101
|
Mochón-Benguigui S, Carneiro-Barrera A, Castillo MJ, Amaro-Gahete FJ. Is Sleep Associated with the S-Klotho Anti-Aging Protein in Sedentary Middle-Aged Adults? The FIT-AGEING Study. Antioxidants (Basel) 2020; 9:antiox9080738. [PMID: 32806634 PMCID: PMC7463654 DOI: 10.3390/antiox9080738] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/08/2020] [Accepted: 08/10/2020] [Indexed: 11/16/2022] Open
Abstract
Sleep and Klotho have both been closely related to the ageing process, both playing a substantial role in the endocrine and immune systems and, thereby, in oxidative stress and chronic inflammation. However, there are no studies elucidating the relationship between sleep and Klotho. Therefore, this study investigated the association of sleep quantity and quality with the shed form of the α-Klotho gene (S-Klotho plasma levels) in sedentary middle-aged adults. A total of 74 volunteers (52.7% women; aged 53.7 ± 5.1) were recruited for the present study. Objective sleep quality parameters (total sleep time (TST), wake after sleep onset (WASO), and sleep efficiency (SE)) were determined using a wrist-worn accelerometer over seven consecutive days, and the subjective sleep quality was assessed by the Pittsburgh Sleep Quality Index (PSQI; higher scores indicate worse sleep quality). The S-Klotho plasma levels were measured in the ethylenediaminetetraacetic acid plasma using a solid-phase sandwich enzyme-linked immunosorbent assay. Objective sleep parameters were associated with the S-Klotho plasma levels only after including the age, fat mass percentage, and lean mass index as covariates. A direct relationship was observed between the subjective sleep quality (inverse of PSQI scores) and the S-Klotho plasma levels in sedentary middle-aged adults. Improving sleep quantity and quality could be considered an anti-aging therapeutic approach for the prevention, slowing, and even reversal of the physiological decline and degenerative pathologies that are certainly related to the aging process.
Collapse
Affiliation(s)
- Sol Mochón-Benguigui
- EFFECTS-262 Research Group, Department of Medical Physiology, School of Medicine, University of Granada, 18016 Granada, Spain;
- Correspondence: (S.M.-B.); (F.J.A.-G.)
| | - Almudena Carneiro-Barrera
- Sleep and Health Promotion Laboratory, Mind, Brain and Behaviour Research Centre (CIMCYC), University of Granada, 18011 Granada, Spain;
| | - Manuel J. Castillo
- EFFECTS-262 Research Group, Department of Medical Physiology, School of Medicine, University of Granada, 18016 Granada, Spain;
| | - Francisco J. Amaro-Gahete
- EFFECTS-262 Research Group, Department of Medical Physiology, School of Medicine, University of Granada, 18016 Granada, Spain;
- Correspondence: (S.M.-B.); (F.J.A.-G.)
| |
Collapse
|
102
|
Xie T, Ye W, Liu J, Zhou L, Song Y. The Emerging Key Role of Klotho in the Hypothalamus-Pituitary-Ovarian Axis. Reprod Sci 2020; 28:322-331. [PMID: 32783104 DOI: 10.1007/s43032-020-00277-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 07/24/2020] [Indexed: 12/16/2022]
Abstract
The hypothalamus-pituitary-ovary axis is the most important system for regulating female reproductive endocrine function. Its dysfunction would lead to the abnormal secretion of gonadotropin-releasing hormone, follicle-stimulating hormone, or luteinizing hormone, and eventually result in the occurrence of reproductive disease, such as congenital hypogonadotropic hypogonadism, polycystic ovary syndrome, and premature ovarian failure. Recently, an anti-aging gene, Klotho, has gained broad attention in female reproductive diseases. Reports have shown that Klotho is closely correlated to the hypothalamus-pituitary-ovary axis and plays a key role in the development and progression of reproductive diseases. With this issue, we generally review the physiological and pathological role of Klotho in the hypothalamus-pituitary-ovary axis. We also review the underlying mechanisms of Klotho in promoting and preventing female reproductive diseases, which involve the dysfunction of the fibroblast growth factor-Klotho endocrine system, the abnormal signaling regulation of Wnt-β-catenin and insulin-like growth factor-1, the accumulation of oxidative stress, and the inhibition of autophagy, eventually affecting the genesis, development, ovulation, or atresia of follicles. The present review would provide new insights and potential therapeutic target strategies for clinical strategies.
Collapse
Affiliation(s)
- Tingting Xie
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave., Guangzhou, 510515, China
| | - Wenting Ye
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave., Guangzhou, 510515, China
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave., Guangzhou, 510515, China
| | - Jing Liu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave., Guangzhou, 510515, China
| | - Lili Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave., Guangzhou, 510515, China.
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China.
| | - Yali Song
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave., Guangzhou, 510515, China.
| |
Collapse
|
103
|
Infante B, Franzin R, Madio D, Calvaruso M, Maiorano A, Sangregorio F, Netti GS, Ranieri E, Gesualdo L, Castellano G, Stallone G. Molecular Mechanisms of AKI in the Elderly: From Animal Models to Therapeutic Intervention. J Clin Med 2020; 9:jcm9082574. [PMID: 32784471 PMCID: PMC7464895 DOI: 10.3390/jcm9082574] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 07/21/2020] [Accepted: 08/05/2020] [Indexed: 12/17/2022] Open
Abstract
Acute kidney injury (AKI), a critical syndrome characterized by a sudden reduction of renal function, is a common disorder among elderly patients particularly in Intensive Care Unit (ICU). AKI is closely associated with both short- and long-term mortality and length of hospital stay and is considered a predictor of chronic kidney disease (CKD). Specific hemodynamic, metabolic, and molecular changes lead to increased susceptibility to injury in the aged kidney; therefore, certain causes of AKI such as the prerenal reduction in renal perfusion or vascular obstructive conditions are more common in the elderly; moreover, AKI is often multifactorial and iatrogenic. Older patients present several comorbidities (diabetes, hypertension, heart failure) and are exposed to multiple medical interventions such as the use of nephrotoxic contrasts media and medications, which can also trigger AKI. Considering the emerging relevance of this condition, prevention and treatment of AKI in the elderly should be crucial in the internist and emergency setting. This review article summarizes the incidence, the risk factors, the pathophysiology, the molecular mechanisms and the strategies of prevention and treatment of AKI in elderly patients.
Collapse
Affiliation(s)
- Barbara Infante
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto Luigi 251, 71122 Foggia, Italy; (B.I.); (D.M.); (A.M.); (F.S.); (G.S.)
| | - Rossana Franzin
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, 70124 Bari, Italy; (R.F.); (L.G.)
| | - Desirèe Madio
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto Luigi 251, 71122 Foggia, Italy; (B.I.); (D.M.); (A.M.); (F.S.); (G.S.)
| | - Martina Calvaruso
- Nephrology, Dialysis and Transplantation Unit, Department of Biomedical Sciences, University of Foggia, 71122 Foggia, Italy;
| | - Annamaria Maiorano
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto Luigi 251, 71122 Foggia, Italy; (B.I.); (D.M.); (A.M.); (F.S.); (G.S.)
| | - Fabio Sangregorio
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto Luigi 251, 71122 Foggia, Italy; (B.I.); (D.M.); (A.M.); (F.S.); (G.S.)
| | - Giuseppe Stefano Netti
- Clinical Pathology, Department of Surgical and Medical Sciences, University of Foggia, Viale Pinto Luigi 251, 71122 Foggia, Italy; (G.S.N.); (E.R.)
| | - Elena Ranieri
- Clinical Pathology, Department of Surgical and Medical Sciences, University of Foggia, Viale Pinto Luigi 251, 71122 Foggia, Italy; (G.S.N.); (E.R.)
| | - Loreto Gesualdo
- Nephrology, Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, 70124 Bari, Italy; (R.F.); (L.G.)
| | - Giuseppe Castellano
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto Luigi 251, 71122 Foggia, Italy; (B.I.); (D.M.); (A.M.); (F.S.); (G.S.)
- Correspondence: ; Tel.: +39-088-173-2610; Fax: +39-088-173-6001
| | - Giovanni Stallone
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto Luigi 251, 71122 Foggia, Italy; (B.I.); (D.M.); (A.M.); (F.S.); (G.S.)
| |
Collapse
|
104
|
Liu QF, Li SS, Yu LX, Feng JH, Xue LL, Lu GY. The prognostic value of soluble Klotho in patients with haemodialysis: a systematic review and meta-analysis. Ther Adv Chronic Dis 2020; 11:2040622320940176. [PMID: 35154625 PMCID: PMC8832317 DOI: 10.1177/2040622320940176] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 06/03/2020] [Indexed: 01/09/2023] Open
Abstract
Aim: The correlation between soluble Klotho (sKlotho) levels and clinical outcomes
remains inconclusive for patients undergoing maintenance haemodialysis
(MHD). We aimed to evaluate the potential predictive significance of sKlotho
in this population by conducting a meta-analysis. Methods: PubMed, Embase, Web of Science and Cochrane Library were comprehensively
searched for studies concerning the association between sKlotho level and
clinical outcomes including cardiovascular (CV) events and all-cause
mortality. The pooled hazard ratios (HR) and 95% confidence intervals (CI)
were generated using either random or fixed effects models. Sensitivity and
subgroup analyses were used to explore heterogeneity sources. Results: Eight prospective studies with 992 MHD participants were included and reduced
sKlotho levels predicted more adverse outcomes in this meta-analysis. The
pooled HRs and 95% CIs related to CV events, mortality, or composite
outcomes were 1.73 (95% CI 1.08–2.76, p = 0.02), 2.34 (95%
CI 1.34–2.07, p = 0.003) or 1.75 (95% CI 1.19–2.57,
p = 0.005). Moderate heterogeneity was observed in the
composite adverse outcomes (I2 = 57%,
p = 0.05). Age and sKlotho level were the main sources
of heterogeneities in the subgroup analysis. Conclusion: Lower sKlotho levels were associated with more CV events and all-cause
mortality, suggesting that sKlotho may have predictive value in CKD patients
receiving haemodialysis.
Collapse
Affiliation(s)
- Qi-Feng Liu
- Department of Nephrology, The First Affiliated Hospital of Soochow University, Jiangsu, China
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, China
| | - Sha-Sha Li
- Clinical Research & Lab Centre, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, China
- Immunology Laboratory, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, China
| | - Li-Xia Yu
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, China
| | - Jian-Hua Feng
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, China
| | - Li-Li Xue
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, China
| | - Guo-Yuan Lu
- Department of Nephrology, The First Affiliated Hospital of Soochow University, 188 Shizi Road Suzhou, Jiangsu, 215006, China
| |
Collapse
|
105
|
Bao JF, Hu PP, She QY, Li A. A Land of Controversy: Fibroblast Growth Factor-23 and Uremic Cardiac Hypertrophy. J Am Soc Nephrol 2020; 31:1423-1434. [PMID: 32527977 PMCID: PMC7351013 DOI: 10.1681/asn.2020010081] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cardiac hypertrophy is a common feature in patients with CKD. Recent studies revealed that two phosphate regulators, fibroblast growth factor-23 and α-Klotho, are highly involved in the pathophysiologic process of CKD-induced cardiac hypertrophy. With decreasing renal function, elevated fibroblast growth factor-23 and decreased α-Klotho may contribute to cardiac hypertrophy by targeting the heart directly or by inducing systemic changes, such as vascular injury, hemodynamic disorders, and inflammation. However, several studies have demonstrated that disturbances in the fibroblast growth factor-23/α-Klotho axis do not lead to cardiac hypertrophy. In this review, we describe the cardiac effects of the fibroblast growth factor-23/α-Klotho axis and summarize recent progress in this field. In addition, we present not only the main controversies in this field but also provide possible directions to resolve these disputes.
Collapse
Affiliation(s)
- Jing-Fu Bao
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Pan-Pan Hu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qin-Ying She
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Aiqing Li
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
106
|
Hung Y, Chen YC, Huang SY, Lu YY, Lin YK, Kao YH, Lin WS, Chen SA, Chen YJ. Klotho modulates electrical activity and calcium homeostasis in pulmonary vein cardiomyocytes via PI3K/Akt signalling. Europace 2020; 22:1132-1141. [DOI: 10.1093/europace/euaa100] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 05/06/2020] [Indexed: 12/11/2022] Open
Abstract
Abstract
Aims
Klotho, a potential antiageing protein has remarkable cardiovascular effects, which is lower in the patients with chronic kidney disease (CKD). Chronic kidney disease increases the risk of atrial fibrillation, majorly triggered by pulmonary vein (PV) arrhythmogenesis. This study investigated whether klotho protein can modulate PV electrical activity and the underlying potential mechanisms.
Methods and results
A conventional microelectrode and whole-cell patch clamp were used to investigate the action potentials and ionic currents in isolated rabbit PV tissue preparations and single cardiomyocytes before and after klotho administration. Phosphoinositide 3-kinase (PI3K)/Akt signalling was studied using western blotting. Klotho significantly reduced PV spontaneous beating rates in PV tissue preparations at 1.0 and 3.0 ng/mL (but not at 0.1 and 0.3 ng/mL). In the presence of the Akt inhibitor (10 µM), klotho (1.0 and 3.0 ng/mL) did not change PV electrical activities. Klotho (1.0 ng/mL) significantly decreased the late sodium current (INa-Late) and L-type calcium current (ICa-L), similar to the Akt inhibitor (10 µM). Western blots demonstrated that klotho (1.0 ng/mL)-treated PV cardiomyocytes had less phosphorylation of Akt (Ser473) compared with klotho-untreated cardiomyocytes. Compared with control PVs, klotho at relatively lower concentrations (0.1 and 0.3 ng/mL) significantly reduced beating rates and decreased the amplitudes of delay afterdepolarizations in CKD PVs.
Conclusion
Klotho modulated PV electrical activity by inhibiting PI3K/Akt signalling, which may provide a novel insight into CKD-induced arrhythmogenesis.
Collapse
Affiliation(s)
- Yuan Hung
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yao-Chang Chen
- Department of Biomedical Engineering, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Yu Huang
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
- Division of Cardiac Electrophysiology, Cardiovascular Center, Cathay General Hospital, Taipei, Taiwan
| | - Yen-Yu Lu
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
- Division of Cardiology, Department of Internal Medicine, Sijhih Cathay General Hospital, New Taipei City, Taiwan
| | - Yung-Kuo Lin
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Hsun Kao
- Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wei-Shiang Lin
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
- Department of Biomedical Engineering, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Ann Chen
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Division of Cardiology and Cardiovascular Research Center, Veterans General Hospital-Taipei, Taipei, Taiwan
| | - Yi-Jen Chen
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
107
|
Miranda Pérez AA, Gutiérrez Pérez ME, Urraza Robledo AI, Delgadillo Guzmán D, Ruíz Flores P, López Márquez FC. Klotho-HIV and Oxidative Stress: The Role of Klotho in Cardiovascular Disease Under HIV Infection-A Review. DNA Cell Biol 2020; 39:1478-1485. [PMID: 32584609 DOI: 10.1089/dna.2020.5444] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Combined antiretroviral therapy has improved quality and life expectancy of people living with human immunodeficiency virus (HIV). However, this therapy increases oxidative stress (OS), which in turn causes alterations in lipid and carbon metabolism, kidney disease, liver cirrhosis, and increased risk of cardiovascular disease. The Klotho gene has been implicated in cardiovascular risk increase. Klotho protein expression at X level decreases the risk of heart disease. HIV-positive people usually present low plasma levels of Klotho; thus, contributing to some extent to an increase in cardiovascular risk for these types of patients, mostly by favoring atherosclerosis. Therefore, our aim is to provide an overview of the effect of OS on Klotho protein and its consequent cardiometabolic alterations in HIV-positive patients on antiretroviral therapy.
Collapse
Affiliation(s)
- Alberto Alejandro Miranda Pérez
- Department of Molecular Immunobiology, Biomedical Research Center, Faculty of Medicine, Autonomous University of Coahuila, Torreon, Mexico
| | - María Elena Gutiérrez Pérez
- Department of Molecular Immunobiology, Biomedical Research Center, Faculty of Medicine, Autonomous University of Coahuila, Torreon, Mexico
| | | | - Dealmy Delgadillo Guzmán
- Department of Pharmacology, Faculty of Medicine, Autonomous University of Coahuila, Torreon, Mexico
| | - Pablo Ruíz Flores
- Department of Genetics, Center for Biomedical Research Faculty of Medicine, Autonomous University of Coahuila, Torreon, Mexico
| | - Francisco Carlos López Márquez
- Department of Molecular Immunobiology, Biomedical Research Center, Faculty of Medicine, Autonomous University of Coahuila, Torreon, Mexico
| |
Collapse
|
108
|
Sachdeva A, Gouge J, Kontovounisios C, Nikolaou S, Ashworth A, Lim K, Chong I. Klotho and the Treatment of Human Malignancies. Cancers (Basel) 2020; 12:cancers12061665. [PMID: 32585905 PMCID: PMC7352559 DOI: 10.3390/cancers12061665] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 06/16/2020] [Indexed: 12/24/2022] Open
Abstract
Klotho was first discovered as an anti-ageing protein linked to a number of age-related disease processes, including cardiovascular, renal, musculoskeletal, and neurodegenerative conditions. Emerging research has also demonstrated a potential therapeutic role for Klotho in cancer biology, which is perhaps unsurprising given that cancer and ageing share similar molecular hallmarks. In addition to functioning as a tumour suppressor in numerous solid tumours and haematological malignancies, Klotho represents a candidate therapeutic target for patients with these diseases, the majority of whom have limited treatment options. Here, we examine contemporary evidence evaluating the anti-neoplastic effects of Klotho and describe the modulation of downstream oncogenic signalling pathways, including Wnt/β-catenin, FGF, IGF1, PIK3K/AKT, TGFβ, and the Unfolded Protein Response. We also discuss possible approaches to developing therapeutic Klotho and consider technological advances that may facilitate the delivery of Klotho through gene therapy.
Collapse
Affiliation(s)
- Aishani Sachdeva
- The Royal Marsden NHS Foundation Trust, London SW6 6JJ, UK; (A.S.); (C.K.)
- Department of Surgery and Cancer, Chelsea and Westminster Hospital, London SW10 9NH, UK;
| | - Jerome Gouge
- Institute of Structural and Molecular Biology, Birkbeck College, London WC1E 7HX, UK;
| | - Christos Kontovounisios
- The Royal Marsden NHS Foundation Trust, London SW6 6JJ, UK; (A.S.); (C.K.)
- Department of Surgery and Cancer, Chelsea and Westminster Hospital, London SW10 9NH, UK;
| | - Stella Nikolaou
- Department of Surgery and Cancer, Chelsea and Westminster Hospital, London SW10 9NH, UK;
| | - Alan Ashworth
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94158, USA;
| | - Kenneth Lim
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202-5181, USA;
| | - Irene Chong
- The Royal Marsden NHS Foundation Trust, London SW6 6JJ, UK; (A.S.); (C.K.)
- The Institute of Cancer Research, London SW3 6JB, UK
- Correspondence:
| |
Collapse
|
109
|
Klotho and fibroblast growth factors 19 and 21 serum concentrations in children and adolescents with normal body weight and obesity and their associations with metabolic parameters. BMC Pediatr 2020; 20:294. [PMID: 32546231 PMCID: PMC7296965 DOI: 10.1186/s12887-020-02199-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 06/11/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Fibroblast growth factor 19 (FGF19), fibroblast growth factor 21 (FGF21) and Klotho are regulators of energy homeostasis. However, in the pediatric population, the relationships between obesity, metabolic disorders and the aforementioned factors have not been clearly investigated. We analyzed the role of FGF19, FGF21 and Klotho protein in children with normal body weight as well as in overweight and obese subjects and explored their associations with insulin resistance (IR) and metabolic syndrome (MS) and its components. METHODS This was a cross-sectional study conducted in a group of hospitalized children and adolescents. Laboratory investigations included serum analysis of FGF19, FGF21, and Klotho with ELISA kits as well as the analysis of the lipid profile and ALT serum concentrations. Moreover, each subject underwent an oral glucose tolerance test (OGTT) with fasting insulinemia measurement to detect glucose tolerance abnormalities and calculate the Homeostatic Model Assessment of Insulin Resistance (HOMA-IR) index. Furthermore, the clinical analysis included blood pressure measurement, body fat percentage estimation and assessment of the prevalence of MS and its components. RESULTS The study was conducted with 174 children/adolescents aged 6-17 years with normal body weight (N = 48), obesity (N = 92) and overweight (N = 34). Klotho concentration was significantly higher in the obese children [median 168.6 pg/ml (90.2 to 375.9)]) than in the overweight [131.3 pg/ml (78.0 to 313.0)] and normal-body-weight subjects [116.6 pg/ml (38.5 to 163.9)] (p = 0.0334) and was also significantly higher in insulin-resistant children than in insulin-sensitive children [185.3 pg/ml (102.1 to 398.2) vs 132.6 pg/ml (63.9 to 275.6), p = 0.0283]. FGF21 was elevated in patients with MS compared to the FGF21 levels in other subjects [136.2 pg/ml (86.5 to 239.9) vs 82.6 pg/ml (41.8 to 152.4), p = 0.0286]. The multivariable model showed that FGF19 was an independent predictor of IR after adjusting for pubertal stage and BMI Z-score. CONCLUSIONS Klotho levels were associated with body weight status in children and adolescents. Moreover, Klotho, FGF19 and FGF21 concentrations correlated with IR status and/or components of MS.
Collapse
|
110
|
Guo X, Kassab GS. Increased Serum Klotho With Age-Related Aortic Stiffness and Peripheral Vascular Resistance in Young and Middle-Aged Swine. Front Physiol 2020; 11:591. [PMID: 32581850 PMCID: PMC7297143 DOI: 10.3389/fphys.2020.00591] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 05/11/2020] [Indexed: 12/20/2022] Open
Abstract
The anti-aging function of Klotho gene has been implicated in age-related diseases. The physiological importance of Klotho in the progression of arterial stiffness with aging, however, remains unclear. The goal of this study is to determine the correlation of circulating Klotho with early age-related aortic stiffening and peripheral hemodynamics. We measured serum Klotho levels in a group of pigs with age ranges of 1.5-9 years and investigated the relationship between Klotho levels and biomarkers of aortic stiffening with aging, including aortic pulse wave velocity (PWV), augmentation index (AIx), and pulse pressure (PP). The effects of aortic stiffening on peripheral vascular resistance, compliance, and function were also evaluated. We found that increased aortic stiffness occurred at middle age (>5 years old), as evidenced by an increase in PWV and AIx (p < 0.001), but with no changes in blood pressure and PP. With advancing age, increased femoral vascular resistance positively correlated with aortic PWV and AIx (p < 0.01). No significant difference in endothelium function and arterial compliance for femoral and small peripheral arteries was observed between young and middle-aged groups. The serum Klotho levels were lower in young and higher in middle-aged pigs (p < 0.001), and a positive correlation was found between Klotho and aortic PWV, AIx, and femoral vascular resistance (p < 0.01). Our findings suggest that early-aged aortic stiffening has adverse effect on peripheral hemodynamics, independent of blood pressure levels. Elevated Klotho secretion was associated with increased aortic stiffness and peripheral vascular resistance with aging.
Collapse
Affiliation(s)
| | - Ghassan S. Kassab
- California Medical Innovations Institute, San Diego, CA, United States
| |
Collapse
|
111
|
Charoenngam N, Ponvilawan B, Ungprasert P. Lower circulating soluble Klotho level is associated with increased risk of all-cause mortality in chronic kidney disease patients: a systematic review and meta-analysis. Int Urol Nephrol 2020; 52:1543-1550. [PMID: 32462356 DOI: 10.1007/s11255-020-02510-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 05/12/2020] [Indexed: 12/18/2022]
Abstract
PURPOSE This study aimed to investigate the association between circulating soluble Klotho level and risk of all-cause mortality in chronic kidney disease (CKD) patients using systematic review and meta-analysis technique. METHODS Potentially eligible studies were identified from Medline and EMBASE databases from inception to March 2020 using a search strategy that consisted of terms for "Klotho" and "Mortality". Eligible study must be a cohort study that consists of one cohort of CKD patients with higher circulating soluble Klotho level and another cohort of CKD patients with lower circulating soluble Klotho level. The study must also report relative risk (RR), incidence rate ratio, hazard risk ratio or standardized incidence ratio with 95% confidence intervals (95% CIs) comparing all-cause mortality between CKD patients with lower circulating soluble Klotho level versus CKD patients with higher circulating soluble Klotho level. If the study divides patients (per circulating soluble Klotho level) into more than two groups, a comparison between the highest and the lowest group would be extracted. Point estimates with standard errors were retrieved from each study and were combined together using the generic inverse variance method. RESULTS A total of 2964 articles were retrieved. After two rounds of an independent review by two investigators, six prospective cohort studies met the eligibility criteria and were included into the meta-analysis. CKD patients with lower circulating soluble Klotho level had a significantly increased risk of all-cause mortality with the pooled risk ratio of 1.88 (95% CI 1.29-2.74; I2 0%). The funnel plot was fairly symmetric and did not reveal any suggestive evidence of publication bias. CONCLUSION The current study found a significant association between lower circulating soluble Klotho level and increased risk of all-cause mortality in CKD patients. However, this meta-analysis carries some limitations, including relatively small sample size, lack of adjustment for potential confounders and between-study heterogeneity in baseline characteristics of the patients and cut-off values used to categorize patients into higher and lower circulating serum Klotho level group.
Collapse
Affiliation(s)
- Nipith Charoenngam
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Prannok Road, Bangkoknoi, Bangkok, 10700, Thailand.
| | - Ben Ponvilawan
- Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Patompong Ungprasert
- Department of Rheumatic and Immunologic Diseases, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
112
|
Ma Z, Liu J, Li J, Jiang H, Kong J. Klotho ameliorates the onset and progression of cataract via suppressing oxidative stress and inflammation in the lens in streptozotocin-induced diabetic rats. Int Immunopharmacol 2020; 85:106582. [PMID: 32438076 DOI: 10.1016/j.intimp.2020.106582] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 05/04/2020] [Accepted: 05/07/2020] [Indexed: 12/25/2022]
Abstract
Increased oxidative stress and inflammation play an important role in the pathogenesis of diabetic cataract. Klotho, known as an anti-ageing protein, has antioxidative and anti-inflammatory properties. Klotho is expressed in limited tissues including the lens. Here we examined whether klotho expression is decreased in diabetic lens and, if so, whether klotho treatment can prevent diabetic cataract formation. Streptozotocin (STZ)-induced diabetic rats and age-matched control rats were treated with vehicle or klotho protein, starting at 1 week after STZ injection. Twelve weeks after treatment, cataract formation was observed in diabetic rats but not control rats. Cataract formation and scores were significantly less in klotho-treated diabetic rats than vehicle-treated diabetic rats. Levels of klotho in plasma, aqueous humor and lens were significantly decreased in vehicle-treated diabetic rats, compared with control rats, but were restored in klotho-treated diabetic rats. Additionally, vehicle-treated diabetic rats had increased oxidative stress and inflammation in the lens, which were associated with decreased antioxidant transcriptional master regulator Nrf2 activity and increased transcription factor NF-κB activity. All of these findings were ameliorated in klotho-treated diabetic rats. Notably, klotho treatment did not alter blood glucose in diabetic rats. These results indicate that klotho reduction may increase susceptibility of the lens to oxidative and inflammatory insults, promoting cataract formation under diabetic conditions. Klotho treatment can ameliorate the onset and progression of diabetic cataract via enhancing Nrf2-mediated antioxidant defense and suppressing NF-κB-mediated inflammatory responses. Klotho in the lens may be a novel therapeutic target for prevention of cataract formation in diabetes.
Collapse
Affiliation(s)
- Zhongxu Ma
- Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Vision Science, Clinical College of Ophthalmology, Tianjin Medical University, China.
| | - Jingjing Liu
- The Fourth Affiliated Hospital of China Medical University, Provincial Key Laboratory of Lens Research, China
| | - Jing Li
- Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Vision Science, Clinical College of Ophthalmology, Tianjin Medical University, China
| | - Hao Jiang
- Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Vision Science, Clinical College of Ophthalmology, Tianjin Medical University, China
| | - Jun Kong
- The Fourth Affiliated Hospital of China Medical University, Provincial Key Laboratory of Lens Research, China.
| |
Collapse
|
113
|
Kanasaki K. N-acetyl-seryl-aspartyl-lysyl-proline is a valuable endogenous antifibrotic peptide for kidney fibrosis in diabetes: An update and translational aspects. J Diabetes Investig 2020; 11:516-526. [PMID: 31997585 PMCID: PMC7232267 DOI: 10.1111/jdi.13219] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 01/23/2020] [Accepted: 01/24/2020] [Indexed: 02/06/2023] Open
Abstract
N-acetyl-seryl-aspartyl-lysyl-proline (AcSDKP) is an endogenous peptide that has been confirmed to show excellent organ-protective effects. Even though originally discovered as a modulator of hemotopoietic stem cells, during the recent two decades, AcSDKP has been recognized as valuable antifibrotic peptide. The antifibrotic mechanism of AcSDKP is not yet clear; we have established that AcSDKP could target endothelial-mesenchymal transition program through the induction of the endothelial fibroblast growth factor receptor signaling pathway. Also, recent reports suggested the clinical significance of AcSDKP. The aim of this review was to update recent advances of the mechanistic action of AcSDKP and discuss translational research aspects.
Collapse
Affiliation(s)
- Keizo Kanasaki
- Internal Medicine 1Faculty of MedicineShimane UniversityIzumoJapan
- Department of Diabetology and EndocrinologyKanazawa Medical UniversityUchinadaJapan
- Division of Anticipatory Molecular Food Science and TechnologyKanazawa Medical UniversityUchinadaJapan
| |
Collapse
|
114
|
Cheikhi A, Barchowsky A, Sahu A, Shinde SN, Pius A, Clemens ZJ, Li H, Kennedy CA, Hoeck JD, Franti M, Ambrosio F. Klotho: An Elephant in Aging Research. J Gerontol A Biol Sci Med Sci 2020; 74:1031-1042. [PMID: 30843026 DOI: 10.1093/gerona/glz061] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Indexed: 12/12/2022] Open
Abstract
The year 2017 marked the 20th anniversary of the first publication describing Klotho. This single protein was and is remarkable in that its absence in mice conferred an accelerated aging, or progeroid, phenotype with a dramatically shortened life span. On the other hand, genetic overexpression extended both health span and life span by an impressive 30%. Not only has Klotho deficiency been linked to a number of debilitating age-related illnesses but many subsequent reports have lent credence to the idea that Klotho can compress the period of morbidity and extend the life span of both model organisms and humans. This suggests that Klotho functions as an integrator of organ systems, making it both a promising tool for advancing our understanding of the biology of aging and an intriguing target for interventional studies. In this review, we highlight advances in our understanding of Klotho as well as key challenges that have somewhat limited our view, and thus translational potential, of this potent protein.
Collapse
Affiliation(s)
- Amin Cheikhi
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh.,Division of Geriatric Medicine, Department of Medicine, University of Pittsburgh
| | - Aaron Barchowsky
- Department of Environmental and Occupational Health, University of Pittsburgh.,Department of Pharmacology and Chemical Biology, University of Pittsburgh
| | - Amrita Sahu
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh.,Department of Environmental and Occupational Health, University of Pittsburgh
| | - Sunita N Shinde
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh
| | - Abish Pius
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh
| | - Zachary J Clemens
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh
| | - Hua Li
- Department of Biotherapeutics Discovery, Research Division, Boehringer Ingelheim Pharmaceuticals, Inc. Ridgefield, Connecticut
| | - Charles A Kennedy
- Department of Research Beyond Borders, Regenerative Medicine, Boehringer Ingelheim Pharmaceuticals, Inc. Ridgefield, Connecticut
| | - Joerg D Hoeck
- Department of Research Beyond Borders, Regenerative Medicine, Boehringer Ingelheim Pharmaceuticals, Inc. Ridgefield, Connecticut
| | - Michael Franti
- Department of Research Beyond Borders, Regenerative Medicine, Boehringer Ingelheim Pharmaceuticals, Inc. Ridgefield, Connecticut
| | - Fabrisia Ambrosio
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh.,Department of Environmental and Occupational Health, University of Pittsburgh.,Department of Bioengineering, University of Pittsburgh, Pennsylvania.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pennsylvania
| |
Collapse
|
115
|
Yang K, Yang J, Bi X, Yu Z, Xiao T, Huang Y, Liu Y, Xiong J, Zhao J. Serum Klotho, Cardiovascular Events, and Mortality in Nondiabetic Chronic Kidney Disease. Cardiorenal Med 2020; 10:175-187. [DOI: 10.1159/000506380] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 02/03/2020] [Indexed: 11/19/2022] Open
Abstract
Background: Experimental studies indicate that Klotho deficiency is a pathogenic factor for CKD-related complications, including cardiovascular disease (CVD). However, the association between serum Klotho and clinical outcomes in nondiabetic CKD patients needs to be further clarified. We aimed to determine whether serum Klotho levels are associated with CVD events and mortality in predialysis CKD patients without diabetes. Methods: A total of 336 CKD stage 2–5 predialysis patients without diabetes were recruited and followed from the end of 2014 to January 2019 for CVD events and overall mortality. Serum Klotho was detected by ELISA and divided into quartiles (lowest, middle, second highest, and highest quartiles) according to their serum Klotho category. Results: After a median follow-up of 3.52 years (IQR 3.34–3.76), Kaplan-Meier analysis showed that, compared to participants with a Klotho level in the highest quartile (the reference category), those in the lowest Klotho quartile were associated with a higher all-cause mortality risk (HR = 7.05; 95% CI 1.59–31.25) and a higher CVD event risk (HR = 3.02; 95% CI 1.45–6.30). In addition, the middle Klotho quartile was also associated with CVD event risk (HR = 2.56; 95% CI 1.21–5.41). Moreover, in the multivariate-adjusted model, the lowest Klotho quartile remained significantly associated with all-cause mortality (HR = 5.17; 95% CI 1.07–24.96), and the middle Klotho quartile maintained a significant association with CVD event risk (HR = 2.32; 95% CI 1.03–5.21). Conclusion: These results suggest that lower serum Klotho levels are independently associated with overall mortality and CVD events in nondiabetic predialysis CKD patients.
Collapse
|
116
|
Moos WH, Faller DV, Glavas IP, Harpp DN, Kanara I, Mavrakis AN, Pernokas J, Pernokas M, Pinkert CA, Powers WR, Sampani K, Steliou K, Vavvas DG, Zamboni RJ, Kodukula K, Chen X. Klotho Pathways, Myelination Disorders, Neurodegenerative Diseases, and Epigenetic Drugs. Biores Open Access 2020; 9:94-105. [PMID: 32257625 PMCID: PMC7133426 DOI: 10.1089/biores.2020.0004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In this review we outline a rationale for identifying neuroprotectants aimed at inducing endogenous Klotho activity and expression, which is epigenetic action, by definition. Such an approach should promote remyelination and/or stimulate myelin repair by acting on mitochondrial function, thereby heralding a life-saving path forward for patients suffering from neuroinflammatory diseases. Disorders of myelin in the nervous system damage the transmission of signals, resulting in loss of vision, motion, sensation, and other functions depending on the affected nerves, currently with no effective treatment. Klotho genes and their single-pass transmembrane Klotho proteins are powerful governors of the threads of life and death, true to the origin of their name, Fates, in Greek mythology. Among its many important functions, Klotho is an obligatory co-receptor that binds, activates, and/or potentiates critical fibroblast growth factor activity. Since the discovery of Klotho a little over two decades ago, it has become ever more apparent that when Klotho pathways go awry, oxidative stress and mitochondrial dysfunction take over, and age-related chronic disorders are likely to follow. The physiological consequences can be wide ranging, potentially wreaking havoc on the brain, eye, kidney, muscle, and more. Central nervous system disorders, neurodegenerative in nature, and especially those affecting the myelin sheath, represent worthy targets for advancing therapies that act upon Klotho pathways. Current drugs for these diseases, even therapeutics that are disease modifying rather than treating only the symptoms, leave much room for improvement. It is thus no wonder that this topic has caught the attention of biomedical researchers around the world.
Collapse
Affiliation(s)
- Walter H. Moos
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of California San Francisco, San Francisco, San Francisco, California
- ShangPharma Innovation, Inc., South San Francisco, California
| | - Douglas V. Faller
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
- Cancer Research Center, Boston University School of Medicine, Boston, Massachusetts
| | - Ioannis P. Glavas
- Department of Ophthalmology, New York University School of Medicine, New York, New York
| | - David N. Harpp
- Department of Chemistry, McGill University, Montreal, Canada
| | | | - Anastasios N. Mavrakis
- Department of Medicine, Tufts University School of Medicine, St. Elizabeth's Medical Center, Boston, Massachusetts
| | - Julie Pernokas
- Advanced Dental Associates of New England, Woburn, Massachusetts
| | - Mark Pernokas
- Advanced Dental Associates of New England, Woburn, Massachusetts
| | - Carl A. Pinkert
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Whitney R. Powers
- Department of Health Sciences, Boston University, Boston, Massachusetts
- Department of Anatomy, Boston University School of Medicine, Boston, Massachusetts
| | - Konstantina Sampani
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
- Beetham Eye Institute, Joslin Diabetes Center, Boston, Massachusetts
| | - Kosta Steliou
- Cancer Research Center, Boston University School of Medicine, Boston, Massachusetts
- PhenoMatriX, Inc., Natick, Massachusetts
| | - Demetrios G. Vavvas
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
- Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts
| | | | | | - Xiaohong Chen
- Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
- Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts
| |
Collapse
|
117
|
Zhong X, Jagarlapudi S, Weng Y, Ly M, Rouse JC, McClure K, Ishino T, Zhang Y, Sousa E, Cohen J, Tzvetkova B, Cote K, Scarcelli JJ, Johnson K, Palandra J, Apgar JR, Yaddanapudi S, Gonzalez-Villalobos RA, Opsahl AC, Lam K, Yao Q, Duan W, Sievers A, Zhou J, Ferguson D, D'Antona A, Zollner R, Zhu HL, Kriz R, Lin L, Clerin V. Structure-function relationships of the soluble form of the antiaging protein Klotho have therapeutic implications for managing kidney disease. J Biol Chem 2020; 295:3115-3133. [PMID: 32005658 PMCID: PMC7062171 DOI: 10.1074/jbc.ra119.012144] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/20/2020] [Indexed: 01/28/2023] Open
Abstract
The fortuitously discovered antiaging membrane protein αKlotho (Klotho) is highly expressed in the kidney, and deletion of the Klotho gene in mice causes a phenotype strikingly similar to that of chronic kidney disease (CKD). Klotho functions as a co-receptor for fibroblast growth factor 23 (FGF23) signaling, whereas its shed extracellular domain, soluble Klotho (sKlotho), carrying glycosidase activity, is a humoral factor that regulates renal health. Low sKlotho in CKD is associated with disease progression, and sKlotho supplementation has emerged as a potential therapeutic strategy for managing CKD. Here, we explored the structure-function relationship and post-translational modifications of sKlotho variants to guide the future design of sKlotho-based therapeutics. Chinese hamster ovary (CHO)- and human embryonic kidney (HEK)-derived WT sKlotho proteins had varied activities in FGF23 co-receptor and β-glucuronidase assays in vitro and distinct properties in vivo Sialidase treatment of heavily sialylated CHO-sKlotho increased its co-receptor activity 3-fold, yet it remained less active than hyposialylated HEK-sKlotho. MS and glycopeptide-mapping analyses revealed that HEK-sKlotho is uniquely modified with an unusual N-glycan structure consisting of N,N'-di-N-acetyllactose diamine at multiple N-linked sites, one of which at Asn-126 was adjacent to a putative GalNAc transfer motif. Site-directed mutagenesis and structural modeling analyses directly implicated N-glycans in Klotho's protein folding and function. Moreover, the introduction of two catalytic glutamate residues conserved across glycosidases into sKlotho enhanced its glucuronidase activity but decreased its FGF23 co-receptor activity, suggesting that these two functions might be structurally divergent. These findings open up opportunities for rational engineering of pharmacologically enhanced sKlotho therapeutics for managing kidney disease.
Collapse
Affiliation(s)
- Xiaotian Zhong
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139.
| | - Srinath Jagarlapudi
- Internal Medicine, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Yan Weng
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Mellisa Ly
- Analytical Research and Development, Biotherapeutics Pharmaceutical Sciences, Pfizer Inc., Andover, Massachusetts 01810
| | - Jason C Rouse
- Analytical Research and Development, Biotherapeutics Pharmaceutical Sciences, Pfizer Inc., Andover, Massachusetts 01810
| | - Kim McClure
- Internal Medicine, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Tetsuya Ishino
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Yan Zhang
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Eric Sousa
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Justin Cohen
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Boriana Tzvetkova
- Analytical Research and Development, Biotherapeutics Pharmaceutical Sciences, Pfizer Inc., Andover, Massachusetts 01810
| | - Kaffa Cote
- Analytical Research and Development, Biotherapeutics Pharmaceutical Sciences, Pfizer Inc., Andover, Massachusetts 01810
| | - John J Scarcelli
- Cell Line Development, Biotherapeutics Pharmaceutical Sciences, Pfizer Inc., Andover, Massachusetts 01810
| | - Keith Johnson
- Analytical Research and Development, Biotherapeutics Pharmaceutical Sciences, Pfizer Inc., Andover, Massachusetts 01810
| | - Joe Palandra
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - James R Apgar
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Suma Yaddanapudi
- Internal Medicine, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | | | - Alan C Opsahl
- Internal Medicine, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Khetemenee Lam
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Qing Yao
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Weili Duan
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Annette Sievers
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Jing Zhou
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Darren Ferguson
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Aaron D'Antona
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Richard Zollner
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Hongli L Zhu
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Ron Kriz
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Laura Lin
- BioMedicine Design, Pfizer Worldwide Research, Cambridge, Massachusetts 02139
| | - Valerie Clerin
- Internal Medicine, Pfizer Worldwide Research, Cambridge, Massachusetts 02139.
| |
Collapse
|
118
|
Munc13 mediates klotho-inhibitable diacylglycerol-stimulated exocytotic insertion of pre-docked TRPC6 vesicles. PLoS One 2020; 15:e0229799. [PMID: 32134975 PMCID: PMC7058344 DOI: 10.1371/journal.pone.0229799] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 02/13/2020] [Indexed: 11/29/2022] Open
Abstract
α-Klotho is a type 1 transmembrane protein that exhibits aging suppression function. The large amino-terminal extracellular domain of α-klotho is shed as soluble klotho (sKlotho) and functions as a circulating cardioprotective hormone. Diacylglycerol (DAG)-activated calcium-permeable TRPC6 channel plays a critical role in stress-induced cardiac remodeling. DAG activates TRPC6 by acting directly on the channel to increase its activity and by stimulation of channel exocytosis. sKlotho protects the heart by inhibiting DAG stimulation of TRPC6 exocytosis. How DAG stimulates TRPC6 exocytosis and thereby inhibition by sKlotho are unknown. Using a compound that directly activates TRPC6 without affecting channel exocytosis, we validate that sKlotho selectively blocks DAG stimulation of channel exocytosis. We further show that DAG stimulates exocytosis of TRPC6-containing vesicles pre-docked to the plasma membrane. Mnuc13 family proteins play important roles in the proper assembly of SNARE proteins and priming the vesicle competent for fusion. We show that DAG stimulates TRPC6 exocytosis by targeting to the C1 domain of Munc13-2. The results provide fresh insights into the molecular mechanism by which DAG regulates vesicle fusion and how sKlotho protects the heart against injury.
Collapse
|
119
|
Prud'homme GJ, Glinka Y, Kurt M, Liu W, Wang Q. Systemic Klotho therapy protects against insulitis and enhances beta-cell mass in NOD mice. Biochem Biophys Res Commun 2020; 525:693-698. [PMID: 32139120 DOI: 10.1016/j.bbrc.2020.02.123] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 02/19/2020] [Indexed: 12/17/2022]
Abstract
The levels of the anti-aging protein α-Klotho, in its soluble form (s-Klotho), are depressed in the circulation of patients with type 1 diabetes (T1D) or type 2 diabetes (T2D). Gene transfer experiments have suggested a protective role for β-cell specific expression of α-Klotho in murine models of T1D and T1D, but these approaches are not easily translatable to clinical therapy. It is unknown whether systemic s-Klotho protein treatment ameliorates disease in T1D, which is characterized by autoimmune destruction of β cells. We previously reported from in vitro experiments with β cells that s-Klotho increases insulin secretion, reduces cells death and promotes β-cell replication. Here, we investigated s-Klotho protein therapy in NOD mice, which have autoimmune T1D. We observed that diabetic NOD mice have significantly lower plasma levels of s-Klotho, compared to their non-diabetic counterparts. To examine in vivo effects of Klotho, we treated NOD mice with s-Klotho protein, or with a Klotho blocking antibody. Systemic treatment with s-Klotho ameliorated diabetes; notably increasing β-cell replication and total β-cell mass. Klotho expression was increased locally in the islets. s-Klotho also markedly reduced immune-cell infiltration of islets (insulitis). In contrast, administration of the Klotho antibody was detrimental, and aggravated the loss of β-cell mass. Thus, s-Klotho has protective effects in this model of T1D, and this appears to depend on a combination of increased β-cell replication and reduced insulitis. These findings suggest that s-Klotho might be effective as a new therapeutic agent for T1D.
Collapse
Affiliation(s)
- Gérald J Prud'homme
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine, Unity Health Toronto (St. Michael's Hospital Site), Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada.
| | - Yelena Glinka
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, Ontario, Canada.
| | - Merve Kurt
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, Ontario, Canada.
| | - Wenjuan Liu
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, Ontario, Canada.
| | - Qinghua Wang
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, Ontario, Canada; Department of Physiology and Department of Medicine, University of Toronto, Toronto, Ontario, Canada; Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China; Division of Endocrinology and Metabolism, Unity Health Toronto (St. Michael's Hospital Site), Toronto, Ontario, Canada.
| |
Collapse
|
120
|
The E, Yao Q, Zhang P, Zhai Y, Ao L, Fullerton DA, Meng X. Mechanistic Roles of Matrilin-2 and Klotho in Modulating the Inflammatory Activity of Human Aortic Valve Cells. Cells 2020; 9:cells9020385. [PMID: 32046115 PMCID: PMC7072362 DOI: 10.3390/cells9020385] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 01/30/2020] [Accepted: 02/05/2020] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Calcific aortic valve disease (CAVD) is a chronic inflammatory disease. Soluble extracellular matrix (ECM) proteins can act as damage-associated molecular patterns and may induce valvular inflammation. Matrilin-2 is an ECM protein and has been found to elevate the pro-osteogenic activity in human aortic valve interstitial cells (AVICs). Klotho, an anti-aging protein, appears to have anti-inflammatory properties. The effect of matrilin-2 and Klotho on AVIC inflammatory responses remains unclear. METHODS AND RESULTS Isolated human AVICs were exposed to matrilin-2. Soluble matrilin-2 induced the production of ICAM-1, MCP-1, and IL-6. It also induced protein kinase R (PKR) activation via Toll-like receptor (TLR) 2 and 4. Pretreatment with PKR inhibitors inhibited NF-κB activation and inflammatory mediator production induced by matrilin-2. Further, recombinant Klotho suppressed PKR and NF-κB activation and markedly reduced the production of inflammatory mediators in human AVICs exposed to matrilin-2. CONCLUSIONS This study revealed that soluble matrilin-2 upregulates AVIC inflammatory activity via activation of the TLR-PKR-NF-κB pathway and that Klotho is potent to suppress AVIC inflammatory responses to a soluble ECM protein through inhibiting PKR. These novel findings indicate that soluble matrilin-2 may accelerate the progression of CAVD by inducing valvular inflammation and that Klotho has the potential to suppress valvular inflammation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xianzhong Meng
- Correspondence: ; Tel.: +1-303-724-6303; Fax: +1-303-724-6330
| |
Collapse
|
121
|
Relationship between plasma S-Klotho and cardiometabolic risk in sedentary adults. Aging (Albany NY) 2020; 12:2698-2710. [PMID: 31958773 PMCID: PMC7041759 DOI: 10.18632/aging.102771] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 01/12/2020] [Indexed: 12/15/2022]
Abstract
This cross-sectional study investigates the relationship between the shed form of the Klotho protein (S-Klotho) in plasma, and cardiometabolic risk in healthy, sedentary adults. The study subjects were 214 healthy, sedentary adults (~64% women). Data were collected during the baseline assessments of two randomized controlled trials: The FIT-AGEING study (n=74 [~50% women] middle-aged adults aged 40-65 years) and the ACTIBATE study (n=140 [~70% women] young adults aged 18-25 years). A sex-specific cardiometabolic risk score was calculated for each subject based on waist circumference, systolic and diastolic blood pressure, and plasma glucose, high-density lipoprotein cholesterol and triglycerides. A significant inverse relationship was detected between S-Klotho and the cardiometabolic risk score of both the middle-aged men and women (β=-0.658, R2=0.433, P<0.001 and β=-0.442, R2=0.195, P=0.007) which persisted after adjusting for actual age, energy intake, and VO2max. No significant association was found between S-Klotho and cardiometabolic risk score for the young, healthy adults (P>0.5), nor for the young, healthy men and women when analysed separately (all P>0.1). In conclusion, in healthy, sedentary, middle-aged adults, but not in young, healthy, sedentary adults, higher plasma S-Klotho concentrations are associated with a lower cardiometabolic risk score.
Collapse
|
122
|
Barbosa AM, Martel F. Targeting Glucose Transporters for Breast Cancer Therapy: The Effect of Natural and Synthetic Compounds. Cancers (Basel) 2020; 12:cancers12010154. [PMID: 31936350 PMCID: PMC7016663 DOI: 10.3390/cancers12010154] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/06/2020] [Accepted: 01/07/2020] [Indexed: 02/07/2023] Open
Abstract
Reprogramming of cellular energy metabolism is widely accepted to be a cancer hallmark. The deviant energetic metabolism of cancer cells-known as the Warburg effect-consists in much higher rates of glucose uptake and glycolytic oxidation coupled with the production of lactic acid, even in the presence of oxygen. Consequently, cancer cells have higher glucose needs and thus display a higher sensitivity to glucose deprivation-induced death than normal cells. So, inhibitors of glucose uptake are potential therapeutic targets in cancer. Breast cancer is the most commonly diagnosed cancer and a leading cause of cancer death in women worldwide. Overexpression of facilitative glucose transporters (GLUT), mainly GLUT1, in breast cancer cells is firmly established, and the consequences of GLUT inhibition and/or knockout are under investigation. Herein we review the compounds, both of natural and synthetic origin, found to interfere with uptake of glucose by breast cancer cells, and the consequences of interference with that mechanism on breast cancer cell biology. We will also present data where the interaction with GLUT is exploited in order to increase the efficiency or selectivity of anticancer agents, in breast cancer cells.
Collapse
Affiliation(s)
- Ana M. Barbosa
- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, 4169-007 Porto, Portugal;
| | - Fátima Martel
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
- Correspondence: ; Tel.: +351-22-042-6654
| |
Collapse
|
123
|
Welc SS, Wehling-Henricks M, Kuro-o M, Thomas KA, Tidball JG. Modulation of Klotho expression in injured muscle perturbs Wnt signalling and influences the rate of muscle growth. Exp Physiol 2020; 105:132-147. [PMID: 31724771 PMCID: PMC6938556 DOI: 10.1113/ep088142] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
NEW FINDINGS What is the central question of this study? Does modulating the expression of Klotho affect myogenesis following acute injury of healthy, non-senescent muscle? What is the main finding and its importance? Klotho can accelerate muscle growth following acute injury of healthy, adult mice, which supports the possibility that increased delivery of Klotho could have therapeutic value for improving repair of damaged muscle. ABSTRACT Skeletal muscle injuries activate a complex programme of myogenesis that can restore normal muscle structure. We tested whether modulating the expression of klotho influenced the response of mouse muscles to acute injury. Our findings show that klotho expression in muscle declines at 3 days post-injury. That reduction in klotho expression coincided with elevated expression of targets of Wnt signalling (Ccnd1; Myc) and increased MyoD+ muscle cell numbers, reflecting the onset of myogenic cell differentiation. klotho expression subsequently increased at 7 days post-injury with elevated expression occurring primarily in inflammatory lesions, which was accompanied by reduced expression of Wnt target genes (Ccnd1: 91%; Myc: 96%). Introduction of a klotho transgene maintained high levels of klotho expression over the course of muscle repair and attenuated the increases in Ccnd1 and Myc expression that occurred at 3 days post-injury. Correspondingly, transgene expression reduced Wnt signalling in Pax7+ cells, reflected by reductions in Pax7+ cells expressing active β-catenin, and reduced the numbers of MyoD+ cells at 3 days post-injury. At 21 days post-injury, muscles in klotho transgenic mice showed increased Pax7+ and decreased myogenin+ cell densities and large increases in myofibre size. Likewise, treating myogenic cells in vitro with Klotho reduced Myod expression but did not affect Pax7 expression. Muscle inflammation was only slightly modulated by increased klotho expression, initially reducing the expression of M2-biased macrophage markers Cd163 and Cd206 at 3 days post-injury and later increasing the expression of pan-macrophage marker F480 and Cd68 at 21 days post-injury. Collectively, our study shows that Klotho modulates myogenesis and that increased expression accelerates muscle growth after injury.
Collapse
Affiliation(s)
- Steven S. Welc
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA
- Indiana University School of Medicine, 635 Barnhill Drive, MS-332, Indianapolis, IN 46202
| | | | - Makoto Kuro-o
- Division of Anti-Aging Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Kyle A. Thomas
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA
| | - James G. Tidball
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, CA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA
| |
Collapse
|
124
|
Rao Z, Zheng L, Huang H, Feng Y, Shi R. α-Klotho Expression in Mouse Tissues Following Acute Exhaustive Exercise. Front Physiol 2019; 10:1498. [PMID: 31920703 PMCID: PMC6919267 DOI: 10.3389/fphys.2019.01498] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 11/25/2019] [Indexed: 12/22/2022] Open
Abstract
α-Klotho, a multifunctional protein, has been demonstrated to protect tissues from injury via anti-oxidation and anti-inflammatory effects. The expression of α-klotho is regulated by several physiological and pathological factors, including acute inflammatory stress, oxidative stress, hypertension, and chronic renal failure. Exhaustive exercise has been reported to result in tissue damage, which is induced by inflammation, oxidative stress, and energy metabolism disturbance. However, little is known about the effects of exhaustive exercise on the expression of α-klotho in various tissues. To determine the effects, the treadmill exhaustion test in mice was performed and the mice were sacrificed at different time points following exhaustive exercise. Our results confirmed that the full-length (130 kDa) and shorter-form (65 kDa) α-klotho were primarily expressed in the kidneys. Moreover, we found that, except for the kidneys and brain, other tissues primarily expressed the shorter-form α-klotho, including liver, which was in contrast to previous reports. Furthermore, the shorter-form α-klotho was decreased immediately following the acute exhaustive exercise and was then restored to the pre-exercise level or even higher levels in the next few days. Our results indicate that α-klotho may play a key role in the body exhaustion and recovery following exhaustive exercise.
Collapse
Affiliation(s)
- Zhijian Rao
- College of Physical Education, Shanghai Normal University, Shanghai, China.,School of Kinesiology, Shanghai University of Sport, Shanghai, China.,Department of Kinesiology and Physiology, East Carolina University, Greenville, NC, United States
| | - Lifang Zheng
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Hu Huang
- Department of Kinesiology and Physiology, East Carolina University, Greenville, NC, United States
| | - Yu Feng
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Rengfei Shi
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
125
|
Xiao Z, King G, Mancarella S, Munkhsaikhan U, Cao L, Cai C, Quarles LD. FGF23 expression is stimulated in transgenic α-Klotho longevity mouse model. JCI Insight 2019; 4:132820. [PMID: 31801907 PMCID: PMC6962016 DOI: 10.1172/jci.insight.132820] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 10/23/2019] [Indexed: 12/21/2022] Open
Abstract
Observations in transgenic α-Klotho (Kl) mice (KlTg) defined the antiaging role of soluble Klotho (sKL130). A genetic translocation that elevates sKL levels in humans is paradoxically associated with increased circulating fibroblast growth factor 23 (FGF23) levels and the potential of both membrane KL (mKL135) and sKL130 to act as coreceptors for FGF23 activation of fibroblast growth factor receptors (FGFRs). Neither FGF23 expression nor the contributions of FGF23, mKL135, and sKL130 codependent and independent functions have been investigated in KlTg mice. In the current study, we examined the effects of Kl overexpression on FGF23 levels and functions in KlTg mice. We found that mKL135 but not sKL130 stimulated FGF23 expression in osteoblasts, leading to elevated Fgf23 bone expression and circulating levels in KlTg mice. Elevated FGF23 suppressed 1,25(OH)2D and parathyroid hormone levels but did not cause hypophosphatemic rickets in KlTg mice. KlTg mice developed low aldosterone-associated hypertension but not left ventricular hypertrophy. Mechanistically, we found that mKL135 and sKL130 are essential cofactors for FGF23-mediated ERK activation but that they inhibited FGF23 stimulation of PLC-γ and PI3K/AKT signaling. Thus, increased longevity in KlTg mice occurs in the presence of excess FGF23 that interacts with mKL and sKL to bias FGFR pathways.
Collapse
Affiliation(s)
- Zhousheng Xiao
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Gwendalyn King
- Department of Biology, Creighton University, Omaha, Nebraska, USA
| | | | - Undral Munkhsaikhan
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Li Cao
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Chun Cai
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Leigh Darryl Quarles
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
126
|
Ewendt F, Föller M. p38MAPK controls fibroblast growth factor 23 (FGF23) synthesis in UMR106-osteoblast-like cells and in IDG-SW3 osteocytes. J Endocrinol Invest 2019; 42:1477-1483. [PMID: 31201665 DOI: 10.1007/s40618-019-01073-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 06/10/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND p38 mitogen-activated protein kinase (p38MAPK) is a serine/threonine kinase activated by cellular stress stimuli including radiation, osmotic shock, and inflammation and influencing apoptosis, cell proliferation, and autophagy. Moreover, p38MAPK induces transcriptional activity of the transcription factor complex NFκB mediating multiple pro-inflammatory cellular responses. Fibroblast growth factor 23 (FGF23) is produced by bone cells, and regulates renal phosphate and vitamin D metabolism as a hormone. FGF23 expression is enhanced by NFκB. Here, we analyzed the relevance of p38MAPK activity for the production of FGF23. METHODS Fgf23 expression was analyzed by qRT-PCR and FGF23 protein by ELISA in UMR106 osteoblast-like cells and in IDG-SW3 osteocytes. RESULTS Inhibition of p38MAPK with SB203580 or SB202190 significantly down-regulated Fgf23 expression and FGF23 protein expression. Conversely, p38MAPK activator anisomycin increased the abundance of Fgf23 mRNA. NFκB inhibitors wogonin and withaferin A abrogated the stimulatory effect of anisomycin on Fgf23 gene expression. CONCLUSION p38MAPK induces FGF23 formation, an effect at least in part dependent on NFκB activity.
Collapse
Affiliation(s)
- F Ewendt
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, 06120, Halle (Saale), Germany
| | - M Föller
- Institute of Physiology, University of Hohenheim, Garbenstraße 30, 70599, Stuttgart, Germany.
| |
Collapse
|
127
|
Recombinant Klotho Protects Human Periodontal Ligament Stem Cells by Regulating Mitochondrial Function and the Antioxidant System during H 2O 2-Induced Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9261565. [PMID: 31885825 PMCID: PMC6914990 DOI: 10.1155/2019/9261565] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 10/29/2019] [Indexed: 01/08/2023]
Abstract
Human periodontal ligament stem cells (hPDLSCs) are a favourable source for tissue engineering, but oxidative stress conditions during cell culture and transplantation could affect stem cell viability and stemness, leading to failed regeneration. The aim of this study was to evaluate the antioxidant and protective effects of Klotho, an antiageing protein, against cell damage and the loss of osteogenesis in hPDLSCs in H2O2-induced oxidative environments. H2O2 was used as an exogenous reactive oxygen species (ROS) to induce oxidative stress. Recombinant human Klotho protein was administered before H2O2 treatment. Multitechniques were used to assess antioxidant activity, cell damage, and osteogenic ability of hPDLSCs in oxidative stress and the effects of Klotho on hPDLSCs. Mitochondrial function was analyzed by an electron microscopy scan of cellular structure, mitochondrial DNA copy number, and cellular oxygen consumption rate (OCR). Furthermore, we explored the pathway by which Klotho may function to regulate the antioxidant system. We found that pretreatment with recombinant human Klotho protein could enhance SOD activity and reduce intracellular oxidative stress levels. Klotho reduced H2O2-induced cellular damage and eventually maintained the osteogenic differentiation potential of hPDLSCs. Notably, Klotho promoted mitochondrial function and activated antioxidants by negatively regulating the PI3K/AKT/FoxO1 pathway. The findings suggest that Klotho protein enhanced the antioxidative ability of hPDLSCs and protected stem cell viability and stemness from H2O2-induced oxidative stress by restoring mitochondrial functions and the antioxidant system.
Collapse
|
128
|
The Signaling of Cellular Senescence in Diabetic Nephropathy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:7495629. [PMID: 31687085 PMCID: PMC6794967 DOI: 10.1155/2019/7495629] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 07/03/2019] [Accepted: 07/23/2019] [Indexed: 12/13/2022]
Abstract
Diabetic nephropathy is the leading cause of chronic kidney disease (CKD) in western countries. Notably, it has a rapidly rising prevalence in China. The patients, commonly complicated with cardiovascular diseases and neurologic disorders, are at high risk to progress into end-stage renal disease (ESRD) and death. However, the pathogenic mechanisms of diabetic nephropathy have not been determined. Cellular senescence, which recently has gained broad attention, is thought to be an important player in the onset and development of diabetic nephropathy. In this issue, we generally review the mechanisms of cellular senescence in diabetic nephropathy, which involve telomere attrition, DNA damage, epigenetic alterations, mitochondrial dysfunction, loss of Klotho, Wnt/β-catenin signaling activation, persistent inflammation, and accumulation of uremic toxins. Moreover, we highlight the potential therapeutic targets of cellular senescence in diabetic nephropathy and provide important clues for clinical strategies.
Collapse
|
129
|
Soluble klotho regulates the function of salivary glands by activating KLF4 pathways. Aging (Albany NY) 2019; 11:8254-8269. [PMID: 31581134 PMCID: PMC6814581 DOI: 10.18632/aging.102318] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 09/21/2019] [Indexed: 12/21/2022]
Abstract
The dysfunction of salivary glands commonly induces dry mouth, infections, and dental caries caused by a lack of saliva. This study was performed to determine the genetic and functional changes in salivary glands using a klotho (-/-) mouse model. Here, we confirmed the attenuation of KLF4 expression in the salivary glands of klotho (-/-) mice. Soluble klotho overexpression induced KLF4 transcription and KLF4-mediated signaling pathways, including mTOR, AMPK, and SOD1/2. Silencing klotho via siRNA significantly down-regulated KLF4 expression. Additionally, we monitored the function of salivary glands and soluble klotho and/or KLF4 responses and demonstrated that soluble klotho increased the expression of KLF4 and markers of salivary gland function (α-amylase, ZO-1, and Aqua5) in primary cultured salivary gland cells from wild type and klotho (-/-) mice. In a 3D culture system, cell sphere aggregates were observed in soluble klotho- or KLF4-expressing cells and exhibited higher expression levels of salivary gland function-related proteins than those in nontransfected cells. These results suggest that activation of the klotho-mediated KLF4 signaling pathway contributes to potentiating the function of salivary glands.
Collapse
|
130
|
Dehghani A, Hafizibarjin Z, Najjari R, Kaseb F, Safari F. Resveratrol and 1,25-dihydroxyvitamin D co-administration protects the heart against D-galactose-induced aging in rats: evaluation of serum and cardiac levels of klotho. Aging Clin Exp Res 2019; 31:1195-1205. [PMID: 30484255 DOI: 10.1007/s40520-018-1075-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 11/14/2018] [Indexed: 01/14/2023]
Abstract
The current study investigates the cooperative cardioprotective effect of calcitriol (active form of vitamin D) combined with resveratrol in a rat model of D-galactose (D.gal)-induced aging. Male Wistar rats received resveratrol (D.gal + Res), calcitriol (D.gal + Cal), or a combination of them (D.gal + Res + Cal). Intact animals served as control (Ctl). Blood pressure (BP) was recorded by cannulation of the left carotid artery. Fibrosis and cell size were assessed by Masson's trichrome and hematoxylin-eosin staining, respectively. Cardiac and serum level of antiaging protein, klotho, was measured by ELISA assay method. Gene expression was evaluated by real-time RT-PCR. Biochemical tests were performed according to the standardized method. In D.gal + Res + Cal group, BP, heart weight-to-body weight ratio, and cardiomyocytes size decreased significantly compared with D-gal group. The cardiac transcription levels of catalase and superoxide dismutase 1 and 2 were upregulated in D.gal + Res + Cal compared to the D.gal group (P < 0.001, P < 0.05, P < 0.05, respectively). Increased level of malondialdehyde was observed in D.gal group (P < 0.01 vs. Ctl) which was normalized partially in D.gal + Res + Cal group (P < 0.05). Catalase and superoxide dismutase activity also increased in D.gal + Res + Cal group (P < 0.01 vs. D.gal). Cardiac Klotho, as the antiaging protein, remained unchanged at mRNA and protein levels among the experimental groups. The serum level of Klotho did not change significantly in D.gal group; however, in D.gal + Res + Cal group, serum klotho concentration was increased (P < 0.05 vs. D.gal). It could be concluded that co-administration of resveratrol and vitamin D protects the heart against aging-induced damage by the modulation of hemodynamic parameters and antioxidant status of the heart. Furthermore, increased serum level of klotho could be a novel mechanism for antiaging effects of resveratrol and vitamin D.
Collapse
Affiliation(s)
- Ali Dehghani
- Department of Elderly Health, Faculty of Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Zeynab Hafizibarjin
- Department of Physiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Razieh Najjari
- Department of Physiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Kaseb
- Faculty of Paramedicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Safari
- Department of Physiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
- Biotechnology Research Center, International Campus, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
131
|
Franklin AD, Saqibuddin J, Stephens K, Birkett R, Marsden L, Ernst LM, Mestan KK. Cord blood alpha klotho is decreased in small for gestational age preterm infants with placental lesions of accelerated aging. Placenta 2019; 87:1-7. [PMID: 31499336 DOI: 10.1016/j.placenta.2019.08.088] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 08/10/2019] [Accepted: 08/29/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND Intrauterine growth restriction is often accompanied by placental vascular disease, of which histologic maternal vascular malperfusion is prominent. Maternal vascular malperfusion is characterized by accelerated villous maturation consistent with placental aging. Alpha klotho is an anti-aging protein produced by the placenta. We hypothesize that cord blood alpha klotho varies with maternal vascular malperfusion and small for gestational age infants through dysregulated angiogenesis. METHODS Nested case-control study of 54 preterm infants (N = 22 small for gestational age infants, 32 appropriate for gestational age infants, mean gestational age = 33.7 ± 2.7 weeks) and validation sample (N = 39) from a longitudinal birth cohort at Prentice Women's Hospital, Chicago, IL. Cord blood alpha klotho was measured via enzyme-linked immunoassay; concentrations were linked to multiplex data of cord blood angiogenic growth factors. RESULTS Median cord blood alpha klotho was decreased in small for gestational age infants (1200 [859, 2083] pg/mL) versus controls (3193 [1703, 3963] pg/mL; p < 0.01) and with severe maternal vascular malperfusion (1170 [760, 2645] pg/mL; P < 0.01), consistent with validation sample. Alpha klotho was decreased with maternal vascular malperfusion sublesions signifying accelerated villous maturation, including increased syncytial knots (1230 [805, 3606] pg/mL; p < 0.05) and distal villous hypoplasia (1170 [770, 3390] pg/mL; p < 0.05). Among 15 angiogenic markers, alpha klotho correlated directly with angiopoietin-2 (beta-coefficient = 2.6, p = 0.01). CONCLUSIONS Cord blood alpha klotho is decreased with small for gestational infants and maternal vascular malperfusion sublesions of accelerated placental villous maturation, and correlated with angiopoietin-2. Alpha klotho may play a role in vascular-mediated accelerated placental aging leading to intrauterine growth restriction.
Collapse
Affiliation(s)
- Andrew D Franklin
- Department of Pediatrics, Division of Neonatology, Northwestern University Feinberg School of Medicine, Ann & Robert H. Lurie Children's Hospital, 225 E Chicago Ave, Chicago, IL, 60611, USA.
| | - Juanita Saqibuddin
- Department of Pediatrics, Division of Neonatology, Northwestern University Feinberg School of Medicine, Ann & Robert H. Lurie Children's Hospital, 225 E Chicago Ave, Chicago, IL, 60611, USA
| | - Kelli Stephens
- Department of Pediatrics, Division of Neonatology, Northwestern University Feinberg School of Medicine, Ann & Robert H. Lurie Children's Hospital, 225 E Chicago Ave, Chicago, IL, 60611, USA
| | - Robert Birkett
- Department of Pediatrics, Division of Neonatology, Northwestern University Feinberg School of Medicine, Ann & Robert H. Lurie Children's Hospital, 225 E Chicago Ave, Chicago, IL, 60611, USA
| | - Lily Marsden
- Utah State Office of the Medical Examiner, 4451 2700 W, Taylorsville, UT, 84129, USA
| | - Linda M Ernst
- Department of Pathology and Laboratory Medicine, NorthShore University HealthSystem, 2560 Ridge Ave, Evanston, IL, 60201, USA
| | - Karen K Mestan
- Department of Pediatrics, Division of Neonatology, Northwestern University Feinberg School of Medicine, Ann & Robert H. Lurie Children's Hospital, 225 E Chicago Ave, Chicago, IL, 60611, USA
| |
Collapse
|
132
|
Scazzone C, Agnello L, Sasso BL, Ragonese P, Bivona G, Realmuto S, Iacolino G, Gambino CM, Bellia C, Salemi G, Ciaccio M. Klotho and vitamin D in multiple sclerosis: an Italian study. Arch Med Sci 2019; 16:842-847. [PMID: 32542086 PMCID: PMC7286339 DOI: 10.5114/aoms.2019.86969] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 05/01/2019] [Indexed: 12/21/2022] Open
Abstract
INTRODUCTION Low vitamin D levels have been recognised as an important risk factor for autoimmune diseases, including multiple sclerosis (MS). MS is a multifactorial disease, the pathogenesis of which contributes both to genetic and environmental factors. Polymorphisms in genes codifying molecules involved in vitamin D homeostasis have been associated with hypovitaminosis D. However, the influence of polymorphisms of Klotho, which codify a protein with a pivotal role in vitamin D metabolism, have never been investigated. The aim of this study was to evaluate the association among genetic variants of Klotho, namely rs1207568 and rs9536314, serum 25(OH)D3 levels, and multiple sclerosis (both risk and disease progression). MATERIAL AND METHODS 107 patients with MS and 133 healthy controls were enrolled in this study. Serum 25(OH)D3 levels and genotyping of Klotho SNPs were evaluated in all participants by high-performance liquid chromatography and real-time polymerase chain reaction, respectively. RESULTS Allelic and genotypic frequencies did not differ between patients and controls. Concerning rs1207568, we found a trend toward lower serum 25(OH)D3 levels in MS patients with A allele (mutant), both in heterozygosis (AG) and in homozygosis (AA), in comparison to MS patients with G allele in homozygosis (GG) (AG + AA 20.5 ±6.3 µg/l; GG 22.5 ±7.5 µg/l, p = 0.07). CONCLUSIONS Our findings did not identify a role of Klotho in the genetic susceptibility to MS.
Collapse
Affiliation(s)
- Concetta Scazzone
- Department of Biomedicine, Neuroscience, and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Laboratory Medicine, University of Palermo, Palermo, Italy
| | - Luisa Agnello
- Department of Biomedicine, Neuroscience, and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Laboratory Medicine, University of Palermo, Palermo, Italy
| | - Bruna Lo Sasso
- Department of Biomedicine, Neuroscience, and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Laboratory Medicine, University of Palermo, Palermo, Italy
| | - Paolo Ragonese
- Department of Experimental Biomedicine and Neuroscience, University of Palermo, Palermo, Italy
| | - Giulia Bivona
- Department of Biomedicine, Neuroscience, and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Laboratory Medicine, University of Palermo, Palermo, Italy
| | - Sabrina Realmuto
- Department of Experimental Biomedicine and Neuroscience, University of Palermo, Palermo, Italy
| | - Giorgia Iacolino
- Department of Biomedicine, Neuroscience, and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Laboratory Medicine, University of Palermo, Palermo, Italy
| | - Caterina Maria Gambino
- Department of Biomedicine, Neuroscience, and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Laboratory Medicine, University of Palermo, Palermo, Italy
| | - Chiara Bellia
- Department of Biomedicine, Neuroscience, and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Laboratory Medicine, University of Palermo, Palermo, Italy
| | - Giuseppe Salemi
- Department of Experimental Biomedicine and Neuroscience, University of Palermo, Palermo, Italy
| | - Marcello Ciaccio
- Department of Biomedicine, Neuroscience, and Advanced Diagnostics, Institute of Clinical Biochemistry, Clinical Molecular Medicine, and Laboratory Medicine, University of Palermo, Palermo, Italy
- Department of Laboratory Medicine, University-Hospital, Palermo, Italy
| |
Collapse
|
133
|
Zhu WS, Naler L, Maul RW, Sallin MA, Sen JM. Immune system development and age-dependent maintenance in Klotho-hypomorphic mice. Aging (Albany NY) 2019; 11:5246-5257. [PMID: 31386628 PMCID: PMC6682518 DOI: 10.18632/aging.102121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 07/20/2019] [Indexed: 12/12/2022]
Abstract
Circulating Klotho peptide hormone has anti-aging activity and affects tissue maintenance. Hypomorphic mutant Klotho [kl/kl] mice on C57BL/6xC3H, BALB/c and 129 genetic backgrounds, show decreased Klotho expression that correlate with accelerated aging including pre-mature death due to abnormally high levels of serum vitamin D. These mice also show multiple impairments in the immune system. However, it remains unresolved if the defects in the immune system stem from decreased Klotho expression or high vitamin D levels in the serum. Transfer of the kl/kl allele to pure C57BL/6 genetic background [B6-kl/kl] significantly reduced expression of Klotho at all ages. Surprisingly, B6-kl/kl mice showed normalized serum vitamin D levels, amelioration of severe aging-related phenotypes and normal lifespan. This paper reports a detailed analysis of the immune system in B6-kl/kl mice in the absence of detrimental levels of serum vitamin D. Remarkably, the data reveal that in the absence of overt systemic stress, such as abnormally high vitamin D levels, reduced expression of Klotho does not have a major effect on the generation and maintenance of the immune system.
Collapse
Affiliation(s)
- Wandi Sandra Zhu
- National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.,Current address: Department of Immunology and Microbiology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Lynette Naler
- National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.,Current address: Department of Chemical Engineering, Virginia Tech, Blacksburg, VA 24061, USA
| | - Robert W Maul
- National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Michelle A Sallin
- National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Jyoti Misra Sen
- National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.,Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
134
|
Nakanishi K, Nishida M, Taneike M, Yamamoto R, Adachi H, Moriyama T, Yamauchi-Takihara K. Implication of alpha-Klotho as the predictive factor of stress. J Investig Med 2019; 67:1082-1086. [PMID: 31324693 DOI: 10.1136/jim-2018-000977] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2019] [Indexed: 11/03/2022]
Abstract
Stress is known as a risk factor for both mental and physical health problems. While stress is known as one of the major health problems in modern society, a biomarker of stress has not yet been well established. In the present study, we focused on the serum levels of α-Klotho (αKl) as a possible objective biomarker of stress. Subjects included apparently healthy individuals who underwent a health examination in the Osaka University Health and Counseling Center. Physical and biochemical parameters were obtained from all subjects. Information regarding the lifestyle of each individual was obtained via questionnaires. Among male subjects, serum levels of soluble αKl (sαKl) were significantly elevated in subjects who had poor stress management and unsatisfactory sleep, suggesting that stress management and sleeping conditions influenced the serum levels of sαKl. The total Kessler Screening Scale for Psychological Distress (K6) score was significantly increased in subjects who reported experiencing considerable stress, had poor stress management and unsatisfactory sleep. Since serum levels of sαKl showed the same tendency as the K6 score in terms of the relationship between stress management and sleeping conditions in male subjects, increased sαKl levels could be associated with considerable psychological stress in healthy men.
Collapse
Affiliation(s)
- Kaori Nakanishi
- Health and Counseling Center, Osaka University, Toyonaka, Osaka, Japan
| | - Makoto Nishida
- Health and Counseling Center, Osaka University, Toyonaka, Osaka, Japan
| | - Manabu Taneike
- Health and Counseling Center, Osaka University, Toyonaka, Osaka, Japan
| | - Ryohei Yamamoto
- Health and Counseling Center, Osaka University, Toyonaka, Osaka, Japan
| | - Hiroyoshi Adachi
- Health and Counseling Center, Osaka University, Toyonaka, Osaka, Japan
| | - Toshiki Moriyama
- Health and Counseling Center, Osaka University, Toyonaka, Osaka, Japan
| | | |
Collapse
|
135
|
Milovanova LY, Kozlovskaya(Lysenko) LV, Androsova TV, Lebedeva MV, Taranova MV, Milovanova SY, Kondratyeva TB, Zubacheva DO, Tchebotareva NV, Kozlov VV, Kuchieva AM, Li OA, Reshetnikov VA. Low protein diet with essential amino acids ketoanalogues combination can affect serum FGF-23 and Klotho levels in chronic kidney disease 3b-4 stages patients: randomized pilot study. TERAPEVT ARKH 2019; 91:47-56. [DOI: 10.26442/00403660.2019.06.000252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Indexed: 11/22/2022]
Abstract
Protein restriction diet (PRD) with ketoanalagues of essential amino acids (KA) combination can improve of chronic kidney disease (CKD) course while, the precise mechanisms of PRD + KAA action in CKD are not known yet. We have conducted a prospective, randomized, controlled study of PRD and KAA patient’s group in compare with PRD without KAA group in regarding to serum Klotho and FGF-23 levels in patients with CKD. Materials and methods. The study included 79 CKD 3b-4 stages patients, non - diabetic etiology, used PRD (0.6 g/kg/day). The patients were randomized in two groups: 42 patients, received PRD + KAA (Group 1) and 37 patients continued the PRD without KAA (Group 2). Serum FGF-23 (Human FGF-23 ELISA kit with antibodies to native FGF-23 molecule, Merk Millipore MILLENZFGF-23-32K), Klotho (Human soluble Klotho with antiKlotho monoclonal antibodies, IBL-Takara 27998-96Well) levels, as well as instrumental examination: bioimpedance analysis [assess of muscle body mass (MBM), fat body mass (FBM), body mass index (BMI) and others]; sphygmography [assess of augmentation (stiffness) indices (AI), central (aortal) blood pressure (CBP) by «Sphygmacor» device]; as well as echocardiography [assess of cardiac (valvular) calcification score (CCS) and left ventricular myocardium mass index (LVMMI)], were studded in addition to conventional examination. Results and discussion. To the end of 14th month of the study the PRD group reached a body mass index (BMI) decrease (p=0.046), including MBM in men (p=0.027) and woman (p=0.044). In addition, higher FGF-23 (p=0.029), and lower Klotho (p=0.037) serum levels were revealed in the PRD group compared to the PRD+KAA group as well as the increase in AI (p=0.034), CCS (p=0.048), and LVMMI (p=0.023). Conclusion. Use of PRD + KAA provides adequate nutrition status and more efficient correction of FGF-23 and Klotho imbalance in CKD progression that may contribute to alleviation of both cardiovascular calcification and cardiac remodeling in CKD. Importantly, a prolonged PRD use without supplementation of KAA may lead to malnutrition signs.
Collapse
|
136
|
The Prognostic Role of Klotho in Patients with Chronic Kidney Disease: A Systematic Review and Meta-analysis. DISEASE MARKERS 2019; 2019:6468729. [PMID: 31275449 PMCID: PMC6589248 DOI: 10.1155/2019/6468729] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/11/2019] [Accepted: 04/30/2019] [Indexed: 12/14/2022]
Abstract
Objective The prognostic role of Klotho in patients with chronic kidney disease is still controversial. Therefore, we performed this meta-analysis to assess the relationship between the low sKlotho level and the risk of adverse kidney outcomes. Materials and Methods We systematically searched medical databases, such as PubMed, Embase, and the Cochrane Library, for eligible publications regarding the relationship between the low sKlotho level and risk of adverse kidney outcomes. The quality of included studies was assessed by using the Newcastle–Ottawa Scale. Combined hazard ratios (HRs) and its 95% confidence intervals (CIs) were calculated using a random- or fixed-effect model. Subgroup analysis was conducted with stratification by age, estimated glomerular filtration rate (eGFR), follow-up interval, region, and study quality. All data was analyzed by RevMan 5.3 analysis software. Results Eight cohort studies with 3586 participants from 3818 studies were included in our final analysis. Levels of sKlotho were significantly correlated with the eGFR, with a summary correlation coefficient r and 95% CI of 0.469 (0.226, 0.658). Additionally, low sKlotho levels were strongly associated with increased adverse kidney outcomes, and the pooled HR and its 95% CI were 1.64 (1.19, 2.26; P = 0.002), despite publication bias and statistical heterogeneity (I2 = 52%, P = 0.07). Furthermore, this positive correlation was still observed in all of the subgroup analyses. However, heterogeneity was present in subgroup analyses stratified by the eGFR and follow-up interval. Conclusion Levels of sKlotho are positively correlated with the eGFR, and low sKlotho levels are significantly associated with an increased risk of poor kidney outcomes. Therefore, sKlotho could be used as a novel biomarker for early diagnosis and prognostic assessment for patients with chronic kidney disease. Studies with a larger sample size and longer follow-up period are warranted to validate our results.
Collapse
|
137
|
Singh A, Verma A, Sallin MA, Lang F, Sen R, Sen JM. Noncoding variations in Cyp24a1 gene are associated with Klotho-mediated aging phenotypes in different strains of mice. Aging Cell 2019; 18:e12949. [PMID: 30920112 PMCID: PMC6516175 DOI: 10.1111/acel.12949] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 02/05/2019] [Accepted: 03/01/2019] [Indexed: 01/04/2023] Open
Abstract
In mutant mice, reduced levels of Klotho promoted high levels of active vitamin D in the serum. Genetic or dietary manipulations that diminished active vitamin D alleviated aging‐related phenotypes caused by Klotho down‐regulation. The hypomorphic Klotho [kl/kl] allele that decreases Klotho expression in C3H, BALB/c, 129, and C57BL/6 genetic backgrounds substantially increases 1,25(OH)2D3 levels in the sera of susceptible C3H, BALB/c, and 129, but not C57BL/6 mice. This may be attributed to increased basal expression of Cyp24a1 in C57BL/6 mice, which promotes inactivation of 1,25(OH)2D3. Decreased expression of Cyp24a1 in susceptible strains was associated with genetic alterations in noncoding regions of Cyp24a1 gene, which were strongly reminiscent of super‐enhancers that regulate gene expression. These observations suggest that higher basal expression of an enzyme required for catabolizing vitamin D renders B6‐kl/kl mice less susceptible to changes in Klotho expression, providing a plausible explanation for the lack of aging phenotypes on C57BL/6 strain.
Collapse
Affiliation(s)
- Amit Singh
- National Institute on Aging National Institutes of Health Baltimore Maryland
| | - Anjali Verma
- National Institute on Aging National Institutes of Health Baltimore Maryland
| | - Michelle A. Sallin
- National Institute on Aging National Institutes of Health Baltimore Maryland
| | - Florian Lang
- Institute of Physiology Eberhard Karls University of Tübingen Tübingen Germany
| | - Ranjan Sen
- National Institute on Aging National Institutes of Health Baltimore Maryland
- Department of Medicine The Johns Hopkins University School of Medicine Baltimore Maryland
| | - Jyoti Misra Sen
- National Institute on Aging National Institutes of Health Baltimore Maryland
- Department of Medicine The Johns Hopkins University School of Medicine Baltimore Maryland
| |
Collapse
|
138
|
Wright JD, An SW, Xie J, Lim C, Huang CL. Soluble klotho regulates TRPC6 calcium signaling via lipid rafts, independent of the FGFR-FGF23 pathway. FASEB J 2019; 33:9182-9193. [PMID: 31063704 DOI: 10.1096/fj.201900321r] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Soluble klotho (sKlotho), the shed ectodomain of α-klotho, protects the heart by down-regulating transient receptor potential canonical isoform 6 (TRPC6)-mediated calcium signaling. Binding to α2-3-sialyllactose moiety of gangliosides in lipid rafts and inhibition of raft-dependent signaling underlies the mechanism. A recent 3-Å X-ray structure of sKlotho in complex with fibroblast growth factor receptor (FGFR) and fibroblast growth factor 23 (FGF23) indicates that its β6α6 loop might block access to the proposed binding site for α2-3-sialyllactose. It was concluded that sKlotho only functions in complex with FGFR and FGF23 and that sKlotho's pleiotropic effects all depend on FGF23. Here, we report that sKlotho can inhibit TRPC6 channels expressed in cells lacking endogenous FGFRs. Structural modeling and molecular docking show that a repositioned β6α6 loop allows sKlotho to bind α2-3-sialyllactose. Molecular dynamic simulations further show the α2-3-sialyllactose-bound sKlotho complex to be stable. Domains mimicking sKlotho's sialic acid-recognizing activity inhibit TRPC6. The results strongly support the hypothesis that sKlotho can exert effects independent of FGF23 and FGFR.-Wright, J. D., An, S.-W., Xie, J., Lim, C., Huang, C.-L. Soluble klotho regulates TRPC6 calcium signaling via lipid rafts, independent of the FGFR-FGF23 pathway.
Collapse
Affiliation(s)
- Jon D Wright
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Sung-Wan An
- Division of Nephrology, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA; and
| | - Jian Xie
- Division of Nephrology, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA; and
| | - Carmay Lim
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.,Department of Chemistry, National Tsing Hua University, Hsinchu, Taiwan
| | - Chou-Long Huang
- Division of Nephrology, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA; and
| |
Collapse
|
139
|
Gao R, Kanasaki K, Li J, Kitada M, Okazaki T, Koya D. βklotho is essential for the anti-endothelial mesenchymal transition effects of N-acetyl-seryl-aspartyl-lysyl-proline. FEBS Open Bio 2019; 9:1029-1038. [PMID: 30972974 PMCID: PMC6487725 DOI: 10.1002/2211-5463.12638] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 03/25/2019] [Accepted: 04/08/2019] [Indexed: 12/12/2022] Open
Abstract
Endothelial–mesenchymal transition (EndMT) has emerged as an essential bioprocess responsible for the development of organ fibrosis. We have previously reported that fibroblast growth factor receptor 1 (FGFR1) is involved in the anti‐EndMT effect of N‐acetyl‐seryl‐aspartyl‐lysyl‐proline (AcSDKP). FGFR1 is expressed on the cell membrane and performs its biological function through interaction with co‐receptors, including βklotho (KLB). However, it remains unknown whether KLB is involved in the anti‐EndMT effects of AcSDKP. Here, we demonstrated that AcSDKP increased KLB expression in an FGFR1‐dependent manner and that KLB deficiency induced AcSDKP‐resistant EndMT via the induction of the mitogen‐activated protein kinase (MAPK) pathway. In cultured endothelial cells, AcSDKP increased KLB protein level in an FGFR1‐dependent manner through induction of the FGFR1–KLB complex. KLB suppression by small interfering RNA transfection did not affect FGFR1 levels and resulted in the induction of EndMT. In contrast to the EndMT observed under FGFR1 deficiency, the EndMT induced by KLB suppression was not accompanied by the induction of Smad3 phosphorylation; instead, KLB‐deficient cells exhibited induced activation of the MAPK/extracellular signal‐regulated kinase (ERK) kinase (MEK) and ERK pathways. Treatment with the specific MEK inhibitor U0126 diminished KLB deficiency‐induced EndMT. Consistent with this finding, AcSDKP did not suppress either EndMT or MEK/ERK activation induced by KLB deficiency. Application of either FGF19 or FGF21 synergistically augmented the anti‐EndMT effects of AcSDKP. Taken together, these results indicate that endogenous peptide AcSDKP exerts its activity through induction of the FGFR1–KLB complex in vascular endothelial cells.
Collapse
Affiliation(s)
- Rongfen Gao
- Department of Diabetology & Endocrinology, Kanazawa Medical University, Uchinada, Japan.,Department of Hematology & Immunology, Kanazawa Medical University, Uchinada, Japan
| | - Keizo Kanasaki
- Department of Diabetology & Endocrinology, Kanazawa Medical University, Uchinada, Japan.,Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute, Kanazawa Medical University, Uchinada, Japan
| | - Jinpeng Li
- Department of Diabetology & Endocrinology, Kanazawa Medical University, Uchinada, Japan
| | - Munehiro Kitada
- Department of Diabetology & Endocrinology, Kanazawa Medical University, Uchinada, Japan.,Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute, Kanazawa Medical University, Uchinada, Japan
| | - Toshiro Okazaki
- Department of Hematology & Immunology, Kanazawa Medical University, Uchinada, Japan
| | - Daisuke Koya
- Department of Diabetology & Endocrinology, Kanazawa Medical University, Uchinada, Japan.,Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute, Kanazawa Medical University, Uchinada, Japan
| |
Collapse
|
140
|
Li S, Yu L, He A, Liu Q. Klotho Inhibits Unilateral Ureteral Obstruction-Induced Endothelial-to-Mesenchymal Transition via TGF-β1/Smad2/Snail1 Signaling in Mice. Front Pharmacol 2019; 10:348. [PMID: 31024315 PMCID: PMC6460315 DOI: 10.3389/fphar.2019.00348] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 03/20/2019] [Indexed: 12/14/2022] Open
Abstract
This study aimed to evaluate repression of Klotho on unilateral ureteral obstruction (UUO)-induced renal fibrosis and endothelial-to-mesenchymal transition (EndoMT) in mice. Renal fibrosis model was established by UUO in C57BL/6 male mice. Recombinant Klotho protein was administered to UUO mice as treatment group, and the mice in sham and UUO group were administered with an equal volume of vehicle. EndoMT biomarkers and TGF-β1/Smad2/Snail1 signaling were examined by immunofluorescence, immunohistochemistry, and western blotting assays. UUO deteriorated kidney function and resulted in increased expression of the mesenchymal marker α-smooth muscle actin and decreased expression of vascular endothelial cadherin, an endothelial marker. Moreover, UUO enhanced TGF-β1, phosphorylated Smad2 (p-Smad2), and Snail1 expression. Interestingly, Klotho treatment suppressed UUO-induced TGF-β1, p-Smad3, and Snail1 expression, which was accompanied by alleviation of the EndoMT process. Our findings demonstrated that Klotho significantly ameliorated EndoMT progression by targeting TGF-β1/Smad/Snail1 signaling in UUO mice, which provides the possibility for Klotho-based therapeutic protection against renal fibrosis.
Collapse
Affiliation(s)
- Shasha Li
- Clinical Research and Lab Centre, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, China
| | - Lixia Yu
- Department of Nephrology, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, China
| | - Aolin He
- Clinical Research and Lab Centre, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, China
| | - Qifeng Liu
- Department of Nephrology, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, China
| |
Collapse
|
141
|
Abstract
Hypophosphatemic rickets, mostly of the X-linked dominant form caused by pathogenic variants of the PHEX gene, poses therapeutic challenges with consequences for growth and bone development and portends a high risk of fractions and poor bone healing, dental problems and nephrolithiasis/nephrocalcinosis. Conventional treatment consists of PO4 supplements and calcitriol requiring monitoring for treatment-emergent adverse effects. FGF23 measurement, where available, has implications for the differential diagnosis of hypophosphatemia syndromes and, potentially, treatment monitoring. Newer therapeutic modalities include calcium sensing receptor modulation (cinacalcet) and biological molecules targeting FGF23 or its receptors. Their long-term effects must be compared with those of conventional treatments.
Collapse
Affiliation(s)
- Martin Bitzan
- Department of Pediatrics, The Montreal Children's Hospital, McGill University Health Centre, 1001 Boulevard Décarie, Room B RC.6164, Montreal, Quebec H4A 3J1, Canada.
| | - Paul R Goodyer
- The Research Institute of the McGill University Health Centre, 1001 Boulevard Décarie, Room EM1.2232, Montreal, Quebec H4A3J1, Canada
| |
Collapse
|
142
|
Kan WC, Hwang JY, Chuang LY, Guh JY, Ye YL, Yang YL, Huang JS. Effect of osthole on advanced glycation end products-induced renal tubular hypertrophy and role of klotho in its mechanism of action. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 53:205-212. [PMID: 30668400 DOI: 10.1016/j.phymed.2018.09.030] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 08/13/2018] [Accepted: 09/03/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Osthole has been widely reported to have pharmacological activities such as anti-cancer, anti-inflammation and anti-hyperlipidemic effects. Klotho was identified as an anti-senescence protein in a variety of tissues. Loss of klotho has been associated with chronic kidney disease. However, potential roles and molecular events for osthole and klotho in diabetic nephropathy remain unclear. PURPOSE In the current study, we undertook to study the effect of osthole on klotho expression in advanced glycation end products (AGE)-cultured human renal proximal tubular cells, and to investigate the molecular mechanisms of osthole and exogenous klotho against AGE-induced renal tubular hypertrophy. METHODS Cell viability was elucidated by MTT assay. Protein expression was measured by Western blotting. mRNA level was analyzed by real-time PCR. Cellular hypertrophy growth was evaluated by hypertrophy index. Relative cell size was detected by flow cytometry. RESULTS We found that raising the ambient AGE concentration causes a dose-dependent decrease in klotho synthesis. Osthole significantly increased AGE-inhibited klotho mRNA and protein expression. Osthole and exogenous klotho treatments significantly attenuated AGE-induced Janus kinase 2 (JAK2)-signal transducers and activators of transcription 1 (STAT1) and STAT3 activation. Moreover, protein levels of suppressor of cytokine signaling 1 (SOCS1) and SOCS3 were augmented by osthole and exogenous klotho. The abilities of osthole and exogenous klotho to reverse AGE-induced cellular hypertrophy were verified by the observation that osthole and exogenous klotho inhibited p21Waf1/Cip1/collagen IV/RAGE expression, total protein content, and cell size. CONCLUSION Consequently, we found that osthole attenuated AGE-induced renal tubular hypertrophy via induction of klotho expression and suppression of the JAK2-STAT1/STAT3 signaling. These results also showed that klotho might be used as a unique molecular target for the treatment of diabetic nephropathy.
Collapse
Affiliation(s)
- Wei-Chih Kan
- Division of Nephrology, Department of Internal Medicine, Chi-Mei Medical Center, Tainan, Taiwan
| | - Jean-Yu Hwang
- Department of Food Nutrition, Chung Hwa University of Medical Technology, Tainan, Taiwan
| | - Lea-Yea Chuang
- Department of Biochemistry, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jinn-Yuh Guh
- Department of Internal Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Ling Ye
- Department of Biotechnology, National Formosa University, Yunlin, Taiwan
| | - Yu-Lin Yang
- Department of Medical Laboratory Science and Biotechnology, Chung Hwa University of Medical Technology, Tainan, Taiwan
| | - Jau-Shyang Huang
- Department of Biomedicine and Health Science, Chung Hwa University of Medical Technology, 89, Wen-Hwa 1st St. Rende Dist., Tainan 71703, Taiwan.
| |
Collapse
|
143
|
Olejnik A, Franczak A, Krzywonos-Zawadzka A, Kałużna-Oleksy M, Bil-Lula I. The Biological Role of Klotho Protein in the Development of Cardiovascular Diseases. BIOMED RESEARCH INTERNATIONAL 2018; 2018:5171945. [PMID: 30671457 PMCID: PMC6323445 DOI: 10.1155/2018/5171945] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 11/09/2018] [Accepted: 11/28/2018] [Indexed: 12/22/2022]
Abstract
Klotho is a membrane-bound or soluble antiaging protein, whose protective activity is essential for a proper function of many organs. In 1997, an accidental insertion of a transgene led to creation of transgenic mice with several age-related disorders. In Klotho-deficient mice, the inherited phenotypes closely resemble human aging, while in an animal model of Klotho overexpression, the lifespan is extended. Klotho protein is detected mainly in the kidneys and brain. It is a coreceptor for fibroblast growth factor and hence is involved in maintaining endocrine system homeostasis. Furthermore, an inhibition of insulin/insulin-like growth factor-1 signaling pathway by Klotho regulates oxidative stress and reduces cell death. The association between serum Klotho and the classic risk factors, as well as the clinical history of cardiovascular disease, was also shown. There are a lot of evidences that Klotho deficiency correlates with the occurrence and development of coronary artery disease, atherosclerosis, myocardial infarction, and left ventricular hypertrophy. Therefore, an involvement of Klotho in the signaling pathways and in regulation of a proper cell metabolism could be a crucial factor in the cardiac and vascular protection. It is also well established that Klotho protein enhances the antioxidative response via augmented production of superoxide dismutase and reduced generation of reactive oxygen species. Recent studies have proven an expression of Klotho in cardiomyocytes and its increased expression in stress-related heart injury. Thus, the antioxidative and antiapoptotic activity of Klotho could be considered as the novel protective factor in cardiovascular disease and heart injury.
Collapse
Affiliation(s)
- Agnieszka Olejnik
- Department of Medical Laboratory Diagnostics, Division of Clinical Chemistry, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Aleksandra Franczak
- Department of Medical Laboratory Diagnostics, Division of Clinical Chemistry, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Anna Krzywonos-Zawadzka
- Department of Medical Laboratory Diagnostics, Division of Clinical Chemistry, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Marta Kałużna-Oleksy
- Department of Cardiology, University Hospital of Lord's Transfiguration, Poznan University of Medical Sciences, 61-848 Poznan, Poland
| | - Iwona Bil-Lula
- Department of Medical Laboratory Diagnostics, Division of Clinical Chemistry, Wroclaw Medical University, 50-556 Wroclaw, Poland
| |
Collapse
|
144
|
Farías-Basulto A, Martínez-Ramírez HR, Gómez-García EF, Cueto-Manzano AM, Cortés-Sanabria L, Hernández-Ramos LE, Ramírez-López G, Mendoza-Carrera F. Circulating Levels of Soluble Klotho and Fibroblast Growth Factor 23 in Diabetic Patients and Its Association with Early Nephropathy. Arch Med Res 2018; 49:451-455. [DOI: 10.1016/j.arcmed.2019.01.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 12/16/2018] [Accepted: 01/18/2019] [Indexed: 10/27/2022]
|
145
|
Milovanova L, Fomin V, Moiseev S, Taranova M, Milovanov Y, Lysenko Kozlovskaya L, Kozlov V, Kozevnikova E, Milovanova S, Lebedeva M, Reshetnikov V. Effect of essential amino acid кetoanalogues and protein restriction diet on morphogenetic proteins (FGF-23 and Кlotho) in 3b-4 stages chronic кidney disease patients: a randomized pilot study. Clin Exp Nephrol 2018; 22:1351-1359. [PMID: 29948444 DOI: 10.1007/s10157-018-1591-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 05/17/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND A low protein diet (LPD) with essential amino acid ketoanalogue supplementation (KA) may contribute in improving of chronic kidney disease (CKD), while the exact mechanisms of KA's effect are not established yet. We have conducted a prospective, randomized, controlled comparative study of LPD + KA and LPD alone in relation to serum Klotho, FGF-23 levels in CKD patients. METHODS 79 non-diabetic CKD 3b-4 stage patients, compliant with LPD diet (0.6 g/kg of body weight/day), had been selected. The patients were randomized into two groups. The first group (42 patients) received LPD + КA. The second group (37 patients) continued the LРD alone. In addition to routine tests, serum Klotho, FGF-23 levels, as well as bioimpedance analysis, sphygmography (stiffness (augmentation) indices (AI), central (aortal) blood pressure) with a «SphygmaCor» device; echocardiography (valvular calcification score (VCS) and LVMMI), were performed. RESULTS There were body mass indices' decrease (p = 0.046), including muscle body mass in men (p = 0.027) and woman (p = 0.044) in the LPD group to the end of study (14th month). In addition, lower FGF-23 (p = 0.029), and higher sKlotho (p = 0.037) were detected in the LPD + KA group compared to the LPD one. The increase in AI (p = 0.034), VCS (p = 0.048), and LVMMI (p = 0.023) was detected more often in the LPD group at the end of study. CONCLUSION LPD + KA provides support for nutrition status and contributes to more efficient correction of FGF-23 and Klotho abnormalities that may result in cardiovascular calcification and cardiac remodeling decreasing in CKD. At the same time, a prolonged LPD alone may lead to malnutrition.
Collapse
Affiliation(s)
- Lyudmila Milovanova
- Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya str. 8, bld.2, Moscow, 119991, Russian Federation. .,Clinic of Nephrology and Internal Diseases, Rossolimo str. 11, bld. 5, Moscow, 119992, Russian Federation.
| | - Victor Fomin
- Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya str. 8, bld.2, Moscow, 119991, Russian Federation.,Department of Faculty Therapy No. 1, Bolshaya Pirogovskaya str., 6, bld 1., Moscow, 119435, Russian Federation
| | - Sergey Moiseev
- Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya str. 8, bld.2, Moscow, 119991, Russian Federation.,Clinic of Nephrology and Internal Diseases, Rossolimo str. 11, bld. 5, Moscow, 119992, Russian Federation
| | - Marina Taranova
- Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya str. 8, bld.2, Moscow, 119991, Russian Federation.,Clinic of Nephrology and Internal Diseases, Rossolimo str. 11, bld. 5, Moscow, 119992, Russian Federation
| | - Yury Milovanov
- Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya str. 8, bld.2, Moscow, 119991, Russian Federation.,Clinic of Nephrology and Internal Diseases, Rossolimo str. 11, bld. 5, Moscow, 119992, Russian Federation
| | - Lidia Lysenko Kozlovskaya
- Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya str. 8, bld.2, Moscow, 119991, Russian Federation.,Clinic of Nephrology and Internal Diseases, Rossolimo str. 11, bld. 5, Moscow, 119992, Russian Federation
| | - Vasiliy Kozlov
- Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya str. 8, bld.2, Moscow, 119991, Russian Federation.,Department of Public Health and Health Care Organization, Bolshaya Pirogovskaya str. 2, bld. 2, Moscow, 119435, Russian Federation
| | - Elena Kozevnikova
- Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya str. 8, bld.2, Moscow, 119991, Russian Federation.,Clinic of Nephrology and Internal Diseases, Rossolimo str. 11, bld. 5, Moscow, 119992, Russian Federation
| | - Svetlana Milovanova
- Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya str. 8, bld.2, Moscow, 119991, Russian Federation.,Clinic of Nephrology and Internal Diseases, Rossolimo str. 11, bld. 5, Moscow, 119992, Russian Federation
| | - Marina Lebedeva
- Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya str. 8, bld.2, Moscow, 119991, Russian Federation.,Clinic of Nephrology and Internal Diseases, Rossolimo str. 11, bld. 5, Moscow, 119992, Russian Federation
| | - Vladimir Reshetnikov
- Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya str. 8, bld.2, Moscow, 119991, Russian Federation.,Department of Public Health and Health Care Organization, Bolshaya Pirogovskaya str. 2, bld. 2, Moscow, 119435, Russian Federation
| |
Collapse
|
146
|
Aroor AR, Manrique-Acevedo C, DeMarco VG. The role of dipeptidylpeptidase-4 inhibitors in management of cardiovascular disease in diabetes; focus on linagliptin. Cardiovasc Diabetol 2018; 17:59. [PMID: 29669555 PMCID: PMC5907287 DOI: 10.1186/s12933-018-0704-1] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 04/12/2018] [Indexed: 12/15/2022] Open
Abstract
Multiple population based analyses have demonstrated a high incidence of cardiovascular disease (CVD) and cardiovascular (CV) mortality in subjects with T2DM that reduces life expectancy by as much as 15 years. Importantly, the CV system is particularly sensitive to the metabolic and immune derangements present in obese pre-diabetic and diabetic individuals; consequently, CV dysfunction is often the initial CV derangement to occur and promotes the progression to end organ/tissue damage in T2DM. Specifically, diabetic CVD can manifest as microvascular complications, such as nephropathy, retinopathy, and neuropathy, as well as, macrovascular impairments, including ischemic heart disease, peripheral vascular disease, and cerebrovascular disease. Despite some progress in prevention and treatment of CVD, mainly via blood pressure and dyslipidemia control strategies, the impact of metabolic disease on CV outcomes is still a major challenge and persists in proportion to the epidemics of obesity and diabetes. There is abundant pre-clinical and clinical evidence implicating the DPP-4-incretin axis in CVD. In this regard, linagliptin is a unique DPP-4 inhibitor with both CV and renal safety profiles. Moreover, it exerts beneficial CV effects beyond glycemic control and beyond class effects. Linagliptin is protective for both macrovascular and microvascular complications of diabetes in preclinical models, as well as clinical models. Given the role of endothelial-immune cell interactions as one of the key events in the initiation and progression of CVD, linagliptin modulates these cell–cell interactions by affecting two important pathways involving stimulation of NO signaling and potent inhibition of a key immunoregulatory molecule.
Collapse
Affiliation(s)
- Annayya R Aroor
- Diabetes and Cardiovascular Center, University of Missouri School of Medicine, Columbia, MO, USA.,Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri-Columbia School of Medicine, One Hospital Drive, Columbia, MO, 65212, USA.,Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA
| | - Camila Manrique-Acevedo
- Diabetes and Cardiovascular Center, University of Missouri School of Medicine, Columbia, MO, USA.,Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri-Columbia School of Medicine, One Hospital Drive, Columbia, MO, 65212, USA.,Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA
| | - Vincent G DeMarco
- Diabetes and Cardiovascular Center, University of Missouri School of Medicine, Columbia, MO, USA. .,Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri-Columbia School of Medicine, One Hospital Drive, Columbia, MO, 65212, USA. .,Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA. .,Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
147
|
Izzo C, Carrizzo A, Alfano A, Virtuoso N, Capunzo M, Calabrese M, De Simone E, Sciarretta S, Frati G, Oliveti M, Damato A, Ambrosio M, De Caro F, Remondelli P, Vecchione C. The Impact of Aging on Cardio and Cerebrovascular Diseases. Int J Mol Sci 2018; 19:E481. [PMID: 29415476 PMCID: PMC5855703 DOI: 10.3390/ijms19020481] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 01/29/2018] [Accepted: 02/01/2018] [Indexed: 01/03/2023] Open
Abstract
A growing number of evidences report that aging represents the major risk factor for the development of cardio and cerebrovascular diseases. Understanding Aging from a genetic, biochemical and physiological point of view could be helpful to design a better medical approach and to elaborate the best therapeutic strategy to adopt, without neglecting all the risk factors associated with advanced age. Of course, the better way should always be understanding risk-to-benefit ratio, maintenance of independence and reduction of symptoms. Although improvements in treatment of cardiovascular diseases in the elderly population have increased the survival rate, several studies are needed to understand the best management option to improve therapeutic outcomes. The aim of this review is to give a 360° panorama on what goes on in the fragile ecosystem of elderly, why it happens and what we can do, right now, with the tools at our disposal to slow down aging, until new discoveries on aging, cardio and cerebrovascular diseases are at hand.
Collapse
Affiliation(s)
- Carmine Izzo
- Departement of Medicine and Surgery, University of Salerno, 84081 Salerno, Italy; (C.I.); (M.C.); (M.O.); (F.D.C.); (P.R.)
| | - Albino Carrizzo
- Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Italy; (A.C.); (S.S.); (G.F.); (A.D.); (M.A.)
| | - Antonia Alfano
- Heart Department, A.O.U. “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy; (A.A.); (E.D.S.)
| | - Nicola Virtuoso
- Department of Cardiovascular Medicine, A.O.U. Federico II, 80131 Naples, Italy;
| | - Mario Capunzo
- Departement of Medicine and Surgery, University of Salerno, 84081 Salerno, Italy; (C.I.); (M.C.); (M.O.); (F.D.C.); (P.R.)
| | - Mariaconsiglia Calabrese
- Rehabilitation Department, A.O.U. “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy;
| | - Eros De Simone
- Heart Department, A.O.U. “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy; (A.A.); (E.D.S.)
| | - Sebastiano Sciarretta
- Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Italy; (A.C.); (S.S.); (G.F.); (A.D.); (M.A.)
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Polo Pontino, 04100 Latina, Italy
| | - Giacomo Frati
- Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Italy; (A.C.); (S.S.); (G.F.); (A.D.); (M.A.)
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Polo Pontino, 04100 Latina, Italy
| | - Marco Oliveti
- Departement of Medicine and Surgery, University of Salerno, 84081 Salerno, Italy; (C.I.); (M.C.); (M.O.); (F.D.C.); (P.R.)
| | - Antonio Damato
- Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Italy; (A.C.); (S.S.); (G.F.); (A.D.); (M.A.)
| | - Mariateresa Ambrosio
- Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Italy; (A.C.); (S.S.); (G.F.); (A.D.); (M.A.)
| | - Francesco De Caro
- Departement of Medicine and Surgery, University of Salerno, 84081 Salerno, Italy; (C.I.); (M.C.); (M.O.); (F.D.C.); (P.R.)
| | - Paolo Remondelli
- Departement of Medicine and Surgery, University of Salerno, 84081 Salerno, Italy; (C.I.); (M.C.); (M.O.); (F.D.C.); (P.R.)
| | - Carmine Vecchione
- Departement of Medicine and Surgery, University of Salerno, 84081 Salerno, Italy; (C.I.); (M.C.); (M.O.); (F.D.C.); (P.R.)
- Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Italy; (A.C.); (S.S.); (G.F.); (A.D.); (M.A.)
| |
Collapse
|
148
|
Iñiguez G, Gallardo P, Castro JJ, Gonzalez R, Garcia M, Kakarieka E, San Martin S, Johnson MC, Mericq V, Cassorla F. Klotho Gene and Protein in Human Placentas According to Birth Weight and Gestational Age. Front Endocrinol (Lausanne) 2018; 9:797. [PMID: 30697189 PMCID: PMC6340928 DOI: 10.3389/fendo.2018.00797] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 12/19/2018] [Indexed: 01/02/2023] Open
Abstract
Introduction: Fetal growth restriction may be the consequence of maternal, fetal, or placental factors. The insulin-like growth factors (IGFs) are major determinants of fetal growth, and are expressed in the mother, fetus and placenta in most species. Previously we reported higher placental protein content of IGF-I, IGF-IR, and AKT in small (SGA) compared with those from appropriate for gestational age (AGA) placentas. The protein Klotho, has been reported in placenta and may regulate IGF-I activity. In this study we determined Klotho gene expression and protein immunostaining in term (T-SGA y T-AGA) and preterm (PT-SGA y PT-AGA) human placentas. In addition, we assessed the effect of Klotho on the IGF-IR and AKT activation induced by IGF-I. Methods: Placentas (n = 1 17) from 32 T-SGA (birth weight (BW) = -1.74 ± 0.08 SDS), 37 T-AGA (BW = 0.12 ± 0.12 SDS), 20 PT-SGA (BW = -2.08 ± 0.14 SDS), and 28 PT-AGA (BW = -0.43 ± 0.13 SDS) newborns were collected. mRNA expression by RT-PCR in the chorionic (CP) and basal (BP) plates of the placentas, and the presence of Klotho was evaluated by immunohistochemistry (integral optical density, IOD). In addition, we developed placental explants that were incubated with IGF-I in the presence or absence of Klotho. Results: We found a lower mRNA expression and protein immunoreactivity of Klotho in the CP of SGA (term and preterm) compared with AGA placentas. We also observed a significant reduction in IGF-IR tyrosine activation induced by IGF-I 10 nM when preincubated with 2.0 nM of Klotho (2.4 ± 0.5 arbitrary units vs. 1.3 ± 0.3 AU), and similar results we observed on AKT and ERK42/44 activation. Conclusion: We describe for the first time that Klotho mRNA and protein varies according to fetal growth and gestational age. In addition, Klotho appears to down-regulate the activation induced by IGF-I on IGF-IR and AKT, suggesting that Klotho may be regulating IGF-I activity in human placentas according to intrauterine fetal growth.
Collapse
Affiliation(s)
- Germán Iñiguez
- School of Medicine, Maternal and Child Research Institute (IDIMI), University of Chile, Santiago, Chile
- *Correspondence: Germán Iñiguez
| | - Pedro Gallardo
- School of Medicine, Maternal and Child Research Institute (IDIMI), University of Chile, Santiago, Chile
| | - Juan Jose Castro
- School of Medicine, Maternal and Child Research Institute (IDIMI), University of Chile, Santiago, Chile
| | - Rene Gonzalez
- Biomedical Research Centre, School of Medicine, University of Valparaíso, Valparaíso, Chile
| | - Mirna Garcia
- Neonatology Unit, San Borja Arriarán Clinical Hospital, Santiago, Chile
| | - Elena Kakarieka
- Pathology Unit, San Borja Arriarán Clinical Hospital, Santiago, Chile
| | - Sebastian San Martin
- Biomedical Research Centre, School of Medicine, University of Valparaíso, Valparaíso, Chile
| | - Maria Cecilia Johnson
- School of Medicine, Maternal and Child Research Institute (IDIMI), University of Chile, Santiago, Chile
| | - Verónica Mericq
- School of Medicine, Maternal and Child Research Institute (IDIMI), University of Chile, Santiago, Chile
| | - Fernando Cassorla
- School of Medicine, Maternal and Child Research Institute (IDIMI), University of Chile, Santiago, Chile
| |
Collapse
|
149
|
Fukumoto S. Targeting Fibroblast Growth Factor 23 Signaling with Antibodies and Inhibitors, Is There a Rationale? Front Endocrinol (Lausanne) 2018; 9:48. [PMID: 29515522 PMCID: PMC5826173 DOI: 10.3389/fendo.2018.00048] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Fibroblast growth factor 23 (FGF23) is a phosphotropic hormone mainly produced by bone. FGF23 reduces serum phosphate by suppressing intestinal phosphate absorption through reducing 1,25-dihydroxyvitamin D and proximal tubular phosphate reabsorption. Excessive actions of FG23 result in several kinds of hypophosphatemic rickets/osteomalacia including X-linked hypophosphatemic rickets (XLH) and tumor-induced osteomalacia. While neutral phosphate and active vitamin D are standard therapies for child patients with XLH, these medications have several limitations both in their effects and adverse events. Several approaches that inhibit FGF23 actions including anti-FGF23 antibodies and inhibitors of FGF signaling have been shown to improve phenotypes of model mice for FG23-related hypophosphatemic diseases. In addition, clinical trials indicated that a humanized anti-FGF23 antibody increased serum phosphate and improved quality of life in patients with XLH. Furthermore, circulatory FGF23 is high in patients with chronic kidney disease (CKD). Many epidemiological studies indicated the association between high FGF23 levels and various adverse events especially in patients with CKD. However, it is not known whether the inhibition of FGF23 activities in patients with CKD is beneficial for these patients. In this review, recent findings concerning the modulation of FGF23 activities are discussed.
Collapse
Affiliation(s)
- Seiji Fukumoto
- Department of Molecular Endocrinology, Fujii Memorial Institute of Medical Sciences, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
- *Correspondence: Seiji Fukumoto,
| |
Collapse
|
150
|
Richter B, Faul C. FGF23 Actions on Target Tissues-With and Without Klotho. Front Endocrinol (Lausanne) 2018; 9:189. [PMID: 29770125 PMCID: PMC5940753 DOI: 10.3389/fendo.2018.00189] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/06/2018] [Indexed: 12/11/2022] Open
Abstract
Fibroblast growth factor (FGF) 23 is a phosphaturic hormone whose physiologic actions on target tissues are mediated by FGF receptors (FGFR) and klotho, which functions as a co-receptor that increases the binding affinity of FGF23 for FGFRs. By stimulating FGFR/klotho complexes in the kidney and parathyroid gland, FGF23 reduces renal phosphate uptake and secretion of parathyroid hormone, respectively, thereby acting as a key regulator of phosphate metabolism. Recently, it has been shown that FGF23 can also target cell types that lack klotho. This unconventional signaling event occurs in an FGFR-dependent manner, but involves other downstream signaling pathways than in "classic" klotho-expressing target organs. It appears that klotho-independent signaling mechanisms are only activated in the presence of high FGF23 concentrations and result in pathologic cellular changes. Therefore, it has been postulated that massive elevations in circulating levels of FGF23, as found in patients with chronic kidney disease, contribute to associated pathologies by targeting cells and tissues that lack klotho. This includes the induction of cardiac hypertrophy and fibrosis, the elevation of inflammatory cytokine expression in the liver, and the inhibition of neutrophil recruitment. Here, we describe the signaling and cellular events that are caused by FGF23 in tissues lacking klotho, and we discuss FGF23's potential role as a hormone with widespread pathologic actions. Since the soluble form of klotho can function as a circulating co-receptor for FGF23, we also discuss the potential inhibitory effects of soluble klotho on FGF23-mediated signaling which might-at least partially-underlie the pleiotropic tissue-protective functions of klotho.
Collapse
|