51
|
Berezin AE, Berezin AA. Impaired function of fibroblast growth factor 23 / Klotho protein axis in prediabetes and diabetes mellitus: Promising predictor of cardiovascular risk. Diabetes Metab Syndr 2019; 13:2549-2556. [PMID: 31405675 DOI: 10.1016/j.dsx.2019.07.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 07/08/2019] [Indexed: 12/12/2022]
Abstract
The discovery of clear molecular mechanisms of early cardiac and vascular complications in patients with prediabetes and known diabetes mellitus are core element of stratification at risk with predictive model creation further. Previous clinical studies have shown a pivotal role of impaired signaling axis of fibroblast growth factor 23 (FGF23), FGF23 receptor isoforms and its co-factor Klotho protein in cardiovascular (CV) complications in prediabetes and diabetes. Although there were data received in clinical studies, which confirmed a causative role of altered function of FGF-23/Klotho protein axis in manifestation of CV disease in prediabetes and type 2 diabetes mellitus (T2DM), the target therapy of these diseases directing on improvement of metabolic profiles, systemic and adipokine-relating inflammation by beneficial restoring of dysregulation in FGF-23/Klotho protein axis remain to be not fully clear. The aim of the review was to summarize findings regarding the role of impaired FGF-23/Klotho protein axis in developing CV complications in patients with prediabetes and type 2 diabetes mellitus. It has been elucidated that elevated levels of FGF-23 and deficiency of Klotho protein in peripheral blood are predictors of CV disease and CV outcomes in patients with (pre) diabetes, while predictive values of dynamic changes of the concentrations of these biomarkers require to be elucidated in detail in the future.
Collapse
Affiliation(s)
- Alexander E Berezin
- Internal Medicine Department, State Medical University, Ministry of Health of Ukraine, Zaporozhye, 69035, Ukraine.
| | - Alexander A Berezin
- Internal Medicine Department, Medical Academy of Post-Graduate Education, Ministry of Health of Ukraine, Zaporozhye, 69096, Ukraine
| |
Collapse
|
52
|
Romero A, San Hipólito‐Luengo Á, Villalobos LA, Vallejo S, Valencia I, Michalska P, Pajuelo‐Lozano N, Sánchez‐Pérez I, León R, Bartha JL, Sanz MJ, Erusalimsky JD, Sánchez‐Ferrer CF, Romacho T, Peiró C. The angiotensin-(1-7)/Mas receptor axis protects from endothelial cell senescence via klotho and Nrf2 activation. Aging Cell 2019; 18:e12913. [PMID: 30773786 PMCID: PMC6516147 DOI: 10.1111/acel.12913] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 12/03/2018] [Accepted: 01/06/2019] [Indexed: 12/25/2022] Open
Abstract
Endothelial cell senescence is a hallmark of vascular aging that predisposes to vascular disease. We aimed to explore the capacity of the renin–angiotensin system (RAS) heptapeptide angiotensin (Ang)‐(1‐7) to counteract human endothelial cell senescence and to identify intracellular pathways mediating its potential protective action. In human umbilical vein endothelial cell (HUVEC) cultures, Ang II promoted cell senescence, as revealed by the enhancement in senescence‐associated galactosidase (SA‐β‐gal+) positive staining, total and telomeric DNA damage, adhesion molecule expression, and human mononuclear adhesion to HUVEC monolayers. By activating the G protein‐coupled receptor Mas, Ang‐(1‐7) inhibited the pro‐senescence action of Ang II, but also of a non‐RAS stressor such as the cytokine IL‐1β. Moreover, Ang‐(1‐7) enhanced endothelial klotho levels, while klotho silencing resulted in the loss of the anti‐senescence action of the heptapeptide. Indeed, both Ang‐(1‐7) and recombinant klotho activated the cytoprotective Nrf2/heme oxygenase‐1 (HO‐1) pathway. The HO‐1 inhibitor tin protoporphyrin IX prevented the anti‐senescence action evoked by Ang‐(1‐7) or recombinant klotho. Overall, the present study identifies Ang‐(1‐7) as an anti‐senescence peptide displaying its protective action beyond the RAS by consecutively activating klotho and Nrf2/HO‐1. Ang‐(1‐7) mimetic drugs may thus prove useful to prevent endothelial cell senescence and its related vascular complications.
Collapse
Affiliation(s)
- Alejandra Romero
- Department of Pharmacology Faculty of Medicine Universidad Autónoma de Madrid Madrid Spain
| | | | - Laura A. Villalobos
- Department of Pharmacology Faculty of Medicine Universidad Autónoma de Madrid Madrid Spain
| | - Susana Vallejo
- Department of Pharmacology Faculty of Medicine Universidad Autónoma de Madrid Madrid Spain
- Instituto de Investigaciones Sanitarias IdiPAZ Madrid Spain
| | - Inés Valencia
- Department of Pharmacology Faculty of Medicine Universidad Autónoma de Madrid Madrid Spain
| | - Patrycja Michalska
- Department of Pharmacology Faculty of Medicine Universidad Autónoma de Madrid Madrid Spain
- Instituto Teófilo Hernando Universidad Autónoma de Madrid Madrid Spain
| | - Natalia Pajuelo‐Lozano
- Department of BiochemistryFaculty of MedicineUniversidad Autónoma de Madrid Madrid Spain
- Instituto de Investigaciones BiomédicasUAM-CSIC Madrid Spain
| | - Isabel Sánchez‐Pérez
- Department of BiochemistryFaculty of MedicineUniversidad Autónoma de Madrid Madrid Spain
- Instituto de Investigaciones BiomédicasUAM-CSIC Madrid Spain
- CIBER for Rare Diseases Valencia Spain
| | - Rafael León
- Instituto Teófilo Hernando Universidad Autónoma de Madrid Madrid Spain
- Servicio de Farmacología ClínicaInstituto de Investigación SanitariaHospital Universitario de la Princesa Madrid Spain
| | - José Luis Bartha
- Instituto de Investigaciones Sanitarias IdiPAZ Madrid Spain
- Department of Obstetrics and GynecologyFaculty of MedicineUniversidad Autónoma de Madrid Madrid Spain
| | - María Jesús Sanz
- Department of PharmacologyUniversidad de Valencia Valencia Spain
- Institute of Health Research INCLIVAUniversity Clinic Hospital of Valencia Valencia Spain
| | | | - Carlos F. Sánchez‐Ferrer
- Department of Pharmacology Faculty of Medicine Universidad Autónoma de Madrid Madrid Spain
- Instituto de Investigaciones Sanitarias IdiPAZ Madrid Spain
| | - Tania Romacho
- Department of Pharmacology Faculty of Medicine Universidad Autónoma de Madrid Madrid Spain
| | - Concepción Peiró
- Department of Pharmacology Faculty of Medicine Universidad Autónoma de Madrid Madrid Spain
- Instituto de Investigaciones Sanitarias IdiPAZ Madrid Spain
| |
Collapse
|
53
|
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: 2.8] [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
|
54
|
Donate-Correa J, Martín-Núñez E, Ferri C, Hernández-Carballo C, Tagua VG, Delgado-Molinos A, López-Castillo Á, Rodríguez-Ramos S, Cerro-López P, López-Tarruella VC, Arévalo-González MA, Pérez-Delgado N, Mora-Fernández C, Navarro-González JF. FGF23 and Klotho Levels are Independently Associated with Diabetic Foot Syndrome in Type 2 Diabetes Mellitus. J Clin Med 2019; 8:jcm8040448. [PMID: 30987161 PMCID: PMC6518424 DOI: 10.3390/jcm8040448] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/30/2019] [Accepted: 04/01/2019] [Indexed: 01/14/2023] Open
Abstract
Background: Diabetic foot syndrome (DFS) is a prevalent complication in the diabetic population and a major cause of hospitalizations. Diverse clinical studies have related alterations in the system formed by fibroblast growth factor (FGF)-23 and Klotho (KL) with vascular damage. In this proof-of-concept study, we hypothesize that the levels of FGF23 and Klotho are altered in DFS patients. Methods: Twenty patients with limb amputation due to DFS, 37 diabetic patients without DFS, and 12 non-diabetic cadaveric organ donors were included in the study. Serum FGF23/Klotho and inflammatory markers were measured by enzyme-linked immunosorbent assay (ELISA). Protein and gene expression levels in the vascular samples were determined by immunohistochemistry and quantitative real-time PCR, respectively. Results: Serum Klotho is significantly reduced and FGF23 is significantly increased in patients with DFS (p < 0.01). Vascular immunoreactivity and gene expression levels for Klotho were decreased in patients with DFS (p < 0.01). Soluble Klotho was inversely related to serum C-reactive protein (r = −0.30, p < 0.05). Vascular immunoreactivities for Klotho and IL6 showed an inverse association (r = −0.29, p < 0.04). Similarly, vascular gene expression of KL and IL6 were inversely associated (r = −0.31, p < 0.05). Logistic regression analysis showed that higher Klotho serum concentrations and vascular gene expression levels were related to a lower risk of DFS, while higher serum FGF23 was associated with a higher risk for this complication. Conclusion: FGF23/Klotho system is associated with DFS, pointing to a new pathophysiological pathway involved in the development and progression of this complication.
Collapse
Affiliation(s)
- Javier Donate-Correa
- Research Unit, University Hospital Nuestra Señora de Candelaria (UHNSC), 38010 Santa Cruz de Tenerife, Spain.
| | - Ernesto Martín-Núñez
- Research Unit, University Hospital Nuestra Señora de Candelaria (UHNSC), 38010 Santa Cruz de Tenerife, Spain.
- Doctoral and Graduate School, University of La Laguna, 38200 San Cristóbal de La Laguna, Spain.
| | - Carla Ferri
- Research Unit, University Hospital Nuestra Señora de Candelaria (UHNSC), 38010 Santa Cruz de Tenerife, Spain.
- Doctoral and Graduate School, University of La Laguna, 38200 San Cristóbal de La Laguna, Spain.
| | - Carolina Hernández-Carballo
- Research Unit, University Hospital Nuestra Señora de Candelaria (UHNSC), 38010 Santa Cruz de Tenerife, Spain.
- Doctoral and Graduate School, University of La Laguna, 38200 San Cristóbal de La Laguna, Spain.
| | - Víctor G Tagua
- Research Unit, University Hospital Nuestra Señora de Candelaria (UHNSC), 38010 Santa Cruz de Tenerife, Spain.
| | - Alejandro Delgado-Molinos
- Vascular Surgery Service, University Hospital Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain.
| | - Ángel López-Castillo
- Vascular Surgery Service, University Hospital Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain.
| | - Sergio Rodríguez-Ramos
- Transplant Coordination, University Hospital Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain.
| | - Purificación Cerro-López
- Transplant Coordination, University Hospital Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain.
| | | | | | - Nayra Pérez-Delgado
- Clinical Analysis Service, University Hospital Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain.
| | - Carmen Mora-Fernández
- Research Unit, University Hospital Nuestra Señora de Candelaria (UHNSC), 38010 Santa Cruz de Tenerife, Spain.
| | - Juan F Navarro-González
- Research Unit, University Hospital Nuestra Señora de Candelaria (UHNSC), 38010 Santa Cruz de Tenerife, Spain.
- Nephrology Service, University Hospital Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain.
- Institute of Biomedical Technologies, University of La Laguna, 38200 San Cristóbal de La Laguna, Spain.
| |
Collapse
|
55
|
Kim HJ, Lee J, Chae DW, Lee KB, Sung SA, Yoo TH, Han SH, Ahn C, Oh KH. Serum klotho is inversely associated with metabolic syndrome in chronic kidney disease: results from the KNOW-CKD study. BMC Nephrol 2019; 20:119. [PMID: 30943913 PMCID: PMC6446407 DOI: 10.1186/s12882-019-1297-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 03/17/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Metabolic syndrome (MS) is prevalent in chronic kidney disease (CKD). Klotho, a protein linked to aging, is closely associated with CKD. Each component of MS and klotho has an association. However, little is known about the association between klotho and MS per se. We investigated the association between serum klotho levels and MS using baseline cross-sectional data obtained from a large Korean CKD cohort. METHODS Of the 2238 subjects recruited in the KoreaN Cohort Study for Outcome in Patients With Chronic Kidney Disease (KNOW-CKD) between 2011 and 2016, 484 patients with missing data on serum klotho and extreme klotho values (values lower than the detectable range or > 6000 pg/mL) or with autosomal dominant polycystic kidney disease patients were excluded. The data of the remaining 1754 subjects were included in the present study. MS was defined using the revised National Cholesterol Education Program Adult Treatment Panel (NCEP-ATP) III criteria. Serum klotho levels were measured using an enzyme-linked immunosorbent assay. RESULTS Mean patient age was 54.9 ± 12.1 years and 1110 (63.3%) were male. The prevalence of MS among all study subjects was 63.7% (n = 1118). The median serum klotho level was 527 pg/mL (interquartile range [IQR]: 418-656 pg/mL). Serum klotho level was significantly lower in MS patients than patients without MS (Median [IQR]; 521 pg/mL [413, 651] vs. 541 pg/mL [427, 676], respectively; P = 0.012). After adjusting for age, sex, estimated glomerular filtration rate, and overt proteinuria, serum klotho was independently associated with MS (adjusted odds ratio [OR], 0.44; 95% confidence interval, 0.23-0.82; P = 0.010). Furthermore, the adjusted OR for MS was found to be significantly increased at serum klotho levels of < 518 pg/mL (receiver operating characteristic curve cut-off value). CONCLUSIONS Serum klotho was inversely associated with the presence of MS in patients with CKD. TRIAL REGISTRATION This trial was registered on ClinicalTrials.gov on 26 June 2012 ( https://clinicaltrials.gov;NCT01630486 ).
Collapse
Affiliation(s)
- Hyo Jin Kim
- Department of Internal Medicine, Dongguk University College of Medicine, Gyeongju-si, Gyeongsangbuk-do, Korea
| | - Joongyub Lee
- School of Medicine, Inha University, Incheon, Korea.,Department of Prevention and Management, Inha University Hospital, Incheon, Korea
| | - Dong-Wan Chae
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnamsi, Gyeonggi-do, Korea
| | - Kyu-Beck Lee
- Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Su Ah Sung
- Department of Internal Medicine, Nowon Eulji Medical Center, Eulji University, Seoul, Korea
| | - Tae-Hyun Yoo
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Seung Hyeok Han
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Curie Ahn
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-Ro, Jongno-Gu, Seoul, 03080, Korea
| | - Kook-Hwan Oh
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-Ro, Jongno-Gu, Seoul, 03080, Korea.
| |
Collapse
|
56
|
Adema AY, de Roij van Zuijdewijn CLM, Hoenderop JG, de Borst MH, Ter Wee PM, Heijboer AC, Vervloet MG. Influence of exogenous growth hormone administration on circulating concentrations of α-klotho in healthy and chronic kidney disease subjects: a prospective, single-center open case-control pilot study. BMC Nephrol 2018; 19:327. [PMID: 30442108 PMCID: PMC6238285 DOI: 10.1186/s12882-018-1114-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 10/22/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The CKD-associated decline in soluble α-Klotho (α-Klotho) levels is considered detrimental. Some studies suggest a direct induction of α-Klotho concentrations by growth hormone (GH). In the present study, the effect of exogenous GH administration on α-Klotho concentrations in a clinical cohort with mild chronic kidney disease (CKD) and healthy subjects was studied. METHODS A prospective, single-center open case-control pilot study was performed involving 8 patients with mild CKD and 8 healthy controls matched for age and sex. All participants received subcutaneous GH injections (Genotropin®, 20 mcg/kg/day) for 7 consecutive days. α-Klotho concentrations were measured at baseline, after 7 days of therapy and 1 week after the intervention was stopped. RESULTS α-Klotho concentrations were not different between CKD-patients and healthy controls at baseline (554 (388-659) vs. 547 (421-711) pg/mL, P = 0.38). Overall, GH therapy increased α-Klotho concentrations from 554 (405-659) to 645 (516-754) pg/mL, P < 0.05). This was accompanied by an increase of IGF-1 concentrations from 26.8 ± 5.0 nmol/L to 61.7 ± 17.7 nmol/L (P < 0.05). GH therapy induced a trend toward increased α-Klotho concentrations both in the CKD group (554 (388-659) to 591 (358-742) pg/mL (P = 0.19)) and the healthy controls (547 (421-711) pg/mL to 654 (538-754) pg/mL (P = 0.13)). The change in α-Klotho concentration was not different for both groups (P for interaction = 0.71). α-Klotho concentrations returned to baseline levels within one week after the treatment (P < 0.05). CONCLUSIONS GH therapy increases α-Klotho concentrations in subjects with normal renal function or stage 3 CKD. A larger follow-up study is needed to determine whether the effect size is different between both groups or in patients with more severe CKD. TRIAL REGISTRATION This trial is registered in EudraCT ( 2013-003354-24 ).
Collapse
Affiliation(s)
- Aaltje Y Adema
- Department of Nephrology, VU University Medical Center, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands
| | | | - Joost G Hoenderop
- Department of Physiology, Radboud University Medical Center Nijmegen, Nijmegen, The Netherlands
| | - Martin H de Borst
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, Groningen, The Netherlands
| | - Piet M Ter Wee
- Department of Nephrology, VU University Medical Center, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands
| | - Annemieke C Heijboer
- Department of Clinical Chemistry, VU University Medical Center, Amsterdam, The Netherlands
| | - Marc G Vervloet
- Department of Nephrology, VU University Medical Center, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands. .,Amsterdam Cardiovascular Sciences (ACS), Amsterdam, The Netherlands.
| | | |
Collapse
|
57
|
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
|
58
|
Bahrami LS, Sezavar Seyedi Jandaghi SH, Janani L, Pahlavan M, Arabi SM, Sadeghi H, Vafa M. Vitamin D supplementation and serum heat shock protein 60 levels in patients with coronary heart disease: a randomized clinical trial. Nutr Metab (Lond) 2018; 15:56. [PMID: 30093913 PMCID: PMC6080550 DOI: 10.1186/s12986-018-0292-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 07/24/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The aim in this study was to investigate the effect of vitamin D (25(OH)D3) supplementation on heat shock protein 60 (HSP 60) and other inflammatory markers (IL-17, TNF-α, PAB) in patients with coronary heart disease (CHD). METHODS In this double-blind, randomized clinical trial, we recruited 80 male and female patients aged 30-60 with CHD and 25(OH)D3 serum levels < 30 ng/ml from Rasool-e-Akram Hospital in Tehran, Iran. Serum levels of HSP 60 as primary outcome, and 25(OH)D3, IL-17, TNF-α, PAB, lipid profiles and parathyroid hormone (PTH) as secondary outcomes were measured at baseline and post-intervention. We randomly assigned eligible participants to a placebo group (N = 40) or an intervention group (N = 40) (50,000 IU/wk. vitamin D supplement) for eight weeks. RESULTS The results demonstrated that vitamin D supplementation resulted in a significant increase in 25(OH) D3 serum levels in the intervention group compared to the placebo group (46.86 vs. 7.28 ng/ml). PTH levels decreased in the intervention group compared to the placebo group (- 19.81 vs. 2.92 pg/ml) after eight weeks of supplementation. Furthermore, we observed a significant change in waist circumference (- 0.97 vs. -0.26 cm), fat percentage (-.13 vs. 0.1%), systolic blood pressure (- 3.85 vs. -2.11 mmHg) and diastolic blood presure (- 4 vs. -1.86 mmHg) in the vitamin D group compared to the placebo group (all P values < 0.05). Other variables did not significantly change after the intervention. CONCLUSION Based on our findings, weekly vitamin D supplementation of 50,000 IU for eight weeks in patients with CHD resulted in decreased systolic and diastolic blood pressure, waist circumference and fat percentage. No significant effect on HSP 60, inflammatory markers or lipid profiles was observed. TRIAL REGISTRATION IRCT, IRCT201612122365N14. Registered 12 December 2016.
Collapse
Affiliation(s)
- Leila Sadat Bahrami
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | | | - Leila Janani
- Department of Biostatistics, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Mina Pahlavan
- Department of Medical Surgical Nursing, Nursing and Midwifery School, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Mostafa Arabi
- Department of Nutrition, Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Homa Sadeghi
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Mohammadreza Vafa
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
59
|
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: 4.7] [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
|
60
|
Chuchana P, Mausset-Bonnefont AL, Mathieu M, Espinoza F, Teigell M, Toupet K, Ripoll C, Djouad F, Noel D, Jorgensen C, Brondello JM. Secreted α-Klotho maintains cartilage tissue homeostasis by repressing NOS2 and ZIP8-MMP13 catabolic axis. Aging (Albany NY) 2018; 10:1442-1453. [PMID: 29920476 PMCID: PMC6046234 DOI: 10.18632/aging.101481] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 06/13/2018] [Indexed: 04/24/2023]
Abstract
Progressive loss of tissue homeostasis is a hallmark of numerous age-related pathologies, including osteoarthritis (OA). Accumulation of senescent chondrocytes in joints contributes to the age-dependent cartilage loss of functions through the production of hypertrophy-associated catabolic matrix-remodeling enzymes and pro-inflammatory cytokines. Here, we evaluated the effects of the secreted variant of the anti-aging hormone α-Klotho on cartilage homeostasis during both cartilage formation and OA development. First, we found that α-Klotho expression was detected during mouse limb development, and transiently expressed during in vitro chondrogenic differentiation of bone marrow-derived mesenchymal stem cells. Genome-wide gene array analysis of chondrocytes from OA patients revealed that incubation with recombinant secreted α-Klotho repressed expression of the NOS2 and ZIP8/MMP13 catabolic remodeling axis. Accordingly, α-Klotho expression was reduced in chronically IL1β-treated chondrocytes and in cartilage of an OA mouse model. Finally, in vivo intra-articular secreted α-Kotho gene transfer delays cartilage degradation in the OA mouse model. Altogether, our results reveal a new tissue homeostatic function for this anti-aging hormone in protecting against OA onset and progression.
Collapse
Affiliation(s)
- Paul Chuchana
- IRMB, INSERM, Montpellier University, Montpellier, France
| | | | - Marc Mathieu
- IRMB, INSERM, Montpellier University, Montpellier, France
| | | | | | - Karine Toupet
- IRMB, INSERM, Montpellier University, Montpellier, France
| | | | - Farida Djouad
- IRMB, INSERM, Montpellier University, Montpellier, France
| | - Danièle Noel
- IRMB, INSERM, Montpellier University, Montpellier, France
| | - Christian Jorgensen
- IRMB, INSERM, Montpellier University, Montpellier, France
- CHU Montpellier, Montpellier, France
| | | |
Collapse
|
61
|
Kim HJ, Kang E, Oh YK, Kim YH, Han SH, Yoo TH, Chae DW, Lee J, Ahn C, Oh KH. The association between soluble klotho and cardiovascular parameters in chronic kidney disease: results from the KNOW-CKD study. BMC Nephrol 2018; 19:51. [PMID: 29506503 PMCID: PMC5838864 DOI: 10.1186/s12882-018-0851-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Accepted: 02/20/2018] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Klotho, a protein linked to aging, has emerged as a pivotal player in mineral bone metabolism and might explain the relationship between chronic kidney disease (CKD) and cardiovascular disease (CVD). The present study aimed to investigate the association between serum klotho and cardiac parameters from a large-scale Korean CKD cohort. METHODS We analyzed 2101 participants from KoreaN Cohort Study for Outcome in Patients With Chronic Kidney Disease (KNOW-CKD) cohort who had been measured for serum klotho levels. Left ventricular hypertrophy evaluated by left ventricular mass index (LVMI) and arterial stiffness measured by brachial-to-ankle pulse wave velocity (baPWV) were explored as cardiovascular parameters. RESULTS Patients were 53.6 ± 12.2 years old and 61.1% were male. The mean estimated glomerular filtration rate (eGFR) was 53.0 ± 30.7 mL/min/1.73m2. The median serum klotho level was 536 (interquartile range [IQR]: 420-667) pg/mL. Advanced CKD stages were associated with lower serum klotho levels (P < 0.001, P for linear trend < 0.001). Ascending quartiles of klotho were significantly associated with decreased LMVI (P < 0.001, P for linear trend< 0.001). A multivariable linear regression model showed serum klotho had a significant inverse association with LVMI (β - 0.04; 95% CI [confidence interval] -0.004, - 0.00007; P = 0.041). However, there was no significant association between serum klotho and baPWV after adjustment (β 0.003; 95% CI -0.04, 0.05; P = 0.876). TRIAL REGISTRATION This trial was registered on ClinicalTrials.gov on 28 June 2012 ( NCT01630486 ). CONCLUSIONS Serum klotho was an independent biomarker of LVMI, but not arterial stiffness.
Collapse
Affiliation(s)
- Hyo Jin Kim
- Department of Internal Medicine, Dongguk University College of Medicine, Gyeongju-si, Gyeongsangbuk-do, South Korea
| | - Eunjeong Kang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Yun Kyu Oh
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Yeong Hoon Kim
- Department of Internal Medicine, Inje University, Busan Paik Hospital, Busan, South Korea
| | - Seung Hyeok Han
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Tae Hyun Yoo
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Dong-Wan Chae
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Joongyub Lee
- Medical Research Collaborating Center, Seoul National University Hospital and Seoul, National University College of Medicine, Seoul, South Korea
| | - Curie Ahn
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Kook-Hwan Oh
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
62
|
Caicedo D, Díaz O, Devesa P, Devesa J. Growth Hormone (GH) and Cardiovascular System. Int J Mol Sci 2018; 19:ijms19010290. [PMID: 29346331 PMCID: PMC5796235 DOI: 10.3390/ijms19010290] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 01/08/2018] [Accepted: 01/12/2018] [Indexed: 01/02/2023] Open
Abstract
This review describes the positive effects of growth hormone (GH) on the cardiovascular system. We analyze why the vascular endothelium is a real internal secretion gland, whose inflammation is the first step for developing atherosclerosis, as well as the mechanisms by which GH acts on vessels improving oxidative stress imbalance and endothelial dysfunction. We also report how GH acts on coronary arterial disease and heart failure, and on peripheral arterial disease, inducing a neovascularization process that finally increases flow in ischemic tissues. We include some preliminary data from a trial in which GH or placebo is given to elderly people suffering from critical limb ischemia, showing some of the benefits of the hormone on plasma markers of inflammation, and the safety of GH administration during short periods of time, even in diabetic patients. We also analyze how Klotho is strongly related to GH, inducing, after being released from the damaged vascular endothelium, the pituitary secretion of GH, most likely to repair the injury in the ischemic tissues. We also show how GH can help during wound healing by increasing the blood flow and some neurotrophic and growth factors. In summary, we postulate that short-term GH administration could be useful to treat cardiovascular diseases.
Collapse
Affiliation(s)
- Diego Caicedo
- Department of Angiology and Vascular Surgery, Complejo Hospitalario Universitario de Pontevedra, 36701 Pontevedra, Spain.
| | - Oscar Díaz
- Department of Cardiology, Complejo Hospitalario Universitario de Pontevedra, 36701 Pontevedra, Spain.
| | - Pablo Devesa
- Research and Development, The Medical Center Foltra, 15886 Teo, Spain.
| | - Jesús Devesa
- Scientific Direction, The Medical Center Foltra, 15886 Teo, Spain.
| |
Collapse
|
63
|
Gazdhar A, Ravikumar P, Pastor J, Heller M, Ye J, Zhang J, Moe OW, Geiser T, Hsia CCW. Alpha-Klotho Enrichment in Induced Pluripotent Stem Cell Secretome Contributes to Antioxidative Protection in Acute Lung Injury. Stem Cells 2017; 36:616-625. [PMID: 29226550 DOI: 10.1002/stem.2752] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 11/07/2017] [Accepted: 11/21/2017] [Indexed: 02/06/2023]
Abstract
Induced pluripotent stem cells (iPSCs) have been reported to alleviate organ injury, although the mechanisms of action remain unclear and administration of intact cells faces many limitations. We hypothesized that cell-free conditioned media (CM) containing the secretome of iPSCs possess antioxidative constituents that can alleviate pulmonary oxidant stress damage. We derived iPSCs from human dermal fibroblasts and harvested the CM. Addition of iPSC CM to cultured human alveolar type-1 epithelial cells mitigated hyperoxia-induced depletion of endogenous total antioxidant capacity while tracheal instillation of iPSC CM into adult rat lungs enhanced hyperoxia-induced increase in TAC. In both the in vitro and in vivo models, iPSC CM ameliorated oxidative damage to DNA, lipid, and protein, and activated the nuclear factor (erythroid 2)-related factor 2 (Nrf2) network of endogenous antioxidant proteins. Compared with control fibroblast-conditioned or cell-free media, iPSC CM is highly enriched with αKlotho at a concentration up to more than 10-fold of that in normal serum. αKlotho is an essential antioxidative cell maintenance and protective factor and an activator of the Nrf2 network. Immunodepletion of αKlotho reduced iPSC CM-mediated cytoprotection by ∼50%. Thus, the abundant αKlotho content significantly contributes to iPSC-mediated antioxidation and cytoprotection. Results uncover a major mechanism of iPSC action, suggest a fundamental role of αKlotho in iPSC maintenance, and support the translational potential of airway delivery of cell-free iPSC secretome for protection against lung injury. The targeted cell-free secretome-based approach may also be applicable to the amelioration of injury in other organs. Stem Cells 2018;36:616-625.
Collapse
Affiliation(s)
- Amiq Gazdhar
- Department of Pulmonary Medicine, University Hospital, Bern, Switzerland.,Department of Clinical Research, University Hospital, Bern, Switzerland
| | - Priya Ravikumar
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Johanne Pastor
- Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Manfred Heller
- Department of Clinical Research, University Hospital, Bern, Switzerland
| | - Jianfeng Ye
- Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jianning Zhang
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Orson W Moe
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Thomas Geiser
- Department of Pulmonary Medicine, University Hospital, Bern, Switzerland.,Department of Clinical Research, University Hospital, Bern, Switzerland
| | - Connie C W Hsia
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
64
|
Soluble levels and endogenous vascular gene expression of KLOTHO are related to inflammation in human atherosclerotic disease. Clin Sci (Lond) 2017; 131:2601-2609. [PMID: 28963437 DOI: 10.1042/cs20171242] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 09/13/2017] [Accepted: 09/25/2017] [Indexed: 01/29/2023]
Abstract
Atherosclerosis is a chronic inflammatory disorder affecting the artery wall. Klotho, an anti-aging factor expressed in the vessel walls that participates in the maintenance of vascular homeostasis, can be down-regulated by inflammation. In this proof-of-concept work we seek to characterize the arterial KLOTHO expression in the vascular wall, as well as the serum concentration of this protein, in a group of patients with clinical atherosclerotic disease. In addition, we aim to analyze the relationship between Klotho and inflammation. Vascular samples were obtained from 27 patients with atherosclerotic disease under an elective vascular surgery procedure, and from 11 control subjects (cadaveric organ donation programme). qRT-PCR was performed to analyze the gene expression of KLOTHO, TNF-α, IL-6, and IL-10 Serum levels of soluble KLOTHO were measured by ELISA. As compared with control subjects, serum concentrations and vascular expression of Klotho were lower in patients with atherosclerotic vascular disease, whereas inflammatory status was significantly higher. There was a negative and significant correlation between inflammatory parameters and Klotho. After controlling for the effect of other variables, partial correlation showed a direct relationship between vascular KLOTHO gene expression and IL-10 mRNA levels, whereas there was a negative association with serum LDL concentrations and vascular TNF-α expression. Our study indicates an inverse interrelationship between inflammation and Klotho in atherosclerosis. Further studies are necessary to elucidate whether the inflammatory state causes Klotho deficiency or, on the contrary, reduction of Klotho could be responsible for greater inflammation, and finally, to investigate the potential clinical relevance of this association.
Collapse
|
65
|
Abstract
PURPOSE Klotho is a newly discovered protein that presumably has an important role in the aging process. The goal of this research is to compare the levels of Klotho and Endothelin-1 (ET-1) in the serum and aqueous humor of patients with pseudoexfoliation syndrome (PES) and pseudoexfoliative glaucoma (PEG). PATIENTS AND METHODS Aqueous humor and serum samples were obtained at the time of cataract surgery from 15 patients with PES, 15 patients with PEG, and 15 control patients. All of the samples were analyzed using enzyme-linked immunosorbent assay to evaluate the levels of ET-1 and Klotho protein. RESULT Aqueous and serum levels of Klotho in PES patients (49.02±10.97, 56.32±10.25 ng/mL) and PEG patients (34.53±4.87, 50.49±2.63 ng/mL) were lower than in control patients (56.31±7.68, 65.06±12.32 ng/mL). Both aqueous and serum levels of Klotho in the PEG group were lower than in the PES group (P=0.001). Mean aqueous and serum levels of ET-1 in the PES (1.28±0.09, 1.65±0.75 pg/mL) and the PEG groups (1.45±0.07, 1.58±0.58 pg/mL) were significantly higher than that measured in the control group (1.17±0.09, 1.16±0.34 pg/mL). Aqueous levels of ET-1 in the PEG group were higher than the PES group (P=0.04), but there were no significant difference in serum levels of ET-1 between the PES and the PEG groups (P=0.83). CONCLUSIONS Aqueous and serum levels of Klotho decreased both in PES and PEG patients, and this decrease in the PEG group was more significant. In contrast, aqueous and serum levels of ET-1 increased in the PES and the PEG patients, and the increase in the aqueous level of ET-1 in PEG patients was more significant.
Collapse
|
66
|
Huang C, Liu L, You Z, Zhao Y, Dong J, Du Y, Ogawa R. Endothelial dysfunction and mechanobiology in pathological cutaneous scarring: lessons learned from soft tissue fibrosis. Br J Dermatol 2017; 177:1248-1255. [PMID: 28403507 DOI: 10.1111/bjd.15576] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2017] [Indexed: 12/13/2022]
Affiliation(s)
- C. Huang
- Department of Dermatology Beijing Tsinghua Changgung Hospital Tsinghua University Beijing 102218 China
- Department of Plastic Surgery Meitan General Hospital Beijing 100028 China
| | - L. Liu
- Department of Biomedical Engineering School of Medicine Tsinghua University Beijing 100084 China
| | - Z. You
- Department of Biomedical Engineering School of Medicine Tsinghua University Beijing 100084 China
| | - Y. Zhao
- Department of Dermatology Beijing Tsinghua Changgung Hospital Tsinghua University Beijing 102218 China
| | - J. Dong
- Department of Hepatobiliary Surgery Beijing Tsinghua Changgung Hospital Tsinghua University Beijing 102218 China
| | - Y. Du
- Department of Biomedical Engineering School of Medicine Tsinghua University Beijing 100084 China
| | - R. Ogawa
- Department of Plastic, Reconstructive and Aesthetic Surgery Nippon Medical School Tokyo 113‐8603 Japan
| |
Collapse
|
67
|
Wu J, Zhao YM, Deng ZK. Tangeretin ameliorates renal failure via regulating oxidative stress, NF-κB–TNF-α/iNOS signalling and improves memory and cognitive deficits in 5/6 nephrectomized rats. Inflammopharmacology 2017; 26:119-132. [DOI: 10.1007/s10787-017-0394-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 08/23/2017] [Indexed: 12/20/2022]
|
68
|
Endothelin-1, α-Klotho, 25(OH) Vit D levels and severity of disease in scleroderma patients. Rheumatol Int 2017; 37:1651-1657. [PMID: 28831601 DOI: 10.1007/s00296-017-3797-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 08/17/2017] [Indexed: 10/19/2022]
Abstract
Considering the role of endothelin-1 (ET-1) in tissue remodeling and fibrosis during the development of scleroderma as well as the effect of α-Klotho in pathogenesis of calcinosis and/or endothelial cell injury and its correlation with severity of disease, this study aimed to evaluate serum ET-1, α-Klotho and 25(OH) vitamin D levels in patients with limited and diffuse scleroderma compared to healthy subjects. In this cross-sectional study, 60 scleroderma patients according to the ACR/EULAR 2013 criteria and 60 age- and sex-matched healthy controls were included. In patients, clinical examination was performed and Medsger severity scale was assessed. Serum ET-1, soluble α-Klotho and 25(OH)D3 levels were measured using ELISA kits. The mean ± SD age of patients and controls was 46.2 ± 9.6 and 47.2 ± 7.0 years, respectively. Compared to healthy controls, serum ET-1 was significantly higher in SSc patients (p = 0.001); whilst serum α-Klotho and 25(OH)D3 were significantly lower in patients (p = 0.001). The most common organs involved in patients were skin, lung, peripheral vascular and gastrointestinal system and the severity of involvement was mainly mild and/or moderate. There were no significant differences in serum ET-1 and α-Klotho levels according to the severity of different organ involvement (p > 0.05). There was no significant correlation between presence or absence of calcinosis and negative or positivity of auto-antibodies with ET-1, α-Klotho and 25(OH)D3 levels. Although our study revealed higher serum ET-1 and lower serum α-Klotho levels in SSc patients compared to healthy controls, there were not any significant correlations between their serum levels with severity of organ involvement.
Collapse
|
69
|
Neyra JA, Hu MC. Potential application of klotho in human chronic kidney disease. Bone 2017; 100:41-49. [PMID: 28115282 PMCID: PMC5474175 DOI: 10.1016/j.bone.2017.01.017] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 01/18/2017] [Accepted: 01/18/2017] [Indexed: 01/13/2023]
Abstract
The extracellular domain of transmembrane alpha-Klotho (αKlotho, hereinafter simply called Klotho) is cleaved by secretases and released into the circulation as soluble Klotho. Soluble Klotho in the circulation starts to decline early in chronic kidney disease (CKD) stage 2 and urinary Klotho possibly even earlier in CKD stage 1. Therefore soluble Klotho could serve as an early and sensitive marker of kidney function decline. Moreover, preclinical animal data support Klotho deficiency is not just merely a biomarker, but a pathogenic factor for CKD progression and extrarenal CKD complications including cardiovascular disease and disturbed mineral metabolism. Prevention of Klotho decline, re-activation of endogenous Klotho production or supplementation of exogenous Klotho are all associated with attenuation of renal fibrosis, retardation of CKD progression, improvement of mineral metabolism, amelioration of cardiomyopathy, and alleviation of vascular calcification in CKD. Therefore Klotho is not only a diagnostic and/or prognostic marker for CKD, but the treatment of Klotho deficiency may be a promising strategy to prevent, retard, and decrease the burden of comorbidity in CKD.
Collapse
Affiliation(s)
- Javier A Neyra
- Department of Internal Medicine, University of Texas Southwestern Medical Center, USA; Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, USA
| | - Ming Chang Hu
- Department of Internal Medicine, University of Texas Southwestern Medical Center, USA; Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, USA.
| |
Collapse
|
70
|
Hsia CCW, Ravikumar P, Ye J. Acute lung injury complicating acute kidney injury: A model of endogenous αKlotho deficiency and distant organ dysfunction. Bone 2017; 100:100-109. [PMID: 28347910 PMCID: PMC5621379 DOI: 10.1016/j.bone.2017.03.047] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 03/23/2017] [Accepted: 03/23/2017] [Indexed: 12/11/2022]
Abstract
The lung interfaces with atmospheric oxygen via a large surface area and is perfused by the entire venous return bearing waste products collected from the whole body. It is logical that the lung is endowed with generous anti-oxidative capacity derived both locally and from the circulation. The single-pass pleiotropic alpha-Klotho (αKlotho) protein was discovered when its genetic disruption led to premature multi-organ degeneration and early death. The extracellular domain of αKlotho is cleaved by secretases and released into circulation as endocrine soluble αKlotho protein, exerting wide-ranging cytoprotective effects including anti-oxidation on distant organs including the lung, which exhibits high sensitivity to circulating αKlotho insufficiency. Because circulating αKlotho is derived mainly from the kidney, acute kidney injury (AKI) leads to systemic αKlotho deficiency that in turn increases the risks of pulmonary complications, i.e., edema and inflammation, culminating in the acute respiratory distress syndrome. Exogenous αKlotho increases endogenous anti-oxidative capacity partly via activation of the Nrf2 pathway to protect lungs against injury caused by direct hyperoxia exposure or AKI. This article reviews the current knowledge of αKlotho antioxidation in the lung in the setting of AKI as a model of circulating αKlotho deficiency, an under-recognized condition that weakens innate cytoprotective defenses and contributes to the dysfunction in distant organs.
Collapse
Affiliation(s)
- Connie C W Hsia
- Department of Internal Medicine, Pulmonary and Critical Care Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-9034, United States of America.
| | - Priya Ravikumar
- Department of Internal Medicine, Pulmonary and Critical Care Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-9034, United States of America; Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX 75390-9034, United States of America
| | - Jianfeng Ye
- Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX 75390-9034, United States of America
| |
Collapse
|
71
|
Abstract
Epidemiological studies have shown that ageing is a major non-reversible risk factor for cardiovascular disease. Vascular ageing starts early in life and is characterized by a gradual change of vascular structure and function resulting in increased arterial stiffening. At the present review we discuss the role of the most important molecular pathways involved in vascular ageing, their association with arterial stiffening and possible novel therapeutic targets that may delay this otherwise irreversible degenerating process. Specifically, we discuss the role of oxidative stress, telomere shortening, and ubiquitin proteasome system in endothelial cell senescence and dysfunction in vascular inflammation and in arterial stiffening. Further, we summarize the most important molecular mechanisms regulating vascular ageing including sirtuin 1, telomerase, klotho, JunD, and amyloid beta 1-40 peptide.
Collapse
Affiliation(s)
- Ageliki Laina
- Department of Clinical Therapeutics, Alexandra Hospital, University of Athens, Athens, Greece
| | - Konstantinos Stellos
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine, Goethe University Frankfurt, Frankfurt, Germany; Department of Cardiology, Center of Internal Medicine, Goethe University Frankfurt, Frankfurt, Germany; German Center of Cardiovascular Research (Deutsches Zentrum für Herz-Kreislaufforschung; DZHK), Rhein-Main Partner Site, Frankfurt, Germany
| | - Kimon Stamatelopoulos
- Department of Clinical Therapeutics, Alexandra Hospital, University of Athens, Athens, Greece.
| |
Collapse
|
72
|
α-Klotho expression determines nitric oxide synthesis in response to FGF-23 in human aortic endothelial cells. PLoS One 2017; 12:e0176817. [PMID: 28463984 PMCID: PMC5413063 DOI: 10.1371/journal.pone.0176817] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 04/18/2017] [Indexed: 01/03/2023] Open
Abstract
Endothelial cells (ECs) express fibroblast growth factor (FGF) receptors and are metabolically active after treatment with FGF-23. It is not known if this effect is α-Klotho independent or mediated by humoral or endogenous endothelial α-Klotho. In the present study, we aimed to characterize EC α-Klotho expression within the human vascular tree and to investigate the potential role of α-Klotho in determining FGF-23 mediated EC regulation. Human tissue and ECs from various organs were used for immunohistochemistry and Western blot. Primary cultures of human aortic endothelial cells (HAECs) and human brain microvascular endothelial cells (HBMECs) were used to generate in vitro cell models. We found endogenous α-Klotho expression in ECs from various organs except in microvascular ECs from human brain. Furthermore, FGF-23 stimulated endothelial nitric oxide synthase (eNOS) expression, nitric oxide (NO) production, and cell proliferation in HAECs. Interestingly, these effects were not observed in our HBMEC model in vitro. High phosphate treatment and endothelial α-Klotho knockdown mitigated FGF-23 mediated eNOS induction, NO production, and cell proliferation in HAECs. Rescue treatment with soluble α-Klotho did not reverse endothelial FGF-23 resistance caused by reduced or absent α-Klotho expression in HAECs. These novel observations provide evidence for differential α-Klotho functional expression in the human endothelium and its presence may play a role in determining the response to FGF-23 in the vascular tree. α-Klotho was not detected in cerebral microvascular ECs and its absence may render these cells nonresponsive to FGF-23.
Collapse
|
73
|
Abstract
Chronic kidney disease (CKD) is associated with an increased risk of cardiovascular mortality, infections, and impaired cognitive function. It is characterized by excessively increased levels of the phosphaturic hormone fibroblast growth factor 23 (FGF23) and a deficiency of its co-receptor Klotho. Despite the important physiological effect of FGF23 in maintaining phosphate homeostasis, there is increasing evidence that higher FGF23 levels are a risk factor for mortality and cardiovascular disease. FGF23 directly induces left ventricular hypertrophy via activation of the FGF receptor 4/calcineurin/nuclear factor of activated T cells signaling pathway. By contrast, the impact of FGF23 on endothelial function and the development of atherosclerosis are poorly understood. The results of recent experimental studies indicate that FGF23 directly impacts on hippocampal neurons and may thereby impair learning and memory function in CKD patients. Finally, it has been shown that FGF23 interferes with the immune system by directly acting on polymorphonuclear leukocytes and macrophages. In this review, we discuss recent data from clinical and experimental studies on the extrarenal effects of FGF23 with respect to the cardiovascular, central nervous, and immune systems.
Collapse
|
74
|
Mencke R, Hillebrands JL. The role of the anti-ageing protein Klotho in vascular physiology and pathophysiology. Ageing Res Rev 2017; 35:124-146. [PMID: 27693241 DOI: 10.1016/j.arr.2016.09.001] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 09/23/2016] [Indexed: 02/08/2023]
Abstract
Klotho is an anti-ageing protein that functions in many pathways that govern ageing, like regulation of phosphate homeostasis, insulin signaling, and Wnt signaling. Klotho expression levels and levels in blood decline during ageing. The vascular phenotype of Klotho deficiency features medial calcification, intima hyperplasia, endothelial dysfunction, arterial stiffening, hypertension, and impaired angiogenesis and vasculogenesis, with characteristics similar to aged human arteries. Klotho-deficient phenotypes can be prevented and rescued by Klotho gene expression or protein supplementation. High phosphate levels are likely to be directly pathogenic and are a prerequisite for medial calcification, but more important determinants are pathways that regulate cellular senescence, suggesting that deficiency of Klotho renders cells susceptible to phosphate toxicity. Overexpression of Klotho is shown to ameliorate medial calcification, endothelial dysfunction, and hypertension. Endogenous vascular Klotho expression is a controversial subject and, currently, no compelling evidence exists that supports the existence of vascular membrane-bound Klotho expression, as expressed in kidney. In vitro, Klotho has been shown to decrease oxidative stress and apoptosis in both SMCs and ECs, to reduce SMC calcification, to maintain the contractile SMC phenotype, and to prevent μ-calpain overactivation in ECs. Klotho has many protective effects with regard to the vasculature and constitutes a very promising therapeutic target. The purpose of this review is to explore the etiology of the vascular phenotype of Klotho deficiency and the therapeutic potential of Klotho in vascular disease.
Collapse
|
75
|
Yao Y, Wang Y, Zhang Y, Liu C. Klotho ameliorates oxidized low density lipoprotein (ox-LDL)-induced oxidative stress via regulating LOX-1 and PI3K/Akt/eNOS pathways. Lipids Health Dis 2017; 16:77. [PMID: 28407763 PMCID: PMC5390438 DOI: 10.1186/s12944-017-0447-0] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 03/09/2017] [Indexed: 12/27/2022] Open
Abstract
Background Atherosclerosis is a common cardiovascular disease that causes myocardial infarction, heart failure, and stroke. Increased oxidized low density lipoprotein (ox-LDL) in the sub-endothelium is the characteristic origin of atherogenesis. Klotho, an anti-aging protein, has been reported to protect against atherosclerosis and ameliorate endothelial dysfunction in vivo. The aim of this study is to investigatethe anti-oxidative activity of Klothoin ox-LDL-treated human umbilical vein endothelial cells (HUVECs). Methods After pre-treatment with 200 pMKlotho for 1 h, HUVECs were stimulated with 50 μg/ml ox-LDL for 24 h. Reactive oxygen species (ROS) and superoxide dismutase (SOD) levels were analyzed in the cells. Nitric oxide (NO) concertation was measured in the medium supernatant. Related proteins or genes were detected with Western blot or real time PCR, respectively, in the cell lysates. Results Initially, oxidative damage in HUVECs was established by adding 50 μg/mL ox-LDL, which resulted in decreased cellular viability, SOD/Cu/Zn-SOD and endothelial NO synthase (eNOS) expression and NO production, as well as increased malondialdehyde (MDA) levels, ROS production, inducible NO synthase (iNOS), phosphatidyl inositol-3 kinase (PI3K), protein kinase B (Akt), gp91 phox, and lectin-like ox-LDL receptor (LOX-1) expression in HUVECs. Pre-incubation with recombinant Klotho (200 pM) significantly prevented all of these alterations. These results suggest that Klotho can attenuate ox-LDL-induced oxidative stress in HUVECs through upregulating oxidative scavengers (SOD and NO) viaactivating the PI3K/Akt/eNOS pathway and depressing LOX-1expression. Conclusions These results suggest that Klotho has a potential therapeutic effect on attenuating endothelial dysfunction and ameliorating atherosclerosis. Electronic supplementary material The online version of this article (doi:10.1186/s12944-017-0447-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yansheng Yao
- Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Yanbing Wang
- Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Yibo Zhang
- Department of Pathogenic Biology, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Chang Liu
- Department of Endocrinology, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China.
| |
Collapse
|
76
|
Mazzotta C, Manetti M, Rosa I, Romano E, Blagojevic J, Bellando-Randone S, Bruni C, Lepri G, Guiducci S, Ibba-Manneschi L, Matucci-Cerinic M. Proangiogenic effects of soluble α-Klotho on systemic sclerosis dermal microvascular endothelial cells. Arthritis Res Ther 2017; 19:27. [PMID: 28183357 PMCID: PMC5301388 DOI: 10.1186/s13075-017-1233-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 01/12/2017] [Indexed: 01/07/2023] Open
Abstract
Background Systemic sclerosis (SSc) is characterized by endothelial cell (EC) apoptosis, impaired angiogenesis and peripheral microvasculopathy. Soluble α-Klotho (sKl) is a pleiotropic molecule with multiple effects on ECs, including antioxidant and vasculoprotective activities. On the EC surface, sKl interacts with vascular endothelial growth factor (VEGF) receptor-2 (VEGFR-2) and transient receptor potential canonical-1 (TRPC-1) cation channel to control EC homeostasis. Here, we investigated whether sKl might act as a protective factor to improve angiogenesis in dermal microvascular endothelial cells (MVECs) from SSc patients (SSc-MVECs). Methods Wound healing assay was performed on healthy dermal MVECs (H-MVECs) challenged with sera from healthy controls or SSc patients with or without the addition of sKl. Capillary morphogenesis on Matrigel was assessed in H-MVECs and SSc-MVECs at basal conditions and treated with sKl, as well as in H-MVECs challenged with healthy or SSc sera in presence or absence of sKl. The expression of α-Klotho, VEGF165b, VEGFR-2, TRPC-1, Ki67 and active caspase-3 in H-MVECs and SSc-MVECs was investigated by western blotting. Immunostaining for α-Klotho was performed in H-MVECs and SSc-MVECs, and in healthy and SSc skin sections. Results Treatment with sKl effectively counteracted the inihibitory effects of SSc sera on wound healing ability and angiogenic performance of H-MVECs. The addition of sKl significantly improved angiogenesis and maintained over time capillary-like tube formation in vitro by SSc-MVECs. Stimulation of SSc-MVECs with sKl resulted in the upregulation of the proliferation marker Ki67 in parallel with the downregulation of proapoptotic active caspase-3. The expression of α-Klotho was significantly lower in SSc-MVECs than in H-MVECs. The expression of TRPC-1 was also significantly decreased, while that of VEGFR-2 and VEGF165b was significantly increased, in SSc-MVECs compared with H-MVECs. Challenge with sKl either significantly increased TRPC-1 or decreased VEGF165b in SSc-MVECs. Ex vivo analyses revealed that α-Klotho immunostaining was almost absent in the dermal microvascular network of SSc skin compared with control skin. Conclusions Our findings provide the first evidence that α-Klotho is significantly decreased in the microvasculature in SSc skin and that sKl administration may effectively improve SSc-MVEC functions in vitro by acting as a powerful proangiogenic factor.
Collapse
Affiliation(s)
- Celestina Mazzotta
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, AOUC, Largo Brambilla 3, 50134, Florence, Italy.
| | - Mirko Manetti
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Largo Brambilla 3, 50134, Florence, Italy
| | - Irene Rosa
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, AOUC, Largo Brambilla 3, 50134, Florence, Italy.,Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Largo Brambilla 3, 50134, Florence, Italy
| | - Eloisa Romano
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, AOUC, Largo Brambilla 3, 50134, Florence, Italy
| | - Jelena Blagojevic
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, AOUC, Largo Brambilla 3, 50134, Florence, Italy
| | - Silvia Bellando-Randone
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, AOUC, Largo Brambilla 3, 50134, Florence, Italy
| | - Cosimo Bruni
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, AOUC, Largo Brambilla 3, 50134, Florence, Italy
| | - Gemma Lepri
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, AOUC, Largo Brambilla 3, 50134, Florence, Italy
| | - Serena Guiducci
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, AOUC, Largo Brambilla 3, 50134, Florence, Italy
| | - Lidia Ibba-Manneschi
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Largo Brambilla 3, 50134, Florence, Italy
| | - Marco Matucci-Cerinic
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, AOUC, Largo Brambilla 3, 50134, Florence, Italy
| |
Collapse
|
77
|
Dalton GD, Xie J, An SW, Huang CL. New Insights into the Mechanism of Action of Soluble Klotho. Front Endocrinol (Lausanne) 2017; 8:323. [PMID: 29250031 PMCID: PMC5715364 DOI: 10.3389/fendo.2017.00323] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 11/02/2017] [Indexed: 12/31/2022] Open
Abstract
The klotho gene encodes a type I single-pass transmembrane protein that contains a large extracellular domain, a membrane spanning segment, and a short intracellular domain. Klotho protein exists in several forms including the full-length membrane form (mKl) and a soluble circulating form [soluble klotho (sKl)]. mKl complexes with fibroblast growth factor receptors to form coreceptors for FGF23, which allows it to participate in FGF23-mediated signal transduction and regulation of phosphate and calcium homeostasis. sKl is present in the blood, urine, and cerebrospinal fluid where it performs a multitude of functions including regulation of ion channels/transporters and growth factor signaling. How sKl exerts these pleiotropic functions is poorly understood. One hurdle in understanding sKl's mechanism of action as a "hormone" has been the inability to identify a receptor that mediates its effects. In the body, the kidneys are a major source of sKl and sKl levels decline during renal disease. sKl deficiency in chronic kidney disease makes the heart susceptible to stress-induced injury. Here, we summarize the current knowledge of mKl's mechanism of action, the mechanistic basis of sKl's protective, FGF23-independent effects on the heart, and provide new insights into the mechanism of action of sKl focusing on recent findings that sKl binds sialogangliosides in membrane lipid rafts to regulate growth factor signaling.
Collapse
Affiliation(s)
- George D. Dalton
- Department of Medicine, Division of Gastroenterology, Duke University Medical Center, Durham, NC, United States
| | - Jian Xie
- Department of Internal Medicine, Division of Nephrology and Hypertension, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Sung-Wan An
- Department of Internal Medicine, Division of Nephrology and Hypertension, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Chou-Long Huang
- Department of Internal Medicine, Division of Nephrology and Hypertension, University of Iowa Carver College of Medicine, Iowa City, IA, United States
- *Correspondence: Chou-Long Huang,
| |
Collapse
|
78
|
Paula RS, Souza VC, Machado-Silva W, Almeida BRS, Daros AC, Gomes L, Ferreira AP, Brito CJ, Córdova C, Moraes CF, Nóbrega OT. Serum Klotho (but not haplotypes) associate with the post-myocardial infarction status of older adults. Clinics (Sao Paulo) 2016; 71:725-732. [PMID: 28076518 PMCID: PMC5175288 DOI: 10.6061/clinics/2016(12)09] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 09/13/2016] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVES: The number of deaths from vascular diseases is incredibly high worldwide, and reliable markers for major events are still needed. The current cross-sectional study investigated the association of Klotho haplotypes and Klotho serum levels with classic risk factors and a clinical history of vascular events. METHODS: Clinical, anthropometric, biochemical and nutritional assessments were conducted with 168 older adults, complemented by genotyping (rs9536314 and rs9527025) and the detection of serum Klotho (ELISA). RESULTS: Klotho levels and haplotypes did not associate with most classic risk factors for vascular events, including markers such as C-reactive protein and homocysteine. A positive association was only found between Klotho levels and the previous occurrence of a myocardial infarction by both correlational (p=0.006) and variance analyses (p<0.001), and these associations were independent of the context. CONCLUSION: Our results suggest that serum Klotho is higher in individuals with a clinical history of myocardial infarction but not with a history of coronary artery disease or stroke. None of the Klotho haplotypes were associated with the variables investigated herein.
Collapse
Affiliation(s)
| | | | | | | | - Andersen C Daros
- Centro Universitário de Brasília (UNICEUB), Brasília/, DF, Brazil
| | - Lucy Gomes
- Universidade Católica de Brasília (UCB-DF), Brasília/, DF, Brazil
| | - Aparecido P Ferreira
- Universidade Católica de Brasília (UCB-DF), Brasília/, DF, Brazil
- Faculdades Promove (ICESP), Brasília/, DF, Brazil
| | - Ciro J Brito
- Universidade Federal de Juiz de Fora (UFJF), Juiz de Fora/, MG, Brazil
| | - Cláudio Córdova
- Universidade Católica de Brasília (UCB-DF), Brasília/, DF, Brazil
| | - Clayton F Moraes
- Universidade de Brasília (UnB), Brasília/, DF, Brazil
- Universidade Católica de Brasília (UCB-DF), Brasília/, DF, Brazil
| | | |
Collapse
|
79
|
Varshney R, Ali Q, Wu C, Sun Z. Monocrotaline-Induced Pulmonary Hypertension Involves Downregulation of Antiaging Protein Klotho and eNOS Activity. Hypertension 2016; 68:1255-1263. [PMID: 27672025 DOI: 10.1161/hypertensionaha.116.08184] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 08/15/2016] [Indexed: 12/20/2022]
Abstract
The objective of this study is to investigate whether stem cell delivery of secreted Klotho (SKL), an aging-suppressor protein, attenuates monocrotaline-induced pulmonary vascular dysfunction and remodeling. Overexpression of SKL in mesenchymal stem cells (MSCs) was achieved by transfecting MSCs with lentiviral vectors expressing SKL-green fluorescent protein (GFP). Four groups of rats were treated with monocrotaline, whereas an additional group was given saline (control). Three days later, 4 monocrotaline-treated groups received intravenous delivery of nontransfected MSCs, MSC-GFP, MSC-SKL-GFP, and PBS, respectively. Ex vivo vascular relaxing responses to acetylcholine were diminished in small pulmonary arteries (PAs) in monocrotaline-treated rats, indicating pulmonary vascular endothelial dysfunction. Interestingly, delivery of MSCs overexpressing SKL (MSC-SKL-GFP) abolished monocrotaline-induced pulmonary vascular endothelial dysfunction and PA remodeling. Monocrotaline significantly increased right ventricular systolic blood pressure, which was attenuated significantly by MSC-SKL-GFP, indicating improved PA hypertension. MSC-SKL-GFP also attenuated right ventricular hypertrophy. Nontransfected MSCs slightly, but not significantly, improved PA hypertension and pulmonary vascular endothelial dysfunction. MSC-SKL-GFP attenuated monocrotaline-induced inflammation, as evidenced by decreased macrophage infiltration around PAs. MSC-SKL-GFP increased SKL levels, which rescued the downregulation of SIRT1 (Sirtuin 1) expression and endothelial NO synthase (eNOS) phosphorylation in the lungs of monocrotaline-treated rats. In cultured endothelial cells, SKL abolished monocrotaline-induced downregulation of eNOS activity and NO levels and enhanced cell viability. Therefore, stem cell delivery of SKL is an effective therapeutic strategy for pulmonary vascular endothelial dysfunction and PA remodeling. SKL attenuates monocrotaline-induced PA remodeling and PA smooth muscle cell proliferation, likely by reducing inflammation and restoring SIRT1 levels and eNOS activity.
Collapse
Affiliation(s)
- Rohan Varshney
- From the Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City
| | - Quaisar Ali
- From the Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City
| | - Chengxiang Wu
- From the Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City
| | - Zhongjie Sun
- From the Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City.
| |
Collapse
|
80
|
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: 2.8] [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
|
81
|
Klotho modulates FGF23-mediated NO synthesis and oxidative stress in human coronary artery endothelial cells. Pflugers Arch 2016; 468:1621-35. [PMID: 27448998 DOI: 10.1007/s00424-016-1858-x] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 06/28/2016] [Accepted: 07/12/2016] [Indexed: 12/27/2022]
Abstract
Chronic kidney disease (CKD) is a state of Klotho deficiency and excess of the phosphaturic hormone fibroblast growth factor 23 (FGF23). Both dysregulations were shown to be associated with endothelial dysfunction in humans, but direct vascular effects of FGF23 remain largely elusive. In vitro experiments were performed to assess the effects of FGF23 (10 ng/mL) in relation to its co-receptor Klotho on nitric oxide (NO) synthesis and reactive oxygen species (ROS) formation and detoxification in human coronary artery endothelial cells (HCAEC). Membrane-bound Klotho is expressed in HCAEC, and FGF23 increases the expression of the Klotho shedding protease ADAM17, and consequently the secretion of soluble Klotho. FGF23 activates FGF receptor 1 and stimulates NO release via Akt-dependent activation of endothelial NO synthase (eNOS). Both FGF receptor (FGFR)-dependent ROS formation via activation of NADPH oxidase 2 (Nox2) as well as ROS degradation via superoxide dismutase 2 (SOD2) and catalase (CAT) is stimulated by FGF23. Pre-incubation with a Klotho inhibitor blunts the FGF23-stimulated Akt-eNOS activation and NO synthesis, and decreases ROS degradation by blocking SOD2 and CAT enzymes, whereas FGF23-stimulated ROS synthesis via Nox2 is unaffected, resulting in low NO bioavailability and increased oxidative stress. Our data indicate that in the presence of Klotho, FGF23 induces NO release in HCAEC and its stimulating effects on ROS production are counterbalanced by increased ROS degradation. In states of Klotho deficiency, e.g., CKD, FGF23-mediated NO synthesis is blunted and ROS formation overrules ROS degradation. Thus, FGF23 excess may primarily promote oxidative stress and thus endothelial dysfunction.
Collapse
|
82
|
Wang N, Bai X, Jin B, Han W, Sun X, Chen X. Association between circulating fibroblast growth factor-23 and age-related cardiovascular-renal parameters in a healthy Chinese population. Geriatr Gerontol Int 2016; 17:1221-1231. [PMID: 27427468 DOI: 10.1111/ggi.12844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 04/29/2016] [Accepted: 05/16/2016] [Indexed: 11/29/2022]
Affiliation(s)
- Nan Wang
- Department of Gerontology and Geriatrics; Shengjing Hospital of China Medical University; Shenyang China
| | - Xiaojuan Bai
- Department of Gerontology and Geriatrics; Shengjing Hospital of China Medical University; Shenyang China
| | - Bo Jin
- Department of Gerontology and Geriatrics; The Second Hospital of Dalian Medical University; Dalian China
| | - Wen Han
- Department of Gerontology and Geriatrics; Shengjing Hospital of China Medical University; Shenyang China
| | - Xuefeng Sun
- Department of Kidney; General Hospital of Chinese People's Liberation Army; Beijing China
| | - Xiangmei Chen
- Department of Kidney; General Hospital of Chinese People's Liberation Army; Beijing China
| |
Collapse
|
83
|
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: 0.4] [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
|
84
|
Abstract
Reactive oxygen species (ROS) and oxidative stress have long been linked to aging and diseases prominent in the elderly such as hypertension, atherosclerosis, diabetes and atrial fibrillation (AF). NADPH oxidases (Nox) are a major source of ROS in the vasculature and are key players in mediating redox signalling under physiological and pathophysiological conditions. In this review, we focus on the Nox-mediated ROS signalling pathways involved in the regulation of 'longevity genes' and recapitulate their role in age-associated vascular changes and in the development of age-related cardiovascular diseases (CVDs). This review is predicated on burgeoning knowledge that Nox-derived ROS propagate tightly regulated yet varied signalling pathways, which, at the cellular level, may lead to diminished repair, the aging process and predisposition to CVDs. In addition, we briefly describe emerging Nox therapies and their potential in improving the health of the elderly population.
Collapse
|
85
|
Kalaitzidis RG, Duni A, Siamopoulos KC. Klotho, the Holy Grail of the kidney: from salt sensitivity to chronic kidney disease. Int Urol Nephrol 2016; 48:1657-66. [PMID: 27215557 DOI: 10.1007/s11255-016-1325-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 05/17/2016] [Indexed: 01/05/2023]
Abstract
The Klotho gene displays an extremely shortened life span with loss of function missense mutations leading to premature multiple organ failure, thus resembling human premature aging syndromes. The transmembrane form of Klotho protein functions as an obligatory co-receptor for FGF23. Klotho and FGF23 are crucial components for the regulation of vitamin D metabolism and subsequently blood phosphate levels. The secreted Klotho protein has multiple regulatory functions, including effects on electrolyte homeostasis, on growth factor pathways as well as on oxidative stress, which are currently the object of extensive research. Klotho protein deficiency is observed in many experimental and clinical disease models. Genetic polymorphisms such as the G-395A polymorphism in the promoter region of the Klotho gene have been associated with the development of essential hypertension. The kidneys are the primary site of Klotho production, and renal Klotho is decreased in CKD, followed by a reduction in plasma Klotho. Klotho deficiency has been both associated with progression of CKD as well as with its cardinal systemic manifestations, including cardiovascular disease. Thus, Klotho has been suggested both as a risk biomarker for early detection of CKD and additionally as a potential therapeutic tool in the future.
Collapse
Affiliation(s)
- Rigas G Kalaitzidis
- Department of Nephrology, University Hospital of Ioannina, Ioannina, Greece.
| | - Anila Duni
- Department of Nephrology, University Hospital of Ioannina, Ioannina, Greece
| | | |
Collapse
|
86
|
Tan SJ, Smith ER, Hewitson TD, Holt SG, Toussaint ND. The importance of klotho in phosphate metabolism and kidney disease. Nephrology (Carlton) 2016; 19:439-49. [PMID: 24750549 DOI: 10.1111/nep.12268] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2014] [Indexed: 12/19/2022]
Abstract
The discovery of fibroblast growth factor-23 (FGF23) and its co-receptor α-klotho has broadened our understanding of mineral metabolism and led to a renewed research focus on phosphate homeostatic pathways in kidney disease. Expanding knowledge of these mechanisms, both in normal physiology and in pathology, identifies targets for potential interventions designed to reduce the complications of renal disease, particularly the cardiovascular sequelae. FGF23 has emerged as a major α-klotho-dependent endocrine regulator of mineral metabolism, functioning to activate vitamin D and as a phosphatonin. However, increasingly there is an appreciation that klotho may act independently as a phosphate regulator, as well as having significant activity in other key biological processes. This review outlines our current understanding of klotho, and its potential contribution to kidney disease and cardiovascular health.
Collapse
Affiliation(s)
- Sven-Jean Tan
- Department of Nephrology, The Royal Melbourne Hospital, Melbourne, Victoria, Australia; Department of Medicine (RMH), The University of Melbourne, Melbourne, Victoria, Australia
| | | | | | | | | |
Collapse
|
87
|
Keles N, Caliskan M, Dogan B, Keles NN, Kalcik M, Aksu F, Kostek O, Aung SM, Isbilen B, Oguz A. Low Serum Level of Klotho Is an Early Predictor of Atherosclerosis. TOHOKU J EXP MED 2016; 237:17-23. [PMID: 26289053 DOI: 10.1620/tjem.237.17] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The Klotho gene, identified as an 'aging suppressor' gene, encodes a single-pass transmembrane protein. The extracellular domain of Klotho is cleaved and released in the blood stream, where it may function as a vasculoprotective hormone. Carotid artery intima-media thickness (CIMT), flow-mediated dilation (FMD) of the brachial artery and epicardial fat thickness (EFT) have been reported as early predictors of atherosclerosis. We aimed to investigate the relationship between serum Klotho levels and early atherosclerotic predictors, including EFT, FMD and CIMT in healthy adults. Fifty healthy volunteers were enrolled in this study, consisting of 21 males and 29 females with median age of 32 years. They were free of known risk factors for cardiovascular diseases. Serum Klotho levels were determined by the ELISA method. The study population was divided into two groups (n = 25 for each) according to the median serum Klotho level (459.4 pg/mL): higher Klotho (HK) group (613.6 pg/mL; ranges of 501.2-772.6 pg/mL) and lower Klotho (LK) group (338.7 pg/mL; ranges of 278.8-430.3 pg/mL). EFT was measured by transthoracic echocardiography, and CIMT and FMD were measured with standard procedures. The LK group showed lower values of FMD (p = 0.012) and larger values of EFT (p = 0.01) and CIMT (p < 0.001), compared to the HK group. Thus, the low serum Klotho levels were associated with increased EFT and CIMT and with the decreased FMD in the study population. We propose that the lower serum Klotho level is a newly identified predictor of atherosclerosis.
Collapse
Affiliation(s)
- Nursen Keles
- Department of Cardiology, Goztepe Training and Research Hospital, Istanbul Medeniyet University
| | | | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Wehling-Henricks M, Li Z, Lindsey C, Wang Y, Welc SS, Ramos JN, Khanlou N, Kuro-O M, Tidball JG. Klotho gene silencing promotes pathology in the mdx mouse model of Duchenne muscular dystrophy. Hum Mol Genet 2016; 25:2465-2482. [PMID: 27154199 DOI: 10.1093/hmg/ddw111] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 03/10/2016] [Accepted: 04/08/2016] [Indexed: 11/12/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a lethal muscle disease involving progressive loss of muscle regenerative capacity and increased fibrosis. We tested whether epigenetic silencing of the klotho gene occurs in the mdx mouse model of DMD and whether klotho silencing is an important feature of the disease. Our findings show that klotho undergoes muscle-specific silencing at the acute onset of mdx pathology. Klotho experiences increased methylation of CpG sites in its promoter region, which is associated with gene silencing, and increases in a repressive histone mark, H3K9me2. Expression of a klotho transgene in mdx mice restored their longevity, reduced muscle wasting, improved function and greatly increased the pool of muscle-resident stem cells required for regeneration. Reductions of fibrosis in late, progressive stages of the mdx pathology achieved by transgene expression were paralleled by reduced expression of Wnt target genes (axin-2), transforming growth factor-beta (TGF-β1) and collagens types 1 and 3, indicating that Klotho inhibition of the profibrotic Wnt/TGFβ axis underlies its anti-fibrotic effect in aging, dystrophic muscle. Thus, epigenetic silencing of klotho during muscular dystrophy contributes substantially to lost regenerative capacity and increased fibrosis of dystrophic muscle during late progressive stages of the disease.
Collapse
Affiliation(s)
| | - Zhenzhi Li
- Department of Integrative Biology and Physiology
| | | | - Ying Wang
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, CA 90095-1606, USA
| | | | | | - Négar Khanlou
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA 90095-1732, USA
| | - Makoto Kuro-O
- Division of Anti-Aging Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - James G Tidball
- Department of Integrative Biology and Physiology .,Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, CA 90095-1606, USA.,Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA 90095-1732, USA
| |
Collapse
|
89
|
Rubinek T, Wolf I. The Role of Alpha-Klotho as a Universal Tumor Suppressor. VITAMINS AND HORMONES 2016; 101:197-214. [PMID: 27125743 DOI: 10.1016/bs.vh.2016.03.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The klotho gene is implicated in many physiological activities, among them aging, glucose metabolism, and phosphate and calcium metabolism. Many cellular activities of klotho were implicated in promoting these activities. Two of them, inhibition of the insulin-like growth factor-1 pathway and of the Wnt signaling pathway, are also major pathways associated with cancer development and progression. These discoveries prompted a surge of research aiming to elucidate the role of klotho in cancer. Studies show that klotho is universally silenced in a wide array of malignancies, including breast, pancreatic, ovarian, lung, colorectal, and melanoma, and that klotho's expression can serve as an invaluable prognostic marker. Epigenetic mechanisms, ie, promoter hypermethylation and histone deacetylation, are mainly associated with klotho's silencing; however, different micro-RNAs were also demonstrated to be involved in the process. The activity of klotho on cancer cells growth was also widely investigated, and accumulating data suggest that klotho forced expression or treatment with the soluble protein can inhibit cancer development and progression. Moreover, studies now aim to reveal the specific region in klotho protein that underlies this anticancer activity in order to develop efficient and safe klotho-based medications.
Collapse
Affiliation(s)
- T Rubinek
- Institute of Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.
| | - I Wolf
- Institute of Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
90
|
Abstract
Alpha-Klotho (αKlotho) protein is encoded by the gene, Klotho, and functions as a coreceptor for endocrine fibroblast growth factor-23. The extracellular domain of αKlotho is cleaved by secretases and released into the circulation where it is called soluble αKlotho. Soluble αKlotho in the circulation starts to decline in chronic kidney disease (CKD) stage 2 and urinary αKlotho in even earlier CKD stage 1. Therefore soluble αKlotho is an early and sensitive marker of decline in kidney function. Preclinical data from numerous animal experiments support αKlotho deficiency as a pathogenic factor for CKD progression and extrarenal CKD complications including cardiac and vascular disease, hyperparathyroidism, and disturbed mineral metabolism. αKlotho deficiency induces cell senescence and renders cells susceptible to apoptosis induced by a variety of cellular insults including oxidative stress. αKlotho deficiency also leads to defective autophagy and angiogenesis and promotes fibrosis in the kidney and heart. Most importantly, prevention of αKlotho decline, upregulation of endogenous αKlotho production, or direct supplementation of soluble αKlotho are all associated with attenuation of renal fibrosis, retardation of CKD progression, improvement of mineral metabolism, amelioration of cardiac function and morphometry, and alleviation of vascular calcification in CKD. Therefore in rodents, αKlotho is not only a diagnostic and prognostic marker for CKD but the enhancement of endogenous or supplement of exogenous αKlotho are promising therapeutic strategies to prevent, retard, and decrease the comorbidity burden of CKD.
Collapse
Affiliation(s)
- J A Neyra
- University of Texas Southwestern Medical Center, Dallas, TX, United States; Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - M C Hu
- University of Texas Southwestern Medical Center, Dallas, TX, United States; Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, United States.
| |
Collapse
|
91
|
Dounousi E, Torino C, Pizzini P, Cutrupi S, Panuccio V, D'Arrigo G, Abd ElHafeez S, Tripepi G, Mallamaci F, Zoccali C. Intact FGF23 and α-Klotho during acute inflammation/sepsis in CKD patients. Eur J Clin Invest 2016; 46:234-41. [PMID: 26728476 DOI: 10.1111/eci.12588] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 12/30/2015] [Indexed: 12/17/2022]
Abstract
BACKGROUND High FGF23 and low α-Klotho levels associate with systemic inflammation and reduced nitric oxide (NO) bioavailability, but the dynamics of this relationship in patients with CKD has not been investigated. METHODS We sequentially measured serum intact FGF23 and carboxyl-terminal (iFGF23, cFGF23), the iFGF23/cFGF23 ratio, αKlotho, biomarkers of inflammation (hs-CRP, IL-6 and TNF-α) and sepsis (procalcitonin), nitrotyrosine (reflecting NO synthesis and oxidative stress), serum iron and ferritin and CKD-MBD biomarkers, PTH, 25(OH)VD, 1,25(OH)2 VD at peak of intercurrent sepsis and after complete resolution in a series of 17 patients with CKD. RESULTS At peak infection, biomarkers of inflammation/sepsis, ferritin and nitrotyrosine were all very high (all P < 0·01) and declined towards the normal range thereafter (P < 0·01). iFGF23 was 191 ± 10 pg/ml (geometric mean, SD) and doubled to 371 ± 8 pg/ml (P = 0·003) after the resolution of infection, while cFGF23 did not change (246 ± 5 pg/mL vs. 248 ± 5 pg/mL, P = 0·50). As a consequence, the iFGF23/cFGF23 ratio, an indicator of the proteolytic cleavage of the FGF23 molecule, was 0·78 ± 3·87 at peak infection and increased to 1·49 ± 3·00 after resolution of infection (P < 0·001). In contrast, serum α-Klotho levels were upregulated at peak infection (peak infection: 526 ± 4 pg/ml, postinfection: 447 ± 4 pg/ml, P = 0·001). The eGFR, PTH and vitamin D did not change significantly throughout. CONCLUSIONS Acute inflammation/sepsis suppresses the active form of FGF23 and activates α-Klotho, the latter effect being likely attributable to enhance proteolysis of FGF23 molecule. iFGF23 downregulation and α-Klotho upregulation during acute sepsis may participate into the counter-regulatory response to severe inflammation in CKD patients with sepsis.
Collapse
Affiliation(s)
- Evangelia Dounousi
- Department of Nephrology, Medical School, University of Ioannina, Ioannina, Greece
| | - Claudia Torino
- Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension, CNR-IFC, Reggio Calabria, Italy
| | - Patrizia Pizzini
- Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension, CNR-IFC, Reggio Calabria, Italy
| | - Sebastiano Cutrupi
- Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension, CNR-IFC, Reggio Calabria, Italy
| | - Vincenzo Panuccio
- Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension, CNR-IFC, Reggio Calabria, Italy.,Renal Unit, Reggio Calabria, Italy
| | - Graziella D'Arrigo
- Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension, CNR-IFC, Reggio Calabria, Italy
| | | | - Giovanni Tripepi
- Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension, CNR-IFC, Reggio Calabria, Italy
| | - Francesca Mallamaci
- Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension, CNR-IFC, Reggio Calabria, Italy.,Renal Unit, Reggio Calabria, Italy
| | - Carmine Zoccali
- Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension, CNR-IFC, Reggio Calabria, Italy
| |
Collapse
|
92
|
Luo L, Hao Q, Dong B, Yang M. The Klotho gene G-395A polymorphism and metabolic syndrome in very elderly people. BMC Geriatr 2016; 16:46. [PMID: 26880028 PMCID: PMC4754834 DOI: 10.1186/s12877-016-0221-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 02/04/2016] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND To investigate the possible association of the Klotho G-395A polymorphism and metabolic syndrome (MetS) among a population of Chinese nonagenarians and centenarians. METHODS Subjects were from the Project of Longevity and Aging in Dujiangyan (PLAD). The genotyping of G-395A (rs1207568) in the promoter region of the Klotho gene was performed using the TaqMan allelic discrimination assay. MetS was diagnosed according to the International Diabetes Federation criteria. RESULTS We included 695 subjects aged 93.5 ± 3.2 years. G and A allele frequencies were 0.852 and 0.148, respectively. In the whole population, the frequency of MetS was 10.8% and 5.9% in the GG and GA + AA genotype group, respectively (p = 0.004). The -395A allele carriers had significantly lower risk of MetS in the whole population (odd ratio [OR] 0.50, 95% confidential interval [CI] 0.25 to 0.98) and in women (OR 0.51, 95% CI 0.24 to 0.97), but not in men (OR 0.42, 95% CI 0.05 to 3.85). In the whole population and women, the relationship between the Klotho G-395A SNP and MetS might due to its influence on high blood pressure (OR 0.48, 95% CI 0.34 to 0.67; OR 0.47, 95% CI 0.31 to 0.71, respectively) and hypertriglyceridemia (OR 0.66, 95% CI 0.39 to 0.95; OR 0.54, 95% CI 0.31 to 0.98, respectively). In men, this relationship might due to its influence on high blood pressure (OR 0.47, 95% CI 0.25 to 0.90) and low HDL-C (OR 0.69, 95% CI 0.27 to 0.93). CONCLUSIONS The -395A allele carriers of the Klotho gene were correlated with lower risk of MetS among Chinese nonagenarians and centenarians, especially in women.
Collapse
Affiliation(s)
- Li Luo
- The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, No.37 Guoxue Lane, Chengdu, Sichuan, China.
| | - Qiukui Hao
- The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, No.37 Guoxue Lane, Chengdu, Sichuan, China.
| | - Birong Dong
- The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, No.37 Guoxue Lane, Chengdu, Sichuan, China.
| | - Ming Yang
- The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, No.37 Guoxue Lane, Chengdu, Sichuan, China.
| |
Collapse
|
93
|
Keles N, Caliskan M, Dogan B, Aksu F, Bulur S, Keles NN, Kostek O, Aung SM, Isbilen B, Demircioglu K, Kalcik M, Oguz A. Is Low Serum Klotho Level Associated with Alterations in Coronary Flow Reserve? Echocardiography 2016; 33:881-8. [PMID: 26791446 DOI: 10.1111/echo.13176] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND The Klotho gene, described as an "aging suppressor" gene, encodes a single-pass transmembrane protein. The extracellular part of Klotho is cleaved and released into the circulation where it may function as a vasculoprotective hormone. Coronary flow reserve (CFR) is accepted as a marker of coronary microvascular dysfunction when epicardial coronary stenosis is absent. There are no data regarding the relationship between serum Klotho levels and disorders in coronary microcirculation in healthy adults. We aimed to investigate the association between serum Klotho levels and alterations in coronary microcirculation in healthy adults using echocardiographic measurements of CFR. METHODS Thirty-four healthy volunteers (median age: 34 [27-39], 14 males) were enrolled in this study. The study population was divided into two subgroups according to the median value of serum Klotho levels: a high Klotho (HK) group (n = 17, median age: 34 [30-38]; 6 males) and a low Klotho (LK) group (n = 17, median age: 32 [26-39]; 8 males). The analysis of coronary flow velocities was performed by transthoracic Doppler echocardiography. RESULTS Hyperemic diastolic peak flow velocities and CFR were significantly higher in the HK group than in the LK group (70 [66-92] versus 61 [47-66], P = 0.003 and 3.0 [2.6-3.8] versus 2.2 [1.7-2.8], respectively, P = 0.001). Serum Klotho levels were positively correlated with CFR (P < 0.001). CONCLUSION Serum Klotho levels correlate with CFR in a healthy population. Low serum Klotho levels may potentially identify patients with impaired CFR.
Collapse
Affiliation(s)
- Nursen Keles
- Department of Cardiology, Istanbul Medeniyet University, Goztepe Training and Research Hospital, Istanbul, Turkey
| | - Mustafa Caliskan
- Department of Cardiology, Istanbul Medeniyet University, Goztepe Training and Research Hospital, Istanbul, Turkey
| | - Burcu Dogan
- Department of Family Physicians, Istanbul Medeniyet University, Goztepe Training and Research Hospital, Istanbul, Turkey
| | - Feyza Aksu
- Department of Cardiology, Istanbul Medeniyet University, Goztepe Training and Research Hospital, Istanbul, Turkey
| | - Serkan Bulur
- Department of Cardiology, Istanbul Medeniyet University, Goztepe Training and Research Hospital, Istanbul, Turkey
| | - Necibe Nur Keles
- Department of Neurology, Uskudar Government Hospital, İstanbul, Turkey
| | - Osman Kostek
- Department of Internal Medicine, Istanbul Medeniyet University, Goztepe Training and Research Hospital, Istanbul, Turkey
| | - Soe Moe Aung
- Department of Cardiology, Fatih University, Istanbul, Turkey
| | - Banu Isbilen
- Department of Biochemistry, Istanbul Medeniyet University, Goztepe Training and Research Hospital, Istanbul, Turkey
| | - Kenan Demircioglu
- Department of Cardiology, Istanbul Medeniyet University, Goztepe Training and Research Hospital, Istanbul, Turkey
| | - Macit Kalcik
- Department of Cardiology, Iskilip Atıf Hoca State Hospital, Corum, Turkey
| | - Aytekin Oguz
- Department of Internal Medicine, Istanbul Medeniyet University, Goztepe Training and Research Hospital, Istanbul, Turkey
| |
Collapse
|
94
|
Semba RD, Ferrucci L, Sun K, Simonsick E, Turner R, Miljkovic I, Harris T, Schwartz AV, Asao K, Kritchevsky S, Newman AB. Low Plasma Klotho Concentrations and Decline of Knee Strength in Older Adults. J Gerontol A Biol Sci Med Sci 2016; 71:103-8. [PMID: 26359247 PMCID: PMC4706099 DOI: 10.1093/gerona/glv077] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 12/18/2014] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Although the "anti-aging hormone" klotho is associated with sarcopenia in mice, the relationship between klotho and muscle strength in older adults is not well known. METHODS Plasma klotho concentrations were measured in 2,734 older adults, aged 71-80 years, who participated in the Health, Aging and Body Composition Study, a prospective observational cohort study conducted in Memphis, TN and Pittsburgh, PA. Knee extension strength was measured using isokinetic dynamometry at baseline and follow-up 2 and 4 years later. Knee extension strength was normalized for weight. RESULTS At baseline, participants in the highest tertile of plasma klotho had higher knee extension strength (β = .72, standard error [SE] = .018, p < .0001) compared with those in the lowest tertile in a multivariable linear regression model adjusting for age, sex, race, smoking, study site, C-reactive protein, interleukin-6, and diabetes. Participants in the highest tertile of plasma klotho at baseline had less of a decline in knee strength over 4 years of follow-up (β = -.025, SE = .011, p = .02) compared with those in the lowest tertile in a multivariable linear regression model adjusting for the same covariates above. CONCLUSIONS Plasma klotho concentrations were an independent predictor of changes in knee strength over time in older adults. Further studies are needed to identify the biological mechanisms by which circulating klotho could modify skeletal muscle strength.
Collapse
Affiliation(s)
- Richard D Semba
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | | | - Kai Sun
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Randi Turner
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Iva Miljkovic
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania
| | - Tamara Harris
- Intramural Research Program, Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIH, Bethesda, Maryland
| | - Ann V Schwartz
- Department of Epidemiology and Biostatistics, University of California San Francisco School of Medicine
| | - Keiko Asao
- Department of Preventive Medicine, University of Tennessee, Memphis
| | - Stephen Kritchevsky
- Sticht Center on Aging, Wake Forest University, Winston-Salem, North Carolina
| | - Anne B Newman
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania
| |
Collapse
|
95
|
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; 20:128-33. [PMID: 26538295 PMCID: PMC4717853 DOI: 10.1111/jcmm.12710] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [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
|
96
|
Donate-Correa J, Martín-Núñez E, Mora-Fernández C, Muros-de-Fuentes M, Pérez-Delgado N, Navarro-González JF. Klotho in cardiovascular disease: Current and future perspectives. World J Biol Chem 2015; 6:351-357. [PMID: 26629318 PMCID: PMC4656911 DOI: 10.4331/wjbc.v6.i4.351] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 07/31/2015] [Accepted: 09/08/2015] [Indexed: 02/05/2023] Open
Abstract
Protein Klotho, beyond its role as a regulator of the phosphatemia, is also involved in the maintaining of the cardiovascular health, being associated its alterations with the development of cardiovascular damage and increased morbi-mortality. For all this, nowadays Klotho is the subject of a thorough research which is focused on uncover its intimate mechanisms of action, and in analyzing the utility of its modulation as a potential strategy with clinical applicability. Molecular mechanisms of Klotho are not well understood but an emerging research area links Klotho deficiency with vascular pathology. Changes in this protein have been associated with cardiovascular-related complications like inflammation, vascular calcification, and endothelial dysfunction. All this is particularly relevant if considering the recent discovery of Klotho expression in vascular tissue.
Collapse
|
97
|
Zhou X, Chen K, Wang Y, Schuman M, Lei H, Sun Z. Antiaging Gene Klotho Regulates Adrenal CYP11B2 Expression and Aldosterone Synthesis. J Am Soc Nephrol 2015; 27:1765-76. [PMID: 26471128 DOI: 10.1681/asn.2015010093] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 08/27/2015] [Indexed: 12/11/2022] Open
Abstract
Deficiency of the antiaging gene Klotho (KL) induces renal damage and hypertension through unknown mechanisms. In this study, we assessed whether KL regulates expression of CYP11B2, a key rate-limiting enzyme in aldosterone synthesis, in adrenal glands. We found that haplodeficiency of KL(+/-) in mice increased the plasma level of aldosterone by 16 weeks of age, which coincided with spontaneous and persistent elevation of BP. Blockade of aldosterone actions by eplerenone reversed KL deficiency-induced hypertension and attenuated the kidney damage. Protein expression of CYP11B2 was upregulated in adrenal cortex of KL(+/-) mice. KL and CYP11B2 proteins colocalized in adrenal zona glomerulosa cells. Silencing of KL upregulated and overexpression of KL downregulated CYP11B2 expression in human adrenocortical cells. Notably, silencing of KL decreased expression of SF-1, a negative transcription factor of CYP11B2, but increased phosphorylation of ATF2, a positive transcription factor of CYP11B2, which may contribute to upregulation of CYP11B2 expression. Therefore, these results show that KL regulates adrenal CYP11B2 expression. KL deficiency-induced spontaneous hypertension and kidney damage may be partially attributed to the upregulation of CYP11B2 expression and aldosterone synthesis.
Collapse
Affiliation(s)
- Xiaoli Zhou
- Department of Cardiology, First Affiliated Hospital, Chongqing Medical University, Chongqing, China; and Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Kai Chen
- Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Yongjun Wang
- Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Mariano Schuman
- Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Han Lei
- Department of Cardiology, First Affiliated Hospital, Chongqing Medical University, Chongqing, China; and
| | - Zhongjie Sun
- Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
98
|
Abstract
The aim of this study was to analyze the placental expression and allele status of promoter region of Klotho in association with preeclampsia, which represents the most common hypertensive disease of pregnancy. Klotho mRNA and protein levels were determined using real-time PCR and Western blot, respectively, in placental tissue samples obtained from 34 patients affected with preeclampsia and 34 controls. A PCR-based genotyping analysis was carried out in the promoter region of Klotho gene. Moreover, expression levels of pluripotency markers, Nanog and Oct4, and telomere length were assessed using real-time PCR. Klotho mRNA and protein levels were reduced in preeclamptic placentas compared with controls. -744delA single-nucleotide polymorphism was significantly associated with preeclampsia. In pathological placentas, there was a downregulation of pluripotency markers and a reduced telomere length. This study is the first to evaluate the placental expression level of Klotho in association with preeclampsia. Further analyses will clarify its role in the pathogenesis of this pregnancy hypertensive disorder.
Collapse
|
99
|
Baldan A, Giusti A, Bosi C, Malaventura C, Musso M, Forni GL, Volpato S, Zuliani G, Borgna-Pignatti C. Klotho, a new marker for osteoporosis and muscle strength in β-thalassemia major. Blood Cells Mol Dis 2015; 55:396-401. [PMID: 26460265 DOI: 10.1016/j.bcmd.2015.08.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 08/20/2015] [Accepted: 08/21/2015] [Indexed: 12/20/2022]
Abstract
Aim of this study was to compare plasma levels of the secreted protein Klotho in β-thalassemia major patients and in healthy controls. Also, we examined the existence of correlations between the protein level and osteoporosis, poor muscle strength and fractures. A total of 106 patients with β-thalassemia major and 95 healthy blood donors were enrolled. Klotho level in plasma was measured by mean of an ELISA test and the hand-grip strength using a dynamometer. Intact parathyroid hormone (PTH), 25-hydroxy vitamin D (Vitamin D), serum calcium (Ca), phosphate (P), total alkaline phosphatase (ALP), ferritin, creatinine were measured by standard clinical techniques. DXA was used to measure bone mineral density (BMD) at the lumbar spine (L2-L4), femoral neck and total hip. We found that the Klotho protein concentration was lower in the blood of patients with β-thalassemia major than in healthy controls, and it was directly correlated to the hand-grip strength. In β-thalassemia major patients, the secreted Klotho was lower than in healthy controls. The preliminary investigation into the correlation between markers of osteo- and sarcopenia and Klotho demonstrated a decreased Klotho concentration in β-TM patients and a higher probability of having had fragility fractures.
Collapse
Affiliation(s)
- Alessandro Baldan
- Department of Medical Sciences, Section of Pediatrics, University of Ferrara, Italy; Department of Life and Reproduction Sciences, Section of Biology and Genetics, University of Verona, Verona, Italy.
| | - Andrea Giusti
- Bone Clinic, Department of Gerontology and Musculoskeletal Sciences, Galliera Hospital, Genoa, Italy
| | - Cristina Bosi
- Department of Medical Sciences, Section of Internal and Cardio-Respiratory Medicine, University of Ferrara, Italy
| | - Cristina Malaventura
- Department of Medical Sciences, Section of Pediatrics, University of Ferrara, Italy
| | - Marco Musso
- Center of Microcitemia and Congenital Anemias, Galliera Hospital, Genoa, Italy
| | - Gian Luca Forni
- Center of Microcitemia and Congenital Anemias, Galliera Hospital, Genoa, Italy
| | - Stefano Volpato
- Department of Medical Sciences, Section of Internal and Cardio-Respiratory Medicine, University of Ferrara, Italy
| | - Giovanni Zuliani
- Department of Medical Sciences, Section of Internal and Cardio-Respiratory Medicine, University of Ferrara, Italy
| | | |
Collapse
|
100
|
Abstract
Aging is an inevitable and progressive biological process involving dysfunction and eventually destruction of every tissue and organ. This process is driven by a tightly regulated and complex interplay between genetic and acquired factors. Klotho is an antiaging gene encoding a single-pass transmembrane protein, klotho, which serves as an aging suppressor through a wide variety of mechanisms, such as antioxidation, antisenescence, antiautophagy, and modulation of many signaling pathways, including insulin-like growth factor and Wnt. Klotho deficiency activates Wnt expression and activity contributing to senescence and depletion of stem cells, which consequently triggers tissue atrophy and fibrosis. In contrast, the klotho protein was shown to suppress Wnt-signaling transduction, and inhibit cell senescence and preserve stem cells. A better understanding of the potential effects of klotho on stem cells could offer novel insights into the cellular and molecular mechanisms of klotho deficiency-related aging and disease. The klotho protein may be a promising therapeutic agent for aging and aging-related disorders.
Collapse
Affiliation(s)
- Ao Bian
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA ; Department of Nephrology, First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Javier A Neyra
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, USA
| | - Ming Zhan
- Methodist Hospital Research Institute, Weill Cornell Medical College, Houston, TX, USA
| | - Ming Chang Hu
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA ; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, USA
| |
Collapse
|