1
|
Pańczyszyn-Trzewik P, Czechowska E, Stachowicz K, Sowa-Kućma M. The Importance of α-Klotho in Depression and Cognitive Impairment and Its Connection to Glutamate Neurotransmission-An Up-to-Date Review. Int J Mol Sci 2023; 24:15268. [PMID: 37894946 PMCID: PMC10607524 DOI: 10.3390/ijms242015268] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/11/2023] [Accepted: 10/14/2023] [Indexed: 10/29/2023] Open
Abstract
Depression is a serious neuropsychiatric disease affecting an increasing number of people worldwide. Cognitive deficits (including inattention, poor memory, and decision-making difficulties) are common in the clinical picture of depression. Cognitive impairment has been hypothesized to be one of the most important components of major depressive disorder (MDD; referred to as clinical depression), although typical cognitive symptoms are less frequent in people with depression than in people with schizophrenia or bipolar disorder (BD; sometimes referred to as manic-depressive disorder). The importance of α-Klotho in the aging process has been well-documented. Growing evidence points to the role of α-Klotho in regulating other biological functions, including responses to oxidative stress and the modulation of synaptic plasticity. It has been proven that a Klotho deficit may contribute to the development of various nervous system pathologies, such as behavioral disorders or neurodegeneration. Given the growing evidence of the role of α-Klotho in depression and cognitive impairment, it is assumed that this protein may be a molecular link between them. Here, we provide a research review of the role of α-Klotho in depression and cognitive impairment. Furthermore, we propose potential mechanisms (related to oxidative stress and glutamatergic transmission) that may be important in α-Klotho-mediated regulation of mental and cognitive function.
Collapse
Affiliation(s)
- Patrycja Pańczyszyn-Trzewik
- Department of Human Physiology, Institute of Medical Sciences, Medical College of Rzeszow University, Kopisto 2a, 35-959 Rzeszow, Poland; (P.P.-T.); (E.C.)
| | - Ewelina Czechowska
- Department of Human Physiology, Institute of Medical Sciences, Medical College of Rzeszow University, Kopisto 2a, 35-959 Rzeszow, Poland; (P.P.-T.); (E.C.)
| | - Katarzyna Stachowicz
- Department of Neurobiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smetna 12, 31-343 Krakow, Poland;
| | - Magdalena Sowa-Kućma
- Department of Human Physiology, Institute of Medical Sciences, Medical College of Rzeszow University, Kopisto 2a, 35-959 Rzeszow, Poland; (P.P.-T.); (E.C.)
- Centre for Innovative Research in Medical and Natural Sciences, Medical College of Rzeszow University, Warzywna Street 1A, 35-595 Rzeszow, Poland
| |
Collapse
|
2
|
Liu Y, Chen M. Emerging role of α-Klotho in energy metabolism and cardiometabolic diseases. Diabetes Metab Syndr 2023; 17:102854. [PMID: 37722166 DOI: 10.1016/j.dsx.2023.102854] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 08/16/2023] [Accepted: 09/08/2023] [Indexed: 09/20/2023]
Abstract
BACKGROUND AND AIM Klotho was first identified as a gene associated with aging and longevity in 1997. α-Klotho is an anti-aging protein and its role in energy metabolism, various cardiovascular diseases (CVDs), and metabolic disorders is increasingly being recognized. In this review, we aimed to outline the potential protective role and therapeutic prospects of α-Klotho in energy metabolism and cardiometabolic diseases (CMDs). METHODS We comprehensively reviewed the relevant literature in PubMed using the keywords 'Klotho', 'metabolism', 'cardiovascular', 'diabetes', 'obesity', 'metabolic syndrome', and 'nonalcoholic fatty liver disease'. RESULTS α-Klotho can be divided into membrane-bound Klotho, secreted Klotho, and the most studied circulating soluble Klotho that can act as a hormone. Klotho gene polymorphisms have been implicated in energy metabolism and CMDs. α-Klotho can inhibit insulin/insulin growth factor-1 signaling and its overexpression can lead to a 'healthy insulin resistance' and may exert beneficial effects on the regulation of glycolipid metabolism and central energy homeostasis. α-Klotho, mainly serum Klotho, has been revealed to be protective against CVDs, diabetes and its complications, obesity, and nonalcoholic fatty liver disease. Human recombinant Klotho protein/Klotho gene delivery, multiple drugs, or natural products, and exercise can increase α-Klotho expression. CONCLUSION Overall, α-Klotho has demonstrated its potential as a promising target for modulating energy metabolism and CMDs, and further research is needed to explore its utilization in clinical practice in the future.
Collapse
Affiliation(s)
- Yuanbin Liu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, No. 99 Zhang Zhidong Road, Wuhan, Hubei, 430000, PR China
| | - Mingkai Chen
- Department of Gastroenterology, Renmin Hospital of Wuhan University, No. 99 Zhang Zhidong Road, Wuhan, Hubei, 430000, PR China.
| |
Collapse
|
3
|
Tyurenkov IN, Perfilova VN, Nesterova AA, Glinka Y. Klotho Protein and Cardio-Vascular System. BIOCHEMISTRY (MOSCOW) 2021; 86:132-145. [PMID: 33832412 DOI: 10.1134/s0006297921020024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Klotho protein affects a number of metabolic pathways essential for pathogenesis of cardio-vascular diseases and their prevention. It inhibits lipid peroxidation and inflammation, as well as prevents endothelial injury and calcification of blood vessels. Klotho decreases rigidity of blood vessels and suppresses development of the heart fibrosis. Low level of its expression is associated with a number of diseases. Cardioprotective effect of klotho is based on its ability to interact with multiple receptors and ion channels. Being a pleiotropic protein, klotho could be a useful target for therapeutic intervention in the treatment of cardio-vascular diseases. In this review we present data on pharmaceuticals that stimulate klotho expression and suggest some promising research directions.
Collapse
Affiliation(s)
- Ivan N Tyurenkov
- Volgograd State Medical University, Ministry of Health of the Russian Federation, Volgograd, 400066, Russia
| | - Valentina N Perfilova
- Volgograd State Medical University, Ministry of Health of the Russian Federation, Volgograd, 400066, Russia.
| | - Alla A Nesterova
- Pyatigorsk Medical and Pharmaceutical Institute, Branch of the Volgograd State Medical University, Ministry of Health of the Russian Federation, Pyatigorsk, 357500, Russia
| | - Yelena Glinka
- Keenan Research Centre, St. Michael's Hospital, Toronto, ON M5B 1W8, Canada
| |
Collapse
|
4
|
Adamska-Tomaszewska D, Wajda J, Wyskida K, Bednarczyk D, Świat M, Owczarek AJ, Puzianowska-Kuźnicka M, Olszanecka-Glinianowicz M, Chudek J. Higher Serum-Soluble α-Klotho Level Does Not Predict Longer Survival after Stroke. BIOMED RESEARCH INTERNATIONAL 2020; 2020:9283651. [PMID: 33376748 PMCID: PMC7738777 DOI: 10.1155/2020/9283651] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 10/10/2020] [Accepted: 11/04/2020] [Indexed: 01/01/2023]
Abstract
RESULTS There were 5 recurrent strokes and 89 deaths during the 36-month follow-up. Even though no significant differences in OS and SFS between soluble α-Klotho level tertile groups were recorded, unexpectedly, OS and SFS were highest in patients with the lowest soluble α-Klotho concentrations. Moreover, the Cox proportional models adjusted for established risk factors, kidney function, and the severity of stroke revealed that each 100 pg/mL increase in soluble α-Klotho levels was associated with decreased OS (HR = 0.951 (0.908-0.995), p < 0.05) and SFS (HR = 0.949 (0.908-0.993), p < 0.05). In addition, the α-Klotho to iFGF23 index was predicting neither OS nor SFS. CONCLUSION Soluble α-Klotho levels in serum were not related to the severity of neurological deficits and long-term outcomes in patients with IS. No neuroprotective effect of soluble α-Klotho levels in patients with IS was demonstrated.
Collapse
Affiliation(s)
| | - Jarosław Wajda
- Dialysis Center in Rybnik, Regional Specialist Hospital No. 3 in Rybnik, Rybnik, Poland
| | - Katarzyna Wyskida
- Health Promotion and Obesity Management Unit, Department of Pathophysiology Medical Faculty in Katowice, Medical University of Silesia, Katowice, Poland
| | - Dawid Bednarczyk
- Department of Neurology with Stroke Unit, Regional Specialist Hospital No. 2 in Jastrzębie-Zdrój, Jastrzębie-Zdrój, Poland
| | - Maciej Świat
- Department of Neurology with Stroke Unit, Regional Specialist Hospital No. 3 in Rybnik, Rybnik, Poland
- Jan Długosz University in Częstochowa, Częstochowa, Poland
| | - Aleksander J. Owczarek
- Department of Statistics, Department of Instrumental Analysis, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Katowice, Poland
| | - Monika Puzianowska-Kuźnicka
- Department of Human Epigenetics, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
- Department of Geriatrics and Gerontology, Medical Centre of Postgraduate Education, Warsaw, Poland
| | - Magdalena Olszanecka-Glinianowicz
- Health Promotion and Obesity Management Unit, Department of Pathophysiology Medical Faculty in Katowice, Medical University of Silesia, Katowice, Poland
| | - Jerzy Chudek
- Pathophysiology Unit, Department of Pathophysiology Medical Faculty in Katowice, Medical University of Silesia, Katowice, Poland
- Department of Internal Medicine and Oncological Chemotherapy, Medical Faculty in Katowice, Medical University of Silesia, Katowice, Poland
| |
Collapse
|
5
|
Pazik J, Rembek K, Sadowska-Jakubowicz A, Sitarek E, Kosieradzki M, Durlik M. Donor Klotho KL-VS Polymorphism Predicts Allograft Glomerulosclerosis and Early Post-Transplant Kidney Function. Transplant Proc 2020; 52:2371-2375. [PMID: 32571701 DOI: 10.1016/j.transproceed.2020.02.086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 02/16/2020] [Accepted: 02/22/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND The Klotho protein, encoded by the KL (Klotho) gene, exerts antiaging and antifibrotic effects. The KL-VS genotype diminishes Klotho expression and correlates with cardiovascular death, heart failure, and chronic kidney disease progression. The aim of this study was to analyze the contribution of donor Klotho rs9536314 and rs9527025 polymorphisms (KL-VS genotype) to renal allograft morphology and function in the early post-transplant period. METHODS Clinical data and biopsy reports of 170 deceased donor transplantations were retrieved from standard medical files. Donor DNA was genotyped for rs9527025 and rs9536314 SNPs using custom TaqMan assays. RESULTS As rs9527025 remained in full linkage with rs9536314, we report results for the latter. The analyses were performed for G dominant model (GG+GT vs TT). We found an association between reported SNP alleles, morphologic changes in the peritransplant biopsy, and kidney function 3 months after engraftment. A chronic glomerulopathy score of >0 was found in 12.2% of GG+GT cases and in 3.2% of TT cases (P = .023). For G allele carriers, the third month's median estimated glomerular filtration rate value was 35.0 (range, 20.4-76.6 mL/min), while for TT haplotype, the value was 46.3 (range, 15.5-96.8 mL/min), P = .001. At the third post-transplant month, proteinuria incidence was higher for organs with G allele than with TT haplotype (24.4% vs 9.5%; P = .030; odds ratio 3.09; 95% confidence interval 1.22-7.69). CONCLUSION Deceased donor KL-VS polymorphism, altering protein dimerization and coreceptor function, predicts early renal transplant glomerular lesions and function. Further analyses for mentioned effect durability are necessary. ETHICS STATEMENT This study complies with the Helsinki Congress and the Istanbul Declaration regarding donor source. Donors were not prisoners, and were not paid or coerced.
Collapse
Affiliation(s)
- Joanna Pazik
- Department of Transplantation Medicine, Nephrology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Karolina Rembek
- Department of Transplantation Medicine, Nephrology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland.
| | - Anna Sadowska-Jakubowicz
- Department of Transplantation Medicine, Nephrology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Elżbieta Sitarek
- Department of Transplantation Medicine, Nephrology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Maciej Kosieradzki
- Department and Clinic of General and Transplantation Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Magdalena Durlik
- Department of Transplantation Medicine, Nephrology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
6
|
The association between Single Nucleotide Polymorphisms of Klotho Gene and Mortality in Elderly Men: The MrOS Sweden Study. Sci Rep 2020; 10:10243. [PMID: 32581247 PMCID: PMC7314825 DOI: 10.1038/s41598-020-66517-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 02/24/2020] [Indexed: 12/12/2022] Open
Abstract
The Klotho (KL) gene is involved in phosphate homeostasis. Polymorphisms in this gene have been reported to be associated with the risk of cardiovascular disease. Here we used computational tools to predict the damage-associated single nucleotide polymorphisms (SNPs) in the human KL gene. We further investigated the association of SNPs in the KL gene and mortality in the Swedish multicenter prospective Osteoporotic Fractures in Men (MrOS) cohort. This study included 2921 men (aged 69-81 years) with mean 4.49 ± 1.03 years follow-up. 18 SNPs in the KL gene were genotyped using Sequenom. These SNPs were identified by in silico tools for the coding and noncoding genome to predict the damaging SNPs. After quality analyses, SNPs were analyzed for mortality risk using two steps approach on logistic regression model screening and then Cox regression model confirmation. Two non-synonymous SNPs rs9536314 and rs9527025 were found to be potentially damaging SNPs that affect KL protein stability and expression. However, these two SNPs were not statistically significantly associated with all-cause mortality (crude Hazard ratio [HR] 1.72, 95% confidence interval [CI] 0.96-3.07 in rs9536314; crude HR 1.82, 95% CI 0.998-3.33 in rs9527025) or cardiovascular mortality (crude HR 1.52, 95% CI 0.56-4.14 in rs9536314; crude HR 1.54, 95% CI 0.55-4.33 in rs9527025) in additive model using Cox regression analysis. In conclusion, these two potentially damaging SNPs (rs9536314 and rs9527025) in the KL gene were not associated with all-cause mortality or cardiovascular mortality in MrOs cohort. Larger scales studies and meta-analysis are needed to confirm the correlation between polymorphisms of the KL gene and mortality.
Collapse
|
7
|
Miranda Pérez AA, Gutiérrez Pérez ME, Urraza Robledo AI, Delgadillo Guzmán D, Ruíz Flores P, López Márquez FC. Klotho-HIV and Oxidative Stress: The Role of Klotho in Cardiovascular Disease Under HIV Infection-A Review. DNA Cell Biol 2020; 39:1478-1485. [PMID: 32584609 DOI: 10.1089/dna.2020.5444] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Combined antiretroviral therapy has improved quality and life expectancy of people living with human immunodeficiency virus (HIV). However, this therapy increases oxidative stress (OS), which in turn causes alterations in lipid and carbon metabolism, kidney disease, liver cirrhosis, and increased risk of cardiovascular disease. The Klotho gene has been implicated in cardiovascular risk increase. Klotho protein expression at X level decreases the risk of heart disease. HIV-positive people usually present low plasma levels of Klotho; thus, contributing to some extent to an increase in cardiovascular risk for these types of patients, mostly by favoring atherosclerosis. Therefore, our aim is to provide an overview of the effect of OS on Klotho protein and its consequent cardiometabolic alterations in HIV-positive patients on antiretroviral therapy.
Collapse
Affiliation(s)
- Alberto Alejandro Miranda Pérez
- Department of Molecular Immunobiology, Biomedical Research Center, Faculty of Medicine, Autonomous University of Coahuila, Torreon, Mexico
| | - María Elena Gutiérrez Pérez
- Department of Molecular Immunobiology, Biomedical Research Center, Faculty of Medicine, Autonomous University of Coahuila, Torreon, Mexico
| | | | - Dealmy Delgadillo Guzmán
- Department of Pharmacology, Faculty of Medicine, Autonomous University of Coahuila, Torreon, Mexico
| | - Pablo Ruíz Flores
- Department of Genetics, Center for Biomedical Research Faculty of Medicine, Autonomous University of Coahuila, Torreon, Mexico
| | - Francisco Carlos López Márquez
- Department of Molecular Immunobiology, Biomedical Research Center, Faculty of Medicine, Autonomous University of Coahuila, Torreon, Mexico
| |
Collapse
|
8
|
Six I, Flissi N, Lenglet G, Louvet L, Kamel S, Gallet M, Massy ZA, Liabeuf S. Uremic Toxins and Vascular Dysfunction. Toxins (Basel) 2020; 12:toxins12060404. [PMID: 32570781 PMCID: PMC7354618 DOI: 10.3390/toxins12060404] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/15/2020] [Accepted: 06/17/2020] [Indexed: 02/07/2023] Open
Abstract
Vascular dysfunction is an essential element found in many cardiovascular pathologies and in pathologies that have a cardiovascular impact such as chronic kidney disease (CKD). Alteration of vasomotricity is due to an imbalance between the production of relaxing and contracting factors. In addition to becoming a determining factor in pathophysiological alterations, vascular dysfunction constitutes the first step in the development of atherosclerosis plaques or vascular calcifications. In patients with CKD, alteration of vasomotricity tends to emerge as being a new, less conventional, risk factor. CKD is characterized by the accumulation of uremic toxins (UTs) such as phosphate, para-cresyl sulfate, indoxyl sulfate, and FGF23 and, consequently, the deleterious role of UTs on vascular dysfunction has been explored. This accumulation of UTs is associated with systemic alterations including inflammation, oxidative stress, and the decrease of nitric oxide production. The present review proposes to summarize our current knowledge of the mechanisms by which UTs induce vascular dysfunction.
Collapse
Affiliation(s)
- Isabelle Six
- UR 7517 UPJV, Pathophysiological Mechanisms and Consequences of Cardiovascular Calcifications (MP3CV), Picardie Jules Verne University, 80025 Amiens, France; (N.F.); (G.L.); (L.L.); (S.K.); (M.G.); (S.L.)
- Correspondence: ; Tel./Fax: +03-22-82-54-25
| | - Nadia Flissi
- UR 7517 UPJV, Pathophysiological Mechanisms and Consequences of Cardiovascular Calcifications (MP3CV), Picardie Jules Verne University, 80025 Amiens, France; (N.F.); (G.L.); (L.L.); (S.K.); (M.G.); (S.L.)
| | - Gaëlle Lenglet
- UR 7517 UPJV, Pathophysiological Mechanisms and Consequences of Cardiovascular Calcifications (MP3CV), Picardie Jules Verne University, 80025 Amiens, France; (N.F.); (G.L.); (L.L.); (S.K.); (M.G.); (S.L.)
| | - Loïc Louvet
- UR 7517 UPJV, Pathophysiological Mechanisms and Consequences of Cardiovascular Calcifications (MP3CV), Picardie Jules Verne University, 80025 Amiens, France; (N.F.); (G.L.); (L.L.); (S.K.); (M.G.); (S.L.)
| | - Said Kamel
- UR 7517 UPJV, Pathophysiological Mechanisms and Consequences of Cardiovascular Calcifications (MP3CV), Picardie Jules Verne University, 80025 Amiens, France; (N.F.); (G.L.); (L.L.); (S.K.); (M.G.); (S.L.)
- Amiens-Picardie University Hospital, Human Biology Center, 80054 Amiens, France
| | - Marlène Gallet
- UR 7517 UPJV, Pathophysiological Mechanisms and Consequences of Cardiovascular Calcifications (MP3CV), Picardie Jules Verne University, 80025 Amiens, France; (N.F.); (G.L.); (L.L.); (S.K.); (M.G.); (S.L.)
| | - Ziad A. Massy
- Service de Néphrologie et Dialyse, Assistance Publique—Hôpitaux de Paris (APHP), Hôpital Universitaire Ambroise Paré, 92100 Boulogne Billancourt, France;
- INSERM U1018, Equipe 5, CESP (Centre de Recherche en Épidémiologie et Santé des Populations), Université Paris Saclay et Université Versailles Saint Quentin en Yvelines, 94800 Villejuif, France
| | - Sophie Liabeuf
- UR 7517 UPJV, Pathophysiological Mechanisms and Consequences of Cardiovascular Calcifications (MP3CV), Picardie Jules Verne University, 80025 Amiens, France; (N.F.); (G.L.); (L.L.); (S.K.); (M.G.); (S.L.)
- Pharmacology Department, Amiens University Hospital, 80025 Amiens, France
| |
Collapse
|
9
|
KLOTHO polymorphisms and age-related outcomes in community-dwelling older subjects: The São Paulo Ageing & Health (SPAH) Study. Sci Rep 2020; 10:8574. [PMID: 32444684 PMCID: PMC7244540 DOI: 10.1038/s41598-020-65441-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 05/05/2020] [Indexed: 02/06/2023] Open
Abstract
Defective KLOTHO gene expression in mice led to a syndrome resembling human ageing. This study evaluated three KLOTHO polymorphisms, namely G395A, C1818T, and C370S, in an elderly population (mean age of 73 years) and their associations with ageing-related outcomes (cardiovascular events, kidney function, osteoporosis, sarcopenia) and mortality. Estimated glomerular filtration rates (eGFR) was lower in subjects with 1818TT (P = 0.047) and 370SS (P = 0.046) genotypes. The 1818TT genotype (P = 0.006) and 1818T allele were associated with higher frequency of myocardial infarction (MI) (CC:1.7% vs. CT + TT:7.0%; P = 0.002). The 370SS genotype was associated with lower stroke frequency (P = 0.001). MI (OR 3.35 [95% CI: 1.29–8.74]) and stroke (OR 3.64 [95% CI: 1.48–8.97]) were associated with mortality. Regarding MI, logistic regression showed 1818T allele was a risk factor for death-related MI (OR 4.29 [95% CI: 1.60–11.52]; P = 0.003), while 370C was protective (OR 0.03 [95% CI: 0.01–0.08]; P < 0.001). Regarding stroke, the 395A and 370C alleles were protective factors (respectively: OR 0.28 [95% CI: 0.20–0.80]; P = 0.018; OR 0.10 [95% CI: 0.05–0.18]; P < 0.001). This is the first study to determine potential associations between common ageing-related outcomes/mortality and KLOTHO polymorphisms. The 1818T allele was a risk factor for MI-related death. The 395A and 370C alleles were protective factors for stroke-related death in elderly from community.
Collapse
|
10
|
Circulating Klotho is linked to prognosis of acute intracerebral hemorrhage. Clin Chim Acta 2019; 497:114-119. [DOI: 10.1016/j.cca.2019.07.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 07/19/2019] [Accepted: 07/19/2019] [Indexed: 12/21/2022]
|
11
|
The goddess who spins the thread of life: Klotho, psychiatric stress, and accelerated aging. Brain Behav Immun 2019; 80:193-203. [PMID: 30872092 PMCID: PMC6660403 DOI: 10.1016/j.bbi.2019.03.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/27/2019] [Accepted: 03/09/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Longevity gene klotho (KL) is associated with age-related phenotypes but has not been evaluated against a direct human biomarker of cellular aging. We examined KL and psychiatric stress, including posttraumatic stress disorder (PTSD), which is thought to potentiate accelerated aging, in association with biomarkers of cellular aging. METHODS The sample comprised 309 white, non-Hispanic genotyped veterans with measures of epigenetic age (DNA methylation age), telomere length (n = 252), inflammation (C-reactive protein), psychiatric symptoms, metabolic function, and white matter neural integrity (diffusion tensor imaging; n = 185). Genotyping and DNA methylation were obtained on epi/genome-wide beadchips. RESULTS In gene by environment analyses, two KL variants (rs9315202 and rs9563121) interacted with PTSD severity (peak corrected p = 0.044) and sleep disturbance (peak corrected p = 0.034) to predict advanced epigenetic age. KL variant, rs398655, interacted with self-reported pain in association with slowed epigenetic age (corrected p = 0.048). A well-studied protective variant, rs9527025, was associated with slowed epigenetic age (p = 0.046). The peak PTSD interaction term (with rs9315202) also predicted C-reactive protein (p = 0.049), and white matter microstructural integrity in two tracts (corrected ps = 0.005 - 0.035). This SNP evidenced a main effect with an index of metabolic syndrome severity (p = 0.015). Effects were generally accentuated in older subjects. CONCLUSIONS Rs9315202 predicted multiple biomarkers of cellular aging such that psychiatric stress was more strongly associated with cellular aging in those with the minor allele. KL genotype may contribute to a synchronized pathological aging response to stress and could be a therapeutic target to alter the pace of cellular aging.
Collapse
|
12
|
Lee JB, Woo HG, Chang Y, Jin YM, Jo I, Kim J, Song TJ. Plasma Klotho concentrations predict functional outcome at three months after acute ischemic stroke patients. Ann Med 2019; 51:262-269. [PMID: 31070492 PMCID: PMC7877889 DOI: 10.1080/07853890.2019.1617434] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background: The Klotho protects the cardiovascular system by protecting against cell apoptosis, inhibiting the production of reactive oxygen species, and modulating inflammation. We aimed to investigate relationship of plasma Klotho concentrations with functional outcome at 3 months after acute cerebral infarction. Methods: We prospectively enrolled 262 first-ever acute cerebral infarction patients from whom a blood sample was acquired within 24 h of admission. An enzyme-linked immunosorbent assay was used for evaluating plasma Klotho concentration. Functional outcome on admission and three months was evaluated. Results: Of the 262 patients, 152 (58.0%) were men. The mean age of these patients was 64.7 years. The mean ± standard deviation of plasma Klotho concentrations was 312.7 ± 153.3 pg/mL. As opposed to patients with good outcome, plasma Klotho levels were lower in the poor outcome group (207.8 ± 96.2 vs. 342.5 ± 153.5 pg/mL, p = .001). In multivariate analysis, increased plasma Klotho concentrations were independently associated with good functional outcome (Odds ratio: 2.42, 95% confidence interval: 1.45-4.04, p < .001). Conclusions: Increased plasma Klotho concentrations were associated with good functional outcome in patients with acute ischemic stroke. We attribute these associations to the pleiotropic effects of Klotho in stroke and vascular diseases. Key message Increased plasma Klotho concentrations were associated with good functional outcome in patients with acute ischemic stroke.
Collapse
Affiliation(s)
- Jun-Beom Lee
- a Department of Neurology , Hongik Hospital , Seoul , Korea
| | - Ho Geol Woo
- b Department of Neurology , College of Medicine, Ewha Womans University , Seoul , Korea
| | - Yoonkyung Chang
- b Department of Neurology , College of Medicine, Ewha Womans University , Seoul , Korea
| | - Yoon Mi Jin
- c Department of Molecular Medicine, College of Medicine , Ewha Womans University , Seoul , Korea
| | - Inho Jo
- c Department of Molecular Medicine, College of Medicine , Ewha Womans University , Seoul , Korea
| | - Jinkwon Kim
- d Department of Neurology , Gangnam Severance Hospital, Yonsei University College of Medicine , Seoul , Korea
| | - Tae-Jin Song
- b Department of Neurology , College of Medicine, Ewha Womans University , Seoul , Korea
| |
Collapse
|
13
|
Wei H, Li H, Song X, Du X, Cai Y, Li C, Dong L, Dong J. Serum klotho: a potential predictor of cerebrovascular disease in hemodialysis patients. BMC Nephrol 2019; 20:63. [PMID: 30791885 PMCID: PMC6385422 DOI: 10.1186/s12882-019-1232-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 01/25/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Hemodialysis patients suffer from a serious threat of cerebrovascular disease. Klotho, as an aging-suppressor gene, contributes to protect on vascular calcification and oxidative stress, which are the risk factors of cerebrovascular disease. The purpose of the present study is to determine the relationship between serum klotho and cerebrovascular disease in patients receiving hemodialysis. METHODS Serum klotho levels of hemodialysis patients were measured by ELISA. Cerebrovascular diseases were diagnosed by CT or MRI scans. The cognitive function of hemodialysis patients with cerebrovascular disease were evaluated with a neuropsychological battery assessing domains of global cognition verbal memory, spatial memory, executive function and verbal fluency. RESULTS Eighty-eight patients were included, 57 ± 14 years, 63.64% male, 52.27% older than 60 years. Twenty-eight participants had cerebrovascular disease (23 cases had cerebral infarction, 5 cases had cerebral hemorrhage). The average level of serum klotho of all participants was 119.10 ± 47.29 pg/ml. The serum klotho level was significantly associated with cerebrovascular disease in hemodialysis patients (HR(95%CI) = 0.975(0.960-0.990), p = 0.001). The optimal cut-off value of serum klotho for predicting cerebrovascular disease in hemodialysis patients was 137.22 pg/ml, with a specificity of 96.4% and a sensitivity of 46.7%. But serum klotho was not an independent risk factor of cognitive impairment for hemodialysis patients with cerebrovascular disease (HR((95%CI) = 1.002(0.986-1.018), p = 0.776) or with cerebral infarction (HR(95%CI) = 1.005(0.987-1.023), p = 0.576). CONCLUSIONS The serum klotho level is a potential predictor of cerebrovascular disease in hemodialysis patients, but it is not an independent risk factor of cognitive impairment for hemodialysis patients with cerebrovascular disease.
Collapse
Affiliation(s)
- Honglan Wei
- Department of Nephrology, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430033, People's Republic of China
| | - Hua Li
- Department of Nephrology, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430033, People's Republic of China
| | - Xiaohong Song
- Department of Nephrology, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430033, People's Republic of China
| | - Xingguo Du
- Department of Nephrology, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430033, People's Republic of China
| | - Yuan Cai
- Department of Nephrology, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430033, People's Republic of China
| | - Chengxu Li
- Department of Nephrology, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430033, People's Republic of China
| | - Liping Dong
- Department of Nephrology, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430033, People's Republic of China
| | - Junwu Dong
- Department of Nephrology, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430033, People's Republic of China.
| |
Collapse
|
14
|
Zhu Z, Xia W, Cui Y, Zeng F, Li Y, Yang Z, Hequn C. Klotho gene polymorphisms are associated with healthy aging and longevity: Evidence from a meta-analysis. Mech Ageing Dev 2019; 178:33-40. [PMID: 30633899 DOI: 10.1016/j.mad.2018.12.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 12/10/2018] [Accepted: 12/30/2018] [Indexed: 01/11/2023]
Abstract
Klotho gene polymorphisms have been implicated in healthy aging, but inconsistences in findings from previous case-control studies have raised concerns regarding the associations between KLOTHO gene polymorphisms and susceptibility to aging-related diseases and longevity. Hence, this meta-analysis was performed. We assessed the associations between two polymorphisms (G-395 A/rs1207568 and F352 V/rs9536314) and five parameters (urolithiasis, cognitive impairment, cardiovascular disease, cancer, and longevity) by calculating pooled odds ratios with 95% confidence intervals. According to the pooled results, the G allele of the G-395 A polymorphism conferred a significantly higher risk of urolithiasis; G-395 A was related to the susceptibility to cardiovascular disease under allele, dominant, and recessive models. There was no significant association between the G-395 A polymorphism and cognitive impairment among the elderly. The F allele of the F352 V polymorphism protected against breast and ovarian cancer susceptibility. Interestingly, based on the results of the subgroup analysis, the F352 V polymorphism was associated with the overall risk of neoplasms in BRCA1 mutation carriers but not in BRCA2 mutation carriers. Moreover, the F allele played a protective role in determining human longevity. In conclusion, Klotho G-395 A polymorphisms were associated with urolithiasis and cardiovascular disease but not with cognitive impairment. Additionally, Klotho F352 V polymorphisms were associated with cancers and longevity.
Collapse
Affiliation(s)
- Zewu Zhu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Weiping Xia
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yu Cui
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Feng Zeng
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yang Li
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Zhongqing Yang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Chen Hequn
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| |
Collapse
|
15
|
Association of Klotho gene polymorphism with hypertension and coronary artery disease in an Iranian population. BMC Cardiovasc Disord 2018; 18:237. [PMID: 30547758 PMCID: PMC6295088 DOI: 10.1186/s12872-018-0971-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 11/27/2018] [Indexed: 01/18/2023] Open
Abstract
Background Klotho, possibly an age-regulating protein, is considered an important factor contributing to the lifespan and pathophysiology of hypertension and coronary artery disease (CAD). The present study was carried out aiming to investigate the association of Klotho-rs564481 (C1818T) gene polymorphism with hypertension and CAD. Methods A total of 286 CAD-suspicious subjects were entered into this case-control study. The polymorphism was investigated in hypertensive patients with no CAD (H-Tens, n = 60); hypertensive patients with CAD (CAD + H-Tens, n = 95); CAD patients with no hypertension (CAD, n = 61); and non-hypertensive non-CAD subjects, which were regarded as the control group (Ctrl, n = 70). Genotype and allele frequencies were assessed using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) method. Results A significant difference was found in allele frequency of Klotho C1818T among the four research groups (P = 0.03). It was also found that wild-type homozygote subjects were negatively associated with hypertension as compared to heterozygote ones (OR = 0.07 [95% CI: 0.008–0.69] P = 0.02). Moreover, in the subgroups older than 57 years old, dominant genetic model demonstrated a negative association with CAD combined with hypertension (OR = 0.31 [95% CI: 0.10–0.95] P = 0.04). Conclusions In conclusion, Klotho C1818T variant may be associated with a decreased risk of hypertension. Moreover, aging enhanced positive effects of the Klotho polymorphism on CAD combined with hypertension, indicating the possibility that the KLOTHO gene might play a part in the age-related occurrence of CAD combined with hypertension.
Collapse
|
16
|
Abstract
Advancing age promotes cardiovascular disease (CVD), the leading cause of death in the United States and many developed nations. Two major age-related arterial phenotypes, large elastic artery stiffening and endothelial dysfunction, are independent predictors of future CVD diagnosis and likely are responsible for the development of CVD in older adults. Not limited to traditional CVD, these age-related changes in the vasculature also contribute to other age-related diseases that influence mammalian health span and potential life span. This review explores mechanisms that influence age-related large elastic artery stiffening and endothelial dysfunction at the tissue level via inflammation and oxidative stress and at the cellular level via Klotho and energy-sensing pathways (AMPK [AMP-activated protein kinase], SIRT [sirtuins], and mTOR [mammalian target of rapamycin]). We also discuss how long-term calorie restriction-a health span- and life span-extending intervention-can prevent many of these age-related vascular phenotypes through the prevention of deleterious alterations in these mechanisms. Lastly, we discuss emerging novel mechanisms of vascular aging, including senescence and genomic instability within cells of the vasculature. As the population of older adults steadily expands, elucidating the cellular and molecular mechanisms of vascular dysfunction with age is critical to better direct appropriate and measured strategies that use pharmacological and lifestyle interventions to reduce risk of CVD within this population.
Collapse
Affiliation(s)
- Anthony J. Donato
- University of Utah, Department of Internal Medicine, Division of Geriatrics, Salt Lake City, Utah
- Veterans Affairs Medical Center-Salt Lake City, Geriatrics Research Education and Clinical Center, Salt Lake City, Utah
| | - Daniel R. Machin
- University of Utah, Department of Internal Medicine, Division of Geriatrics, Salt Lake City, Utah
- Veterans Affairs Medical Center-Salt Lake City, Geriatrics Research Education and Clinical Center, Salt Lake City, Utah
| | - Lisa A. Lesniewski
- University of Utah, Department of Internal Medicine, Division of Geriatrics, Salt Lake City, Utah
- Veterans Affairs Medical Center-Salt Lake City, Geriatrics Research Education and Clinical Center, Salt Lake City, Utah
| |
Collapse
|
17
|
Wu CH, Chang HM, Wang CY, Chen L, Chen LW, Lai CH, Kuo SW, Wang HC, Wu VC. Long-Term Outcomes in Patients with Incident Chronic Obstructive Pulmonary Disease after Acute Kidney Injury: A Competing-Risk Analysis of a Nationwide Cohort. J Clin Med 2018; 7:237. [PMID: 30149499 PMCID: PMC6162866 DOI: 10.3390/jcm7090237] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 08/16/2018] [Accepted: 08/17/2018] [Indexed: 12/24/2022] Open
Abstract
Both acute kidney injury (AKI) and chronic obstructive pulmonary disease (COPD) are associated with increased morbidity and mortality. However, the incidence of de novo COPD in patients with AKI, and the impact of concurrent COPD on the outcome during post-AKI care is unclear. Patients who recovered from dialysis-requiring AKI (AKI-D) during index hospitalizations between 1998 and 2010 were identified from nationwide administrative registries. A competing risk analysis was conducted to predict the incidence of adverse cardiovascular events and mortality. Among the 14,871 patients who recovered from temporary dialysis, 1535 (10.7%) were identified as having COPD (COPD group) one year after index discharge and matched with 1473 patients without COPD (non-COPD group) using propensity scores. Patients with acute kidney disease superimposed withs COPD were associated with a higher risk of incident ischemic stroke (subdistribution hazard ratio (sHR), 1.52; 95% confidence interval (95% CI), 1.17 to 1.97; p = 0.002) and congestive heart failure (CHF; sHR, 1.61; (95% CI), 1.39 to 1.86; p < 0.001). The risks of incident hemorrhagic stroke, myocardial infarction, end-stage renal disease, and mortality were not statistically different between the COPD and non-COPD groups. This observation adds another dimension to accumulating evidence regarding pulmo-renal consequences after AKI.
Collapse
Affiliation(s)
- Che-Hsiung Wu
- Division of Nephrology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan.
- School of Medicine, Tzu Chi University, Hualien 970, Taiwan.
| | - Huang-Ming Chang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 100, Taiwan.
| | - Cheng-Yi Wang
- Department of Internal Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan.
| | - Likwang Chen
- Institute of Population Health Sciences, National Health Research Institutes, Zhunan 350, Taiwan.
| | - Liang-Wen Chen
- Department of Surgery, National Taiwan University Hospital, National Taiwan University, Taipei 100, Taiwan.
| | - Chien-Heng Lai
- Department of Surgery, National Taiwan University Hospital, National Taiwan University, Taipei 100, Taiwan.
| | - Shuenn-Wen Kuo
- Department of Surgery, National Taiwan University Hospital, National Taiwan University, Taipei 100, Taiwan.
| | - Hao-Chien Wang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 100, Taiwan.
| | - Vin-Cent Wu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 100, Taiwan.
| |
Collapse
|
18
|
Pan HC, Chou KM, Lee CC, Yang NI, Sun CY. Circulating Klotho levels can predict long-term macrovascular outcomes in type 2 diabetic patients. Atherosclerosis 2018; 276:83-90. [PMID: 30048945 DOI: 10.1016/j.atherosclerosis.2018.07.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 06/11/2018] [Accepted: 07/05/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND AIMS Type 2 diabetes is a global health problem that is associated with a wide variety of vascular complications and associated morbidity and mortality. Klotho is an enzyme and transmembrane protein, and increasing evidence suggests that Klotho may contribute to reduced oxidative stress, improved endothelial function, and vasoprotection. To date, the physiological role of Klotho in vascular complications associated with diabetes is unclear. METHODS We prospectively recruited 252 patients with type 2 diabetes, who visited an outpatient clinic at our hospital between 2009 and 2011. Patients in the top and bottom tertiles of circulating Klotho levels were enrolled for analysis. RESULTS Of the 168 patients enrolled, 45.8% were male, the mean age was 57.2 years, and the average duration of diabetes was 7.58 years. In multiple regression analysis, a high Klotho level was associated with a reduced risk of developing coronary artery disease and cerebrovascular accidents. Klotho level was also an independent predictor for the development of macroangiopathies within the 7-year study period. CONCLUSIONS Our results suggest that circulating Klotho level is a predictor of long-term macrovascular outcomes in patients with type 2 diabetes.
Collapse
Affiliation(s)
- Heng-Chih Pan
- Division of Nephrology, Department of Internal Medicine, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Kuei-Mei Chou
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Chin-Chan Lee
- Division of Nephrology, Department of Internal Medicine, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan; Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Ning-I Yang
- Chang Gung University College of Medicine, Taoyuan, Taiwan; Division of Cardiology, Department of Internal Medicine, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Chiao-Yin Sun
- Division of Nephrology, Department of Internal Medicine, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan; Chang Gung University College of Medicine, Taoyuan, Taiwan.
| |
Collapse
|
19
|
Klotho G-395A gene polymorphism: impact on progression of end-stage renal disease and development of cardiovascular complications in children on dialysis. Pediatr Nephrol 2018; 33:1019-1027. [PMID: 29313136 DOI: 10.1007/s00467-017-3877-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 11/08/2017] [Accepted: 12/06/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND Klotho G-395-A gene polymorphism may impact children with end-stage renal disease (ESRD). We investigated the relevance of Klotho G-395-A on ESRD development and progression, and its relationship with evolution of cardiovascular complications in pediatric dialysis patients. METHODS Fifty-five children with chronic kidney disease (CKD) and seventy healthy children were genotyped for Klotho G-395A. RESULTS Incidence of GA/AA genotypes and A allele were higher in ESRD patients compared with controls (54.5 vs. 7.1%, P < 0.001; 30.9 vs. 13.6%, P = 0.001, respectively). Also, children with GA/AA genotypes were 15.6 times more likely to develop ESRD than with GG genotype (95% CI 5.4-44.7, P < 0.001). A allele carriers have 2.8 times higher risk of developing ESRD than those with G allele (95% CI 1.5-5.35, P = 0.001). Also, the A allele could be considered a predictor of cardiovascular disease (CVD), as carriers have 161 times higher risk of cardiovascular complications than non-carriers (95% CI 21-1233, P < 0.001). All ESRD patients with CVD presented with left ventricular hypertrophy (LVH) and the frequency of A allele was significantly higher among ESRD children with LVH, whereas G allele frequency was significantly higher among ESRD children without LVH. CONCLUSIONS The A allele of the G-395A Klotho gene polymorphism shows a significantly higher frequency among children with CKD and those with CVD and LVH. This mutant allele could be used as a risk marker for the development of ESRD as well as a predictor of CVD in these children.
Collapse
|
20
|
Skrzypkowska M, Słomiński B, Ryba-Stanisławowska M, Gutknecht P, Siebert J. Circulating CD34+ and CD34+VEGFR2+ progenitor cells are associated with KLOTHO KL-VS polymorphism. Microvasc Res 2018; 119:1-6. [PMID: 29604296 DOI: 10.1016/j.mvr.2018.03.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 03/26/2018] [Accepted: 03/26/2018] [Indexed: 11/30/2022]
Abstract
BACKGROUND KLOTHO is a regulator of endothelial cells activity and integrity. It has been described for the first time because of its anti-aging properties. KLOTHO encoding gene is present in many functional variants in humans, including "KL-VS" variant that has been connected with longevity and cardiovascular disease development. Few mechanisms have been proposed to explain these associations, but none of them focused on cells from CD34+ population. The aim of our study was to investigate influence of KLOTHO KL-VS polymorphism on populations of CD34+ and CD34+VEGFR2+ cells. METHODS AND RESULTS We examined 167 Polish subjects from Pomeranian region. The analysis concerned KL-VS polymorphism, flow cytometry evaluation of whole blood cells and determination of endothelium-associated serum/plasma factors. Our results indicate that individuals possessing at least one KL-VS allele are characterized by greater number of CD34+ and CD34+VEGFR2+ and their various subpopulations (CD34+CD133+, CD34+c-Kit+, CD34+CXCR4+ and CD34+VEGFR2+c-Kit+) than wild-type volunteers. This group also exhibited more favorable lipid profile and statistically insignificant decrease of vWF and angiotensin II in their blood, whereas VEGF levels were elevated. CONCLUSION One of the mechanisms that are responsible for previously described KL-VS heterozygote advantage may be connected with maintaining greater size of hematopoietic and endothelial progenitor cells population.
Collapse
Affiliation(s)
- Maria Skrzypkowska
- Department of Immunology, Medical University of Gdańsk, Dębinki 1, 80-211 Gdańsk, Poland.
| | - Bartosz Słomiński
- Department of Immunology, Medical University of Gdańsk, Dębinki 1, 80-211 Gdańsk, Poland
| | | | - Piotr Gutknecht
- University Center for Cardiology Department of Family Medicine, Medical University of Gdansk, M. Skłodowskiej-Curie 3a, 80-210, Gdańsk, Poland
| | - Janusz Siebert
- University Center for Cardiology Department of Family Medicine, Medical University of Gdansk, M. Skłodowskiej-Curie 3a, 80-210, Gdańsk, Poland
| |
Collapse
|
21
|
Systemic klotho is associated with KLOTHO variation and predicts intrinsic cortical connectivity in healthy human aging. Brain Imaging Behav 2018; 11:391-400. [PMID: 27714549 PMCID: PMC5382127 DOI: 10.1007/s11682-016-9598-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Cognitive decline is a major biomedical challenge as the global population ages. Elevated levels of the longevity factor klotho suppress aging, enhance cognition, and promote synaptic plasticity and neural resilience against aging and Alzheimer’s disease (AD)-related pathogenic proteins. Here, we examined the relationship between human genetic variants of KLOTHO and systemic klotho levels – and assessed neuroanatomic correlates of serum klotho in a cohort of healthy older adults. Serum klotho levels were increased with KL-VS heterozygosity, as anticipated. We report, for the first time, that serum klotho levels were paradoxically decreased with KL-VS homozygosity. Further, we found that higher serum klotho levels were associated with measures of greater intrinsic connectivity in key functional networks of the brain vulnerable to aging and AD such as the fronto-parietal and default mode networks. Our findings suggest that elevated klotho promotes a resilient brain, possibly through increased network connectivity of critical brain regions.
Collapse
|
22
|
Long FY, Shi MQ, Zhou HJ, Liu DL, Sang N, Du JR. Klotho upregulation contributes to the neuroprotection of ligustilide against cerebral ischemic injury in mice. Eur J Pharmacol 2017; 820:198-205. [PMID: 29233659 DOI: 10.1016/j.ejphar.2017.12.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Revised: 12/02/2017] [Accepted: 12/08/2017] [Indexed: 12/30/2022]
Abstract
Klotho, an aging-suppressor gene, encodes a protein that potentially acts as a neuroprotective factor. Our previous studies showed that ligustilide minimizes the cognitive dysfunction and brain damage induced by cerebral ischemia; however, the underlying mechanisms remain unclear. This study aims to investigate whether klotho is involved in the protective effects of ligustilide against cerebral ischemic injury in mice. Cerebral ischemia was induced by bilateral common carotid arterial occlusion. Neurobehavioral tests as well as Nissl and Fluoro-Jade B staining were used to evaluate the protective effects of ligustilide in cerebral ischemia, and Western blotting and ELISA approaches were used to investigate the underlying mechanisms. Administration of ligustilide prevented the development of neurological deficits and reduced neuronal loss in the hippocampal CA1 region and the caudate putamen after cerebral ischemia. The protective effects were associated with inhibition of the RIG-I/NF-κB p65 and Akt/FoxO1 pathways and with prevention of inflammation and oxidative stress in the brain. Further, downregulation of klotho could attenuate the neuroprotection of ligustilide against cerebral ischemic injury. Ligustilide exerted neuroprotective effects in mice after cerebral ischemia by regulating anti-inflammatory and anti-oxidant signaling pathways. Furthermore, klotho upregulation contributes to the neuroprotection of LIG against cerebral ischemic injury. These results indicated that ligustilide may be a promising therapeutic agent for the treatment of cerebral ischemia.
Collapse
Affiliation(s)
- Fang-Yi Long
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu 610041, China; Department of Pharmacy, Sichuan Provincial Hospital for Women and Children, Women and Children's Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu 610041, China
| | - Meng-Qi Shi
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Hong-Jing Zhou
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Dong-Ling Liu
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Na Sang
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Jun-Rong Du
- Department of Pharmacology, Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
23
|
Abstract
The vertebrate endoskeleton is not a mere frame for muscle attachment to facilitate locomotion, but is a massive organ integrated with many physiologic functions including mineral and energy metabolism. Mineral balance is maintained by tightly controlled ion fluxes that are external (intestine and kidney) and internal (between bone and other organs), and are regulated and coordinated by many endocrine signals between these organs. The endocrine fibroblast growth factors (FGFs) and Klotho gene families are complex systems that co-evolved with the endoskeleton. In particular, FGF23 and αKlotho which are primarily derived from bone and kidney respectively, are critical in maintaining mineral metabolism where each of these proteins serving highly diverse roles; abound with many unanswered questions regarding their upstream regulation and downstream functions. Genetic lesions of components of this network produce discreet disturbances in many facets of mineral metabolism. One acquired condition with colossal elevations of FGF23 and suppression of αKlotho is chronic kidney disease where multiple organ dysfunction contributes to the morbidity and mortality. However, the single most important group of derangements that encompasses the largest breadth of complications is mineral metabolism disorders. Mineral metabolic disorders in CKD impact negatively and significantly on the progression of renal disease as well as extra-renal complications. Knowledge of the origin, nature, and impact of phosphate, FGF23, and αKlotho derangements is pivotal to understanding the pathophysiology and treatment of CKD.
Collapse
Affiliation(s)
- Makoto Kuro-O
- Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan; Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Orson W Moe
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
24
|
Lau WL, Huisa BN, Fisher M. The Cerebrovascular-Chronic Kidney Disease Connection: Perspectives and Mechanisms. Transl Stroke Res 2016; 8:67-76. [PMID: 27628245 PMCID: PMC5241336 DOI: 10.1007/s12975-016-0499-x] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 08/21/2016] [Accepted: 08/25/2016] [Indexed: 12/12/2022]
Abstract
Chronic kidney disease (CKD) is an independent risk factor for the development of cerebrovascular disease, particularly small vessel disease which can manifest in a variety of phenotypes ranging from lacunes to microbleeds. Small vessel disease likely contributes to cognitive dysfunction in the CKD population. Non-traditional risk factors for vascular injury in uremia include loss of calcification inhibitors, hyperphosphatemia, increased blood pressure variability, elastinolysis, platelet dysfunction, and chronic inflammation. In this review, we discuss the putative pathways by which these mechanisms may promote cerebrovascular disease and thus increase risk of future stroke in CKD patients.
Collapse
Affiliation(s)
- Wei Ling Lau
- Department of Medicine, Division of Nephrology, University of California, Irvine, CA, USA
| | - Branko N Huisa
- Department of Neurology, University of California, San Diego, CA, USA
| | - Mark Fisher
- Departments of Neurology, Anatomy & Neurobiology, and Pathology & Laboratory Medicine, University of California, Irvine, CA, USA. .,Department of Neurology, UC Irvine Medical Center, 101 The City Drive South, Shanbrom Hall, Room 121, Orange, CA, 92868, USA.
| |
Collapse
|
25
|
Klotho: a humeral mediator in CSF and plasma that influences longevity and susceptibility to multiple complex disorders, including depression. Transl Psychiatry 2016; 6:e876. [PMID: 27576165 PMCID: PMC5022081 DOI: 10.1038/tp.2016.135] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 03/24/2015] [Indexed: 01/04/2023] Open
Abstract
Klotho is a hormone secreted into human cerebrospinal fluid (CSF), plasma and urine that promotes longevity and influences the onset of several premature senescent phenotypes in mice and humans, including atherosclerosis, cardiovascular disease, stroke and osteoporosis. Preliminary studies also suggest that Klotho possesses tumor suppressor properties. Klotho's roles in these phenomena were first suggested by studies demonstrating that a defect in the Klotho gene in mice results in a significant decrease in lifespan. The Klotho-deficient mouse dies prematurely at 8-9 weeks of age. At 4-5 weeks of age, a syndrome resembling human ageing emerges consisting of atherosclerosis, osteoporosis, cognitive disturbances and alterations of hippocampal architecture. Several deficits in Klotho-deficient mice are likely to contribute to these phenomena. These include an inability to defend against oxidative stress in the central nervous system and periphery, decreased capacity to generate nitric oxide to sustain normal endothelial reactivity, defective Klotho-related mediation of glycosylation and ion channel regulation, increased insulin/insulin-like growth factor signaling and a disturbed calcium and phosphate homeostasis accompanied by altered vitamin D levels and ectopic calcification. Identifying the mechanisms by which Klotho influences multiple important pathways is an emerging field in human biology that will contribute significantly to understanding basic physiologic processes and targets for the treatment of complex diseases. Because many of the phenomena seen in Klotho-deficient mice occur in depressive illness, major depression and bipolar disorder represent illnesses potentially associated with Klotho dysregulation. Klotho's presence in CSF, blood and urine should facilitate its study in clinical populations.
Collapse
|
26
|
Kim HK, Jeong BH. Lack of functional KL-VS polymorphism of the KLOTHO gene in the Korean population. Genet Mol Biol 2016; 39:370-3. [PMID: 27560364 PMCID: PMC5004824 DOI: 10.1590/1678-4685-gmb-2015-0160] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 12/04/2015] [Indexed: 05/29/2023] Open
Abstract
The functional variant of the Klotho "KL-VS" stretch, which includes six
polymorphisms in linkage disequilibrium, is reportedly associated with healthy aging
and longevity in European and American populations. Among Asian populations, this
variant has been observed in the Indian population but not in the Iranian population.
An association between KL-VS polymorphism and aging has not been reported in Koreans.
To investigate whether the KL-VS polymorphism could be associated with healthy aging
and longevity in a Korean population, we analyzed genotype and allele frequencies of
the KL-VS variant in a large Korean population sample. The KL-VS variant was not
found in 874 Korean individuals. Thus, it is not possible to test its association to
aging in the East Asian populations.
Collapse
Affiliation(s)
- Hee-Kwon Kim
- Korea Zoonosis Research Institute, Chonbuk National University, Iksan, Jeonbuk, Republic of Korea.,Department of Nuclear Medicine, Molecular Imaging & Therapeutic Medicine Research Center, Biomedical Research Institute, Chonbuk National University Medical School and Hospital, Jeonju, Republic of Korea
| | - Byung-Hoon Jeong
- Korea Zoonosis Research Institute, Chonbuk National University, Iksan, Jeonbuk, Republic of Korea.,Department of Bioactive Material Sciences, Chonbuk National University, Jeonju, Jeonbuk Republic of Korea
| |
Collapse
|
27
|
Abstract
Maintenance of a normal serum phosphate level depends on absorption in the gut, reabsorption and excretion by the kidney, and the flux between the extracellular and skeletal pools. Phosphate homeostasis is a coordinated, complex system of crosstalk between the bone, intestine, kidney, and parathyroid gland. Dysfunction of this system has serious clinical consequences in healthy individuals and those with conditions, such as CKD, in which hyperphosphatemia is associated with increased risks of cardiovascular morbidity and mortality. The last half-century of renal research has helped define the contribution of the parathyroid hormone, calcitriol, fibroblast growth factor 23, and Klotho in the regulation of phosphate. However, despite new discoveries and insights gained during this time, what remains unchanged is the recognition that phosphate retention is the initiating factor for the development of many of the complications observed in CKD, namely secondary hyperparathyroidism and bone and cardiovascular diseases. Controlling phosphate load remains the primary goal in the treatment of CKD. This review discusses the clinical effects of dysregulated phosphate metabolism, particularly in CKD, and its association with cardiovascular disease. The importance of early control of phosphate load in the treatment of CKD is emphasized, and the latest research in the treatment of phosphate retention is discussed.
Collapse
Affiliation(s)
- Cynthia S Ritter
- Renal Division, Washington University School of Medicine, St. Louis, Missouri
| | | |
Collapse
|
28
|
Donate-Correa J, Martín-Núñez E, Martínez-Sanz R, Muros-de-Fuentes M, Mora-Fernández C, Pérez-Delgado N, Navarro-González JF. Influence of Klotho gene polymorphisms on vascular gene expression and its relationship to cardiovascular disease. J Cell Mol Med 2016; 20:128-133. [PMID: 26538295 PMCID: PMC4717853 DOI: 10.1111/jcmm.12710] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 09/01/2015] [Indexed: 12/13/2022] Open
Abstract
Klotho protein has been associated with beneficial effects that contribute to the maintenance of cardiovascular health. Diverse studies suggest that alterations in the levels of this molecule may be associated with pathophysiological abnormalities that result in increased cardiovascular risk. The primary aim of this proof-of-concept study was to analyse the existence of a potential link between Klotho gene polymorphisms and the expression level of this gene in the vascular wall, and additionally with the incidence of cardiovascular disease and cardiovascular risk factors. Our results indicate that the variant G-395A, located in the promoter region, influences Klotho gene vascular expression and is associated with the incidence of diabetes. Similarly, the exonic variant KL-VS was associated with the incidence of atherosclerotic vascular disease and coronary artery disease. Moreover, vascular expression levels of Klotho were related with the incidence of diabetes mellitus and coronary artery disease. These findings, which need to be confirmed in larger studies, suggest a potential role of Klotho in the pathogenesis of vascular damage.
Collapse
Affiliation(s)
- Javier Donate-Correa
- Research Unit, University Hospital Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Ernesto Martín-Núñez
- Research Unit, University Hospital Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Rafael Martínez-Sanz
- Cardiovascular Surgery Service, Hospital Universitario de Canarias, Santa Cruz de Tenerife, Spain
| | - Mercedes Muros-de-Fuentes
- Clinical Biochemistry Service, University Hospital Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Carmen Mora-Fernández
- Research Unit, University Hospital Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Nayra Pérez-Delgado
- Clinical Biochemistry Service, University Hospital Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Juan F Navarro-González
- Research Unit, University Hospital Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
- Nephrology Service, University Hospital Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| |
Collapse
|
29
|
Lim K, Groen A, Molostvov G, Lu T, Lilley KS, Snead D, James S, Wilkinson IB, Ting S, Hsiao LL, Hiemstra TF, Zehnder D. α-Klotho Expression in Human Tissues. J Clin Endocrinol Metab 2015; 100:E1308-18. [PMID: 26280509 PMCID: PMC4596032 DOI: 10.1210/jc.2015-1800] [Citation(s) in RCA: 148] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT α-Klotho has emerged as a powerful regulator of the aging process. To date, the expression profile of α-Klotho in human tissues is unknown, and its existence in some human tissue types is subject to much controversy. OBJECTIVE This is the first study to characterize systemwide tissue expression of transmembrane α-Klotho in humans. We have employed next-generation targeted proteomic analysis using parallel reaction monitoring in parallel with conventional antibody-based methods to determine the expression and spatial distribution of human α-Klotho expression in health. RESULTS The distribution of α-Klotho in human tissues from various organ systems, including arterial, epithelial, endocrine, reproductive, and neuronal tissues, was first identified by immunohistochemistry. Kidney tissues showed strong α-Klotho expression, whereas liver did not reveal a detectable signal. These results were next confirmed by Western blotting of both whole tissues and primary cells. To validate our antibody-based results, α-Klotho-expressing tissues were subjected to parallel reaction monitoring mass spectrometry (data deposited at ProteomeXchange, PXD002775) identifying peptides specific for the full-length, transmembrane α-Klotho isoform. CONCLUSIONS The data presented confirm α-Klotho expression in the kidney tubule and in the artery and provide evidence of α-Klotho expression across organ systems and cell types that has not previously been described in humans.
Collapse
|
30
|
Ding HY, Ma HX. Significant roles of anti-aging protein klotho and fibroblast growth factor23 in cardiovascular disease. J Geriatr Cardiol 2015; 12:439-47. [PMID: 26347327 PMCID: PMC4554784 DOI: 10.11909/j.issn.1671-5411.2015.04.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Revised: 11/06/2014] [Accepted: 02/26/2015] [Indexed: 02/07/2023] Open
Abstract
The klotho gene has been identified as an aging suppressor that encodes a protein involved in cardiovascular disease (CVD). The inactivation of the klotho gene causes serious systemic disorders resembling human aging, such as atherosclerosis, diffuse vascular calcification and shortened life span. Klotho has been demonstrated to ameliorate vascular endothelial dysfunction and delay vascular calcification. Furthermore, klotho gene polymorphisms in the human are associated with various cardiovascular events. Recent experiments show that klotho may reduce transient receptor potential canonical6 (TRPC6) channels, resulting in protecting the heart from hypertrophy and systolic dysfunction. Fibroblast growth factor23 (FGF23) is a bone-derived hormone that plays an important role in the regulation of phosphate and vitamin D metabolism. FGF23 accelerates urinary phosphate excretion and suppresses 1,25-dihydroxy vitaminD3 (1,25(OH)2D3) synthesis in the presence of FGF receptor1 (FGFR1) and its co-receptor klotho, principally in the kidney. The hormonal affects of circulating klotho protein and FGF23 on vascular and heart have contributed to an understanding of their roles in the pathophysiology of arterial stiffness and left ventricular hypertrophy. Klotho and FGF23 appear to play a critical role in the pathogenesis of vascular disease, and may represent a novel potential therapeutic strategy for clinical intervention.
Collapse
Affiliation(s)
- Hong-Ying Ding
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hou-Xun Ma
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
31
|
Yokoyama JS, Sturm VE, Bonham LW, Klein E, Arfanakis K, Yu L, Coppola G, Kramer JH, Bennett DA, Miller BL, Dubal DB. Variation in longevity gene KLOTHO is associated with greater cortical volumes. Ann Clin Transl Neurol 2015; 2:215-30. [PMID: 25815349 PMCID: PMC4369272 DOI: 10.1002/acn3.161] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 11/24/2014] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVE Identifying genetic variation associated with brain structures in aging may elucidate new biologic mechanisms underlying resilience to cognitive decline. We investigated whether carrying one copy of the protective haplotype "KL-VS" in longevity gene KLOTHO (KL) is associated with greater gray matter volume in healthy human aging compared to carrying no copies. METHODS We performed unbiased whole-brain analysis in cognitively normal older adults from two independent cohorts to assess the relationship between KL-VS and gray matter volume using voxel-based morphometry. RESULTS We found that KL-VS heterozygosity was associated with greater volume in right dorsolateral prefrontal cortex (rDLPFC). Because rDLPFC is important for executive function, we analyzed working memory and processing speed in individuals. KL-VS heterozygosity was associated with enhanced executive function. Larger rDLPFC volume correlated with better executive function across the lifespan examined. Statistical analysis suggested that volume partially mediates the effect of genotype on cognition. INTERPRETATION These results suggest that variation in KL is associated with bigger brain volume and better function.
Collapse
Affiliation(s)
- Jennifer S Yokoyama
- Department of Neurology, University of California San FranciscoSan Francisco, California, 94158
| | - Virginia E Sturm
- Department of Neurology, University of California San FranciscoSan Francisco, California, 94158
| | - Luke W Bonham
- Department of Neurology, University of California San FranciscoSan Francisco, California, 94158
| | - Eric Klein
- Department of Neurology and Semel Institute for Neuroscience and Human Behavior, The David Geffen School of Medicine at University of California Los AngelesLos Angeles, California, 90095
| | - Konstantinos Arfanakis
- Department of Biomedical Engineering, Illinois Institute of TechnologyChicago, Illinois, 60616
- Rush Alzheimer's Disease Center, Rush University Medical CenterChicago, Illinois, 60612
| | - Lei Yu
- Rush Alzheimer's Disease Center, Rush University Medical CenterChicago, Illinois, 60612
| | - Giovanni Coppola
- Department of Neurology and Semel Institute for Neuroscience and Human Behavior, The David Geffen School of Medicine at University of California Los AngelesLos Angeles, California, 90095
| | - Joel H Kramer
- Department of Neurology, University of California San FranciscoSan Francisco, California, 94158
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical CenterChicago, Illinois, 60612
| | - Bruce L Miller
- Department of Neurology, University of California San FranciscoSan Francisco, California, 94158
| | - Dena B Dubal
- Department of Neurology, University of California San FranciscoSan Francisco, California, 94158
| |
Collapse
|
32
|
Dubal DB, Yokoyama JS, Zhu L, Broestl L, Worden K, Wang D, Sturm VE, Kim D, Klein E, Yu GQ, Ho K, Eilertson KE, Yu L, Kuro-o M, De Jager PL, Coppola G, Small GW, Bennett DA, Kramer JH, Abraham CR, Miller BL, Mucke L. Life extension factor klotho enhances cognition. Cell Rep 2014; 7:1065-76. [PMID: 24813892 PMCID: PMC4176932 DOI: 10.1016/j.celrep.2014.03.076] [Citation(s) in RCA: 215] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 01/10/2014] [Accepted: 03/31/2014] [Indexed: 12/22/2022] Open
Abstract
Aging is the primary risk factor for cognitive decline, an emerging health threat to aging societies worldwide. Whether anti-aging factors such as klotho can counteract cognitive decline is unknown. We show that a lifespan-extending variant of the human KLOTHO gene, KL-VS, is associated with enhanced cognition in heterozygous carriers. Because this allele increased klotho levels in serum, we analyzed transgenic mice with systemic overexpression of klotho. They performed better than controls in multiple tests of learning and memory. Elevating klotho in mice also enhanced long-term potentiation, a form of synaptic plasticity, and enriched synaptic GluN2B, an N-methyl-D-aspartate receptor (NMDAR) subunit with key functions in learning and memory. Blockade of GluN2B abolished klotho-mediated effects. Surprisingly, klotho effects were evident also in young mice and did not correlate with age in humans, suggesting independence from the aging process. Augmenting klotho or its effects may enhance cognition and counteract cognitive deficits at different life stages.
Collapse
Affiliation(s)
- Dena B Dubal
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA.
| | - Jennifer S Yokoyama
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Lei Zhu
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Lauren Broestl
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Kurtresha Worden
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Dan Wang
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Virginia E Sturm
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Daniel Kim
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | - Eric Klein
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA 90024, USA
| | - Gui-Qiu Yu
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | - Kaitlyn Ho
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | | | - Lei Yu
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - Makoto Kuro-o
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Center for Molecular Medicine, Jichi Medical University, Tochigi 329-0498, Japan
| | - Philip L De Jager
- Program in Translational NeuroPsychiatric Genomics, Institute for Neurosciences, Departments of Neurology & Psychiatry, Brigham and Women's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA; Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142, USA
| | - Giovanni Coppola
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA 90024, USA
| | - Gary W Small
- Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA 90024, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - Joel H Kramer
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Carmela R Abraham
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Bruce L Miller
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Lennart Mucke
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
33
|
Di Bona D, Accardi G, Virruso C, Candore G, Caruso C. Association of Klotho polymorphisms with healthy aging: a systematic review and meta-analysis. Rejuvenation Res 2014; 17:212-6. [PMID: 24164579 DOI: 10.1089/rej.2013.1523] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Today it is clearly evident that genetic background constitutes an integral part of aging and longevity. Many studies on long-lived people have been conducted emphasizing the role of certain genes in long life. Classic case-control studies, genome-wide association studies, and high-throughput sequencing have permitted identification of a variety of genetic variants seemingly associated with longevity. Over the years, aging research has focused on the insulin/insulin-like growth factor-1 (IGF-1) signaling pathway because of its evolutionarily conserved correlation with life-span extension in model animals. Indeed, many single-nucleotide polymorphisms (SNPs) associated with longevity were identified in genes encoding proteins that take part in this metabolic pathway. Closely related to this pathway is the Klotho gene. It encodes a type-I membrane protein expressed in two forms, membrane and secreted. The latter form suppresses oxidative stress and growth factor signaling and regulates ion channels and transporters. In particular, its over-expression seems to be able to suppress insulin/IGF-1 signaling extending life span. Thus, our aim was to assemble the results in the literature concerning the association between the functional variant of the Klotho "KL-VS" stretch, which contains six polymorphisms in linkage disequilibrium, and successful aging to quantify the possible effect of the variants. The results of our systematic review indicate that the Klotho KL-VS variant is associated with healthy aging.
Collapse
Affiliation(s)
- Danilo Di Bona
- 1 Immunosenescence Unit, Department of Pathobiology and Medical and Forensic Biotechnologies, University Hospital, University of Palermo , Palermo, Italy
| | | | | | | | | |
Collapse
|
34
|
Kestenbaum B, Sachs MC, Hoofnagle AN, Siscovick DS, Ix JH, Robinson-Cohen C, Lima JAC, Polak JF, Blondon M, Ruzinski J, Rock D, de Boer IH. Fibroblast growth factor-23 and cardiovascular disease in the general population: the Multi-Ethnic Study of Atherosclerosis. Circ Heart Fail 2014; 7:409-17. [PMID: 24668259 DOI: 10.1161/circheartfailure.113.000952] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Fibroblast growth factor-23 (FGF-23) is a phosphate regulatory hormone that directly stimulates left ventricular hypertrophy in experimental models. The role of FGF-23 in cardiovascular disease development in the general population is unclear. We tested associations of FGF-23 with major subclinical and clinical cardiovascular disease outcomes in a large prospective cohort. METHODS AND RESULTS We evaluated 6547 participants from the Multi-Ethnic Study of Atherosclerosis (MESA) who were initially free of cardiovascular disease. We measured serum FGF-23 using the Kainos immunoassay. The MESA measured left ventricular mass by MRI, coronary calcium by computed tomography, and carotid intima-media thickness by ultrasound. The MESA adjudicated incident heart failure, coronary heart disease, and stroke by medical record review. After adjustment, the highest FGF-23 quartile was associated with an estimated 2.4-g greater left ventricular mass (95% confidence interval, 0.4-4.5 greater) and a 26% greater odds of higher coronary calcium scores (95% confidence interval, 9%-46% greater) compared with the lowest quartile. During 7.5-year follow-up, each 20-pg/mL higher FGF-23 concentration was associated with a 19% greater risk of heart failure (95% confidence interval, 3%-37% greater) and a 14% greater risk of coronary heart disease (95% confidence interval, 1%-28% greater). FGF-23 was not associated with carotid intima-media thickness or stroke. CONCLUSIONS Higher serum FGF-23 concentrations are associated with subclinical cardiac disease and with new heart failure and coronary disease events, but not with carotid intima-media thickness or stroke. FGF-23 may be a novel cardiovascular risk factor in the general population.
Collapse
Affiliation(s)
- Bryan Kestenbaum
- From the Kidney Research Institute, Department of Medicine, Division of Nephrology (B.K., M.C.S., C.R.-C., J.R., D.R., I.H.d.B.), Department of Laboratory Medicine (A.N.H.), Cardiovascular Health Research Unit, Departments of Medicine and Epidemiology (D.S.S.), and Department of Epidemiology (M.B.), University of Washington, Seattle; Division of Nephrology, Department of Medicine, University of California at San Diego (J.H.I.); Nephrology Section, Veterans Affairs San Diego Healthcare System, CA (J.H.I.); and Division of Preventive Medicine, Department of Family and Preventive Medicine, University of California at San Diego (J.H.I.); Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (J.A.C.L.); and Department of Radiology, Tufts-New England Medical Center, Boston, MA (J.F.P.).
| | - Michael C Sachs
- From the Kidney Research Institute, Department of Medicine, Division of Nephrology (B.K., M.C.S., C.R.-C., J.R., D.R., I.H.d.B.), Department of Laboratory Medicine (A.N.H.), Cardiovascular Health Research Unit, Departments of Medicine and Epidemiology (D.S.S.), and Department of Epidemiology (M.B.), University of Washington, Seattle; Division of Nephrology, Department of Medicine, University of California at San Diego (J.H.I.); Nephrology Section, Veterans Affairs San Diego Healthcare System, CA (J.H.I.); and Division of Preventive Medicine, Department of Family and Preventive Medicine, University of California at San Diego (J.H.I.); Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (J.A.C.L.); and Department of Radiology, Tufts-New England Medical Center, Boston, MA (J.F.P.)
| | - Andy N Hoofnagle
- From the Kidney Research Institute, Department of Medicine, Division of Nephrology (B.K., M.C.S., C.R.-C., J.R., D.R., I.H.d.B.), Department of Laboratory Medicine (A.N.H.), Cardiovascular Health Research Unit, Departments of Medicine and Epidemiology (D.S.S.), and Department of Epidemiology (M.B.), University of Washington, Seattle; Division of Nephrology, Department of Medicine, University of California at San Diego (J.H.I.); Nephrology Section, Veterans Affairs San Diego Healthcare System, CA (J.H.I.); and Division of Preventive Medicine, Department of Family and Preventive Medicine, University of California at San Diego (J.H.I.); Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (J.A.C.L.); and Department of Radiology, Tufts-New England Medical Center, Boston, MA (J.F.P.)
| | - David S Siscovick
- From the Kidney Research Institute, Department of Medicine, Division of Nephrology (B.K., M.C.S., C.R.-C., J.R., D.R., I.H.d.B.), Department of Laboratory Medicine (A.N.H.), Cardiovascular Health Research Unit, Departments of Medicine and Epidemiology (D.S.S.), and Department of Epidemiology (M.B.), University of Washington, Seattle; Division of Nephrology, Department of Medicine, University of California at San Diego (J.H.I.); Nephrology Section, Veterans Affairs San Diego Healthcare System, CA (J.H.I.); and Division of Preventive Medicine, Department of Family and Preventive Medicine, University of California at San Diego (J.H.I.); Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (J.A.C.L.); and Department of Radiology, Tufts-New England Medical Center, Boston, MA (J.F.P.)
| | - Joachim H Ix
- From the Kidney Research Institute, Department of Medicine, Division of Nephrology (B.K., M.C.S., C.R.-C., J.R., D.R., I.H.d.B.), Department of Laboratory Medicine (A.N.H.), Cardiovascular Health Research Unit, Departments of Medicine and Epidemiology (D.S.S.), and Department of Epidemiology (M.B.), University of Washington, Seattle; Division of Nephrology, Department of Medicine, University of California at San Diego (J.H.I.); Nephrology Section, Veterans Affairs San Diego Healthcare System, CA (J.H.I.); and Division of Preventive Medicine, Department of Family and Preventive Medicine, University of California at San Diego (J.H.I.); Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (J.A.C.L.); and Department of Radiology, Tufts-New England Medical Center, Boston, MA (J.F.P.)
| | - Cassianne Robinson-Cohen
- From the Kidney Research Institute, Department of Medicine, Division of Nephrology (B.K., M.C.S., C.R.-C., J.R., D.R., I.H.d.B.), Department of Laboratory Medicine (A.N.H.), Cardiovascular Health Research Unit, Departments of Medicine and Epidemiology (D.S.S.), and Department of Epidemiology (M.B.), University of Washington, Seattle; Division of Nephrology, Department of Medicine, University of California at San Diego (J.H.I.); Nephrology Section, Veterans Affairs San Diego Healthcare System, CA (J.H.I.); and Division of Preventive Medicine, Department of Family and Preventive Medicine, University of California at San Diego (J.H.I.); Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (J.A.C.L.); and Department of Radiology, Tufts-New England Medical Center, Boston, MA (J.F.P.)
| | - Joao A C Lima
- From the Kidney Research Institute, Department of Medicine, Division of Nephrology (B.K., M.C.S., C.R.-C., J.R., D.R., I.H.d.B.), Department of Laboratory Medicine (A.N.H.), Cardiovascular Health Research Unit, Departments of Medicine and Epidemiology (D.S.S.), and Department of Epidemiology (M.B.), University of Washington, Seattle; Division of Nephrology, Department of Medicine, University of California at San Diego (J.H.I.); Nephrology Section, Veterans Affairs San Diego Healthcare System, CA (J.H.I.); and Division of Preventive Medicine, Department of Family and Preventive Medicine, University of California at San Diego (J.H.I.); Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (J.A.C.L.); and Department of Radiology, Tufts-New England Medical Center, Boston, MA (J.F.P.)
| | - Joseph F Polak
- From the Kidney Research Institute, Department of Medicine, Division of Nephrology (B.K., M.C.S., C.R.-C., J.R., D.R., I.H.d.B.), Department of Laboratory Medicine (A.N.H.), Cardiovascular Health Research Unit, Departments of Medicine and Epidemiology (D.S.S.), and Department of Epidemiology (M.B.), University of Washington, Seattle; Division of Nephrology, Department of Medicine, University of California at San Diego (J.H.I.); Nephrology Section, Veterans Affairs San Diego Healthcare System, CA (J.H.I.); and Division of Preventive Medicine, Department of Family and Preventive Medicine, University of California at San Diego (J.H.I.); Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (J.A.C.L.); and Department of Radiology, Tufts-New England Medical Center, Boston, MA (J.F.P.)
| | - Marc Blondon
- From the Kidney Research Institute, Department of Medicine, Division of Nephrology (B.K., M.C.S., C.R.-C., J.R., D.R., I.H.d.B.), Department of Laboratory Medicine (A.N.H.), Cardiovascular Health Research Unit, Departments of Medicine and Epidemiology (D.S.S.), and Department of Epidemiology (M.B.), University of Washington, Seattle; Division of Nephrology, Department of Medicine, University of California at San Diego (J.H.I.); Nephrology Section, Veterans Affairs San Diego Healthcare System, CA (J.H.I.); and Division of Preventive Medicine, Department of Family and Preventive Medicine, University of California at San Diego (J.H.I.); Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (J.A.C.L.); and Department of Radiology, Tufts-New England Medical Center, Boston, MA (J.F.P.)
| | - John Ruzinski
- From the Kidney Research Institute, Department of Medicine, Division of Nephrology (B.K., M.C.S., C.R.-C., J.R., D.R., I.H.d.B.), Department of Laboratory Medicine (A.N.H.), Cardiovascular Health Research Unit, Departments of Medicine and Epidemiology (D.S.S.), and Department of Epidemiology (M.B.), University of Washington, Seattle; Division of Nephrology, Department of Medicine, University of California at San Diego (J.H.I.); Nephrology Section, Veterans Affairs San Diego Healthcare System, CA (J.H.I.); and Division of Preventive Medicine, Department of Family and Preventive Medicine, University of California at San Diego (J.H.I.); Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (J.A.C.L.); and Department of Radiology, Tufts-New England Medical Center, Boston, MA (J.F.P.)
| | - Denise Rock
- From the Kidney Research Institute, Department of Medicine, Division of Nephrology (B.K., M.C.S., C.R.-C., J.R., D.R., I.H.d.B.), Department of Laboratory Medicine (A.N.H.), Cardiovascular Health Research Unit, Departments of Medicine and Epidemiology (D.S.S.), and Department of Epidemiology (M.B.), University of Washington, Seattle; Division of Nephrology, Department of Medicine, University of California at San Diego (J.H.I.); Nephrology Section, Veterans Affairs San Diego Healthcare System, CA (J.H.I.); and Division of Preventive Medicine, Department of Family and Preventive Medicine, University of California at San Diego (J.H.I.); Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (J.A.C.L.); and Department of Radiology, Tufts-New England Medical Center, Boston, MA (J.F.P.)
| | - Ian H de Boer
- From the Kidney Research Institute, Department of Medicine, Division of Nephrology (B.K., M.C.S., C.R.-C., J.R., D.R., I.H.d.B.), Department of Laboratory Medicine (A.N.H.), Cardiovascular Health Research Unit, Departments of Medicine and Epidemiology (D.S.S.), and Department of Epidemiology (M.B.), University of Washington, Seattle; Division of Nephrology, Department of Medicine, University of California at San Diego (J.H.I.); Nephrology Section, Veterans Affairs San Diego Healthcare System, CA (J.H.I.); and Division of Preventive Medicine, Department of Family and Preventive Medicine, University of California at San Diego (J.H.I.); Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD (J.A.C.L.); and Department of Radiology, Tufts-New England Medical Center, Boston, MA (J.F.P.)
| |
Collapse
|
35
|
Tucker Zhou TB, King GD, Chen C, Abraham CR. Biochemical and functional characterization of the klotho-VS polymorphism implicated in aging and disease risk. J Biol Chem 2013; 288:36302-11. [PMID: 24217253 PMCID: PMC3868745 DOI: 10.1074/jbc.m113.490052] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 11/06/2013] [Indexed: 12/14/2022] Open
Abstract
Klotho (KL) is an age-regulating protein named after the Greek goddess who spins the thread of life. Mice deficient in KL are normal throughout development, but rapidly degenerate and display a variety of aging-associated abnormalities that eventually lead to decreased life expectancy. While multiple genetic association studies have identified KL polymorphisms linked with changes in disease risk, there is a paucity of concrete mechanistic data to explain how these amino acid substitutions alter KL protein function. The KLVS polymorphism is suggested to lead to changes in protein trafficking although the mechanism is unclear. Our studies have sought to further investigate the functional differences in the KLVS variant that result in increased risk of many age-related diseases. Our findings suggest that the F352V and C370S substitutions lead to alterations in processing as seen by differences in shedding and half-life. Their co-expression in KLVS results in a phenotype resembling wild-type, but despite this intragenic complementation there are still changes in homodimerization and interactions with FGFR1c. Taken together, these studies suggest that KLVS leads to altered homodimerization that indirectly leads to changes in processing and FGFR1c interactions. These findings help elucidate the functional differences that result from the VS polymorphism, which will help clarify how alterations in KL function can lead to human disease and affect cognition and lifespan.
Collapse
Affiliation(s)
- Tracey B. Tucker Zhou
- From the Departments of Pharmacology and Experimental Therapeutics and
- Biochemistry, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Gwendalyn D. King
- From the Departments of Pharmacology and Experimental Therapeutics and
| | - CiDi Chen
- From the Departments of Pharmacology and Experimental Therapeutics and
| | - Carmela R. Abraham
- From the Departments of Pharmacology and Experimental Therapeutics and
- Biochemistry, Boston University School of Medicine, Boston, Massachusetts 02118
| |
Collapse
|
36
|
Abstract
Acromegaly is characterized by excessively high GH and IGF1 levels. Recent data suggest that soluble Klotho (sKlotho) is also elevated in patients with active acromegaly. sKlotho decreases towards normal following removal of the GH-producing pituitary adenoma. The Klotho gene was identified in mice following its accidental disruption by ectopic DNA. It is an ageing suppressor gene of restricted expression (mainly in kidneys, brain, and parathyroid and pituitary glands) encoding a transmembrane protein, mKlotho. mKlotho serves as a co-receptor in fibroblast growth factor 23 (FGF23) signalling. FGF23 promotes urinary phosphate excretion and inhibits the synthesis of calcitriol. The ectodomain of mKlotho is enzymatically released to result in a humoral factor, sKlotho, which exerts systemic effects (on ion channels and signalling pathways), possibly by working as an enzyme that modifies glycans of cell surface glycoproteins. GH enhances renal phosphate reabsorption and calcitriol production, i.e. exerts effects in the proximal tubule opposing those attributed to mKlotho, and attenuates calciuria in the distal tubule similar to sKlotho. sKlotho can be measured in extracellular fluids (serum, urine and cerebrospinal fluid (CSF)) by an ELISA. In line with predominant expression of Klotho in kidneys and choroid plexus, concentrations of sKlotho are particularly high in urine and CSF. Determination of sKlotho in serum and urine (both presumably reflecting GH action on the kidneys) could be used as a supplementary tool in the diagnosis and follow-up of patients with acromegaly. The question arises whether GH exerts selected actions via modifying activities of Klotho.
Collapse
Affiliation(s)
- Christoph Schmid
- Division of Endocrinology and Diabetology, University Hospital Zurich, Rämistrasse 100, CH-8091 Zurich, Switzerland Department of Neurosurgery, University Hospital Zurich, Frauenklinikstrasse 10, 8091 Zurich, Switzerland Division of Endocrinology and Diabetology, Kantonsspital St Gallen, 9007 St Gallen, Switzerland Department of Neurosurgery, Clinic Hirslanden, Witellikerstrasse 40, 8032 Zurich, Switzerland
| | | | | | | | | |
Collapse
|
37
|
Donate-Correa J, Mora-Fernández C, Martínez-Sanz R, Muros-de-Fuentes M, Pérez H, Meneses-Pérez B, Cazaña-Pérez V, Navarro-González JF. Expression of FGF23/KLOTHO system in human vascular tissue. Int J Cardiol 2013; 165:179-183. [PMID: 21945708 DOI: 10.1016/j.ijcard.2011.08.850] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2011] [Revised: 08/26/2011] [Accepted: 08/30/2011] [Indexed: 12/26/2022]
Abstract
BACKGROUND Fibroblast growth factor (FGF)-23 levels have been associated with impaired vasoreactivity, increased arterial stiffness, and cardiovascular morbi-mortality, whereas a protective function of KLOTHO against endothelial dysfunction has been reported. Since expression of the FGF23-KLOTHO system in human vascular tissue remains unproved, we aimed to study the expression of FGF23, FGF receptors (FGFR) and KLOTHO in human aorta. In addition, we analyzed the FGF23-KLOTHO expression in occlusive coronary thrombi. METHODS Thoracic aorta specimens from 44 patients underwent elective cardiac surgery, and thrombus material from 2 patients with acute coronary syndrome (ACS), were tested for FGF23-KLOTHO system expression. RESULTS Expression of KLOTHO (mean expression level 4.85 ± 5.43, arbitrary units) and two of the three cognate FGFR (FGFR-1 and -3) were detected and confirmed by RT-PCR, sequencing and qRT-PCR. KLOTHO expression was confirmed within occlusive coronary thrombi from patients with ACS. However, expression of FGF23 and FGFR4 was not observed. We also detected the aortic expression of membrane-anchored A Desintegrin and Metalloproteinases (ADAM)-17, the enzyme responsible for the shedding of KLOTHO from the cell surface, and the anti-inflammatory cytokine interleukin (IL)-10. Interestingly, in aortic samples there was a direct association between KLOTHO mRNA levels and those of ADAM-17 and IL-10 (r = 0.54, P<0.001; r = 0.51, P<0.01, respectively). CONCLUSIONS Human vascular tissue expresses members of the FGF23-KLOTHO system, indicating that it can be a direct target organ for FGF23. In addition, KLOTHO expression is also detected in occlusive coronary thrombi. These findings suggest a putative role of FGF23-KLOTHO axis in human vascular pathophysiology and cardiovascular disease.
Collapse
Affiliation(s)
- Javier Donate-Correa
- Research Unit, University Hospital Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Yadav S, Hasan N, Marjot T, Khan MS, Prasad K, Bentley P, Sharma P. Detailed analysis of gene polymorphisms associated with ischemic stroke in South Asians. PLoS One 2013; 8:e57305. [PMID: 23505425 PMCID: PMC3591429 DOI: 10.1371/journal.pone.0057305] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Accepted: 01/20/2013] [Indexed: 11/30/2022] Open
Abstract
The burden of stroke is disproportionately high in the South Asian subcontinent with South Asian ethnicity conferring a greater risk of ischemic stroke than European ancestry regardless of country inhabited. While genes associated with stroke in European populations have been investigated, they remain largely unknown in South Asians. We conducted a comprehensive meta-analysis of known genetic polymorphisms associated with South Asian ischemic stroke, and compared effect size of the MTHFR C677T-stroke association with effect sizes predicted from homocysteine-stroke association. Electronic databases were searched up to August 2012 for published case control studies investigating genetic polymorphisms associated with ischemic stroke in South Asians. Pooled odds ratios (OR) for each gene-disease association were calculated using a random-effects model. We identified 26 studies (approximately 2529 stroke cases and 2881 controls) interrogating 33 independent genetic polymorphisms in 22 genes. Ten studies described MTHFR C677T (108 with TT genotype and 2018 with CC genotype) -homocysteine relationship and six studies (735 stroke cases and 713 controls) described homocysteine-ischemic stroke relationship. Risk association ORs were calculated for ACE I/D (OR 5.00; 95% CI, 1.17–21.37; p = 0.03), PDE4D SNP 83 (OR 2.20; 95% CI 1.21–3.99; p = 0.01), PDE4D SNP 32 (OR 1.57; 95% CI 1.01–2.45, p = 0.045) and IL10 G1082A (OR 1.44; 95% CI, 1.09–1.91, p = 0.01). Significant association was observed between elevated plasma homocysteine levels and MTHFR/677 TT genotypes in healthy South Asians (Mean difference (ΔX) 5.18 µmol/L; 95% CI 2.03–8.34: p = 0.001). Our results demonstrate that the genetic etiology of ischemic stroke in South Asians is broadly similar to the risk conferred in Europeans, although the dataset is considerably smaller and warrants the same clinical considerations for risk profiling.
Collapse
Affiliation(s)
- Sunaina Yadav
- Imperial College Cerebrovascular Research Unit (ICCRU), Imperial College London, London, United Kingdom
| | - Nazeeha Hasan
- Imperial College Cerebrovascular Research Unit (ICCRU), Imperial College London, London, United Kingdom
| | - Thomas Marjot
- Imperial College Cerebrovascular Research Unit (ICCRU), Imperial College London, London, United Kingdom
| | - Muhammad S. Khan
- Imperial College Cerebrovascular Research Unit (ICCRU), Imperial College London, London, United Kingdom
| | - Kameshwar Prasad
- Department of Neurology, All India Institute of Medical Sciences, New Delhi, India
| | - Paul Bentley
- Imperial College Cerebrovascular Research Unit (ICCRU), Imperial College London, London, United Kingdom
| | - Pankaj Sharma
- Imperial College Cerebrovascular Research Unit (ICCRU), Imperial College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
39
|
Abstract
A disproportionate expansion of white adipose tissue and abnormal recruitment of adipogenic precursor cells can not only lead to obesity but also impair glucose metabolism, which are both common causes of insulin resistance and diabetes mellitus. The development of novel and effective therapeutic strategies to slow the progression of obesity, diabetes mellitus and their associated complications will require improved understanding of adipogenesis and glucose metabolism. Klotho might have a role in adipocyte maturation and systemic glucose metabolism. Klotho increases adipocyte differentiation in vitro, and mice that lack Klotho activity are lean owing to reduced white adipose tissue accumulation; moreover, mice that lack the Kl gene (which encodes Klotho) are resistant to obesity induced by a high-fat diet. Knockout of Kl in leptin-deficient Lep(ob/ob) mice reduces obesity and increases insulin sensitivity, which lowers blood glucose levels. Energy metabolism might also be influenced by Klotho. However, further studies are needed to explore the possibility that Klotho could be a novel therapeutic target to reduce obesity and related complications, and to determine whether and how Klotho might influence the regulation and function of a related protein, β-Klotho, which is also involved in energy metabolism.
Collapse
Affiliation(s)
- M Shawkat Razzaque
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Research and Education Building, Room 304, 190 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
40
|
Crasto CL, Semba RD, Sun K, Cappola AR, Bandinelli S, Ferrucci L. Relationship of low-circulating "anti-aging" klotho hormone with disability in activities of daily living among older community-dwelling adults. Rejuvenation Res 2012; 15:295-301. [PMID: 22530731 PMCID: PMC3388499 DOI: 10.1089/rej.2011.1268] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2011] [Accepted: 11/10/2011] [Indexed: 01/08/2023] Open
Abstract
The aging suppressor gene klotho encodes a single-pass transmembrane protein klotho that in mice is known to extend life span when overexpressed and to resemble accelerated aging, with skeletal muscle atrophy and decreased bone mineral density, when expression is disrupted. We sought to examine the relationship between plasma klotho and disability in activities of daily living (ADL) in older community-dwelling adults. In a cross-sectional study, plasma klotho was measured in a population-based sample of 802 adults, ≥ 65 years, who participated in the "Invecchiare in Chianti" (Aging in the Chianti Area) (InCHIANTI) study in Tuscany, Italy. The overall proportion of adults with ADL disability was 11.9%. Mean (standard deviation) klotho concentrations were 689 (238) pg/mL. From the lowest to the highest tertile of plasma klotho, 16.1%, 9.7%, and 5.6% of participants, respectively, had ADL disability (p=0.0004). Plasma klotho, per 1 standard deviation increase, was associated with ADL disability (odds ratio=0.57, 95% confidence interval 0.35-0.93, p=0.02) in a multivariate logistic regression model adjusting for age, education, cognition, physical activity, physical performance, total cholesterol, alcohol and tobacco use, and chronic diseases. Low plasma klotho concentrations were independently associated with ADL disability among older community-dwelling men and women.
Collapse
Affiliation(s)
- Candace L Crasto
- Department of Ophthalmology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | |
Collapse
|
41
|
Majumdar V, Christopher R. Association of exonic variants of Klotho with metabolic syndrome in Asian Indians. Clin Chim Acta 2011; 412:1116-21. [PMID: 21376714 DOI: 10.1016/j.cca.2011.02.034] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2010] [Revised: 02/25/2011] [Accepted: 02/26/2011] [Indexed: 01/01/2023]
Abstract
BACKGROUND Klotho, an anti-aging gene, is a functional candidate for metabolic syndrome. We conducted a cross-sectional study to evaluate the association of the genetic variants of Klotho with metabolic syndrome and surrogates of insulin resistance in Asian Indians. METHODS We recruited 428 clinically normal subjects for the study. Genotyping was done by polymerase chain reaction and restriction fragment length polymorphism. RESULTS Significant and borderline associations of the KL-VS (OR=15.88 [95%CI, 2.56-98.70], p=0.003) and C1818T (OR=0.28 [95%CI, 0.07-1.07], p=0.063) variants of the Klotho gene, respectively, were observed with metabolic syndrome. The association of the KL-VS variant with metabolic syndrome could be linked to its observed influence on high blood glucose (OR=6.92 [95% CI=1.75-27.44], p=0.006), high blood pressure (OR=5.21 [95%CI=1.00-38.43], p=0.046), insulin resistance (OR=3.59, [95%CI=1.01-12.79], p=0.048) and trend towards its association with hypertriglyceridemia (OR=3.69 [95%CI=0.92-14.77], p=0.065). CONCLUSIONS The genetic variants of Klotho might predict risk for metabolic syndrome and insulin resistance in Asian Indians. However, larger studies in other ethnic populations are warranted to determine the role of these gene variants in the etiology of metabolic syndrome.
Collapse
Affiliation(s)
- Vijaya Majumdar
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences, Bangalore-560029, India
| | | |
Collapse
|