and improved object recognition, object location and passive avoidance memory. Moreover, α-Klotho overexpression increased hippocampal synaptic transmission in response to standardized stimulation strengths, altered paired-pulse facilitation of synaptic transmission, and enhanced activity-dependent synaptic plasticity. These results indicate that memory formation benefits from an augmented level of secreted α-Klotho.
Collapse
Affiliation(s)
- Dongxue Li
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Dongqing Jing
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Ziyang Liu
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Ying Chen
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Fang Huang
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Thomas Behnisch
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
20
|
Zhu Z, Xia W, Cui Y, Zeng F, Li Y, Yang Z, Hequn C. Klotho gene polymorphisms are associated with healthy aging and longevity: Evidence from a meta-analysis. Mech Ageing Dev 2019; 178:33-40. [PMID: 30633899 DOI: 10.1016/j.mad.2018.12.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 12/10/2018] [Accepted: 12/30/2018] [Indexed: 01/11/2023]
Abstract
Klotho gene polymorphisms have been implicated in healthy aging, but inconsistences in findings from previous case-control studies have raised concerns regarding the associations between KLOTHO gene polymorphisms and susceptibility to aging-related diseases and longevity. Hence, this meta-analysis was performed. We assessed the associations between two polymorphisms (G-395 A/rs1207568 and F352 V/rs9536314) and five parameters (urolithiasis, cognitive impairment, cardiovascular disease, cancer, and longevity) by calculating pooled odds ratios with 95% confidence intervals. According to the pooled results, the G allele of the G-395 A polymorphism conferred a significantly higher risk of urolithiasis; G-395 A was related to the susceptibility to cardiovascular disease under allele, dominant, and recessive models. There was no significant association between the G-395 A polymorphism and cognitive impairment among the elderly. The F allele of the F352 V polymorphism protected against breast and ovarian cancer susceptibility. Interestingly, based on the results of the subgroup analysis, the F352 V polymorphism was associated with the overall risk of neoplasms in BRCA1 mutation carriers but not in BRCA2 mutation carriers. Moreover, the F allele played a protective role in determining human longevity. In conclusion, Klotho G-395 A polymorphisms were associated with urolithiasis and cardiovascular disease but not with cognitive impairment. Additionally, Klotho F352 V polymorphisms were associated with cancers and longevity.
Collapse
Affiliation(s)
- Zewu Zhu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Weiping Xia
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yu Cui
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Feng Zeng
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yang Li
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Zhongqing Yang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Chen Hequn
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| |
Collapse
|
21
|
Behringer V, Stevens JMG, Deschner T, Sonnweber R, Hohmann G. Aging and sex affect soluble alpha klotho levels in bonobos and chimpanzees. Front Zool 2018; 15:35. [PMID: 30250491 PMCID: PMC6146871 DOI: 10.1186/s12983-018-0282-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 09/07/2018] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Throughout life, physiological homeostasis is challenged and the capacity to cope with such challenges declines with increasing age. In many species, sex differences exist in life expectancy. Sex-specific differences have been related to extrinsic factors like mate competition and/or intrinsic proximate mechanisms such as hormonal changes. In humans, an intrinsic factor related to aging is soluble alpha klotho (α-Kl). Both sexes show an age-related decline in α-Kl, but throughout life women have higher levels than men of the same age. Sex differences in α-Kl have been linked to a shorter lifespan, as well as to specific morbidity factors such as atherosclerosis and arteries calcifications. In non-human animals, information on α-Kl levels is rare and restricted to experimental work. Our cross-sectional study is the first on α-Kl levels in two long-lived species: bonobos (Pan paniscus) and chimpanzees (Pan troglodytes). As in most mammals, female bonobos and chimpanzees have longer life expectancy than males. METHODS We measured serum α-Kl levels of 140 subjects from 16 zoos with an ELISA to examine if α-Kl levels reflect this difference in life expectancy. RESULTS In both species and in both sexes, α-Kl levels declined with age suggesting that this marker has potential for aging studies beyond humans. We also found species-specific differences. Adult female bonobos had higher α-Kl levels than males, a difference that corresponds to the pattern found in humans. In chimpanzees, we found the opposite: males had higher α-Kl levels than females. CONCLUSION We suggest that contrasting sex differences in adult α-Kl levels mirror the dominance relations between females and males of the two Pan species; and that this might be related to corresponding sex differences in their exposure to stress. In humans, higher cortisol levels were found to be related to lower α-Kl levels. We conclude that there is great potential for studying aging processes in hominoids, and perhaps also in other non-human primates, by measuring α-Kl levels. To better understand the causes for sex differences in this aging marker, consideration of behavioural parameters such as competition and stress exposure will be required as well as other physiological markers.
Collapse
Affiliation(s)
- V. Behringer
- Department of Primatology, Max Planck Institute for Evolutionary Anthropology, Deutscher Platz 6, 04103 Leipzig, Germany
| | - J. M. G. Stevens
- Antwerp Zoo Centre for Research and Conservation, Royal Zoological Society of Antwerp, K. Astridplein 26, 2018 Antwerp, Belgium
- Behavioral Ecology and Ecophysiology, Department of Biology, University of Antwerp, 2610 Wilrijk, Belgium
| | - T. Deschner
- Department of Primatology, Max Planck Institute for Evolutionary Anthropology, Deutscher Platz 6, 04103 Leipzig, Germany
| | - R. Sonnweber
- Department of Primatology, Max Planck Institute for Evolutionary Anthropology, Deutscher Platz 6, 04103 Leipzig, Germany
| | - G. Hohmann
- Department of Primatology, Max Planck Institute for Evolutionary Anthropology, Deutscher Platz 6, 04103 Leipzig, Germany
| |
Collapse
|
22
|
Revelas M, Thalamuthu A, Oldmeadow C, Evans TJ, Armstrong NJ, Kwok JB, Brodaty H, Schofield PR, Scott RJ, Sachdev PS, Attia JR, Mather KA. Review and meta-analysis of genetic polymorphisms associated with exceptional human longevity. Mech Ageing Dev 2018; 175:24-34. [PMID: 29890178 DOI: 10.1016/j.mad.2018.06.002] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 06/01/2018] [Accepted: 06/07/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND Many factors contribute to exceptional longevity, with genetics playing a significant role. However, to date, genetic studies examining exceptional longevity have been inconclusive. This comprehensive review seeks to determine the genetic variants associated with exceptional longevity by undertaking meta-analyses. METHODS Meta-analyses of genetic polymorphisms previously associated with exceptional longevity (85+) were undertaken. For each variant, meta-analyses were performed if there were data from at least three independent studies available, including two unpublished additional cohorts. RESULTS Five polymorphisms, ACE rs4340, APOE ε2/3/4, FOXO3A rs2802292, KLOTHO KL-VS and IL6 rs1800795 were significantly associated with exceptional longevity, with the pooled effect sizes (odds ratios) ranging from 0.42 (APOE ε4) to 1.45 (FOXO3A males). CONCLUSION In general, the observed modest effect sizes of the significant variants suggest many genes of small influence play a role in exceptional longevity, which is consistent with results for other polygenic traits. Our results also suggest that genes related to cardiovascular health may be implicated in exceptional longevity. Future studies should examine the roles of gender and ethnicity and carefully consider study design, including the selection of appropriate controls.
Collapse
Affiliation(s)
- Mary Revelas
- Centre for Healthy Brain Ageing, School of Psychiatry, UNSW Medicine, University of New South Wales, Sydney, Australia
| | - Anbupalam Thalamuthu
- Centre for Healthy Brain Ageing, School of Psychiatry, UNSW Medicine, University of New South Wales, Sydney, Australia
| | | | | | - Nicola J Armstrong
- Centre for Healthy Brain Ageing, School of Psychiatry, UNSW Medicine, University of New South Wales, Sydney, Australia; Mathematics and Statistics, Murdoch University, Perth, Australia
| | - John B Kwok
- Neuroscience Research Australia, Randwick, Australia; School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Henry Brodaty
- Centre for Healthy Brain Ageing, School of Psychiatry, UNSW Medicine, University of New South Wales, Sydney, Australia; Dementia Centre for Research Collaboration, University of New South Wales, Sydney, Australia
| | - Peter R Schofield
- Neuroscience Research Australia, Randwick, Australia; School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Rodney J Scott
- Faculty of Health, University of Newcastle, Newcastle NSW, Australia; Hunter Area Pathology Service, John Hunter Hospital, Newcastle, New South Wales, Australia
| | - Perminder S Sachdev
- Centre for Healthy Brain Ageing, School of Psychiatry, UNSW Medicine, University of New South Wales, Sydney, Australia; Neuropsychiatric Institute, Prince of Wales Hospital, Barker Street, Randwick, NSW, Australia
| | - John R Attia
- Hunter Medical Research Institute, Newcastle, Australia; Hunter Area Pathology Service, John Hunter Hospital, Newcastle, New South Wales, Australia
| | - Karen A Mather
- Centre for Healthy Brain Ageing, School of Psychiatry, UNSW Medicine, University of New South Wales, Sydney, Australia.
| |
Collapse
|
23
|
Skrzypkowska M, Słomiński B, Ryba-Stanisławowska M, Gutknecht P, Siebert J. Circulating CD34+ and CD34+VEGFR2+ progenitor cells are associated with KLOTHO KL-VS polymorphism. Microvasc Res 2018; 119:1-6. [PMID: 29604296 DOI: 10.1016/j.mvr.2018.03.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 03/26/2018] [Accepted: 03/26/2018] [Indexed: 11/30/2022]
Abstract
BACKGROUND KLOTHO is a regulator of endothelial cells activity and integrity. It has been described for the first time because of its anti-aging properties. KLOTHO encoding gene is present in many functional variants in humans, including "KL-VS" variant that has been connected with longevity and cardiovascular disease development. Few mechanisms have been proposed to explain these associations, but none of them focused on cells from CD34+ population. The aim of our study was to investigate influence of KLOTHO KL-VS polymorphism on populations of CD34+ and CD34+VEGFR2+ cells. METHODS AND RESULTS We examined 167 Polish subjects from Pomeranian region. The analysis concerned KL-VS polymorphism, flow cytometry evaluation of whole blood cells and determination of endothelium-associated serum/plasma factors. Our results indicate that individuals possessing at least one KL-VS allele are characterized by greater number of CD34+ and CD34+VEGFR2+ and their various subpopulations (CD34+CD133+, CD34+c-Kit+, CD34+CXCR4+ and CD34+VEGFR2+c-Kit+) than wild-type volunteers. This group also exhibited more favorable lipid profile and statistically insignificant decrease of vWF and angiotensin II in their blood, whereas VEGF levels were elevated. CONCLUSION One of the mechanisms that are responsible for previously described KL-VS heterozygote advantage may be connected with maintaining greater size of hematopoietic and endothelial progenitor cells population.
Collapse
Affiliation(s)
- Maria Skrzypkowska
- Department of Immunology, Medical University of Gdańsk, Dębinki 1, 80-211 Gdańsk, Poland.
| | - Bartosz Słomiński
- Department of Immunology, Medical University of Gdańsk, Dębinki 1, 80-211 Gdańsk, Poland
| | | | - Piotr Gutknecht
- University Center for Cardiology Department of Family Medicine, Medical University of Gdansk, M. Skłodowskiej-Curie 3a, 80-210, Gdańsk, Poland
| | - Janusz Siebert
- University Center for Cardiology Department of Family Medicine, Medical University of Gdansk, M. Skłodowskiej-Curie 3a, 80-210, Gdańsk, Poland
| |
Collapse
|
24
|
Morar B, Badcock JC, Phillips M, Almeida OP, Jablensky A. The longevity gene Klotho is differentially associated with cognition in subtypes of schizophrenia. Schizophr Res 2018; 193:348-353. [PMID: 28673754 DOI: 10.1016/j.schres.2017.06.054] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 06/22/2017] [Accepted: 06/27/2017] [Indexed: 01/10/2023]
Abstract
Cognitive impairment is a core feature of schizophrenia and impacts negatively the functioning of affected individuals. Cognitive decline correlates with aging, and is the primary cause of loss of independence and reduced quality of life. The klotho gene is a key modulator of aging, with expression deficiency resulting in premature aging, while overexpression extends lifespan and enhances cognition. A haplotype and functional human variant of the gene, KL-VS, increases expression and promotes longevity. KL-VS heterozygosity is associated with enhanced cognition and a larger volume of the right dorsolateral prefrontal cortex, a region involved in planning and decision-making, which is especially susceptible to shrinkage with age. We examined the effect of KL-VS heterozygosity on cognition in 497 schizophrenia patients and 316 healthy controls from the Western Australian Family Study of Schizophrenia (WAFSS) who had been comprehensively characterised by neurocognitive tests and classified into cognitively deficient (CD) and cognitively "spared" (CS) clusters. An older, cognitively normal population sample from the Health in Men Study (HIMS) was included to allow assessment of heterozygosity and memory in aged individuals. We show that heterozygosity is associated with better learning and memory in the younger WAFSS healthy controls but not in the aging HIMS sample. However, in schizophrenia patients, KL-VS has a selective effect on memory, with heterozygotes in CD and CS clusters performing worse than non-carriers. This effect was significant and more severe in the CD cluster, reinforcing the utility of subtyping patients into CD and CS clusters that may differ in their genetic underpinnings.
Collapse
Affiliation(s)
- Bharti Morar
- Centre for Clinical Research in Neuropsychiatry, School of Psychiatry and Clinical Neurosciences, University of Western Australia, MRF Building, 50 Murray Street, Perth 6000, Australia; Cooperative Research Centre for Mental Health, Carlton South, Victoria, Australia; Harry Perkins Institute of Medical Research and Centre for Medical Research, The University of Western Australia, 6 Verdun Street, Nedlands, WA 6009, Australia.
| | - Johanna C Badcock
- Centre for Clinical Research in Neuropsychiatry, School of Psychiatry and Clinical Neurosciences, University of Western Australia, MRF Building, 50 Murray Street, Perth 6000, Australia; Cooperative Research Centre for Mental Health, Carlton South, Victoria, Australia
| | - Michael Phillips
- Harry Perkins Institute of Medical Research and Centre for Medical Research, The University of Western Australia, 6 Verdun Street, Nedlands, WA 6009, Australia
| | - Osvaldo P Almeida
- WA Centre for Health and Ageing, Centre for Medical Research, Perth, Australia
| | - Assen Jablensky
- Centre for Clinical Research in Neuropsychiatry, School of Psychiatry and Clinical Neurosciences, University of Western Australia, MRF Building, 50 Murray Street, Perth 6000, Australia; Cooperative Research Centre for Mental Health, Carlton South, Victoria, Australia
| |
Collapse
|
25
|
Systemic klotho is associated with KLOTHO variation and predicts intrinsic cortical connectivity in healthy human aging. Brain Imaging Behav 2018; 11:391-400. [PMID: 27714549 PMCID: PMC5382127 DOI: 10.1007/s11682-016-9598-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Cognitive decline is a major biomedical challenge as the global population ages. Elevated levels of the longevity factor klotho suppress aging, enhance cognition, and promote synaptic plasticity and neural resilience against aging and Alzheimer’s disease (AD)-related pathogenic proteins. Here, we examined the relationship between human genetic variants of KLOTHO and systemic klotho levels – and assessed neuroanatomic correlates of serum klotho in a cohort of healthy older adults. Serum klotho levels were increased with KL-VS heterozygosity, as anticipated. We report, for the first time, that serum klotho levels were paradoxically decreased with KL-VS homozygosity. Further, we found that higher serum klotho levels were associated with measures of greater intrinsic connectivity in key functional networks of the brain vulnerable to aging and AD such as the fronto-parietal and default mode networks. Our findings suggest that elevated klotho promotes a resilient brain, possibly through increased network connectivity of critical brain regions.
Collapse
|
26
|
Lang F, Leibrock C, Pelzl L, Gawaz M, Pieske B, Alesutan I, Voelkl J. Therapeutic Interference With Vascular Calcification-Lessons From Klotho-Hypomorphic Mice and Beyond. Front Endocrinol (Lausanne) 2018; 9:207. [PMID: 29780355 PMCID: PMC5945862 DOI: 10.3389/fendo.2018.00207] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 04/13/2018] [Indexed: 12/13/2022] Open
Abstract
Medial vascular calcification, a major pathophysiological process associated with cardiovascular disease and mortality, involves osteo-/chondrogenic transdifferentiation of vascular smooth muscle cells (VSMCs). In chronic kidney disease (CKD), osteo-/chondrogenic transdifferentiation of VSMCs and, thus, vascular calcification is mainly driven by hyperphosphatemia, resulting from impaired elimination of phosphate by the diseased kidneys. Hyperphosphatemia with subsequent vascular calcification is a hallmark of klotho-hypomorphic mice, which are characterized by rapid development of multiple age-related disorders and early death. In those animals, hyperphosphatemia results from unrestrained formation of 1,25(OH)2D3 with subsequent retention of calcium and phosphate. Analysis of klotho-hypomorphic mice and mice with vitamin D3 overload uncovered several pathophysiological mechanisms participating in the orchestration of vascular calcification and several therapeutic opportunities to delay or even halt vascular calcification. The present brief review addresses the beneficial effects of bicarbonate, carbonic anhydrase inhibition, magnesium supplementation, mineralocorticoid receptor (MR) blockage, and ammonium salts. The case is made that bicarbonate is mainly effective by decreasing intestinal phosphate absorption, and that carbonic anhydrase inhibition leads to metabolic acidosis, which counteracts calcium-phosphate precipitation and VSMC transdifferentiation. Magnesium supplementation, MR blockage and ammonium salts are mainly effective by interference with osteo-/chondrogenic signaling in VSMCs. It should be pointed out that the, by far, most efficient substances are ammonium salts, which may virtually prevent vascular calcification. Future research will probably uncover further therapeutic options and, most importantly, reveal whether these observations in mice can be translated into treatment of patients suffering from vascular calcification, such as patients with CKD.
Collapse
Affiliation(s)
- Florian Lang
- Department of Physiology I, Eberhard Karls-University, Tübingen, Germany
- *Correspondence: Florian Lang,
| | - Christina Leibrock
- Department of Physiology I, Eberhard Karls-University, Tübingen, Germany
- Fresenius Kabi Deutschland GmbH, Bad Homburg, Germany
| | - Lisann Pelzl
- Department of Physiology I, Eberhard Karls-University, Tübingen, Germany
| | - Meinrad Gawaz
- Department of Internal Medicine III, Eberhard Karls-University, Tübingen, Germany
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité-Universität Medizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Partner Site Berlin, German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Ioana Alesutan
- Department of Internal Medicine and Cardiology, Charité-Universität Medizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Partner Site Berlin, German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Jakob Voelkl
- Department of Internal Medicine and Cardiology, Charité-Universität Medizin Berlin, Berlin, Germany
- Partner Site Berlin, German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| |
Collapse
|
27
|
Leon J, Moreno AJ, Garay BI, Chalkley RJ, Burlingame AL, Wang D, Dubal DB. Peripheral Elevation of a Klotho Fragment Enhances Brain Function and Resilience in Young, Aging, and α-Synuclein Transgenic Mice. Cell Rep 2017; 20:1360-1371. [PMID: 28793260 PMCID: PMC5816951 DOI: 10.1016/j.celrep.2017.07.024] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 05/28/2017] [Accepted: 07/12/2017] [Indexed: 01/24/2023] Open
Abstract
Cognitive dysfunction and decreased mobility from aging and neurodegenerative conditions, such as Parkinson and Alzheimer diseases, are major biomedical challenges in need of more effective therapies. Increasing brain resilience may represent a new treatment strategy. Klotho, a longevity factor, enhances cognition when genetically and broadly overexpressed in its full, wild-type form over the mouse lifespan. Whether acute klotho treatment can rapidly enhance cognitive and motor functions or induce resilience is a gap in our knowledge of its therapeutic potential. Here, we show that an α-klotho protein fragment (αKL-F), administered peripherally, surprisingly induced cognitive enhancement and neural resilience despite impermeability to the blood-brain barrier in young, aging, and transgenic α-synuclein mice. αKL-F treatment induced cleavage of the NMDAR subunit GluN2B and also enhanced NMDAR-dependent synaptic plasticity. GluN2B blockade abolished αKL-F-mediated effects. Peripheral αKL-F treatment is sufficient to induce neural enhancement and resilience in mice and may prove therapeutic in humans.
Collapse
Affiliation(s)
- Julio Leon
- Department of Neurology, Biomedical Sciences Graduate Program, and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Arturo J Moreno
- Department of Neurology, Biomedical Sciences Graduate Program, and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Bayardo I Garay
- Department of Neurology, Biomedical Sciences Graduate Program, and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Robert J Chalkley
- Department of Chemistry and Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Alma L Burlingame
- Department of Chemistry and Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Dan Wang
- Department of Neurology, Biomedical Sciences Graduate Program, and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Dena B Dubal
- Department of Neurology, Biomedical Sciences Graduate Program, and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
28
|
Klotho, APOEε4, cognitive ability, brain size, atrophy, and survival: a study in the Aberdeen Birth Cohort of 1936. Neurobiol Aging 2017; 55:91-98. [DOI: 10.1016/j.neurobiolaging.2017.02.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 02/24/2017] [Accepted: 02/27/2017] [Indexed: 01/03/2023]
|
29
|
Abstract
The vertebrate endoskeleton is not a mere frame for muscle attachment to facilitate locomotion, but is a massive organ integrated with many physiologic functions including mineral and energy metabolism. Mineral balance is maintained by tightly controlled ion fluxes that are external (intestine and kidney) and internal (between bone and other organs), and are regulated and coordinated by many endocrine signals between these organs. The endocrine fibroblast growth factors (FGFs) and Klotho gene families are complex systems that co-evolved with the endoskeleton. In particular, FGF23 and αKlotho which are primarily derived from bone and kidney respectively, are critical in maintaining mineral metabolism where each of these proteins serving highly diverse roles; abound with many unanswered questions regarding their upstream regulation and downstream functions. Genetic lesions of components of this network produce discreet disturbances in many facets of mineral metabolism. One acquired condition with colossal elevations of FGF23 and suppression of αKlotho is chronic kidney disease where multiple organ dysfunction contributes to the morbidity and mortality. However, the single most important group of derangements that encompasses the largest breadth of complications is mineral metabolism disorders. Mineral metabolic disorders in CKD impact negatively and significantly on the progression of renal disease as well as extra-renal complications. Knowledge of the origin, nature, and impact of phosphate, FGF23, and αKlotho derangements is pivotal to understanding the pathophysiology and treatment of CKD.
Collapse
Affiliation(s)
- Makoto Kuro-O
- Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan; Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Orson W Moe
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
30
|
Kim HK, Jeong BH. Lack of functional KL-VS polymorphism of the KLOTHO gene in the Korean population. Genet Mol Biol 2016; 39:370-3. [PMID: 27560364 PMCID: PMC5004824 DOI: 10.1590/1678-4685-gmb-2015-0160] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 12/04/2015] [Indexed: 05/29/2023] Open
Abstract
The functional variant of the Klotho "KL-VS" stretch, which includes six
polymorphisms in linkage disequilibrium, is reportedly associated with healthy aging
and longevity in European and American populations. Among Asian populations, this
variant has been observed in the Indian population but not in the Iranian population.
An association between KL-VS polymorphism and aging has not been reported in Koreans.
To investigate whether the KL-VS polymorphism could be associated with healthy aging
and longevity in a Korean population, we analyzed genotype and allele frequencies of
the KL-VS variant in a large Korean population sample. The KL-VS variant was not
found in 874 Korean individuals. Thus, it is not possible to test its association to
aging in the East Asian populations.
Collapse
Affiliation(s)
- Hee-Kwon Kim
- Korea Zoonosis Research Institute, Chonbuk National University, Iksan, Jeonbuk, Republic of Korea.,Department of Nuclear Medicine, Molecular Imaging & Therapeutic Medicine Research Center, Biomedical Research Institute, Chonbuk National University Medical School and Hospital, Jeonju, Republic of Korea
| | - Byung-Hoon Jeong
- Korea Zoonosis Research Institute, Chonbuk National University, Iksan, Jeonbuk, Republic of Korea.,Department of Bioactive Material Sciences, Chonbuk National University, Jeonju, Jeonbuk Republic of Korea
| |
Collapse
|
31
|
Leibrock CB, Voelkl J, Kuro-O M, Lang F, Lang UE. 1,25(OH)2D3 dependent overt hyperactivity phenotype in klotho-hypomorphic mice. Sci Rep 2016; 6:24879. [PMID: 27109615 PMCID: PMC4843009 DOI: 10.1038/srep24879] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 04/05/2016] [Indexed: 02/06/2023] Open
Abstract
Klotho, a protein mainly expressed in kidney and cerebral choroid plexus, is a powerful regulator of 1,25(OH)2D3 formation. Klotho-deficient mice (kl/kl) suffer from excessive plasma 1,25(OH)2D3-, Ca2+- and phosphate-concentrations, leading to severe soft tissue calcification and accelerated aging. NH4Cl treatment prevents tissue calcification and premature ageing without affecting 1,25(OH)2D3-formation. The present study explored the impact of excessive 1,25(OH)2D3 formation in NH4Cl-treated kl/kl-mice on behavior. To this end kl/kl-mice and wild-type mice were treated with NH4Cl and either control diet or vitamin D deficient diet (LVD). As a result, plasma 1,25(OH)2D3-, Ca2+- and phosphate-concentrations were significantly higher in untreated and in NH4Cl-treated kl/kl-mice than in wild-type mice, a difference abrogated by LVD. In each, open field, dark-light box, and O-maze NH4Cl-treated kl/kl-mice showed significantly higher exploratory behavior than untreated wild-type mice, a difference abrogated by LVD. The time of floating in the forced swimming test was significantly shorter in NH4Cl treated kl/kl-mice compared to untreated wild-type mice and to kl/kl-mice on LVD. In wild-type animals, NH4Cl treatment did not significantly alter 1,25(OH)2D3, calcium and phosphate concentrations or exploratory behavior. In conclusion, the excessive 1,25(OH)2D3 formation in klotho-hypomorphic mice has a profound effect on murine behavior.
Collapse
Affiliation(s)
- Christina B Leibrock
- Department of Physiology, Cardiology &Vascular Medicine, University of Tübingen, Gmelinstr. 5, 72076 Tübingen, Germany
| | - Jakob Voelkl
- Department of Physiology, Cardiology &Vascular Medicine, University of Tübingen, Gmelinstr. 5, 72076 Tübingen, Germany
| | - Makoto Kuro-O
- Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Florian Lang
- Department of Physiology, Cardiology &Vascular Medicine, University of Tübingen, Gmelinstr. 5, 72076 Tübingen, Germany
| | - Undine E Lang
- Department of Psychiatry, University of Basel, Wilhelm Klein-Strasse 27, CH-4012 Basel, Switzerland
| |
Collapse
|
32
|
Mengel-From J, Soerensen M, Nygaard M, McGue M, Christensen K, Christiansen L. Genetic Variants in KLOTHO Associate With Cognitive Function in the Oldest Old Group. J Gerontol A Biol Sci Med Sci 2015; 71:1151-9. [PMID: 26405063 DOI: 10.1093/gerona/glv163] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 08/28/2015] [Indexed: 12/19/2022] Open
Abstract
Decline in cognitive abilities is a major concern in aging individuals. A potential important factor for functioning of the central nervous system in late-life stages is the KLOTHO (KL) gene. KL is expressed in various organs including the brain and is involved in multiple biological processes, for example, growth factor signaling. In the present study, 19 tagging gene variants in KL were studied in relation to 2 measures of cognitive function, a 5-item cognitive composite score and the Mini Mental State Examination, in 1,480 Danes 92-100 years of age. We found that heterozygotes for the previously reported KL-VS had poorer cognitive function than noncarriers. Two other variants positioned in the 5' end of the gene, rs398655 and rs562020, were associated with better cognitive function independently of KL-VS, and the common haplotype AG was associated with poorer cognition, consistently across two cognitive measures in two cohort strata. The haplotype effect was stronger than that of KL-VS. Two variants, rs2283368 and rs9526984, were the only variants significantly associated with cognitive decline over 7 years. We discuss an age-dependent effect of KL and the possibility that multiple gene variants in KL are important for cognitive function among the oldest old participants.
Collapse
Affiliation(s)
- Jonas Mengel-From
- The Danish Aging Research Center and The Danish Twin Registry, Epidemiology, Biostatistics and Biodemography Unit, Department of Public Health, University of Southern Denmark, Odense, Denmark. Department of Clinical Genetics, Odense University Hospital, Odense, Denmark.
| | - Mette Soerensen
- The Danish Aging Research Center and The Danish Twin Registry, Epidemiology, Biostatistics and Biodemography Unit, Department of Public Health, University of Southern Denmark, Odense, Denmark. Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | - Marianne Nygaard
- The Danish Aging Research Center and The Danish Twin Registry, Epidemiology, Biostatistics and Biodemography Unit, Department of Public Health, University of Southern Denmark, Odense, Denmark. Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | - Matt McGue
- The Danish Aging Research Center and The Danish Twin Registry, Epidemiology, Biostatistics and Biodemography Unit, Department of Public Health, University of Southern Denmark, Odense, Denmark. Department of Psychology, University of Minnesota, Minneapolis
| | - Kaare Christensen
- The Danish Aging Research Center and The Danish Twin Registry, Epidemiology, Biostatistics and Biodemography Unit, Department of Public Health, University of Southern Denmark, Odense, Denmark. Department of Clinical Genetics, Odense University Hospital, Odense, Denmark. Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
| | - Lene Christiansen
- The Danish Aging Research Center and The Danish Twin Registry, Epidemiology, Biostatistics and Biodemography Unit, Department of Public Health, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
33
|
Berridge MJ. Vitamin D cell signalling in health and disease. Biochem Biophys Res Commun 2015; 460:53-71. [PMID: 25998734 DOI: 10.1016/j.bbrc.2015.01.008] [Citation(s) in RCA: 142] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 01/05/2015] [Indexed: 12/13/2022]
Abstract
Vitamin D deficiency has been linked to many human diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), hypertension and cardiovascular disease. A Vitamin D phenotypic stability hypothesis, which is developed in this review, attempts to describe how this vital hormone acts to maintain healthy cellular functions. This role of Vitamin D as a guardian of phenotypic stability seems to depend on its ability to maintain the redox and Ca(2+) signalling systems. It is argued that its primary action is to maintain the expression of those signalling components responsible for stabilizing the low resting state of these two signalling pathways. This phenotypic stability role is facilitated through the ability of vitamin D to increase the expression of both Nrf2 and the anti-ageing protein Klotho, which are also major regulators of Ca(2+) and redox signalling. A decline in Vitamin D levels will lead to a decline in the stability of this regulatory signalling network and may account for why so many of the major diseases in man, which have been linked to vitamin D deficiency, are associated with a dysregulation in both ROS and Ca(2+) signalling.
Collapse
|
34
|
Prather AA, Epel ES, Arenander J, Broestl L, Garay BI, Wang D, Dubal DB. Longevity factor klotho and chronic psychological stress. Transl Psychiatry 2015; 5:e585. [PMID: 26080320 PMCID: PMC4490291 DOI: 10.1038/tp.2015.81] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 05/07/2015] [Indexed: 12/31/2022] Open
Abstract
Chronic psychological stress is associated with accelerated aging and premature morbidity and mortality; however, the biology linking chronic psychological stress and its maladaptive effects remains largely unknown. Klotho is a pleiotropic hormone that regulates the aging process and promotes better brain and body health. Whether klotho is linked to psychosocial stress or its negative impact in humans has not been investigated. To address this gap, we recruited 178 healthy women who were either chronically high-stress maternal caregivers for a child with autism spectrum disorder (n = 90) or low-stress control mothers of a typically developing child (n = 88). We found that women under high chronic stress displayed significantly lower levels of the longevity hormone klotho compared with low-stress controls (t(176) = 2.92, P = 0.004; d = 0.44), and the decrease among those under high stress was age-dependent. In addition, high-stress caregivers who reported more depressive symptoms displayed even lower klotho levels compared with low-stress participants. These findings provide the first evidence that klotho levels are sensitive to psychosocial stressors and raise the possibility that klotho may serve as a novel biological link connecting stress, depression and risk for accelerated disease development. Furthermore, these findings have important implications for understanding the plasticity of the aging process and may represent a therapeutic target for mitigating the deleterious effects of chronic psychological stress on health and well-being.
Collapse
Affiliation(s)
- A A Prather
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA, USA,Department of Psychiatry, University of California, San Francisco, 3333 California Street, Suite 465, San Francisco, CA 94118, USA E-mail:
| | - E S Epel
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA, USA
| | - J Arenander
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA, USA
| | - L Broestl
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - B I Garay
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - D Wang
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - D B Dubal
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA,Department of Neurology, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA. E-mail:
| |
Collapse
|
35
|
Demographic characteristics of Sardinian centenarian genealogies: Preliminary results of the AKeA2 study. DEMOGRAPHIC RESEARCH 2015. [DOI: 10.4054/demres.2015.32.37] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
36
|
Yokoyama JS, Sturm VE, Bonham LW, Klein E, Arfanakis K, Yu L, Coppola G, Kramer JH, Bennett DA, Miller BL, Dubal DB. Variation in longevity gene KLOTHO is associated with greater cortical volumes. Ann Clin Transl Neurol 2015; 2:215-30. [PMID: 25815349 PMCID: PMC4369272 DOI: 10.1002/acn3.161] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 11/24/2014] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVE Identifying genetic variation associated with brain structures in aging may elucidate new biologic mechanisms underlying resilience to cognitive decline. We investigated whether carrying one copy of the protective haplotype "KL-VS" in longevity gene KLOTHO (KL) is associated with greater gray matter volume in healthy human aging compared to carrying no copies. METHODS We performed unbiased whole-brain analysis in cognitively normal older adults from two independent cohorts to assess the relationship between KL-VS and gray matter volume using voxel-based morphometry. RESULTS We found that KL-VS heterozygosity was associated with greater volume in right dorsolateral prefrontal cortex (rDLPFC). Because rDLPFC is important for executive function, we analyzed working memory and processing speed in individuals. KL-VS heterozygosity was associated with enhanced executive function. Larger rDLPFC volume correlated with better executive function across the lifespan examined. Statistical analysis suggested that volume partially mediates the effect of genotype on cognition. INTERPRETATION These results suggest that variation in KL is associated with bigger brain volume and better function.
Collapse
Affiliation(s)
- Jennifer S Yokoyama
- Department of Neurology, University of California San FranciscoSan Francisco, California, 94158
| | - Virginia E Sturm
- Department of Neurology, University of California San FranciscoSan Francisco, California, 94158
| | - Luke W Bonham
- Department of Neurology, University of California San FranciscoSan Francisco, California, 94158
| | - Eric Klein
- Department of Neurology and Semel Institute for Neuroscience and Human Behavior, The David Geffen School of Medicine at University of California Los AngelesLos Angeles, California, 90095
| | - Konstantinos Arfanakis
- Department of Biomedical Engineering, Illinois Institute of TechnologyChicago, Illinois, 60616
- Rush Alzheimer's Disease Center, Rush University Medical CenterChicago, Illinois, 60612
| | - Lei Yu
- Rush Alzheimer's Disease Center, Rush University Medical CenterChicago, Illinois, 60612
| | - Giovanni Coppola
- Department of Neurology and Semel Institute for Neuroscience and Human Behavior, The David Geffen School of Medicine at University of California Los AngelesLos Angeles, California, 90095
| | - Joel H Kramer
- Department of Neurology, University of California San FranciscoSan Francisco, California, 94158
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical CenterChicago, Illinois, 60612
| | - Bruce L Miller
- Department of Neurology, University of California San FranciscoSan Francisco, California, 94158
| | - Dena B Dubal
- Department of Neurology, University of California San FranciscoSan Francisco, California, 94158
| |
Collapse
|
37
|
Dubal DB, Zhu L, Sanchez PE, Worden K, Broestl L, Johnson E, Ho K, Yu GQ, Kim D, Betourne A, Kuro-O M, Masliah E, Abraham CR, Mucke L. Life extension factor klotho prevents mortality and enhances cognition in hAPP transgenic mice. J Neurosci 2015; 35:2358-71. [PMID: 25673831 PMCID: PMC4323521 DOI: 10.1523/jneurosci.5791-12.2015] [Citation(s) in RCA: 148] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 12/06/2014] [Accepted: 12/21/2014] [Indexed: 12/11/2022] Open
Abstract
Aging is the principal demographic risk factor for Alzheimer disease (AD), the most common neurodegenerative disorder. Klotho is a key modulator of the aging process and, when overexpressed, extends mammalian lifespan, increases synaptic plasticity, and enhances cognition. Whether klotho can counteract deficits related to neurodegenerative diseases, such as AD, is unknown. Here we show that elevating klotho expression decreases premature mortality and network dysfunction in human amyloid precursor protein (hAPP) transgenic mice, which simulate key aspects of AD. Increasing klotho levels prevented depletion of NMDA receptor (NMDAR) subunits in the hippocampus and enhanced spatial learning and memory in hAPP mice. Klotho elevation in hAPP mice increased the abundance of the GluN2B subunit of NMDAR in postsynaptic densities and NMDAR-dependent long-term potentiation, which is critical for learning and memory. Thus, increasing wild-type klotho levels or activities improves synaptic and cognitive functions, and may be of therapeutic benefit in AD and other cognitive disorders.
Collapse
Affiliation(s)
- Dena B Dubal
- Gladstone Institute of Neurological Disease, San Francisco, California 94158, Department of Neurology, University of California, San Francisco, California 94158,
| | - Lei Zhu
- Gladstone Institute of Neurological Disease, San Francisco, California 94158, Department of Neurology, University of California, San Francisco, California 94158
| | - Pascal E Sanchez
- Gladstone Institute of Neurological Disease, San Francisco, California 94158, Department of Neurology, University of California, San Francisco, California 94158
| | - Kurtresha Worden
- Gladstone Institute of Neurological Disease, San Francisco, California 94158, Department of Neurology, University of California, San Francisco, California 94158
| | - Lauren Broestl
- Department of Neurology, University of California, San Francisco, California 94158
| | - Erik Johnson
- Gladstone Institute of Neurological Disease, San Francisco, California 94158, Department of Neurology, University of California, San Francisco, California 94158
| | - Kaitlyn Ho
- Gladstone Institute of Neurological Disease, San Francisco, California 94158
| | - Gui-Qiu Yu
- Gladstone Institute of Neurological Disease, San Francisco, California 94158
| | - Daniel Kim
- Gladstone Institute of Neurological Disease, San Francisco, California 94158
| | - Alexander Betourne
- Department of Neurology, University of California, San Francisco, California 94158
| | - Makoto Kuro-O
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Eliezer Masliah
- Departments of Neurosciences and Pathology, University of California, San Diego, San Diego, California 92093, and
| | - Carmela R Abraham
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Lennart Mucke
- Gladstone Institute of Neurological Disease, San Francisco, California 94158, Department of Neurology, University of California, San Francisco, California 94158,
| |
Collapse
|
38
|
Leibrock CB, Alesutan I, Voelkl J, Pakladok T, Michael D, Schleicher E, Kamyabi-Moghaddam Z, Quintanilla-Martinez L, Kuro-o M, Lang F. NH4Cl Treatment Prevents Tissue Calcification in Klotho Deficiency. J Am Soc Nephrol 2015; 26:2423-33. [PMID: 25644113 DOI: 10.1681/asn.2014030230] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 11/30/2014] [Indexed: 11/03/2022] Open
Abstract
Klotho, a cofactor in suppressing 1,25(OH)2D3 formation, is a powerful regulator of mineral metabolism. Klotho-hypomorphic mice (kl/kl) exhibit excessive plasma 1,25(OH)2D3, Ca(2+), and phosphate concentrations, severe tissue calcification, volume depletion with hyperaldosteronism, and early death. Calcification is paralleled by overexpression of osteoinductive transcription factor Runx2/Cbfa1, Alpl, and senescence-associated molecules Tgfb1, Pai-1, p21, and Glb1. Here, we show that NH4Cl treatment in drinking water (0.28 M) prevented soft tissue and vascular calcification and increased the life span of kl/kl mice >12-fold in males and >4-fold in females without significantly affecting extracellular pH or plasma concentrations of 1,25(OH)2D3, Ca(2+), and phosphate. NH4Cl treatment significantly decreased plasma aldosterone and antidiuretic hormone concentrations and reversed the increase of Runx2/Cbfa1, Alpl, Tgfb1, Pai-1, p21, and Glb1 expression in aorta of kl/kl mice. Similarly, in primary human aortic smooth muscle cells (HAoSMCs), NH4Cl treatment reduced phosphate-induced mRNA expression of RUNX2/CBFA1, ALPL, and senescence-associated molecules. In both kl/kl mice and phosphate-treated HAoSMCs, levels of osmosensitive transcription factor NFAT5 and NFAT5-downstream mediator SOX9 were higher than in controls and decreased after NH4Cl treatment. Overexpression of NFAT5 in HAoSMCs mimicked the effect of phosphate and abrogated the effect of NH4Cl on SOX9, RUNX2/CBFA1, and ALPL mRNA expression. TGFB1 treatment of HAoSMCs upregulated NFAT5 expression and prevented the decrease of phosphate-induced NFAT5 expression after NH4Cl treatment. In conclusion, NH4Cl treatment prevents tissue calcification, reduces vascular senescence, and extends survival of klotho-hypomorphic mice. The effects of NH4Cl on vascular osteoinduction involve decrease of TGFB1 and inhibition of NFAT5-dependent osteochondrogenic signaling.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Makoto Kuro-o
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas
| | | |
Collapse
|
39
|
Zhang WG, Bai XJ, Chen DP, Lv Y, Sun XF, Cai GY, Bai XY, Chen XM. Association of Klotho and interleukin 6 gene polymorphisms with aging in Han Chinese population. J Nutr Health Aging 2014; 18:900-4. [PMID: 25470806 DOI: 10.1007/s12603-014-0470-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Certain gene polymorphisms are associated with human aging. This study investigated polymorphisms of a metabolism-related gene, Klotho, and an inflammatory gene, IL6, for association with the aging process in a healthy Han Chinese population. A total of 482 healthy subjects were recruited and divided into aging and young groups according to chronological age and biological age. Snapshots were used to detect a Klotho gene tag SNP (rs571118) and the F-SNPs rs9536314 (F352V) and rs9527025 (C370S), and an interleukin 6 (IL-6) gene tag SNP (rs1524107) and the F-SNPs rs1800795 (-174G/C) and rs1800796 (-572G/C). Klotho F352V and IL-6-174G/C was G homozygous, C370S was T homozygous while IL-6-572G/C MAF less than 5%. There was a statistically significant difference in the Klotho rs571118 SNP between chronological age groups, but not biological age groups. However, other SNPs, including IL-6 gene SNPs, didn't correlate with age in the Han Chinese population. Human aging is a complex process that includes chronological and biological aging. Our current data showed that Klotho gene rs571118 SNP was associated with chronological aging, but not biological aging, in a Han Chinese population. Further study will investigate genetic build up for the difference between chronological and biological aging.
Collapse
Affiliation(s)
- W-G Zhang
- Xiang-Mei Chen, Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center of Kidney Diseases, Beijing 100853, China. Phone: 86-010-66937463; Fax: 86-010-68130297;
| | | | | | | | | | | | | | | |
Collapse
|
40
|
|
41
|
We are ageing. BIOMED RESEARCH INTERNATIONAL 2014; 2014:808307. [PMID: 25045704 PMCID: PMC4090574 DOI: 10.1155/2014/808307] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/11/2014] [Revised: 05/27/2014] [Accepted: 05/27/2014] [Indexed: 02/01/2023]
Abstract
Ageing and longevity is unquestioningly complex. Several thoughts and mechanisms of ageing such as pathways involved in oxidative stress, lipid and glucose metabolism, inflammation, DNA damage and repair, growth hormone axis and insulin-like growth factor (GH/IGF), and environmental exposure have been proposed. Also, some theories of ageing were introduced. To date, the most promising leads for longevity are caloric restriction, particularly target of rapamycin (TOR), sirtuins, hexarelin and hormetic responses. This review is an attempt to analyze the mechanisms and theories of ageing and achieving longevity.
Collapse
|
42
|
Dubal DB, Yokoyama JS, Zhu L, Broestl L, Worden K, Wang D, Sturm VE, Kim D, Klein E, Yu GQ, Ho K, Eilertson KE, Yu L, Kuro-o M, De Jager PL, Coppola G, Small GW, Bennett DA, Kramer JH, Abraham CR, Miller BL, Mucke L. Life extension factor klotho enhances cognition. Cell Rep 2014; 7:1065-76. [PMID: 24813892 PMCID: PMC4176932 DOI: 10.1016/j.celrep.2014.03.076] [Citation(s) in RCA: 215] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 01/10/2014] [Accepted: 03/31/2014] [Indexed: 12/22/2022] Open
Abstract
Aging is the primary risk factor for cognitive decline, an emerging health threat to aging societies worldwide. Whether anti-aging factors such as klotho can counteract cognitive decline is unknown. We show that a lifespan-extending variant of the human KLOTHO gene, KL-VS, is associated with enhanced cognition in heterozygous carriers. Because this allele increased klotho levels in serum, we analyzed transgenic mice with systemic overexpression of klotho. They performed better than controls in multiple tests of learning and memory. Elevating klotho in mice also enhanced long-term potentiation, a form of synaptic plasticity, and enriched synaptic GluN2B, an N-methyl-D-aspartate receptor (NMDAR) subunit with key functions in learning and memory. Blockade of GluN2B abolished klotho-mediated effects. Surprisingly, klotho effects were evident also in young mice and did not correlate with age in humans, suggesting independence from the aging process. Augmenting klotho or its effects may enhance cognition and counteract cognitive deficits at different life stages.
Collapse
Affiliation(s)
- Dena B Dubal
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA.
| | - Jennifer S Yokoyama
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Lei Zhu
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Lauren Broestl
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Kurtresha Worden
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Dan Wang
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Virginia E Sturm
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Daniel Kim
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | - Eric Klein
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA 90024, USA
| | - Gui-Qiu Yu
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | - Kaitlyn Ho
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | | | - Lei Yu
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - Makoto Kuro-o
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Center for Molecular Medicine, Jichi Medical University, Tochigi 329-0498, Japan
| | - Philip L De Jager
- Program in Translational NeuroPsychiatric Genomics, Institute for Neurosciences, Departments of Neurology & Psychiatry, Brigham and Women's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA; Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142, USA
| | - Giovanni Coppola
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA 90024, USA
| | - Gary W Small
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA 90024, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - Joel H Kramer
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Carmela R Abraham
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Bruce L Miller
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Lennart Mucke
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
43
|
Di Bona D, Accardi G, Virruso C, Candore G, Caruso C. Association of Klotho polymorphisms with healthy aging: a systematic review and meta-analysis. Rejuvenation Res 2014; 17:212-6. [PMID: 24164579 DOI: 10.1089/rej.2013.1523] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Today it is clearly evident that genetic background constitutes an integral part of aging and longevity. Many studies on long-lived people have been conducted emphasizing the role of certain genes in long life. Classic case-control studies, genome-wide association studies, and high-throughput sequencing have permitted identification of a variety of genetic variants seemingly associated with longevity. Over the years, aging research has focused on the insulin/insulin-like growth factor-1 (IGF-1) signaling pathway because of its evolutionarily conserved correlation with life-span extension in model animals. Indeed, many single-nucleotide polymorphisms (SNPs) associated with longevity were identified in genes encoding proteins that take part in this metabolic pathway. Closely related to this pathway is the Klotho gene. It encodes a type-I membrane protein expressed in two forms, membrane and secreted. The latter form suppresses oxidative stress and growth factor signaling and regulates ion channels and transporters. In particular, its over-expression seems to be able to suppress insulin/IGF-1 signaling extending life span. Thus, our aim was to assemble the results in the literature concerning the association between the functional variant of the Klotho "KL-VS" stretch, which contains six polymorphisms in linkage disequilibrium, and successful aging to quantify the possible effect of the variants. The results of our systematic review indicate that the Klotho KL-VS variant is associated with healthy aging.
Collapse
Affiliation(s)
- Danilo Di Bona
- 1 Immunosenescence Unit, Department of Pathobiology and Medical and Forensic Biotechnologies, University Hospital, University of Palermo , Palermo, Italy
| | | | | | | | | |
Collapse
|
44
|
Zhang WG, Bai XJ, Sun XF, Cai GY, Bai XY, Zhu SY, Zhang M, Chen XM. Construction of an integral formula of biological age for a healthy Chinese population using principle component analysis. J Nutr Health Aging 2014; 18:137-42. [PMID: 24522464 DOI: 10.1007/s12603-013-0345-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND Whereas chronological age (CA) cannot distinguish functional differences among individuals of the same age, the biological age (BA) may be used to reflect the functional state of the body. The purpose of this study was to construct an integral formula of the BA, by using principle component analysis (PCA). METHODS The vital organ function of 505 healthy individuals of Han origin (age 35-91 years) was examined. A total of 114 indicators of cardiovascular, pulmonary, and brain functions, and clinical, inflammatory, genetic, psychological, and life habit factors were assessed as candidate indicators of aging. Candidate indicators were submitted with CA to correlation and redundancy analyses. The PCA method was used to build an integral formula of the BA for the population. RESULTS Seven biomarkers were selected in accordance with a certain load standard. These biomarkers included the trail making test (TMT), pulse pressure (PP), mitral valve annulus ventricular septum of the peak velocity of early filling (MVES), minimum carotid artery intimal-medial thickness (IMTmin), maximum internal diameter of the carotid artery (Dmax), maximal midexpiratory flow rate 75/25 (MMEF75/25), and Cystatin C (CysC). The formula for the BA was: BA = 0.0685 (TMT) + 0.267 (PP) - 1.375 (MVES) + 22.443 (IMTmin) + 2.962 (Dmax) - 2.332 (MMEF75/25) + 16.104 (CysC) + 0.137 (CA) + 0.492. CONCLUSION Several genetic and lifestyle indicators were considered as candidate markers of aging. However, ultimately, only markers reflecting the function of the vital organs were included in the BA formula. This study represents a useful attempt to employ multiple indicators to build a comprehensive BA evaluation formula of aging populations.
Collapse
Affiliation(s)
- W-G Zhang
- Xiang-Mei Chen, Department of Nephrology, Kidney Institute of Chinese PLA, Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, Beijing, 100853, People's Republic of China, , Phone: 86-010-66937463, Fax: 86-010-68130297
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Tucker Zhou TB, King GD, Chen C, Abraham CR. Biochemical and functional characterization of the klotho-VS polymorphism implicated in aging and disease risk. J Biol Chem 2013; 288:36302-11. [PMID: 24217253 PMCID: PMC3868745 DOI: 10.1074/jbc.m113.490052] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 11/06/2013] [Indexed: 12/14/2022] Open
Abstract
Klotho (KL) is an age-regulating protein named after the Greek goddess who spins the thread of life. Mice deficient in KL are normal throughout development, but rapidly degenerate and display a variety of aging-associated abnormalities that eventually lead to decreased life expectancy. While multiple genetic association studies have identified KL polymorphisms linked with changes in disease risk, there is a paucity of concrete mechanistic data to explain how these amino acid substitutions alter KL protein function. The KLVS polymorphism is suggested to lead to changes in protein trafficking although the mechanism is unclear. Our studies have sought to further investigate the functional differences in the KLVS variant that result in increased risk of many age-related diseases. Our findings suggest that the F352V and C370S substitutions lead to alterations in processing as seen by differences in shedding and half-life. Their co-expression in KLVS results in a phenotype resembling wild-type, but despite this intragenic complementation there are still changes in homodimerization and interactions with FGFR1c. Taken together, these studies suggest that KLVS leads to altered homodimerization that indirectly leads to changes in processing and FGFR1c interactions. These findings help elucidate the functional differences that result from the VS polymorphism, which will help clarify how alterations in KL function can lead to human disease and affect cognition and lifespan.
Collapse
Affiliation(s)
- Tracey B. Tucker Zhou
- From the Departments of Pharmacology and Experimental Therapeutics and
- Biochemistry, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Gwendalyn D. King
- From the Departments of Pharmacology and Experimental Therapeutics and
| | - CiDi Chen
- From the Departments of Pharmacology and Experimental Therapeutics and
| | - Carmela R. Abraham
- From the Departments of Pharmacology and Experimental Therapeutics and
- Biochemistry, Boston University School of Medicine, Boston, Massachusetts 02118
| |
Collapse
|
46
|
Abstract
Through alternative splicing, Klotho protein exists both as a secreted and a membrane form whose extracellular domain could be shed from the cell surface by secretases and released into the circulation to act as endocrine factor. Unlike membrane Klotho which functions as a coreceptor for fibroblast growth factor-23 (FGF23) to modulate FGF23 signal transduction, soluble Klotho is a multifunction protein present in the biological fluids including blood, urine and cerebrospinal fluid and plays important roles in antiaging, energy metabolism, inhibition of Wnt signaling, antioxidation, modulation of ion transport, control of parathyroid hormone and 1,25(OH)2VD3 production, and antagonism of renin-angiotensin-aldosterone system. Emerging evidence from clinical and basic studies reveal that chronic kidney disease is a state of endocrine and renal Klotho deficiency, which may serve as an early biomarker and a pathogenic contributor to chronic progression and complications in chronic kidney disease including vascular calcification, cardiac hypertrophy, and secondary hyperparathyroidism. Supplementation of exogenous Klotho and/or upregulation of endogenous Klotho production by using rennin angiotensin system inhibitors, HMG CoA reductase inhibitors, vitamin D analogues, peroxisome proliferator-activated receptors-gamma agonists, or anti-oxidants may confer renoprotection from oxidation and suppression of renal fibrosis, and also on prevention or alleviation of complications in chronic kidney disease. Therefore, Klotho is a highly promising candidate on the horizon as an early biomarker, and as a novel therapeutic agent for chronic kidney disease.
Collapse
Affiliation(s)
- Ming Chang Hu
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Makoto Kuro-o
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Orson W. Moe
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
47
|
Łoniewska B, Adler G, Gumprecht J, Grzeszczak W, Kaczmarczyk M, Bińczak-Kuleta A, Ciechanowicz A, Clark JS. 1936A→G (I646 V) polymorphism in the AKAP10 gene encoding A-kinase-anchoring protein 10 in very long-lived poles is similar to that in newborns. Exp Aging Res 2013; 38:584-92. [PMID: 23092224 DOI: 10.1080/0361073x.2012.726177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
UNLABELLED BACKGROUND/STUDY CONTEXT: The common 1936A→G transition (rs203462) in the AKAP10 gene encoding the A-kinase-anchoring protein 10 has been recently associated with negative prognosis in the aging European American population (60 to 79 years old). The aim of this study was to see the effects of this transition on allele frequency in very long-lived Poles. METHODS AKAP10 genotype and allele distributions were analyzed in Polish subjects: 148 nonagenarians (95 to 103 years old) and 200 healthy newborn controls, using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). Distributions were separated according to gender and χ(2) tests used to analyze possible differences. RESULTS No significant differences were found in genotype or allele distribution between the age groups, for either gender. Percentages of GG AKAP10 homozygotes were slightly greater in the very old subjects than in the newborns (12.2% vs. 9.0%, respectively), and the G allele percentages were very similar (males, 30.7% and 33.0%; females, 34.1% and 35.8%; respectively). CONCLUSION The authors conclude that differences in study results between European Americans (60 to 79 years old) and Poles (≥95 years old) result from either (1) geographical location; or (2) the influence of this polymorphism on groups of people differing in genetic background or environmental history; or (3) the time window affected, including extreme age. Further studies with full age-frequency distributions are needed to clarify these results.
Collapse
Affiliation(s)
- Beata Łoniewska
- Department of Neonatal Diseases, Pomeranian Medical University, Szczecin, Poland
| | | | | | | | | | | | | | | |
Collapse
|
48
|
King GD, Rosene DL, Abraham CR. Promoter methylation and age-related downregulation of Klotho in rhesus monkey. AGE (DORDRECHT, NETHERLANDS) 2012; 34:1405-19. [PMID: 21922250 PMCID: PMC3528360 DOI: 10.1007/s11357-011-9315-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2011] [Accepted: 08/26/2011] [Indexed: 05/31/2023]
Abstract
While overall DNA methylation decreases with age, CpG-rich areas of the genome can become hypermethylated. Hypermethylation near transcription start sites typically decreases gene expression. Klotho (KL) is important in numerous age-associated pathways including insulin/IGF1 and Wnt signaling and naturally decreases with age in brain, heart, and liver across species. Brain tissues from young and old rhesus monkeys were used to determine whether epigenetic modification of the KL promoter underlies age-related decreases in mRNA and protein levels of KL. The KL promoter in genomic DNA from brain white matter did not show evidence of oxidation in vivo but did exhibit an increase in methylation with age. Further analysis identified individual CpG motifs across the region of interest with increased methylation in old animals. In vitro methyl modification of these individual cytosine residues confirmed that methylation of the promoter can decrease gene transcription. These results provide evidence that changes in KL gene expression with age may, at least in part, be the result of epigenetic changes to the 5' regulatory region.
Collapse
Affiliation(s)
- Gwendalyn D. King
- />Department of Biochemistry, Boston University School of Medicine, 72 East Concord Street K304, Boston, MA 02118 USA
- />Department of Neurobiology, University of Alabama, Birmingham, AL USA
| | - Douglas L. Rosene
- />Department of Anatomy and Neurobiology, Boston University School of Medicine, 700 Albany Street W701, Boston, MA 02118 USA
- />Yerkes National Primate Research Center, Emory University, 954 Gatewood, Atlanta, GA 30322 USA
| | - Carmela R. Abraham
- />Department of Biochemistry, Boston University School of Medicine, 72 East Concord Street K304, Boston, MA 02118 USA
| |
Collapse
|
49
|
Paroni G, Seripa D, Panza F, Addante F, Copetti M, D’Onofrio G, Pellegrini F, Fontana L, Pilotto A. Klotho locus, metabolic traits, and serum hemoglobin in hospitalized older patients: a genetic association analysis. AGE (DORDRECHT, NETHERLANDS) 2012; 34:949-968. [PMID: 21695423 PMCID: PMC3682056 DOI: 10.1007/s11357-011-9273-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Accepted: 05/23/2011] [Indexed: 05/30/2023]
Abstract
Klotho (KL) gene has been involved in severe alterations of physiological biochemical parameters leading to premature aging-like phenotypes and strikingly shortening lifespan. KL participates to the regulation of a number of intracellular biochemical pathways, including lipid profile and glucose metabolism. Aim of this study was to investigate the possible association between KL locus and biological parameters commonly accepted as indicators of the clinical status in hospitalized older patients. We genotyped the single-nucleotide polymorphisms (SNPs) rs9536314, rs1207568, and rs564481 at the KL locus in 594 hospitalized older patients (65-99 years), consecutively attending a geriatric ward, and tested the association of these KL variants with biological quantitative traits using analyses of covariance and genetic risk score models. Significant associations of rs9536314 with serum levels of hemoglobin, albumin, and high-density lipoprotein cholesterol (HDL-C) as well as significant associations of rs564481 with serum levels of hemoglobin, fasting insulin, and fasting glucose were observed. Gender-segregated analyses confirmed these associations, and suggested that the associations of KL genotypes with HDL-C, fasting glucose and fasting insulin levels may be driven by the female gender, while the association with serum levels of hemoglobin may be driven by the male gender. The association of KL genotypes with creatinine levels was found only in females, while the association with insulin-like growth factor-1 (IGF-1) and lymphocytes count (LC) was found only in males. The genetic risk score (GRS) models further confirmed significant associations among KL SNPs and hemoglobin, total cholesterol, and HDL-C. Gender-segregated analyses with the GRS-tagged approach confirmed the associations with HDL-C, fasting glucose, and fasting insulin levels in females, and with hemoglobin and LC in males. Our findings suggested that KL locus may influence quantitative traits such as serum levels of lipid, fasting glucose, albumin and hemoglobin in hospitalized older patients, with some gender differences suggested for creatinine, IGF-1 levels, and LC, thus being one of the genetic factors possibly contributing to age-related diseases and longevity.
Collapse
Affiliation(s)
- Giulia Paroni
- />Geriatric Unit and Gerontology-Geriatric Research Laboratory, Department of Medical Sciences, I.R.C.C.S. “Casa Sollievo della Sofferenza, Viale Cappuccini 1, 71013 San Giovanni Rotondo, Foggia Italy
| | - Davide Seripa
- />Geriatric Unit and Gerontology-Geriatric Research Laboratory, Department of Medical Sciences, I.R.C.C.S. “Casa Sollievo della Sofferenza, Viale Cappuccini 1, 71013 San Giovanni Rotondo, Foggia Italy
| | - Francesco Panza
- />Geriatric Unit and Gerontology-Geriatric Research Laboratory, Department of Medical Sciences, I.R.C.C.S. “Casa Sollievo della Sofferenza, Viale Cappuccini 1, 71013 San Giovanni Rotondo, Foggia Italy
| | - Filomena Addante
- />Geriatric Unit and Gerontology-Geriatric Research Laboratory, Department of Medical Sciences, I.R.C.C.S. “Casa Sollievo della Sofferenza, Viale Cappuccini 1, 71013 San Giovanni Rotondo, Foggia Italy
| | - Massimiliano Copetti
- />Unit of Biostatistic, I.R.C.C.S. “Casa Sollievo della Sofferenza”, San Giovanni Rotondo, Foggia Italy
| | - Grazia D’Onofrio
- />Geriatric Unit and Gerontology-Geriatric Research Laboratory, Department of Medical Sciences, I.R.C.C.S. “Casa Sollievo della Sofferenza, Viale Cappuccini 1, 71013 San Giovanni Rotondo, Foggia Italy
| | - Fabio Pellegrini
- />Unit of Biostatistic, I.R.C.C.S. “Casa Sollievo della Sofferenza”, San Giovanni Rotondo, Foggia Italy
- />Laboratory of Clinical Epidemiology of Diabetes and Chronic Diseases, Consorzio Mario Negri Sud, Chieti, Italy
| | - Luigi Fontana
- />Division of Nutrition and Aging, Istituto Superiore di Sanità, Rome, Italy
| | - Alberto Pilotto
- />Geriatric Unit and Gerontology-Geriatric Research Laboratory, Department of Medical Sciences, I.R.C.C.S. “Casa Sollievo della Sofferenza, Viale Cappuccini 1, 71013 San Giovanni Rotondo, Foggia Italy
| |
Collapse
|
50
|
Montesanto A, Dato S, Bellizzi D, Rose G, Passarino G. Epidemiological, genetic and epigenetic aspects of the research on healthy ageing and longevity. IMMUNITY & AGEING 2012; 9:6. [PMID: 22524317 PMCID: PMC3349521 DOI: 10.1186/1742-4933-9-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Accepted: 04/23/2012] [Indexed: 12/23/2022]
Abstract
Healthy ageing and longevity in humans result from a number of factors, including genetic background, favorable environmental and social factors and chance. In this article we aimed to overview the research on the biological basis of human healthy ageing and longevity, discussing the role of epidemiological, genetic and epigenetic factors in the variation of quality of ageing and lifespan, including the most promising candidate genes investigated so far. Moreover, we reported the methodologies applied for their identification, discussing advantages and disadvantages of the different approaches and possible solutions that can be taken to overcome them. Finally, we illustrated the recent approaches to define healthy ageing and underlined the role that the emerging field of epigenetics is gaining in the search for the determinants of healthy ageing and longevity.
Collapse
Affiliation(s)
- Alberto Montesanto
- Department of Cell Biology, University of Calabria, Ponte Pietro Bucci cubo 4 C, 87036 Rende, CS, Italy
| | - Serena Dato
- Department of Cell Biology, University of Calabria, Ponte Pietro Bucci cubo 4 C, 87036 Rende, CS, Italy
| | - Dina Bellizzi
- Department of Cell Biology, University of Calabria, Ponte Pietro Bucci cubo 4 C, 87036 Rende, CS, Italy
| | - Giuseppina Rose
- Department of Cell Biology, University of Calabria, Ponte Pietro Bucci cubo 4 C, 87036 Rende, CS, Italy
| | - Giuseppe Passarino
- Department of Cell Biology, University of Calabria, Ponte Pietro Bucci cubo 4 C, 87036 Rende, CS, Italy
| |
Collapse
|