1
|
Katonova A, Andel R, Jurasova V, Veverova K, Angelucci F, Matoska V, Hort J. Associations of KLOTHO-VS heterozygosity and α-Klotho protein with cerebrospinal fluid Alzheimer's disease biomarkers. J Alzheimers Dis 2025; 105:159-171. [PMID: 40112321 DOI: 10.1177/13872877251326199] [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: 03/22/2025]
Abstract
BackgroundKLOTHO-VS heterozygosity (KL-VSHET) and soluble α-Klotho (sαKl) protein interfere with Alzheimer's disease (AD) pathophysiology, but the specific relationships remain unclear. This study explored these associations across the AD continuum, focusing on core AD biomarkers and markers of neurodegeneration, neuroinflammation, and synaptic dysfunction.ObjectiveWe investigated whether 1) KL-VSHET is associated with lower AD biomarker burden (Aβ42, Aβ42/40 ratio, P-tau181, T-tau) and neurodegeneration (NfL); 2) sαKl relates to AD biomarkers, neurodegeneration (NfL), neuroinflammation (GFAP), and synaptic dysfunction (Ng); 3) associations vary by APOE ε4 status and clinical subgroup.MethodsParticipants (n = 223) were categorized as cognitively healthy (n = 38), aMCI-AD (n = 94), and AD dementia (n = 91). KLOTHO genotyping was available for 128 participants; 138 had cerebrospinal fluid (CSF) and serum sαKl measurements; and 42 had both. Multiple linear regression evaluated associations between KL-VSHET, sαKl levels, and biomarkers, stratified by APOE ε4 status and clinical subgroup.ResultsOverall, the associations between KL-VSHET and higher CSF Aβ42 and Aβ42/40 ratio were non-significant (ps ≥ 0.059) except when restricted to APOE ε4 carriers only (β = 0.11, p = 0.008 and β = 0.16, p = 0.033, respectively). Within clinical subgroups, KL-VSHET was positively associated with Aβ42/40 ratio only in aMCI-AD (β = 0.23, p = 0.034). No significant associations were observed between KL-VSHET and tau biomarkers or NfL. For sαKl, associations with biomarkers were non-significant except for a negative association of serum sαKl with P-tau181 in aMCI-AD (β = -0.25, p = 0.036) and a positive association with Aβ42/40 ratio in APOE ε4 non-carriers (β = 0.24 p = 0.047).ConclusionsKL-VSHET may help protect against amyloid pathology, particularly in the presence of APOE ε4, and regardless of APOE status in aMCI-AD.
Collapse
Affiliation(s)
- Alzbeta Katonova
- Memory Clinic, Department of Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Ross Andel
- Memory Clinic, Department of Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
- Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, AZ, USA
| | - Vanesa Jurasova
- Memory Clinic, Department of Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Katerina Veverova
- Memory Clinic, Department of Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Francesco Angelucci
- Memory Clinic, Department of Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Vaclav Matoska
- Department of Clinical Biochemistry, Hematology and Immunology, Homolka Hospital, Prague, Czech Republic
| | - Jakub Hort
- Memory Clinic, Department of Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| |
Collapse
|
2
|
Blum EG, Edmunds KJ, Breidenbach B, Cook N, Driscoll I, Lose SR, Bendlin BB, Ma Y, Christian B, Betthauser TJ, Sager M, Asthana S, Johnson SC, Cook DB, Okonkwo OC. Physical activity and APOE neuropathology score modify the association of age and [ 11C]-PiB-PET amyloid burden in a cohort enriched with risk for Alzheimer's disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.01.25323157. [PMID: 40093261 PMCID: PMC11908305 DOI: 10.1101/2025.03.01.25323157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Background Physical activity (PA) is a protective factor against amyloid-β (Aβ) accumulation in adults at risk for Alzheimer's disease (AD). This association, however, may differ by apolipoprotein E (APOE) genotype. This work examines interactions between age, PA, and neuropathology-based genetic risk for AD (APOE np ) on Aβ burden in cortical regions sensitive to its accumulation. Materials and Methods Included were 388 cognitively unimpaired, older (mean age ± SD = 68.10 ± 7.09; 66% female) participants from the Wisconsin Registry for Alzheimer's Prevention (WRAP) study. The cohort was enriched with both family history of AD at enrollment and a higher overall prevalence of APOE ε4 allele carriage than typically observed in the general population. PA was assessed using a self-reported questionnaire. Aβ burden was measured using Pittsburg Compound B (11C-PiB) PET imaging, which allowed us to derive volume corrected distribution volume ratio (DVR) maps from nine bilateral regions of interest (ROIs) and a global cortical composite score. Linear regression models examined the interactions between age, PA, and APOE np on Aβ burden. Finally, APOE np scores were aggregated according to estimated risk to illustrate the differential effects between active (weekly moderate PA ≥ 150 minutes) and inactive individuals. Results Three-way interactions (Age × PA × APOE np ) were significant (all P's ≤ 0.05) for the global cortical composite and six of the examined ROIs (the PPC, ACC, mOFC, SMG, MTG, and STG). Models stratified by APOE np and PA showed greater levels of age-related Aβ accumulation in each of these ROIs, with the greatest effects in inactive participants with high APOE np scores. Conclusion Individuals with high APOE np scores who concomitantly engage in suboptimal weekly moderate-intensity PA have greater Aβ burden. These findings underscore how both PA and APOE haplotype play intersect in modifying age-related Aβ burden in brain regions susceptible to its deposition in cognitively unimpaired, older adults at risk for AD.
Collapse
Affiliation(s)
- Eli G Blum
- Wisconsin Alzheimer's Disease Research Center and Department of Geriatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792
| | - Kyle J Edmunds
- Wisconsin Alzheimer's Disease Research Center and Department of Geriatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792
- Institute of Biomedical and Neural Engineering (IBNE), Reykjavík University, 101 Reykjavík, Iceland
| | - Brianne Breidenbach
- Wisconsin Alzheimer's Disease Research Center and Department of Geriatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792
| | - Noah Cook
- Wisconsin Alzheimer's Disease Research Center and Department of Geriatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, 63110
| | - Ira Driscoll
- Wisconsin Alzheimer's Disease Research Center and Department of Geriatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792
| | - Sarah R Lose
- Wisconsin Alzheimer's Disease Research Center and Department of Geriatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792
| | - Barbara B Bendlin
- Wisconsin Alzheimer's Disease Research Center and Department of Geriatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792
| | - Yue Ma
- Wisconsin Alzheimer's Disease Research Center and Department of Geriatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792
| | - Bradley Christian
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792
| | - Tobey J Betthauser
- Wisconsin Alzheimer's Disease Research Center and Department of Geriatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792
| | - Mark Sager
- Wisconsin Alzheimer's Disease Research Center and Department of Geriatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792
| | - Sanjay Asthana
- Wisconsin Alzheimer's Disease Research Center and Department of Geriatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705
| | - Sterling C Johnson
- Wisconsin Alzheimer's Disease Research Center and Department of Geriatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792
| | - Dane B Cook
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705
- Department of Kinesiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792
| | - Ozioma C Okonkwo
- Wisconsin Alzheimer's Disease Research Center and Department of Geriatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792
| |
Collapse
|
3
|
Jarchow M, Driscoll I, Breidenbach BM, Cook N, Gallagher CL, Johnson SC, Asthana S, Hermann BP, Sager MA, Blennow K, Zetterberg H, Carlsson CM, Kollmorgen G, Quijano-Rubio C, Cook DB, Dubal DB, Okonkwo OC. Older more fit KL-VS heterozygotes have more favorable AD-relevant biomarker profiles. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.27.25323056. [PMID: 40093256 PMCID: PMC11908295 DOI: 10.1101/2025.02.27.25323056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
INTRODUCTION While hallmarked by the accumulation of β-amyloid plaques (Aβ) and neurofibrillary tangles (tau) in the brain, Alzheimer's disease (AD) is a multifactorial disorder that involves additional pathological events, including neuroinflammation, neurodegeneration and synaptic dysfunction. AD-associated biomolecular changes seem to be attenuated in carriers of the functionally advantageous variant of the KLOTHO gene (KL-VSHET). Independently, better cardiorespiratory fitness (CRF) is associated with better health outcomes, both in general and specifically with regard to AD pathology. Here we investigate whether the relationships between CRF (peak oxygen consumption (VO2peak)) and cerebrospinal fluid (CSF) core AD biomarkers and those of neuroinflammation, neurodegeneration, and synaptic dysfunction differ for KL-VSHET compared to non-carriers (KL-VSNC). METHODS The cohort, enriched for AD risk, consisted of cognitively unimpaired adults (N=136; MeanAGE(SD)=62.5(6.7)) from the Wisconsin Registry for Alzheimer's Prevention and the Wisconsin Alzheimer's Disease Research Center. Covariate-adjusted (age, sex, parental AD history, APOE4+ status, and age difference between CSF sampling and exercise test) linear models examined the interaction between VO2peak and KLOTHO genotype on core AD biomarker levels in CSF [phosphorylated tau 181 (pTau181), Aβ42/Aβ40, pTau181/Aβ42]. Analyses were repeated for CSF biomarkers of neurodegeneration [total tau (tTau), α-synuclein (α-syn), neurofilament light polypeptide (NfL)], synaptic dysfunction [neurogranin (Ng)], and neuroinflammation [glial fibrillary acidic protein (GFAP), soluble triggering receptor expressed in myeloid cells (sTREM2), chitinase-3-like protein 1 (YKL-40), interleukin 6 (IL-6), S100 calcium-binding protein B (S100B)]. RESULTS The interaction between VO2peak and KL-VSHET was significant for tTau (P=0.05), pTau181 (P=0.03), Ng (P=0.02), sTREM2 (P=0.03), and YKL-40 (P=0.03), such that lower levels of each biomarker were observed for KL-VSHET who were more fit. No significant KL-VSxVO2peak interactions were observed for Aβ42/Aβ40, pTau181/Aβ42, α-syn, NfL, GFAP, IL-6 or S100B (all Ps>0.09). CONCLUSIONS We report a synergistic relationship between KL-VSHET and CRF with regard to pTau181, tTau, Ng, sTREM2 and YKL-40, suggesting a protective role for both KL-VSHET and better cardiovascular fitness against unfavorable AD-related changes. Their potentially shared biological mechanisms will require future investigations.
Collapse
Affiliation(s)
- Mackenzie Jarchow
- Wisconsin Alzheimer’s Disease Research Center, Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Ira Driscoll
- Wisconsin Alzheimer’s Disease Research Center, Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer’s Institute Madison, WI, USA
| | - Brianne M. Breidenbach
- Wisconsin Alzheimer’s Disease Research Center, Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer’s Institute Madison, WI, USA
| | - Noah Cook
- Wisconsin Alzheimer’s Disease Research Center, Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Catherine L. Gallagher
- Geriatric Research Education and Clinical Center, William S. Middleton VA Hospital, Madison, WI, USA
- Department of Neurology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Sterling C. Johnson
- Wisconsin Alzheimer’s Disease Research Center, Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer’s Institute Madison, WI, USA
| | - Sanjay Asthana
- Wisconsin Alzheimer’s Disease Research Center, Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer’s Institute Madison, WI, USA
- Geriatric Research Education and Clinical Center, William S. Middleton VA Hospital, Madison, WI, USA
| | - Bruce P. Hermann
- Wisconsin Alzheimer’s Disease Research Center, Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer’s Institute Madison, WI, USA
- Department of Neurology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Mark A. Sager
- Wisconsin Alzheimer’s Disease Research Center, Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer’s Institute Madison, WI, USA
| | - 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
- Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, China
| | - Henrik Zetterberg
- Wisconsin Alzheimer’s Disease Research Center, Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- 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
- Roche Diagnostics GmbH, Penzberg, Germany
| | - Cynthia M. Carlsson
- Wisconsin Alzheimer’s Disease Research Center, Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer’s Institute Madison, WI, USA
- Department of Neurology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | | | | | - Dane B. Cook
- Research Service, William S. Middleton VA Hospital, Madison, WI, USA
- Department of Kinesiology, School of Education, University of Wisconsin-Madison, Madison, WI, USA
| | - Dena B. Dubal
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, California, USA
| | - Ozioma C. Okonkwo
- Wisconsin Alzheimer’s Disease Research Center, Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer’s Institute Madison, WI, USA
| |
Collapse
|
4
|
Jia B, Xu Y, Zhu X. Cognitive resilience in Alzheimer's disease: Mechanism and potential clinical intervention. Ageing Res Rev 2025; 106:102711. [PMID: 40021093 DOI: 10.1016/j.arr.2025.102711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
Alzheimer's disease (AD) is a globally recognized neurodegenerative disorder that severely impairs cognitive function and imposes substantial psychological and financial burdens on patients and their families. The hallmark pathological features of AD include progressive neurodegeneration, extracellular beta-amyloid (Aβ) plaque accumulation, and intracellular hyperphosphorylated tau protein tangles. However, recent studies have identified a subset of patients exhibiting cognitive resilience, characterized by a slower cognitive decline or the preservation of high cognitive function despite the presence of AD pathology. Cognitive resilience is influenced by a complex interplay of genetic, environmental, and lifestyle factors. In addition, cognitive resilience contributes to the new perspectives on the diagnosis and personalized treatment of AD. This review aims to provide a comprehensive analysis of current studies on cognitive resilience in AD and to explore future research directions of AD diagnosis and treatment.
Collapse
Affiliation(s)
- Bin Jia
- Department of Neurology, Nanjing Drum Tower Hospital, School of Medicine, Jiangsu University, Nanjing, Jiangsu, China; Department of Neurology, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital, School of Medicine, Jiangsu University, Nanjing, Jiangsu, China; Jiangsu Key Laboratory for Molecular Medicine and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China; Nanjing Neurology Clinical Medical Center, and Nanjing Drum Tower Hospital Brain Disease and Brain Science Center, Nanjing, China
| | - Xiaolei Zhu
- Department of Neurology, Nanjing Drum Tower Hospital, School of Medicine, Jiangsu University, Nanjing, Jiangsu, China; Jiangsu Key Laboratory for Molecular Medicine and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China; Nanjing Neurology Clinical Medical Center, and Nanjing Drum Tower Hospital Brain Disease and Brain Science Center, Nanjing, China.
| |
Collapse
|
5
|
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
|
6
|
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
|
7
|
Jackson RJ, Hyman BT, Serrano-Pozo A. Multifaceted roles of APOE in Alzheimer disease. Nat Rev Neurol 2024; 20:457-474. [PMID: 38906999 DOI: 10.1038/s41582-024-00988-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2024] [Indexed: 06/23/2024]
Abstract
For the past three decades, apolipoprotein E (APOE) has been known as the single greatest genetic modulator of sporadic Alzheimer disease (AD) risk, influencing both the average age of onset and the lifetime risk of developing AD. The APOEε4 allele significantly increases AD risk, whereas the ε2 allele is protective relative to the most common ε3 allele. However, large differences in effect size exist across ethnoracial groups that are likely to depend on both global genetic ancestry and local genetic ancestry, as well as gene-environment interactions. Although early studies linked APOE to amyloid-β - one of the two culprit aggregation-prone proteins that define AD - in the past decade, mounting work has associated APOE with other neurodegenerative proteinopathies and broader ageing-related brain changes, such as neuroinflammation, energy metabolism failure, loss of myelin integrity and increased blood-brain barrier permeability, with potential implications for longevity and resilience to pathological protein aggregates. Novel mouse models and other technological advances have also enabled a number of therapeutic approaches aimed at either attenuating the APOEε4-linked increased AD risk or enhancing the APOEε2-linked AD protection. This Review summarizes this progress and highlights areas for future research towards the development of APOE-directed therapeutics.
Collapse
Affiliation(s)
- Rosemary J Jackson
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, USA.
| | - Alberto Serrano-Pozo
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, USA.
| |
Collapse
|
8
|
Driscoll IF, Lose S, Ma Y, Bendlin BB, Gallagher C, Johnson SC, Asthana S, Hermann B, Sager MA, Blennow K, Zetterberg H, Carlsson C, Kollmorgen G, Quijano‐Rubio C, Dubal D, Okonkwo OC. KLOTHO KL-VS heterozygosity is associated with diminished age-related neuroinflammation, neurodegeneration, and synaptic dysfunction in older cognitively unimpaired adults. Alzheimers Dement 2024; 20:5347-5356. [PMID: 39030746 PMCID: PMC11350058 DOI: 10.1002/alz.13912] [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: 01/24/2024] [Revised: 04/12/2024] [Accepted: 04/27/2024] [Indexed: 07/22/2024]
Abstract
INTRODUCTION We examined whether the aging suppressor KLOTHO gene's functionally advantageous KL-VS variant (KL-VS heterozygosity [KL-VSHET]) confers resilience against deleterious effects of aging indexed by cerebrospinal fluid (CSF) biomarkers of neuroinflammation (interleukin-6 [IL-6], S100 calcium-binding protein B [S100B], triggering receptor expressed on myeloid cells [sTREM2], chitinase-3-like protein 1 [YKL-40], glial fibrillary acidic protein [GFAP]), neurodegeneration (total α-synuclein [α-Syn], neurofilament light chain protein), and synaptic dysfunction (neurogranin [Ng]). METHODS This Alzheimer disease risk-enriched cohort consisted of 454 cognitively unimpaired adults (Mage = 61.5 ± 7.75). Covariate-adjusted multivariate regression examined relationships between age (mean-split[age ≥ 62]) and CSF biomarkers (Roche/NeuroToolKit), and whether they differed between KL-VSHET (N = 122) and non-carriers (KL-VSNC; N = 332). RESULTS Older age was associated with a poorer biomarker profile across all analytes (Ps ≤ 0.03). In age-stratified analyses, KL-VSNC exhibited this same pattern (Ps ≤ 0.05) which was not significant for IL-6, S100B, Ng, and α-Syn (Ps ≥ 0.13) in KL-VSHET. Although age-related differences in GFAP, sTREM2, and YKL-40 were evident for both groups (Ps ≤ 0.01), the effect magnitude was markedly stronger for KL-VSNC. DISCUSSION Higher levels of neuroinflammation, neurodegeneration, and synaptic dysfunction in older adults were attenuated in KL-VSHET. HIGHLIGHTS Older age was associated with poorer profiles across all cerebrospinal fluid biomarkers of neuroinflammation, neurodegeneration, and synaptic dysfunction. KLOTHO KL-VS non-carriers exhibit this same pattern, which is does not significantly differ between younger and older KL-VS heterozygotes for interleukin-6, S100 calcium-binding protein B, neurogranin, and total α-synuclein. Although age-related differences in glial fibrillary acidic protein, triggering receptor expressed on myeloid cells, and chitinase-3-like protein 1 are evident for both KL-VS groups, the magnitude of the effect is markedly stronger for KL-VS non-carriers. Higher levels of neuroinflammation, neurodegeneration, and synaptic dysfunction in older adults are attenuated in KL-VS heterozygotes.
Collapse
Affiliation(s)
- Ira Frahmand Driscoll
- Wisconsin Alzheimer's Disease Research Center and Department of GeriatricsUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Sarah Lose
- Wisconsin Alzheimer's Disease Research Center and Department of GeriatricsUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Yue Ma
- Wisconsin Alzheimer's Disease Research Center and Department of GeriatricsUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Barbara B. Bendlin
- Wisconsin Alzheimer's Disease Research Center and Department of GeriatricsUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteMadisonWisconsinUSA
- Geriatric Research Education and Clinical CenterWilliam S. Middleton VA HospitalMadisonWisconsinUSA
| | - Catherine Gallagher
- Geriatric Research Education and Clinical CenterWilliam S. Middleton VA HospitalMadisonWisconsinUSA
- Department of NeurologyUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Sterling C. Johnson
- Wisconsin Alzheimer's Disease Research Center and Department of GeriatricsUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteMadisonWisconsinUSA
| | - Sanjay Asthana
- Wisconsin Alzheimer's Disease Research Center and Department of GeriatricsUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteMadisonWisconsinUSA
- Geriatric Research Education and Clinical CenterWilliam S. Middleton VA HospitalMadisonWisconsinUSA
| | - Bruce Hermann
- Wisconsin Alzheimer's Disease Research Center and Department of GeriatricsUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteMadisonWisconsinUSA
- Department of NeurologyUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Mark A. Sager
- Wisconsin Alzheimer's Disease Research Center and Department of GeriatricsUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteMadisonWisconsinUSA
| | - Kaj Blennow
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologySahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalGöteborgSweden
- Paris Brain InstituteICMPitié‐Salpêtrière HospitalSorbonne UniversityParisFrance
- Neurodegenerative Disorder Research CenterDivision of Life Sciences and Medicineand Department of NeurologyInstitute on Aging and Brain DisordersUniversity of Science and Technology of China and First Affiliated Hospital of USTCHefeiPR China
| | - Henrik Zetterberg
- Wisconsin Alzheimer's Disease Research Center and Department of GeriatricsUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologySahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalGöteborgSweden
- Department of Neurodegenerative DiseaseUCL Institute of Neurology, Queen SquareLondonUK
- UK Dementia Research Institute at UCLLondonUK
- Hong Kong Center for Neurodegenerative DiseasesClear Water BayHong KongPR China
| | - Cynthia Carlsson
- Wisconsin Alzheimer's Disease Research Center and Department of GeriatricsUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteMadisonWisconsinUSA
- Geriatric Research Education and Clinical CenterWilliam S. Middleton VA HospitalMadisonWisconsinUSA
| | | | | | - Dena Dubal
- Department of Neurology and Weill Institute for NeurosciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Ozioma C. Okonkwo
- Wisconsin Alzheimer's Disease Research Center and Department of GeriatricsUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteMadisonWisconsinUSA
- Geriatric Research Education and Clinical CenterWilliam S. Middleton VA HospitalMadisonWisconsinUSA
| |
Collapse
|
9
|
Guo X, Huang X, Yang Y, Dong L, Kong D, Zhang J. FNDC5/Irisin in dementia and cognitive impairment: update and novel perspective. Braz J Med Biol Res 2024; 57:e13447. [PMID: 38985081 PMCID: PMC11249199 DOI: 10.1590/1414-431x2024e13447] [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: 10/26/2023] [Accepted: 05/23/2024] [Indexed: 07/11/2024] Open
Abstract
Epidemiological surveys show that the incidence of age-related dementia and cognitive impairment is increasing and it has been a heavy burden for society, families, and healthcare systems, making the preservation of cognitive function in an increasingly aging population a major challenge. Exercise is beneficial for brain health, and FDNC5/irisin, a new exercise-induced myokine, is thought to be a beneficial mediator to cognitive function and plays an important role in the crosstalk between skeletal muscle and brain. This review provides a critical assessment of the recent progress in both fundamental and clinical research of FDNC5/irisin in dementia and cognitive impairment-related disorders. Furthermore, we present a novel perspective on the therapeutic effectiveness of FDNC5/irisin in alleviating these conditions.
Collapse
Affiliation(s)
- Xiaofeng Guo
- Department of Endocrinology and Metabolism, The Second School of Clinical Medicine of Binzhou Medical University, Yantai, Shandong, China
| | - Xiaocheng Huang
- Department of Health Examination, Weihai Municipal Hospital affiliated to Shandong University, Weihai, Shandong, China
| | - Yachao Yang
- Department of Endocrinology and Metabolism, Weihai Municipal Hospital affiliated to Shandong University, Weihai, Shandong, China
| | - Luying Dong
- Department of Health Examination, Weihai Municipal Hospital affiliated to Shandong University, Weihai, Shandong, China
| | - Dehuan Kong
- Department of Endocrinology and Metabolism, Taian City Central Hospital, Taian, Shandong, China
| | - Jianmei Zhang
- Department of Endocrinology and Metabolism, Weihai Municipal Hospital affiliated to Shandong University, Weihai, Shandong, China
- Department of Geriatrics, Weihai Municipal Hospital Affiliated to Shandong University, Weihai, Shandong, China
| |
Collapse
|
10
|
de Vries LE, Huitinga I, Kessels HW, Swaab DF, Verhaagen J. The concept of resilience to Alzheimer's Disease: current definitions and cellular and molecular mechanisms. Mol Neurodegener 2024; 19:33. [PMID: 38589893 PMCID: PMC11003087 DOI: 10.1186/s13024-024-00719-7] [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: 08/23/2023] [Accepted: 03/20/2024] [Indexed: 04/10/2024] Open
Abstract
Some individuals are able to maintain their cognitive abilities despite the presence of significant Alzheimer's Disease (AD) neuropathological changes. This discrepancy between cognition and pathology has been labeled as resilience and has evolved into a widely debated concept. External factors such as cognitive stimulation are associated with resilience to AD, but the exact cellular and molecular underpinnings are not completely understood. In this review, we discuss the current definitions used in the field, highlight the translational approaches used to investigate resilience to AD and summarize the underlying cellular and molecular substrates of resilience that have been derived from human and animal studies, which have received more and more attention in the last few years. From these studies the picture emerges that resilient individuals are different from AD patients in terms of specific pathological species and their cellular reaction to AD pathology, which possibly helps to maintain cognition up to a certain tipping point. Studying these rare resilient individuals can be of great importance as it could pave the way to novel therapeutic avenues for AD.
Collapse
Affiliation(s)
- Luuk E de Vries
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA, Amsterdam, The Netherlands.
| | - Inge Huitinga
- Department of Neuroimmunology, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA, Amsterdam, The Netherlands
| | - Helmut W Kessels
- Swammerdam Institute for Life Sciences, Amsterdam Neuroscience, University of Amsterdam, 1098 XH, Amsterdam, the Netherlands
| | - Dick F Swaab
- Department of Neuropsychiatric Disorders, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA, Amsterdam, Netherlands
| | - Joost Verhaagen
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA, Amsterdam, The Netherlands
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, Boelelaan 1085, 1081 HV, Amsterdam, The Netherlands
| |
Collapse
|
11
|
Feng Y, Jiang X, Liu W, Lu H. The location, physiology, pathology of hippocampus Melatonin MT 2 receptor and MT 2-selective modulators. Eur J Med Chem 2023; 262:115888. [PMID: 37866336 DOI: 10.1016/j.ejmech.2023.115888] [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: 08/27/2023] [Revised: 10/06/2023] [Accepted: 10/17/2023] [Indexed: 10/24/2023]
Abstract
Melatonin, a neurohormone secreted by the pineal gland and regulated by the suprachiasmatic nucleus (SCN) of the hypothalamus, is synthesized and directly released into the cerebrospinal fluid (CSF) of the third ventricle (3rdv), where it undergoes rapid absorption by surrounding tissues to exert its physiological function. The hippocampus, a vital structure in the limbic system adjacent to the ventricles, plays a pivotal role in emotional response and memory formation. Melatonin MT1 and MT2 receptors are G protein-coupled receptors (GPCRs) that primarily mediate melatonin's receptor-dependent effects. In comparison to the MT1 receptor, the widely expressed MT2 receptor is crucial for mediating melatonin's biological functions within the hippocampus. Specifically, MT2 receptor is implicated in hippocampal synaptic plasticity and memory processes, as well as neurogenesis and axogenesis. Numerous studies have demonstrated the involvement of MT2 receptors in the pathophysiology and pharmacology of Alzheimer's disease, depression, and epilepsy. This review focuses on the anatomical localization of MT2 receptor in the hippocampus, their physiological function in this region, and their signal transduction and pharmacological roles in neurological disorders. Additionally, we conducted a comprehensive review of MT2 receptor ligands used in psychopharmacology and other MT2-selective ligands over recent years. Ultimately, we provide an outlook on future research for selective MT2 receptor drug candidates.
Collapse
Affiliation(s)
- Yueqin Feng
- Department of Ultrasound, the First Affiliated Hospital of China Medical University, Shenyang, PR China
| | - Xiaowen Jiang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Wenwu Liu
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, PR China
| | - Hongyuan Lu
- Department of Clinical Pharmacology, China Medical University, Shenyang, PR China.
| |
Collapse
|
12
|
Ranjbar N, Raeisi M, Barzegar M, Ghorbanihaghjo A, Shiva S, Sadeghvand S, Negargar S, Poursistany H, Raeisi S. The possible anti-seizure properties of Klotho. Brain Res 2023; 1820:148555. [PMID: 37634687 DOI: 10.1016/j.brainres.2023.148555] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 07/30/2023] [Accepted: 08/23/2023] [Indexed: 08/29/2023]
Abstract
Recurrent seizures in epilepsy may lead to progressive neuronal damage, which can diminish health-related quality of life. Evaluation and control of pathological processes in the brain is valuable. It seems imperative that new markers and approaches for seizure alleviation be discovered. Klotho (Kl), an antiaging protein, has protective effects in the brain against neurological disorders. It may also have antiseizure effects by improving creatine transfer to the brain, upregulating excitatory amino acid transporters, and inhibiting insulin/insulin-like growth factor-1 (IGF-1), Wingless (Wnt), transforming growth factor-beta (TGF-β), and retinoic-acid-inducible gene-I (RIG-I)/nuclear translocation of nuclear factor-κB (NF-κB) pathways. Stimulation and activation of the nuclear factor erythroid 2-related factor 2 (Nrf2) and apoptosis signal-regulating kinase 1 (ASK1)/p38 mitogen‑activated protein kinase (MAPK) signaling pathways could also be considered other possible antiseizure mechanisms of Kl. In the present review, the roles of Kl in the central nervous system as well as its possible anti-seizure properties are discussed for the first time.
Collapse
Affiliation(s)
- Nasrin Ranjbar
- Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammadreza Raeisi
- Student Research Committee, Ahvaz Jondishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Barzegar
- Pediatric Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Ghorbanihaghjo
- Biothechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Siamak Shiva
- Pediatric Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shahram Sadeghvand
- Pediatric Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sohrab Negargar
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Haniyeh Poursistany
- Department of Clinical Biochemistry and Laboratory Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sina Raeisi
- Pediatric Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Clinical Research Development Unit of Zahra Mardani Azari Children Educational and Treatment Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
13
|
Kosik KS. Search Strategies for Alzheimer Protector Genes. Ann Neurol 2023; 94:613-617. [PMID: 37574772 DOI: 10.1002/ana.26764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/31/2023] [Accepted: 08/05/2023] [Indexed: 08/15/2023]
Abstract
Large families with autosomal dominant mutations leading to Alzheimer's disease and related conditions can show putative protective gene variants; however, no systematic search strategy exists to find these genes. Described here are the unique demographic circumstances and genetic setting in which the discovery of protective variants is likely. The identification of these genes may reveal pathways with therapeutic implications. ANN NEUROL 2023;94:613-617.
Collapse
Affiliation(s)
- Kenneth S Kosik
- The Neuroscience Research Institute and The Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, California, USA
| |
Collapse
|
14
|
Castner SA, Gupta S, Wang D, Moreno AJ, Park C, Chen C, Poon Y, Groen A, Greenberg K, David N, Boone T, Baxter MG, Williams GV, Dubal DB. Longevity factor klotho enhances cognition in aged nonhuman primates. NATURE AGING 2023; 3:931-937. [PMID: 37400721 PMCID: PMC10432271 DOI: 10.1038/s43587-023-00441-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 05/23/2023] [Indexed: 07/05/2023]
Abstract
Cognitive dysfunction in aging is a major biomedical challenge. Whether treatment with klotho, a longevity factor, could enhance cognition in human-relevant models such as in nonhuman primates is unknown and represents a major knowledge gap in the path to therapeutics. We validated the rhesus form of the klotho protein in mice showing it increased synaptic plasticity and cognition. We then found that a single administration of low-dose, but not high-dose, klotho enhanced memory in aged nonhuman primates. Systemic low-dose klotho treatment may prove therapeutic in aging humans.
Collapse
Affiliation(s)
- Stacy A Castner
- Department of Psychiatry and VA Connecticut Healthcare System, Yale School of Medicine, West Haven, CT, USA
| | - Shweta Gupta
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Dan Wang
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Arturo J Moreno
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Cana Park
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Chen Chen
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Yan Poon
- Unity Biotechnology, Brisbane, CA, USA
| | | | | | | | - Tom Boone
- Tom Boone Consulting, Newbury Park, CA, USA
| | - Mark G Baxter
- Section on Comparative Medicine, Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Graham V Williams
- Department of Psychiatry and VA Connecticut Healthcare System, Yale School of Medicine, West Haven, CT, USA
| | - Dena B Dubal
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, CA, USA.
| |
Collapse
|
15
|
Birdi A, Tomo S, Yadav D, Sharma P, Nebhinani N, Mitra P, Banerjee M, Purohit P. Role of Klotho Protein in Neuropsychiatric Disorders: A Narrative Review. Indian J Clin Biochem 2023; 38:13-21. [PMID: 36684492 PMCID: PMC9852376 DOI: 10.1007/s12291-022-01078-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 07/20/2022] [Indexed: 01/25/2023]
Abstract
Neuropsychiatric disorders are comprised of diseases having both the neurological and psychiatric manifestations. The increasing burden of the disease on the population worldwide makes it necessary to adopt measures to decrease the prevalence. The Klotho is a single pass transmembrane protein that decreases with age, has been associated with various pathological diseases, like reduced bone mineral density, cardiac problems and cognitive impairment. However, multiple studies have explored its role in different neuropsychiatric disorders. A comprehensive search was undertaken in the Pubmed database for articles with the keywords "Klotho" and "neuropsychiatric disorders". The available literature, based on the above search strategy, has been compiled in this brief narrative review to describe the emerging role of Klotho in various neuropsychiatric disorders. The Klotho levels were decreased in various neuropsychiatric disorders except for bipolar disorder. A suppressed Klotho protein levels induced oxidative stress and incited pro-inflammatory conditions significantly contributing to the pathophysiology of neuropsychiatric disorder. The increasing evidence of altered Klotho protein levels in cognition-decrement-related disorders warrants its consideration as a biomarker in various neuropsychiatric diseases. However, further evidence is required to understand its role as a therapeutic target.
Collapse
Affiliation(s)
- Amandeep Birdi
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan India
| | - Sojit Tomo
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan India
| | - Dharmveer Yadav
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan India
| | - Praveen Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan India
| | - Naresh Nebhinani
- Department of Psychiatry, All India Institute of Medical Sciences, Jodhpur, Rajasthan India
| | - Prasenjit Mitra
- Department of Biochemistry, Post Graduate Institute of Medical Sciences, Chandigarh, Punjab India
| | - Mithu Banerjee
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan India
| | - Purvi Purohit
- Department of Biochemistry, All India Institute of Medical Sciences, Jodhpur, Rajasthan India
| |
Collapse
|
16
|
APOE Allele Frequency in Southern Greece: Exploring the Role of Geographical Gradient in the Greek Population. Geriatrics (Basel) 2022; 8:geriatrics8010001. [PMID: 36648906 PMCID: PMC9844375 DOI: 10.3390/geriatrics8010001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 12/09/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND the apolipoprotein e4 allele (APOE4) constitutes an established genetic risk factor for Alzheimer's Disease Dementia (ADD). We aimed to explore the frequency of the APOE isoforms in the Greek population of Southern Greece. METHODS peripheral blood from 175 Greek AD patients, 113 with mild cognitive impairment (MCI), and 75 healthy individuals. DNA isolation was performed with a High Pure PCR Template Kit (Roche), followed by amplification with a real-time qPCR kit (TIB MolBiol) in Roche's Light Cycler PCR platform. RESULTS APOE4 allele frequency was 20.57% in the ADD group, 17.69% in the MCI group, and 6.67% in the control group. APOE3/3 homozygosity was the most common genotype, while the frequency of APOE4/4 homozygosity was higher in the AD group (8.60%). APOE4 carrier status was associated with higher odds for ADD and MCI (OR: 4.49, 95% CI: [1.90-10.61] and OR: 3.82, 95% CI: [1.59-9.17], respectively). CONCLUSION this study examines the APOE isoforms and is the first to report a higher APOE frequency in MCI compared with healthy controls in southern Greece. Importantly, we report the occurrence of the APOE4 allele, related to ADD, as amongst the lowest globally reported, even within the nation, thus enhancing the theory of ethnicity and latitude contribution.
Collapse
|
17
|
Driscoll I, Ma Y, Lose SR, Gallagher CL, Johnson SC, Asthana S, Hermann BP, Sager MA, Blennow K, Zetterberg H, Carlsson CM, Engelman CD, Dubal DB, Okonkwo OC. AD-associated CSF biomolecular changes are attenuated in KL-VS heterozygotes. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2022; 14:e12383. [PMID: 36505396 PMCID: PMC9728548 DOI: 10.1002/dad2.12383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 12/12/2022]
Abstract
Introduction Dementia as an inevitable aging consequence has been challenged and underscores the need for investigations of the factors that confer resilience. We examine whether the functionally advantageous KL-VS variant of the putative aging suppressor KLOTHO gene attenuates age-related cognitive decline and deleterious biomolecular changes. Methods Trajectories of change in memory and executive function (N = 360; 2-12 visits) and cerebrospinal fluid (CSF) Alzheimer's disease (AD) biomarkers-amyloid beta (Aβ)42, total tau (t-tau), phosphorylated tau (p-tau) (N = 112; 2-4 samplings)-were compared between KL-VS non-carriers and heterozygotes in middle-aged and older adults from the Wisconsin Registry for Alzheimer's Prevention and the Wisconsin Alzheimer's Disease Research Center studies. Results Memory and executive function declined (p's≤ 0.001) and CSF t-tau, p-tau, t-tau/Aβ42, and p-tau/Aβ42 levels increased (all p's≤ 0.004) with age. The rate of p-tau accumulation was attenuated for KL-VS heterozygotes (p = 0.03). Discussion KL-VS heterozygosity may confer resilience to AD-associated biomolecular changes.
Collapse
Affiliation(s)
- Ira Driscoll
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteMadisonWisconsinUSA
- Department of PsychologyUniversity of Wisconsin‐MilwaukeeMilwaukeeWisconsinUSA
| | - Yue Ma
- Wisconsin Alzheimer's InstituteMadisonWisconsinUSA
| | - Sarah R. Lose
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteMadisonWisconsinUSA
| | - Catherine L. Gallagher
- Geriatric Research Education and Clinical CenterWilliam S. Middleton VA HospitalMadisonWisconsinUSA
- Department of NeurologyUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Sterling C. Johnson
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteMadisonWisconsinUSA
- Geriatric Research Education and Clinical CenterWilliam S. Middleton VA HospitalMadisonWisconsinUSA
| | - Sanjay Asthana
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteMadisonWisconsinUSA
- Geriatric Research Education and Clinical CenterWilliam S. Middleton VA HospitalMadisonWisconsinUSA
| | - Bruce P. Hermann
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteMadisonWisconsinUSA
- Department of NeurologyUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Mark A. Sager
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteMadisonWisconsinUSA
| | - Kaj Blennow
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologySahlgrenska Academy at the University of GothenburgGöteborgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologySahlgrenska Academy at the University of GothenburgGöteborgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyQueen SquareLondonUK
- UK Dementia Research Institute at UCLLondonUK
| | - Cynthia M. Carlsson
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteMadisonWisconsinUSA
- Geriatric Research Education and Clinical CenterWilliam S. Middleton VA HospitalMadisonWisconsinUSA
| | - Corinne D. Engelman
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteMadisonWisconsinUSA
- Departments of Population Health SciencesUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Dena B. Dubal
- Department of Neurology and Weill Institute for NeurosciencesUniversity of CaliforniaCaliforniaSan FranciscoUSA
| | - Ozioma C. Okonkwo
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Wisconsin Alzheimer's InstituteMadisonWisconsinUSA
- Geriatric Research Education and Clinical CenterWilliam S. Middleton VA HospitalMadisonWisconsinUSA
| |
Collapse
|
18
|
Grøntvedt GR, Sando SB, Lauridsen C, Bråthen G, White LR, Salvesen Ø, Aarsland D, Hessen E, Fladby T, Waterloo K, Scheffler K. Association of Klotho Protein Levels and KL-VS Heterozygosity With Alzheimer Disease and Amyloid and Tau Burden. JAMA Netw Open 2022; 5:e2243232. [PMID: 36413367 PMCID: PMC9682425 DOI: 10.1001/jamanetworkopen.2022.43232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
IMPORTANCE Identification of proteins and genetic factors that reduce Alzheimer disease (AD) pathology is of importance when searching for novel AD treatments. Heterozygosity of the KL-VS haplotype has been associated with reduced amyloid and tau burden. Whether this association is mediated by the Klotho protein remains unclear. OBJECTIVES To assess concentrations of Klotho in cerebrospinal fluid (CSF) and plasma among cognitively healthy controls and patients with AD and to correlate these findings with KL-VS heterozygosity status and amyloid and tau burden. DESIGN, SETTING, AND PARTICIPANTS This case-control study combined 2 independent case-control AD cohorts consisting of 243 referred patients with AD and volunteer controls recruited from January 1, 2009, to December 31, 2018. Klotho levels were measured in CSF and plasma and correlated with KL-VS heterozygosity status and levels of CSF amyloid-β 42 (Aβ42), total tau, and phosphorylated tau. Statistical analysis was performed from January 1, 2021, to March 1, 2022. MAIN OUTCOMES AND MEASURES Associations of Klotho levels in CSF and plasma with levels of CSF biomarkers were analyzed using linear regression. Association analyses were stratified separately by clinical groups, APOE4 status, and KL-VS heterozygosity. Pearson correlation was used to assess the correlation between CSF and plasma Klotho levels. RESULTS A total of 243 participants were included: 117 controls (45 men [38.5%]; median age, 65 years [range, 41-84 years]), 102 patients with mild cognitive impairment due to AD (AD-MCI; 59 men [57.8%]; median age, 66 years [range, 46-80 years]), and 24 patients with dementia due to AD (AD-dementia; 12 men [50.0%]; median age, 64.5 years [range, 54-75 years]). Median CSF Klotho levels were higher in controls (1236.4 pg/mL [range, 20.4-1726.3 pg/mL]; β = 0.103; 95% CI, 0.023-0.183; P = .01) and patients with AD-MCI (1188.1 pg/mL [range, 756.3-1810.3 pg/mL]; β = 0.095; 95% CI, 0.018-0.172; P = .02) compared with patients with AD-dementia (1073.3 pg/mL [range, 698.2-1661.4 pg/mL]). Higher levels of CSF Klotho were associated with lower CSF Aβ42 burden (β = 0.519; 95% CI, 0.201-0.836; P < .001) and tau burden (CSF total tau levels: β = -0.884; 95% CI, 0.223 to -0.395; P < .001; CSF phosphorylated tau levels: β = -0.672; 95% CI, -1.022 to -0.321; P < .001) independent of clinical, KL-VS heterozygosity, or APOE4 status. There was a weak correlation between Klotho CSF and plasma levels among the entire cohort (Pearson correlation r = 0.377; P < .001). CONCLUSIONS AND RELEVANCE The findings of this case-control study suggest that Klotho protein levels were associated with clinical stages of AD, cognitive decline, and amyloid and tau burden and that these outcomes were more clearly mediated by the protein directly rather than the KL-VS heterozygosity variant. When selecting individuals at risk for clinical trials, the Klotho protein level and not only the genetic profile should be considered.
Collapse
Affiliation(s)
- Gøril Rolfseng Grøntvedt
- Department of Neurology and Clinical Neurophysiology, University Hospital of Trondheim, Trondheim, Norway
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- KG Jebsen Centre for Alzheimer’s Disease, Kavli Institute for Systems Neuroscience, Trondheim, Norway
| | - Sigrid Botne Sando
- Department of Neurology and Clinical Neurophysiology, University Hospital of Trondheim, Trondheim, Norway
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- KG Jebsen Centre for Alzheimer’s Disease, Kavli Institute for Systems Neuroscience, Trondheim, Norway
| | - Camilla Lauridsen
- Department of Neurology and Clinical Neurophysiology, University Hospital of Trondheim, Trondheim, Norway
| | - Geir Bråthen
- Department of Neurology and Clinical Neurophysiology, University Hospital of Trondheim, Trondheim, Norway
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- KG Jebsen Centre for Alzheimer’s Disease, Kavli Institute for Systems Neuroscience, Trondheim, Norway
| | - Linda R. White
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Øyvind Salvesen
- Unit for Applied Clinical Research, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Dag Aarsland
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Erik Hessen
- Department of Psychology, University of Oslo, Oslo, Norway
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway
| | - Tormod Fladby
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, Campus Ahus, University of Oslo, Oslo, Norway
| | - Knut Waterloo
- Department of Neurology, University Hospital of North Norway, Tromsø, Norway
| | - Katja Scheffler
- Department of Neurology and Clinical Neurophysiology, University Hospital of Trondheim, Trondheim, Norway
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- KG Jebsen Centre for Alzheimer’s Disease, Kavli Institute for Systems Neuroscience, Trondheim, Norway
| |
Collapse
|
19
|
Niotis K, Akiyoshi K, Carlton C, Isaacson R. Dementia Prevention in Clinical Practice. Semin Neurol 2022; 42:525-548. [PMID: 36442814 DOI: 10.1055/s-0042-1759580] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Over 55 million people globally are living with dementia and, by 2050, this number is projected to increase to 131 million. This poses immeasurable challenges for patients and their families and a significant threat to domestic and global economies. Given this public health crisis and disappointing results from disease-modifying trials, there has been a recent shift in focus toward primary and secondary prevention strategies. Approximately 40% of Alzheimer's disease (AD) cases, which is the most common form of dementia, may be prevented or at least delayed. Success of risk reduction studies through addressing modifiable risk factors, in addition to the failure of most drug trials, lends support for personalized multidomain interventions rather than a "one-size-fits-all" approach. Evolving evidence supports early intervention in at-risk patients using individualized interventions directed at modifiable risk factors. Comprehensive risk stratification can be informed by emerging principals of precision medicine, and include expanded clinical and family history, anthropometric measurements, blood biomarkers, neurocognitive evaluation, and genetic information. Risk stratification is key in differentiating subtypes of dementia and identifies targetable areas for intervention. This article reviews a clinical approach toward dementia risk stratification and evidence-based prevention strategies, with a primary focus on AD.
Collapse
Affiliation(s)
- Kellyann Niotis
- Department of Neurology, Weill Cornell Medicine and New York - Presbyterian, New York, New York
| | - Kiarra Akiyoshi
- Department of Neurology, Weill Cornell Medicine and New York - Presbyterian, New York, New York
| | - Caroline Carlton
- Department of Neurology, Weill Cornell Medicine and New York - Presbyterian, New York, New York
| | - Richard Isaacson
- Department of Neurology, Weill Cornell Medicine and New York - Presbyterian, New York, New York.,Department of Neurology, Florida Atlantic University, Charles E. Schmidt College of Medicine, Boca Raton, Florida
| |
Collapse
|
20
|
Copur S, Berkkan M, Sarafidis P, Kanbay M. Intensive blood pressure control on dementia in patients with chronic kidney disease: Potential reduction in disease burden. Eur J Intern Med 2022; 101:8-13. [PMID: 35465970 DOI: 10.1016/j.ejim.2022.04.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/10/2022] [Accepted: 04/16/2022] [Indexed: 12/17/2022]
Abstract
Chronic kidney disease (CKD) and dementia are both common comorbidities creating considerable morbidity and mortality, especially in the elderly population with potential interactions. Even though various hypothetical mechanisms underlying the pathophysiology of increased risk of dementia and cognitive impairment in CKD patients have been implicated, no consensus has been reached so far. Recent clinical trials have investigated the therapeutic role of intensive blood pressure control on the risk of dementia in CKD patients with potentially improved outcomes. However, such trials have significant limitations that may influence the outcome and lack specific management guidelines. We reviewed the role of blood pressure and other factors on the risk of dementia in CKD patients which is an issue with high potential for clinical implications that may improve morbidity, mortality, and health expenditures along with its' potential pathophysiological mechanisms and future guidance.
Collapse
Affiliation(s)
- Sidar Copur
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Metehan Berkkan
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Pantelis Sarafidis
- Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, Greece
| | - Mehmet Kanbay
- Department of Medicine, Division of Nephrology, Koc University School of Medicine, Istanbul, Turkey.
| |
Collapse
|
21
|
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
|
22
|
Abstract
An increasing amount of very diverse scholarship self-identifies as belonging to the field of neuroethics, illuminating a need to provide some reference points for what that field actually entails. We argue that neuroethics is a single field with distinct perspectives, roles, and subspecialties. We propose that-in addition to the three traditional perspectives delineated by Eric Racine-a fourth, socio-political perspective, must be recognized in neuroethics. The socio-political perspective in neuroethics focuses on the interplay between the behavioral as well as the brain sciences and the socio-political system; this interplay includes social regulation in addition to all other realistic elements of social and political neurodiscourses. Thus, defining what-if any-roles the socio-political perspective in neuroethics might have is a pressing issue. Doing so could provide guidance for defining the criteria for prospective ethical evaluations in neuroethics. A promising approach to doing this could be by describing the roles of neuroethics in terms of the more concrete examples of the roles of political philosophy in general, as in the tradition of John Rawls. We take klotho, the supposed "longevity protein," as a modern neuroethics case to exemplify the obstacles faced in securing neuroethics' legitimacy and how the Rawlsian framework we propose may be applied to handle cases such as this. Ultimately, the socio-political perspective in neuroethics should not be swayed by the media hype and ought to offer useful ethical guidance and articulation of genuine ethical concerns to policy makers and the public alike.
Collapse
|
23
|
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
|
24
|
Kundu P, Zimmerman B, Quinn JF, Kaye J, Mattek N, Westaway SK, Raber J. Serum Levels of α-Klotho Are Correlated with Cerebrospinal Fluid Levels and Predict Measures of Cognitive Function. J Alzheimers Dis 2022; 86:1471-1481. [PMID: 35213382 PMCID: PMC9108571 DOI: 10.3233/jad-215719] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2022] [Indexed: 02/05/2023]
Abstract
BACKGROUND α-klotho might play a role in neurodegenerative diseases. OBJECTIVE To determine levels of α-klotho and apoE in serum and cerebrospinal fluid (CSF) samples and their relationship with the Mini-Mental State Examination (MMSE) and Clinical Dementia Rating (CDR). METHODS All subjects were between age 39 to 83+ (n = 94). CDR and MMSE were administered to all participants. CSF was collected in the early afternoon by lumbar puncture. RESULTS Serum and CSF levels of α-klotho are positively correlated and both predict scores on the MMSE and CDR, regardless of sex or apoE4 status. CONCLUSION Our results demonstrate that α-klotho may be an important biomarker of cognitive health and neurodegeneration, and that relatively non-invasive sampling of α-klotho from serum is likely highly reflective of CSF levels.
Collapse
Affiliation(s)
- Payel Kundu
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, USA
| | - Benjamin Zimmerman
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, USA
- Advanced Imaging Research Center, Oregon Health and Science University, Portland, OR, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Joseph F. Quinn
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
- Department of Neurology, Veterans Affairs Medical Center, Portland, OR, USA
| | - Jeffrey Kaye
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
| | - Nora Mattek
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
| | - Shawn K. Westaway
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, USA
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
- Departments of Psychiatry and Radiation Medicine, Division of Neuroscience, ONPRC, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
25
|
Viggiano D, Bruchfeld A, Carriazo S, de Donato A, Endlich N, Ferreira AC, Figurek A, Fouque D, Franssen CFM, Giannakou K, Goumenos D, Hoorn EJ, Nitsch D, Arduan AO, Pešić V, Rastenyté D, Soler MJ, Rroji M, Trepiccione F, Unwin RJ, Wagner CA, Wiecek A, Zacchia M, Zoccali C, Capasso G. Brain dysfunction in tubular and tubulointerstitial kidney diseases. Nephrol Dial Transplant 2021; 37:ii46-ii55. [PMID: 34792176 PMCID: PMC8713153 DOI: 10.1093/ndt/gfab276] [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: 05/26/2021] [Indexed: 11/14/2022] Open
Abstract
Kidney function has two important elements: glomerular filtration and tubular function (secretion and reabsorption). A persistent decrease in glomerular filtration rate (GFR), with or without proteinuria, is diagnostic of chronic kidney disease (CKD). While glomerular injury or disease is a major cause of CKD and usually associated with proteinuria, predominant tubular injury, with or without tubulointerstitial disease, is typically non-proteinuric. CKD has been linked with cognitive impairment, but it is unclear how much this depends on a reduced GFR, altered tubular function or the presence of proteinuria. Since CKD is often accompanied by tubular and interstitial dysfunction, we explore here for the first time the potential role of the tubular and tubulointerstitial compartments in cognitive dysfunction. To help address this issue, we have selected a group of primary tubular diseases with preserved GFR, in which to review the evidence for any association with brain dysfunction. Cognition, mood, neurosensory, and motor disturbances are not well characterized in tubular diseases, possibly because they are subclinical and less prominent than other clinical manifestations. The available literature suggests that brain dysfunction in tubular and tubulointerstitial diseases is usually mild and is more often seen in disorders of water handling. Brain dysfunction may occur when severe electrolyte and water disorders in young children persist over a long period of time before the diagnosis is made. We have chosen as examples to highlight this topic, Bartter and Gitelman syndromes and nephrogenic diabetes insipidus. We discuss current published findings, some unanswered questions, and propose topics for future research.
Collapse
Affiliation(s)
- Davide Viggiano
- Department of Translational Medical Sciences, Univ. Campania "L.Vanvitelli", Naples, Italy. BIOGEM, Institute of Molecular Biology and Genetics, Ariano Irpino. Italy
| | - Annette Bruchfeld
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden. Department of Renal Medicine, Karolinska University Hospital and CLINTEC Karolinska Institutet, Stockholm, Sweden
| | - Sol Carriazo
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | - Antonio de Donato
- Department of Translational Medical Sciences, Univ. Campania "L.Vanvitelli", Naples, Italy. BIOGEM, Institute of Molecular Biology and Genetics, Ariano Irpino. Italy
| | - Nicole Endlich
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Germany
| | - Ana Carina Ferreira
- Nephrology Department, Centro Hospitalar E Universitário de Lisboa Central, Lisbon, Portugal; Universidade Nova de Lisboa
- Faculdade de Ciências Médicas, Lisbon, Portugal
| | - Andreja Figurek
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Denis Fouque
- Department of Nephrology, Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Pierre-Benite, France; University of Lyon, France
| | - Casper F M Franssen
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Konstantinos Giannakou
- Department of Health Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus
| | - Dimitrios Goumenos
- Department of Nephrology and Renal Transplantation, Patras University Hospital, Patras, Greece
| | - Ewout J Hoorn
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Dorothea Nitsch
- Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Alberto Ortiz Arduan
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz-UAM, Madrid, Spain
| | - Vesna Pešić
- Department of Physiology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Daiva Rastenyté
- Medical Academy, Department of Neurology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Maria José Soler
- Nephrology Department, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Merita Rroji
- Department of Nephrology, University Hospital Center "Mother Tereza", Tirana, Albania
| | - Francesco Trepiccione
- Department of Translational Medical Sciences, Univ. Campania "L.Vanvitelli", Naples, Italy. BIOGEM, Institute of Molecular Biology and Genetics, Ariano Irpino, Italy
| | - Robert J Unwin
- Department of Renal Medicine, Division of Medicine, University College London, UK
| | - Carsten A Wagner
- Institute of Physiology, University of Zürich, Zurich, Switzerland
| | - Andrzej Wiecek
- Department of Nephrology, Transplantation and Internal Medicine, Medical University of Silesia, Katowice, Poland
| | - Miriam Zacchia
- Department of Translational Medical Sciences, Univ. Campania "L.Vanvitelli", Naples, Italy. BIOGEM, Institute of Molecular Biology and Genetics, Ariano Irpino, Italy
| | - Carmine Zoccali
- Renal Research Institute, New York, USA and Associazione Ipertensione, Nefrologia, Trapianto Renale (IPNET), Italy
| | - Giovambattista Capasso
- Department of Translational Medical Sciences, Univ. Campania "L.Vanvitelli", Naples, Italy. BIOGEM, Institute of Molecular Biology and Genetics, Ariano Irpino, Italy
| | | |
Collapse
|
26
|
Walker CK, Herskowitz JH. Dendritic Spines: Mediators of Cognitive Resilience in Aging and Alzheimer's Disease. Neuroscientist 2021; 27:487-505. [PMID: 32812494 PMCID: PMC8130863 DOI: 10.1177/1073858420945964] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cognitive resilience is often defined as the ability to remain cognitively normal in the face of insults to the brain. These insults can include disease pathology, such as plaques and tangles associated with Alzheimer's disease, stroke, traumatic brain injury, or other lesions. Factors such as physical or mental activity and genetics may contribute to cognitive resilience, but the neurobiological underpinnings remain ill-defined. Emerging evidence suggests that dendritic spine structural plasticity is one plausible mechanism. In this review, we highlight the basic structure and function of dendritic spines and discuss how spine density and morphology change in aging and Alzheimer's disease. We note evidence that spine plasticity mediates resilience to stress, and we tackle dendritic spines in the context of cognitive resilience to Alzheimer's disease. Finally, we examine how lifestyle and genetic factors may influence dendritic spine plasticity to promote cognitive resilience before discussing evidence for actin regulatory kinases as therapeutic targets for Alzheimer's disease.
Collapse
Affiliation(s)
- Courtney K. Walker
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, USA
| | - Jeremy H. Herskowitz
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, USA
| |
Collapse
|
27
|
Sohn HY, Kim SI, Park JY, Park SH, Koh YH, Kim J, Jo C. ApoE4 attenuates autophagy via FoxO3a repression in the brain. Sci Rep 2021; 11:17604. [PMID: 34475505 PMCID: PMC8413297 DOI: 10.1038/s41598-021-97117-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 08/19/2021] [Indexed: 02/07/2023] Open
Abstract
Apolipoprotein E (ApoE) plays multiple roles in lipid transport, neuronal signaling, glucose metabolism, mitochondrial function, and inflammation in the brain. It is also associated with neurodegenerative diseases, and its influence differs depending on the isoform. In particular, the ε4 allele of APOE is the highest genetic risk factor for developing late-onset Alzheimer's disease (AD). However, the mechanism by which ApoE4 contributes to the pathogenesis of AD remains unclear. We investigated the effect of ApoE4 on autophagy in the human brains of ApoE4 carriers. Compared to non-carriers, the expression of FoxO3a regulating autophagy-related genes was significantly reduced in ApoE4 carriers, and the phosphorylation level of FoxO3a at Ser253 increased in ApoE4 carriers, indicating that FoxO3a is considerably repressed in ApoE4 carriers. As a result, the protein expression of FoxO3a downstream genes, such as Atg12, Beclin-1, BNIP3, and PINK1, was significantly decreased, likely leading to dysfunction of both autophagy and mitophagy in ApoE4 carriers. In addition, phosphorylated tau accumulated more in ApoE4 carriers than in non-carriers. Taken together, our results suggest that ApoE4 might attenuate autophagy via the repression of FoxO3a in AD pathogenesis. The regulation of the ApoE4-FoxO3a axis may provide a novel therapeutic target for the prevention and treatment of AD with the APOE4 allele.
Collapse
Affiliation(s)
- Hee-Young Sohn
- grid.415482.e0000 0004 0647 4899Division of Brain Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, 187 Osongsaengmyeong2-ro, Osong-eup, Cheongju-si, Chungcheongbuk-do 28159 Republic of Korea ,grid.222754.40000 0001 0840 2678Laboratory of Biochemistry, Division of Life Sciences, Korea University, Seoul, 02841 Republic of Korea
| | - Seong-Ik Kim
- grid.31501.360000 0004 0470 5905Department of Pathology, Seoul National University College of Medicine, Seoul, 03080 Republic of Korea
| | - Jee-Yun Park
- grid.415482.e0000 0004 0647 4899Division of Brain Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, 187 Osongsaengmyeong2-ro, Osong-eup, Cheongju-si, Chungcheongbuk-do 28159 Republic of Korea
| | - Sung-Hye Park
- grid.31501.360000 0004 0470 5905Department of Pathology, Seoul National University College of Medicine, Seoul, 03080 Republic of Korea
| | - Young Ho Koh
- grid.415482.e0000 0004 0647 4899Division of Brain Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, 187 Osongsaengmyeong2-ro, Osong-eup, Cheongju-si, Chungcheongbuk-do 28159 Republic of Korea
| | - Joon Kim
- grid.222754.40000 0001 0840 2678Laboratory of Biochemistry, Division of Life Sciences, Korea University, Seoul, 02841 Republic of Korea
| | - Chulman Jo
- grid.415482.e0000 0004 0647 4899Division of Brain Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, 187 Osongsaengmyeong2-ro, Osong-eup, Cheongju-si, Chungcheongbuk-do 28159 Republic of Korea
| |
Collapse
|
28
|
Exercise-Linked Irisin Prevents Mortality and Enhances Cognition in a Mice Model of Cerebral Ischemia by Regulating Klotho Expression. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:1697070. [PMID: 34306305 PMCID: PMC8282383 DOI: 10.1155/2021/1697070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/13/2021] [Accepted: 06/13/2021] [Indexed: 01/04/2023]
Abstract
Irisin, which can be released in the hippocampus after physical exercise, is demonstrated to have beneficial effects on neurovascular diseases. This study investigated the impact of exercise linked-irisin on mortality and cognition in a mice model of cerebral ischemia and further explored its underlying mechanism. The cerebrospinal concentrations of irisin and klotho from ischemic stroke patients were measured with an enzyme-linked immunosorbent assay (ELISA). The cognitive function of mice was evaluated by a series of behavioural experiments. The expressions of klotho, MnSOD, and FOXO3a in the hippocampus of mice were detected by Western blot. Superoxide production in the brain tissue of mice was evaluated with the dihydroethidium (DHE) dying. The results demonstrated that stroke patients showed a positive correlation between their CSF irisin concentration and klotho concentration. In addition, when mice subjected to cerebral ischemia, their cognitive function was impaired, the protein expressions of klotho, MnSOD, and FOXO3a downregulated, and the production of reactive oxygen species (ROS) increased compared with the sham group. After pretreatment with exogenous irisin, improved cognitive impairment, upregulated protein expressions of klotho, MnSOD, and FOXO3a, and reduced ROS generation were observed in mice with MCAO. However, the neuroprotective effects of irisin compromised with the evidence of severe cognitive impairment, decreased protein expressions of MnSOD and FOXO3a, and increased ROS production in klotho knockout mice. Thus, our results indicated that exercise-linked irisin could prevent mortality and improve cognitive impairment after cerebral ischemia by regulating klotho expression.
Collapse
|
29
|
KL-VS heterozygosity is associated with lower amyloid-dependent tau accumulation and memory impairment in Alzheimer's disease. Nat Commun 2021; 12:3825. [PMID: 34158479 PMCID: PMC8219708 DOI: 10.1038/s41467-021-23755-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 05/12/2021] [Indexed: 11/08/2022] Open
Abstract
Klotho-VS heterozygosity (KL-VShet) is associated with reduced risk of Alzheimer’s disease (AD). However, whether KL-VShet is associated with lower levels of pathologic tau, i.e., the key AD pathology driving neurodegeneration and cognitive decline, is unknown. Here, we assessed the interaction between KL-VShet and levels of beta-amyloid, a key driver of tau pathology, on the levels of PET-assessed neurofibrillary tau in 551 controls and patients across the AD continuum. KL-VShet showed lower cross-sectional and longitudinal increase in tau-PET per unit increase in amyloid-PET when compared to that of non-carriers. This association of KL-VShet on tau-PET was stronger in Klotho mRNA-expressing brain regions mapped onto a gene expression atlas. KL-VShet was related to better memory functions in amyloid-positive participants and this association was mediated by lower tau-PET. Amyloid-PET levels did not differ between KL-VShet carriers versus non-carriers. Together, our findings provide evidence to suggest a protective role of KL-VShet against amyloid-related tau pathology and tau-related memory impairments in elderly humans at risk of AD dementia. The KL-VS haplotype of the Klotho gene has been associated with reduced risk of Alzheimer’s disease and dementia. Here the authors show an association between the KL-VS haplotype and amyloid-dependent tau accumulation using PET data.
Collapse
|
30
|
Hanson K, Fisher K, Hooper N. Exploiting the neuroprotective effects of α-klotho to tackle ageing- and neurodegeneration-related cognitive dysfunction. Neuronal Signal 2021; 5:NS20200101. [PMID: 34194816 PMCID: PMC8204227 DOI: 10.1042/ns20200101] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 12/26/2022] Open
Abstract
Cognitive dysfunction is a key symptom of ageing and neurodegenerative disorders, such as Alzheimer's disease (AD). Strategies to enhance cognition would impact the quality of life for a significant proportion of the ageing population. The α-klotho protein may protect against cognitive decline through multiple mechanisms: such as promoting optimal synaptic function via activation of N-methyl-d-aspartate (NMDA) receptor signalling; stimulating the antioxidant defence system; reducing inflammation; promoting autophagy and enhancing clearance of amyloid-β. However, the molecular and cellular pathways by which α-klotho mediates these neuroprotective functions have yet to be fully elucidated. Key questions remain unanswered: which form of α-klotho (transmembrane, soluble or secreted) mediates its cognitive enhancing properties; what is the neuronal receptor for α-klotho and which signalling pathways are activated by α-klotho in the brain to enhance cognition; how does peripherally administered α-klotho mediate neuroprotection; and what is the molecular basis for the beneficial effect of the VS variant of α-klotho? In this review, we summarise the recent research on neuronal α-klotho and discuss how the neuroprotective properties of α-klotho could be exploited to tackle age- and neurodegeneration-associated cognitive dysfunction.
Collapse
Affiliation(s)
- Kelsey Hanson
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, U.K
| | - Kate Fisher
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, U.K
| | - Nigel M. Hooper
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, U.K
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance and University of Manchester, Manchester, U.K
| |
Collapse
|
31
|
Gaitán JM, Moon HY, Stremlau M, Dubal DB, Cook DB, Okonkwo OC, van Praag H. Effects of Aerobic Exercise Training on Systemic Biomarkers and Cognition in Late Middle-Aged Adults at Risk for Alzheimer's Disease. Front Endocrinol (Lausanne) 2021; 12:660181. [PMID: 34093436 PMCID: PMC8173166 DOI: 10.3389/fendo.2021.660181] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/30/2021] [Indexed: 12/11/2022] Open
Abstract
Increasing evidence indicates that physical activity and exercise training may delay or prevent the onset of Alzheimer's disease (AD). However, systemic biomarkers that can measure exercise effects on brain function and that link to relevant metabolic responses are lacking. To begin to address this issue, we utilized blood samples of 23 asymptomatic late middle-aged adults, with familial and genetic risk for AD (mean age 65 years old, 50% female) who underwent 26 weeks of supervised treadmill training. Systemic biomarkers implicated in learning and memory, including the myokine Cathepsin B (CTSB), brain-derived neurotrophic factor (BDNF), and klotho, as well as metabolomics were evaluated. Here we show that aerobic exercise training increases plasma CTSB and that changes in CTSB, but not BDNF or klotho, correlate with cognitive performance. BDNF levels decreased with exercise training. Klotho levels were unchanged by training, but closely associated with change in VO2peak. Metabolomic analysis revealed increased levels of polyunsaturated free fatty acids (PUFAs), reductions in ceramides, sphingo- and phospholipids, as well as changes in gut microbiome metabolites and redox homeostasis, with exercise. Multiple metabolites (~30%) correlated with changes in BDNF, but not CSTB or klotho. The positive association between CTSB and cognition, and the modulation of lipid metabolites implicated in dementia, support the beneficial effects of exercise training on brain function. Overall, our analyses indicate metabolic regulation of exercise-induced plasma BDNF changes and provide evidence that CTSB is a marker of cognitive changes in late middle-aged adults at risk for dementia.
Collapse
Affiliation(s)
- Julian M. Gaitán
- Wisconsin Alzheimer’s Disease Research Center and Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Hyo Youl Moon
- Lab of Neurosciences, National Institute on Aging (NIA), Baltimore, MD, United States
- Department of Education, Seoul National University, Seoul, South Korea
- Institute of Sport Science, Seoul National University, Seoul, South Korea
- Institute on Aging, Seoul National University, Seoul, South Korea
| | - Matthew Stremlau
- Lab of Neurosciences, National Institute on Aging (NIA), Baltimore, MD, United States
| | - Dena B. Dubal
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, United States
| | - Dane B. Cook
- Department of Kinesiology, University of Wisconsin School of Education, Madison, WI, United States
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI, United States
| | - Ozioma C. Okonkwo
- Wisconsin Alzheimer’s Disease Research Center and Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, United States
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Henriette van Praag
- Lab of Neurosciences, National Institute on Aging (NIA), Baltimore, MD, United States
- Brain Institute and Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, United States
| |
Collapse
|
32
|
Seto M, Weiner RL, Dumitrescu L, Hohman TJ. Protective genes and pathways in Alzheimer's disease: moving towards precision interventions. Mol Neurodegener 2021; 16:29. [PMID: 33926499 PMCID: PMC8086309 DOI: 10.1186/s13024-021-00452-5] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 04/20/2021] [Indexed: 12/29/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive, neurodegenerative disorder that is characterized by neurodegeneration, cognitive impairment, and an eventual inability to perform daily tasks. The etiology of Alzheimer's is complex, with numerous environmental and genetic factors contributing to the disease. Late-onset AD is highly heritable (60 to 80%), and over 40 risk loci for AD have been identified via large genome-wide association studies, most of which are common variants with small effect sizes. Although these discoveries have provided novel insight on biological contributors to AD, disease-modifying treatments remain elusive. Recently, the concepts of resistance to pathology and resilience against the downstream consequences of pathology have been of particular interest in the Alzheimer's field as studies continue to identify individuals who evade the pathology of the disease even into late life and individuals who have all of the neuropathological features of AD but evade downstream neurodegeneration and cognitive impairment. It has been hypothesized that a shift in focus from Alzheimer's risk to resilience presents an opportunity to uncover novel biological mechanisms of AD and to identify promising therapeutic targets for the disease. This review will highlight a selection of genes and variants that have been reported to confer protection from AD within the literature and will also discuss evidence for the biological underpinnings behind their protective effect with a focus on genes involved in lipid metabolism, cellular trafficking, endosomal and lysosomal function, synaptic function, and inflammation. Finally, we offer some recommendations in areas where the field can rapidly advance towards precision interventions that leverage the ideas of protection and resilience for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Mabel Seto
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, 1207 17th Ave S, Nashville, TN 37212 USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN USA
- Department of Pharmacology, Vanderbilt University, Nashville, TN USA
| | - Rebecca L. Weiner
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, 1207 17th Ave S, Nashville, TN 37212 USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN USA
- Department of Pharmacology, Vanderbilt University, Nashville, TN USA
| | - Logan Dumitrescu
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, 1207 17th Ave S, Nashville, TN 37212 USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN USA
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN USA
| | - Timothy J. Hohman
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, 1207 17th Ave S, Nashville, TN 37212 USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN USA
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN USA
| |
Collapse
|
33
|
Wolf EJ, Chen CD, Zhao X, Zhou Z, Morrison FG, Daskalakis NP, Stone A, Schichman S, Grenier JG, Fein-Schaffer D, Huber BR, Abraham CR, Miller MW, Logue MW. Klotho, PTSD, and advanced epigenetic age in cortical tissue. Neuropsychopharmacology 2021; 46:721-730. [PMID: 33096543 PMCID: PMC8027437 DOI: 10.1038/s41386-020-00884-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 09/12/2020] [Accepted: 09/29/2020] [Indexed: 01/04/2023]
Abstract
This study examined the klotho (KL) longevity gene polymorphism rs9315202 and psychopathology, including posttraumatic stress disorder (PTSD), depression, and alcohol-use disorders, in association with advanced epigenetic age in three postmortem cortical tissue regions: dorsolateral and ventromedial prefrontal cortices and motor cortex. Using data from the VA National PTSD Brain Bank (n = 117), we found that rs9315202 interacted with PTSD to predict advanced epigenetic age in motor cortex among the subset of relatively older (>=45 years), white non-Hispanic decedents (corrected p = 0.014, n = 42). An evaluation of 211 additional common KL variants revealed that only variants in linkage disequilibrium with rs9315202 showed similarly high levels of significance. Alcohol abuse was nominally associated with advanced epigenetic age in motor cortex (p = 0.039, n = 114). The rs9315202 SNP interacted with PTSD to predict decreased KL expression via DNAm age residuals in motor cortex among older white non-Hispanics decedents (indirect β = -0.198, p = 0.027). Finally, in dual-luciferase enhancer reporter system experiments, we found that inserting the minor allele of rs9315202 in a human kidney cell line HK-2 genomic DNA resulted in a change in KL transcriptional activities, likely operating via long noncoding RNA in this region. This was the first study to examine multiple forms of psychopathology in association with advanced DNA methylation age across several brain regions, to extend work concerning the association between rs9315202 and advanced epigenetic to brain tissue, and to identify the effects of rs9315202 on KL gene expression. KL augmentation holds promise as a therapeutic intervention to slow the pace of cellular aging, disease onset, and neuropathology, particularly in older, stressed populations.
Collapse
Affiliation(s)
- Erika J Wolf
- National Center for PTSD at VA Boston Healthcare System, Boston, MA, USA.
- Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA.
| | - Ci-Di Chen
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Xiang Zhao
- National Center for PTSD at VA Boston Healthcare System, Boston, MA, USA
- Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Zhenwei Zhou
- National Center for PTSD at VA Boston Healthcare System, Boston, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Filomene G Morrison
- National Center for PTSD at VA Boston Healthcare System, Boston, MA, USA
- Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | | | - Annjanette Stone
- Pharmacogenomics Analysis Laboratory, Research Service, Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Steven Schichman
- Pharmacogenomics Analysis Laboratory, Research Service, Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Jaclyn Garza Grenier
- Brigham and Women's Hospital, Channing Division of Network Medicine, Boston, MA, USA
| | - Dana Fein-Schaffer
- National Center for PTSD at VA Boston Healthcare System, Boston, MA, USA
| | - Bertrand R Huber
- Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Carmela R Abraham
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Mark W Miller
- National Center for PTSD at VA Boston Healthcare System, Boston, MA, USA
- Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
| | - Mark W Logue
- National Center for PTSD at VA Boston Healthcare System, Boston, MA, USA
- Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Biomedical Genetics, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
34
|
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
|
35
|
Zimmermann M, Köhler L, Kovarova M, Lerche S, Schulte C, Wurster I, Machetanz G, Deuschle C, Hauser AK, Gasser T, Berg D, Schleicher E, Maetzler W, Brockmann K. The longevity gene Klotho and its cerebrospinal fluid protein profiles as a modifier for Parkinson´s disease. Eur J Neurol 2021; 28:1557-1565. [PMID: 33449400 DOI: 10.1111/ene.14733] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 01/08/2021] [Accepted: 01/11/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Parkinson´s disease (PD) has a large phenotypic variability, which may, at least partly, be genetically driven including alterations of gene products. Candidates might not only be proteins associated with disease risk but also pathways that play a role in aging. OBJECTIVE To evaluate phenotype-modifying effects of genetic variants in Klotho, a longevity gene. METHODS We analyzed two longitudinal cohorts: one local cohort comprising 459 PD patients who underwent genotyping for the KL-VS haplotype in Klotho including a subgroup of 125 PD patients and 50 healthy controls who underwent biochemical cerebrospinal fluid (CSF) analyses of Klotho and fibroblast growth factor 23 as well as vitamin D metabolites. The second cohort comprised 297 patients from the Parkinson's Progression Markers Initiative (PPMI) for validation of genetic-clinical findings. RESULTS PD patients carrying the KL-VS haplotype demonstrated a shorter interval between PD onset and onset of cognitive impairment (both cohorts) and higher Unified Parkinson´s Disease Rating Scale part III (UPDRS III) scores (PPMI). CSF protein levels of Klotho and fibroblast growth factor 23 were lower in PD patients irrespective of gender compared to controls. Moreover, low CSF levels of Klotho were associated with higher scores in the UPDRS III and Hoehn and Yahr Scale. CONCLUSIONS Our results indicate that genetic variants in Klotho together with its corresponding CSF protein profiles are associated with aspects of disease severity in PD. These findings suggest that pathways associated with aging might be targets for future biomarker research in PD.
Collapse
Affiliation(s)
- Milan Zimmermann
- Center of Neurology, Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany.,German Center for Neurodegenerative Diseases (DZNE, University of Tuebingen, Tuebingen, Germany
| | - Leonie Köhler
- Center of Neurology, Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany
| | - Marketa Kovarova
- Department of Internal Medicine, University of Tuebingen, Tuebingen, Germany
| | - Stefanie Lerche
- Center of Neurology, Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany.,German Center for Neurodegenerative Diseases (DZNE, University of Tuebingen, Tuebingen, Germany
| | - Claudia Schulte
- Center of Neurology, Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany.,German Center for Neurodegenerative Diseases (DZNE, University of Tuebingen, Tuebingen, Germany
| | - Isabel Wurster
- Center of Neurology, Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany.,German Center for Neurodegenerative Diseases (DZNE, University of Tuebingen, Tuebingen, Germany
| | - Gerrit Machetanz
- Center of Neurology, Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany.,German Center for Neurodegenerative Diseases (DZNE, University of Tuebingen, Tuebingen, Germany
| | - Christian Deuschle
- Center of Neurology, Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany.,German Center for Neurodegenerative Diseases (DZNE, University of Tuebingen, Tuebingen, Germany
| | - Ann-Kathrin Hauser
- Center of Neurology, Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany.,German Center for Neurodegenerative Diseases (DZNE, University of Tuebingen, Tuebingen, Germany
| | - Thomas Gasser
- Center of Neurology, Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany.,German Center for Neurodegenerative Diseases (DZNE, University of Tuebingen, Tuebingen, Germany
| | - Daniela Berg
- Center of Neurology, Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany.,Department of Neurology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Erwin Schleicher
- Department of Internal Medicine, University of Tuebingen, Tuebingen, Germany
| | - Walter Maetzler
- Department of Neurology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Kathrin Brockmann
- Center of Neurology, Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research, University of Tuebingen, Tuebingen, Germany.,German Center for Neurodegenerative Diseases (DZNE, University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
36
|
Driscoll I, Ma Y, Gallagher CL, Johnson SC, Asthana S, Hermann BP, Sager MA, Blennow K, Zetterberg H, Carlsson CM, Engelman CD, Dubal DB, Okonkwo OC. Age-Related Tau Burden and Cognitive Deficits Are Attenuated in KLOTHO KL-VS Heterozygotes. J Alzheimers Dis 2021; 79:1297-1305. [PMID: 33427737 DOI: 10.3233/jad-200944] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Identification of new genetic variants that modify Alzheimer's disease (AD) risk will elucidate novel targets for curbing the disease progression or delaying symptom onset. OBJECTIVE To examine whether the functionally advantageous KLOTHO gene KL-VS variant attenuates age-related alteration in cerebrospinal fluid (CSF) biomarkers or cognitive function in middle-aged and older adults enriched for AD risk. METHODS Sample included non-demented adults (N = 225, mean age = 63±8, 68% women) from the Wisconsin Registry for Alzheimer's Prevention and the Wisconsin Alzheimer's Disease Research Center who were genotyped for KL-VS, underwent CSF sampling and had neuropsychological testing data available proximal to CSF draw. Covariate-adjusted multivariate regression examined relationships between age group (Younger versus Older; mean split at 63 years), AD biomarkers, and neuropsychological performance tapping memory and executive function, and whether these relationships differed between KL-VS non-carriers (KL-VSNC) and heterozygote (KL-VSHET). RESULTS In the pooled analyses, older age was associated with higher levels of total tau (tTau), phosphorylated tau (pTau), and their respective ratios to amyloid-β (Aβ)42 (ps ≤ 0.002), and with poorer performance on neuropsychological tests (ps ≤ 0.001). In the stratified analyses, KL-VSNC exhibited this age-related pattern of associations with CSF biomarkers (all ps ≤ 0.001), and memory and executive function (ps ≤ 0.003), which were attenuated in KL-VSHET (ps ≥ 0.14). CONCLUSION Worse memory and executive function, and higher tau burden with age were attenuated in carriers of a functionally advantageous KLOTHO variant. KL-VS heterozygosity seems to be protective against age-related cognitive and biomolecular alterations that confer risk for AD.
Collapse
Affiliation(s)
- Ira Driscoll
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, Madison, WI, USA.,Wisconsin Alzheimer's Institute, Madison, WI, USA.,Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Yue Ma
- Wisconsin Alzheimer's Institute, Madison, WI, USA
| | - Catherine L Gallagher
- Geriatric Research Education and Clinical Center, William S. Middleton VA Hospital, Madison, WI, USA.,Department of Neurology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Sterling C Johnson
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, Madison, WI, USA.,Wisconsin Alzheimer's Institute, Madison, WI, USA.,Geriatric Research Education and Clinical Center, William S. Middleton VA Hospital, Madison, WI, USA
| | - Sanjay Asthana
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, Madison, WI, USA.,Wisconsin Alzheimer's Institute, Madison, WI, USA.,Geriatric Research Education and Clinical Center, William S. Middleton VA Hospital, Madison, WI, USA
| | - Bruce P Hermann
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, Madison, WI, USA.,Wisconsin Alzheimer's Institute, Madison, WI, USA.,Department of Neurology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Mark A Sager
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, Madison, WI, USA.,Wisconsin Alzheimer's Institute, Madison, WI, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Göteborg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Göteborg, 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
| | - Cynthia M Carlsson
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, Madison, WI, USA.,Wisconsin Alzheimer's Institute, Madison, WI, USA.,Geriatric Research Education and Clinical Center, William S. Middleton VA Hospital, Madison, WI, USA
| | - Corinne D Engelman
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, Madison, WI, USA.,Wisconsin Alzheimer's Institute, Madison, WI, USA.,Departments of Population Health Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Dena B Dubal
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Ozioma C Okonkwo
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin-Madison, 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
|
37
|
Could α-Klotho Unlock the Key Between Depression and Dementia in the Elderly: from Animal to Human Studies. Mol Neurobiol 2021; 58:2874-2885. [PMID: 33527303 DOI: 10.1007/s12035-021-02313-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 01/25/2021] [Indexed: 10/22/2022]
Abstract
α-Klotho is known for its aging-related functions and is associated with neurodegenerative diseases, accelerated aging, premature morbidity, and mortality. Recent literature suggests that α-Klotho is also involved in the regulation of mental functions, such as cognition and psychosis. While most of studies of α-Klotho are focusing on its anti-aging functions and protective role in dementia, increasing evidence showed many shared symptoms between depression and dementia, while depression has been proposed as the preclinical stage of dementia such as Alzheimer's disease (AD). To see whether and how α-Klotho can be a key biological link between depression and dementia, in this review, we first gathered the evidence on biological distribution and function of α-Klotho in psychiatric functions from animal studies to human clinical investigations with a focus on the regulation of cognition and mood. Then, we discussed and highlighted the potential common underlying mechanisms of α-Klotho between psychiatric diseases and cognitive impairment. Finally, we hypothesized that α-Klotho might serve as a neurobiological link between depression and dementia through the regulation of oxidative stress and inflammation.
Collapse
|
38
|
Belloy ME, Eger SJ, Le Guen Y, Napolioni V, Deters KD, Yang HS, Scelsi MA, Porter T, James SN, Wong A, Schott JM, Sperling RA, Laws SM, Mormino EC, He Z, Han SS, Altmann A, Greicius MD. KL∗VS heterozygosity reduces brain amyloid in asymptomatic at-risk APOE∗4 carriers. Neurobiol Aging 2021; 101:123-129. [PMID: 33610961 DOI: 10.1016/j.neurobiolaging.2021.01.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/30/2020] [Accepted: 01/09/2021] [Indexed: 11/15/2022]
Abstract
KLOTHO∗VS heterozygosity (KL∗VSHET+) was recently shown to be associated with reduced risk of Alzheimer's disease (AD) in APOE∗4 carriers. Additional studies suggest that KL∗VSHET+ protects against amyloid burden in cognitively normal older subjects, but sample sizes were too small to draw definitive conclusions. We performed a well-powered meta-analysis across 5 independent studies, comprising 3581 pre-clinical participants ages 60-80, to investigate whether KL∗VSHET+ reduces the risk of having an amyloid-positive positron emission tomography scan. Analyses were stratified by APOE∗4 status. KL∗VSHET+ reduced the risk of amyloid positivity in APOE∗4 carriers (odds ratio = 0.67 [0.52-0.88]; p = 3.5 × 10-3), but not in APOE∗4 non-carriers (odds ratio = 0.94 [0.73-1.21]; p = 0.63). The combination of APOE∗4 and KL∗VS genotypes should help enrich AD clinical trials for pre-symptomatic subjects at increased risk of developing amyloid aggregation and AD. KL-related pathways may help elucidate protective mechanisms against amyloid accumulation and merit exploration for novel AD drug targets. Future investigation of the biological mechanisms by which KL interacts with APOE∗4 and AD are warranted.
Collapse
Affiliation(s)
- Michael E Belloy
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA.
| | - Sarah J Eger
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Yann Le Guen
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Valerio Napolioni
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Kacie D Deters
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Hyun-Sik Yang
- Department of Neurology, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Marzia A Scelsi
- Centre for Medical Image Computing (CMIC), University College London, London, UK
| | - Tenielle Porter
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia; School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Sarah-Naomi James
- Medical Research Council Unit for Lifelong Health and Ageing, University College London, London, UK
| | - Andrew Wong
- Medical Research Council Unit for Lifelong Health and Ageing, University College London, London, UK
| | - Jonathan M Schott
- Dementia Research Centre, University College London Queen Square Institute of Neurology, University College London, London, UK; UK Dementia Research Institute, University College London, London, UK
| | - Reisa A Sperling
- Department of Neurology, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Simon M Laws
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia; School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Elisabeth C Mormino
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Zihuai He
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA; Department of Medicine, Quantitative Sciences Unit, Stanford University, Stanford, CA, USA
| | - Summer S Han
- Department of Medicine, Quantitative Sciences Unit, Stanford University, Stanford, CA, USA; Department of Neurosurgery, Stanford University, Stanford, CA, USA
| | - Andre Altmann
- Centre for Medical Image Computing (CMIC), University College London, London, UK
| | - Michael D Greicius
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | | | | | | | | |
Collapse
|
39
|
Cambray S, Bermudez-Lopez M, Bozic M, Valdivielso JM. Association of a single nucleotide polymorphism combination pattern of the Klotho gene with non-cardiovascular death in patients with chronic kidney disease. Clin Kidney J 2021; 13:1017-1024. [PMID: 33391745 PMCID: PMC7769551 DOI: 10.1093/ckj/sfaa014] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 01/13/2020] [Indexed: 12/20/2022] Open
Abstract
Background Chronic kidney disease (CKD) is associated with an elevated risk of all-cause mortality, with cardiovascular death being extensively investigated. However, non-cardiovascular mortality represents the biggest percentage, showing an evident increase in recent years. Klotho is a gene highly expressed in the kidney, with a clear influence on lifespan. Low levels of Klotho have been linked to CKD progression and adverse outcomes. Single nucleotide polymorphisms (SNPs) of the Klotho gene have been associated with several diseases, but studies investigating the association of Klotho SNPs with non-cardiovascular death in CKD populations are lacking. Methods The main aim of this study was to assess whether 11 Klotho SNPs were associated with non-cardiovascular death in a subpopulation of the National Observatory of Atherosclerosis in Nephrology (NEFRONA) study (n = 2185 CKD patients). Results After 48 months of follow-up, 62 cardiovascular deaths and 108 non-cardiovascular deaths were recorded. We identified a high non-cardiovascular death risk combination of SNPs corresponding to individuals carrying the most frequent allele (G) at rs562020, the rare allele (C) at rs2283368 and homozygotes for the rare allele (G) at rs2320762 (rs562020 GG/AG + rs2283368 CC/CT + rs2320762 GG). Among the patients with the three SNPs genotyped (n = 1016), 75 (7.4%) showed this combination. Furthermore, 95 (9.3%) patients showed a low-risk combination carrying all the opposite genotypes (rs562020 AA + rs2283368 TT + rs2320762 GT/TT). All the other combinations [n = 846 (83.3%)] were considered as normal risk. Using competing risk regression analysis, we confirmed that the proposed combinations are independently associated with a higher {hazard ratio [HR] 3.28 [confidence interval (CI) 1.51–7.12]} and lower [HR 6 × 10−6 (95% CI 3.3 × 10−7–1.1 × 10−5)] risk of suffering a non-cardiovascular death in the CKD population of the NEFRONA cohort compared with patients with the normal-risk combination. Conclusions Determination of three SNPs of the Klotho gene could help in the prediction of non-cardiovascular death in CKD.
Collapse
Affiliation(s)
- Serafi Cambray
- Vascular and Renal Translational Research Group, Institute for Biomedical Research Dr. Pifarré Foundation, IRBLleida and RedinRen RETIC, ISCIII, Lleida, Spain
| | - Marcelino Bermudez-Lopez
- Vascular and Renal Translational Research Group, Institute for Biomedical Research Dr. Pifarré Foundation, IRBLleida and RedinRen RETIC, ISCIII, Lleida, Spain
| | - Milica Bozic
- Vascular and Renal Translational Research Group, Institute for Biomedical Research Dr. Pifarré Foundation, IRBLleida and RedinRen RETIC, ISCIII, Lleida, Spain
| | - Jose M Valdivielso
- Vascular and Renal Translational Research Group, Institute for Biomedical Research Dr. Pifarré Foundation, IRBLleida and RedinRen RETIC, ISCIII, Lleida, Spain
| | | |
Collapse
|
40
|
Ho WY, Navakkode S, Liu F, Soong TW, Ling SC. Deregulated expression of a longevity gene, Klotho, in the C9orf72 deletion mice with impaired synaptic plasticity and adult hippocampal neurogenesis. Acta Neuropathol Commun 2020; 8:155. [PMID: 32887666 PMCID: PMC7473815 DOI: 10.1186/s40478-020-01030-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 08/29/2020] [Indexed: 01/17/2023] Open
Abstract
Hexanucleotide repeat expansion of C9ORF72 is the most common genetic cause of amyotrophic lateral sclerosis and frontotemporal dementia. Synergies between loss of C9ORF72 functions and gain of toxicities from the repeat expansions contribute to C9ORF72-mediated pathogenesis. However, how loss of C9orf72 impacts neuronal and synaptic functions remains undetermined. Here, we showed that long-term potentiation at the dentate granule cells and long-term depression at the Schaffer collateral/commissural synapses at the area CA1 were reduced in the hippocampus of C9orf72 knockout mice. Using unbiased transcriptomic analysis, we identified that Klotho, a longevity gene, was selectively dysregulated in an age-dependent manner. Specifically, Klotho protein expression in the hippocampus of C9orf72 knockout mice was incorrectly enriched in the dendritic regions of CA1 with concomitant reduction in granule cell layer of dentate gyrus at 3-month of age followed by an accelerating decline during aging. Furthermore, adult hippocampal neurogenesis was reduced in C9orf72 knockout mice. Taken together, our data suggest that C9ORF72 is required for synaptic plasticity and adult neurogenesis in the hippocampus and Klotho deregulations may be part of C9ORF72-mediated toxicity.
Collapse
|
41
|
Viggiano D, Wagner CA, Martino G, Nedergaard M, Zoccali C, Unwin R, Capasso G. Mechanisms of cognitive dysfunction in CKD. Nat Rev Nephrol 2020; 16:452-469. [PMID: 32235904 DOI: 10.1038/s41581-020-0266-9] [Citation(s) in RCA: 182] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/02/2020] [Indexed: 02/07/2023]
Abstract
Cognitive impairment is an increasingly recognized major cause of chronic disability and is commonly found in patients with chronic kidney disease (CKD). Knowledge of the relationship between kidney dysfunction and impaired cognition may improve our understanding of other forms of cognitive dysfunction. Patients with CKD are at an increased risk (compared with the general population) of both dementia and its prodrome, mild cognitive impairment (MCI), which are characterized by deficits in executive functions, memory and attention. Brain imaging in patients with CKD has revealed damage to white matter in the prefrontal cortex and, in animal models, in the subcortical monoaminergic and cholinergic systems, accompanied by widespread macrovascular and microvascular damage. Unfortunately, current interventions that target cardiovascular risk factors (such as anti-hypertensive drugs, anti-platelet agents and statins) seem to have little or no effect on CKD-associated MCI, suggesting that the accumulation of uraemic neurotoxins may be more important than disturbed haemodynamic factors or lipid metabolism in MCI pathogenesis. Experimental models show that the brain monoaminergic system is susceptible to uraemic neurotoxins and that this system is responsible for the altered sleep pattern commonly observed in patients with CKD. Neural progenitor cells and the glymphatic system, which are important in Alzheimer disease pathogenesis, may also be involved in CKD-associated MCI. More detailed study of CKD-associated MCI is needed to fully understand its clinical relevance, underlying pathophysiology, possible means of early diagnosis and prevention, and whether there may be novel approaches and potential therapies with wider application to this and other forms of cognitive decline.
Collapse
Affiliation(s)
- Davide Viggiano
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy.,Biogem Scarl, Ariano Irpino, Italy
| | - Carsten A Wagner
- Institute of Physiology, University of Zurich, Zurich, Switzerland, and National Center of Competence in Research NCCR Kidney.CH, Zurich, Switzerland
| | - Gianvito Martino
- IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Maiken Nedergaard
- University of Rochester Medical Center, School of Medicine and Dentistry, Rochester, NY, USA
| | - Carmine Zoccali
- Institute of Clinical Physiology, National Research Council (CNR), Reggio Calabria Unit, Reggio Calabria, Italy
| | - Robert Unwin
- Department of Renal Medicine, University College London (UCL), Royal Free Campus, London, UK.,Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Giovambattista Capasso
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy. .,Biogem Scarl, Ariano Irpino, Italy.
| |
Collapse
|
42
|
Moos WH, Faller DV, Glavas IP, Harpp DN, Kanara I, Mavrakis AN, Pernokas J, Pernokas M, Pinkert CA, Powers WR, Sampani K, Steliou K, Vavvas DG, Zamboni RJ, Kodukula K, Chen X. Klotho Pathways, Myelination Disorders, Neurodegenerative Diseases, and Epigenetic Drugs. Biores Open Access 2020; 9:94-105. [PMID: 32257625 PMCID: PMC7133426 DOI: 10.1089/biores.2020.0004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In this review we outline a rationale for identifying neuroprotectants aimed at inducing endogenous Klotho activity and expression, which is epigenetic action, by definition. Such an approach should promote remyelination and/or stimulate myelin repair by acting on mitochondrial function, thereby heralding a life-saving path forward for patients suffering from neuroinflammatory diseases. Disorders of myelin in the nervous system damage the transmission of signals, resulting in loss of vision, motion, sensation, and other functions depending on the affected nerves, currently with no effective treatment. Klotho genes and their single-pass transmembrane Klotho proteins are powerful governors of the threads of life and death, true to the origin of their name, Fates, in Greek mythology. Among its many important functions, Klotho is an obligatory co-receptor that binds, activates, and/or potentiates critical fibroblast growth factor activity. Since the discovery of Klotho a little over two decades ago, it has become ever more apparent that when Klotho pathways go awry, oxidative stress and mitochondrial dysfunction take over, and age-related chronic disorders are likely to follow. The physiological consequences can be wide ranging, potentially wreaking havoc on the brain, eye, kidney, muscle, and more. Central nervous system disorders, neurodegenerative in nature, and especially those affecting the myelin sheath, represent worthy targets for advancing therapies that act upon Klotho pathways. Current drugs for these diseases, even therapeutics that are disease modifying rather than treating only the symptoms, leave much room for improvement. It is thus no wonder that this topic has caught the attention of biomedical researchers around the world.
Collapse
Affiliation(s)
- Walter H. Moos
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of California San Francisco, San Francisco, San Francisco, California
- ShangPharma Innovation, Inc., South San Francisco, California
| | - Douglas V. Faller
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
- Cancer Research Center, Boston University School of Medicine, Boston, Massachusetts
| | - Ioannis P. Glavas
- Department of Ophthalmology, New York University School of Medicine, New York, New York
| | - David N. Harpp
- Department of Chemistry, McGill University, Montreal, Canada
| | | | - Anastasios N. Mavrakis
- Department of Medicine, Tufts University School of Medicine, St. Elizabeth's Medical Center, Boston, Massachusetts
| | - Julie Pernokas
- Advanced Dental Associates of New England, Woburn, Massachusetts
| | - Mark Pernokas
- Advanced Dental Associates of New England, Woburn, Massachusetts
| | - Carl A. Pinkert
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Whitney R. Powers
- Department of Health Sciences, Boston University, Boston, Massachusetts
- Department of Anatomy, Boston University School of Medicine, Boston, Massachusetts
| | - Konstantina Sampani
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
- Beetham Eye Institute, Joslin Diabetes Center, Boston, Massachusetts
| | - Kosta Steliou
- Cancer Research Center, Boston University School of Medicine, Boston, Massachusetts
- PhenoMatriX, Inc., Natick, Massachusetts
| | - Demetrios G. Vavvas
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
- Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts
| | | | | | - Xiaohong Chen
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
- Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts
| |
Collapse
|
43
|
|