1
|
Wang B, Xu W, Mei Z, Yang W, Meng X, An G. Association between serum Klotho levels and estimated pulse wave velocity in postmenopausal women: a cross-sectional study of NHANES 2007-2016. Front Endocrinol (Lausanne) 2024; 15:1471548. [PMID: 39329104 PMCID: PMC11424431 DOI: 10.3389/fendo.2024.1471548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 08/26/2024] [Indexed: 09/28/2024] Open
Abstract
Background Postmenopausal women are at an increased risk of arterial stiffness, which can be assessed using estimated pulse wave velocity (ePWV). This study aimed to investigate the relationship between serum klotho levels and ePWV in postmenopausal women. Methods This cross-sectional study used data from postmenopausal women who participated in the National Health and Nutrition Examination Survey (NHANES) between 2007 and 2016. Participants were divided into two groups based on the presence of hypertension. Weighted multivariate linear regression was used to analyze the relationship between serum Klotho levels and ePWV in each group. Restricted cubic spline models with multivariable adjustments were employed to examine nonlinear associations within each group. Results Our analysis included 4,468 postmenopausal women from the NHANES database, with 1,671 in the non-hypertensive group and 2,797 in the hypertensive group. In all regression models, serum Klotho (ln-transformed) levels were significantly and independently negatively correlated with ePWV in the non-hypertensive group. After fully adjusting for confounders, a 1-unit increase in ln(Klotho) was associated with a 0.13 m/s decrease in ePWV (β = -0.13, 95% CI -0.23 to -0.03; p = 0.008). Additionally, in the fully adjusted model, participants in the highest quartile of ln(Klotho) had an ePWV value 0.14 m/s lower than those in the lowest quartile (p for trend = 0.017; 95% CI -0.23 to -0.05; p = 0.002). This negative correlation was consistent across subgroups and was particularly significant among women aged < 60 years, nonsmokers, and non-Hispanic Black women. However, no association was observed between serum Klotho levels and ePWV in the hypertensive group. Conclusion Hypertension may affect the relationship between serum Klotho level and ePWV in postmenopausal women. Increased serum Klotho levels may reduce arterial stiffness in postmenopausal women. Further studies are required to confirm these findings.
Collapse
Affiliation(s)
- Baiqiang Wang
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, China
- Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Wenqu Xu
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, China
- Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Zeyuan Mei
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, China
- Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Wei Yang
- Department of Cardiology, People's Hospital of Rizhao, Rizhao, China
| | - Xiao Meng
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, China
- Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Guipeng An
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, China
- Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
2
|
Crosswhite P, Sun Z. TNFα Induces DNA and Histone Hypomethylation and Pulmonary Artery Smooth Muscle Cell Proliferation Partly via Excessive Superoxide Formation. Antioxidants (Basel) 2024; 13:677. [PMID: 38929115 PMCID: PMC11200563 DOI: 10.3390/antiox13060677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/17/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Objective: The level of tumor necrosis factor-α (TNF-α) is upregulated during the development of pulmonary vascular remodeling and pulmonary hypertension. A hallmark of pulmonary arterial (PA) remodeling is the excessive proliferation of PA smooth muscle cells (PASMCs). The purpose of this study is to investigate whether TNF-α induces PASMC proliferation and explore the potential mechanisms. Methods: PASMCs were isolated from 8-week-old male Sprague-Dawley rats and treated with 0, 20, or 200 ng/mL TNF-α for 24 or 48 h. After treatment, cell number, superoxide production, histone acetylation, DNA methylation, and histone methylation were assessed. Results: TNF-α treatment increased NADPH oxidase activity, superoxide production, and cell numbers compared to untreated controls. TNF-α-induced PASMC proliferation was rescued by a superoxide dismutase mimetic tempol. TNF-α treatment did not affect histone acetylation at either dose but did significantly decrease DNA methylation. DNA methyltransferase 1 activity was unchanged by TNF-α treatment. Further investigation using QRT-RT-PCR revealed that GADD45-α, a potential mediator of DNA demethylation, was increased after TNF-α treatment. RNAi inhibition of GADD45-α alone increased DNA methylation. TNF-α impaired the epigenetic mechanism leading to DNA hypomethylation, which can be abolished by a superoxide scavenger tempol. TNF-α treatment also decreased H3-K4 methylation. TNF-α-induced PASMC proliferation may involve the H3-K4 demethylase enzyme, lysine-specific demethylase 1 (LSD1). Conclusions: TNF-α-induced PASMC proliferation may be partly associated with excessive superoxide formation and histone and DNA methylation.
Collapse
Affiliation(s)
- Patrick Crosswhite
- Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Department of Human Physiology, Gonzaga University, Spokane, WA 99205, USA
| | - Zhongjie Sun
- Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Department of Physiology, College of Medicine, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA
| |
Collapse
|
3
|
Han X, Akinseye L, Sun Z. KDM6A Demethylase Regulates Renal Sodium Excretion and Blood Pressure. Hypertension 2024; 81:541-551. [PMID: 38164755 PMCID: PMC10922853 DOI: 10.1161/hypertensionaha.123.22026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 12/13/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND KDM6A (Lysine-Specific Demethylase 6A) is a specific demethylase for histone 3 lysine (K) 27 trimethylation (H3K27me3). The purpose of this study is to investigate whether KDM6A in renal tubule cells plays a role in the regulation of kidney function and blood pressure. METHODS We first crossed Ksp-Cre+/- and KDM6Aflox/flox mice for generating inducible kidney-specific deletion of KDM6A gene. RESULTS Notably, conditional knockout of KDM6A gene in renal tubule cells (KDM6A-cKO) increased H3K27me3 levels which leads to a decrease in Na excretion and elevation of blood pressure. Further analysis showed that the expression of NKCC2 (Na-K-2Cl cotransporter 2) and NCC (Na-Cl cotransporters) was upregulated which contributes to impaired Na excretion in KDM6A-cKO mice. The expression of AQP2 (aquaporin 2) was also increased in KDM6A-cKO mice, which may facilitate water reabsorption in KDM6A-cKO mice. The expression of Klotho was downregulated while expression of aging markers including p53, p21, and p16 was upregulated in kidneys of KDM6A-cKO mice, indicating that deletion of KDM6A in the renal tubule cells promotes kidney aging. Interestingly, KDM6A-cKO mice developed salt-sensitive hypertension which can be rescued by treatment with Klotho. KDM6A deficiency induced salt-sensitive hypertension likely through downregulation of the Klotho/ERK (extracellular signal-regulated kinase) signaling and upregulation of the WNK (with-no-lysine kinase) signaling. CONCLUSIONS This study provides the first evidence that KDM6A plays an essential role in maintaining normal tubular function and blood pressure. Renal tubule cell specific KDM6A deficiency causes hypertension due to increased H3K27me3 levels and the resultant downregulation of Klotho gene expression which disrupts the Klotho/ERK/NCC/NKCC2 signaling.
Collapse
Affiliation(s)
- Xiaobin Han
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Leah Akinseye
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Zhongjie Sun
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
4
|
Zhang J, Zhang A. Relationships between serum Klotho concentrations and cognitive performance among older chronic kidney disease patients with albuminuria in NHANES 2011-2014. Front Endocrinol (Lausanne) 2023; 14:1215977. [PMID: 37560310 PMCID: PMC10407554 DOI: 10.3389/fendo.2023.1215977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 07/07/2023] [Indexed: 08/11/2023] Open
Abstract
Background The potential relationship between Klotho and cognitive function is limited and controversial. This study aimed to quantify the association of Klotho and cognitive impairment in chronic kidney disease (CKD) patients with albuminuria. Methods Serum Klotho was measured by enzyme-linked immunosorbent assay. Patients with urine albumin to creatinine ratio (UACR) > 30mg/g from the National Health and Nutrition Survey (NHANES) 2011-2014 were divided into 4 groups according to the quartile of Klotho. Cognitive function was examined using the Consortium to Establish a Registry for Alzheimer's Disease (CERAD), Digit Symbol Substitution Test (DSST), and Animal Fluency Test. The relationship between Klotho and cognitive function was analyzed by multivariable regression and subgroup analysis. Results Among 368 CKD patients with albuminuria, we found that Klotho was negatively associated with creatinine, and positively associated with hemoglobin, and estimated glomerular filtration rate. No significant linear relationship was showed between Klotho (as a continuous variable) and cognitive function. When regarded Klotho as a category variable, patients in the quartile 3 group were at a better cognitive performance for CEARD-word learning subset and DSST, especially in the CKD patients with 30 mg/g < UACR <300 mg/g, but not in participants with UACR > 300 mg/g. Conclusions The increased Klotho was associated with an increased cognitive function in CKD patients with microalbuminuria. Further studies are needed to demonstrate whether Klotho may be a beneficial biomarker of cognitive health and neurodegeneration.
Collapse
Affiliation(s)
- Jialing Zhang
- Department of Nephrology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Aihua Zhang
- Department of Nephrology, Xuanwu Hospital, Capital Medical University, Beijing, China
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
5
|
Fountoulakis N, Psefteli PM, Maltese G, Gnudi L, Siow RC, Karalliedde J. Reduced Levels of the Antiaging Hormone Klotho are Associated With Increased Aortic Stiffness in Diabetic Kidney Disease. Kidney Int Rep 2023; 8:1380-1388. [PMID: 37441489 PMCID: PMC10334399 DOI: 10.1016/j.ekir.2023.04.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 04/17/2023] [Accepted: 04/24/2023] [Indexed: 07/15/2023] Open
Abstract
Introduction Aortic pulse wave velocity (Ao-PWV) predicts cardiovascular and kidney disease in type 2 diabetes (T2D). Klotho is a circulating antiaging hormone (sKlotho) with putative cardiorenal protective effects. The relationship between sKlotho and Ao-PWV in diabetic kidney disease (DKD) is unknown. Methods In a cross-sectional cohort study, the correlation of sKlotho measured by a validated immunoassay, and Ao-PWV measured by applanation tonometry, was investigated in 172 participants with T2D and early stage DKD (all had estimated glomerular filtration rate [eGFR] >45 ml/min) on stable renin angiotensin system (RAS) inhibition. In cultured human aortic smooth muscle cells (HASMCs) stimulated with angiotensin II (AngII), the effects of recombinant human sKlotho pretreatment were assessed on intracellular calcium ([Ca2+]i) responses and expression of proteins associated with proosteogenic HASMC phenotypes. Results Mean (range) age of the cohort was 61.3 years (40-82) and 65% were male. Mean (±SD) Ao-PWV was 11.4 (±2.3) m/s, eGFR 78.8 (±23.5) and median (interquartile range) sKlotho of 358.5 (194.2-706.3) pg/ml. In multivariable linear regression analyses, we observed a statistically significant inverse relationship between sKlotho and Ao-PWV, which was independent of clinical risk factors for cardiorenal disease. Pretreatment of cultured HASMC with sKlotho significantly attenuated AngII-stimulated [Ca2+]i transients and reduced osteogenic collagen (Col1a2) expression. Conclusions In individuals with T2D and early DKD, lower levels of sKlotho are associated with increased Ao-PWV. Taken together with the direct effect of sKlotho on mediators of aortic wall stiffness in vitro, these findings may explain the enhanced risk of cardiorenal disease in DKD.
Collapse
Affiliation(s)
- Nikolaos Fountoulakis
- Unit for Metabolic Medicine, School of Cardiovascular and Metabolic Medicine and Sciences, Faculty of Life Sciences and Medicine, King’s British Heart Foundation Center of Research Excellence, King’s College London, London, UK
| | - Paraskevi-Maria Psefteli
- Unit for Metabolic Medicine, School of Cardiovascular and Metabolic Medicine and Sciences, Faculty of Life Sciences and Medicine, King’s British Heart Foundation Center of Research Excellence, King’s College London, London, UK
| | - Giuseppe Maltese
- Unit for Metabolic Medicine, School of Cardiovascular and Metabolic Medicine and Sciences, Faculty of Life Sciences and Medicine, King’s British Heart Foundation Center of Research Excellence, King’s College London, London, UK
| | - Luigi Gnudi
- Unit for Metabolic Medicine, School of Cardiovascular and Metabolic Medicine and Sciences, Faculty of Life Sciences and Medicine, King’s British Heart Foundation Center of Research Excellence, King’s College London, London, UK
| | - Richard C. Siow
- Unit for Metabolic Medicine, School of Cardiovascular and Metabolic Medicine and Sciences, Faculty of Life Sciences and Medicine, King’s British Heart Foundation Center of Research Excellence, King’s College London, London, UK
| | - Janaka Karalliedde
- Unit for Metabolic Medicine, School of Cardiovascular and Metabolic Medicine and Sciences, Faculty of Life Sciences and Medicine, King’s British Heart Foundation Center of Research Excellence, King’s College London, London, UK
| |
Collapse
|
6
|
Alhodieb FS, Rahman MA, Barkat MA, Alanezi AA, Barkat HA, Hadi HA, Harwansh RK, Mittal V. Nanomedicine-driven therapeutic interventions of autophagy and stem cells in the management of Alzheimer's disease. Nanomedicine (Lond) 2023; 18:145-168. [PMID: 36938800 DOI: 10.2217/nnm-2022-0108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2023] Open
Abstract
Drug-loaded, brain-targeted nanocarriers could be a promising tool in overcoming the challenges associated with Alzheimer's disease therapy. These nanocargoes are enormously flexible to functionalize and facilitate the delivery of drugs to brain cells by bridging the blood-brain barrier and into brain cells. To date, modifications have included nanoparticles (NPs) coating with tunable surfactants/phospholipids, covalently attaching polyethylene glycol chains (PEGylation), and tethering different targeting ligands to cell-penetrating peptides in a manner that facilitates their entry across the BBB and downregulates various pathological hallmarks as well as intra- and extracellular signaling pathways. This review provides a brief update on drug-loaded, multifunctional nanocarriers and the therapeutic intervention of autophagy and stem cells in the management of Alzheimer's disease.
Collapse
Affiliation(s)
- Fahad Saad Alhodieb
- Department of Clinical Nutrition, College of Applied Health Sciences in Arras, Qassim University, Ar Rass, 51921, Saudi Arabia
| | | | - Muhammad Abul Barkat
- Department of Pharmaceutics, College of Pharmacy, University of Hafr Al-Batin, Al Jamiah, Hafr Al Batin, 39524, Saudi Arabia
| | - Abdulkareem A Alanezi
- Department of Pharmaceutics, College of Pharmacy, University of Hafr Al-Batin, Al Jamiah, Hafr Al Batin, 39524, Saudi Arabia
| | - Harshita Abul Barkat
- Department of Pharmaceutics, College of Pharmacy, University of Hafr Al-Batin, Al Jamiah, Hafr Al Batin, 39524, Saudi Arabia.,Dermatopharmaceutics Research Group, Faculty of Pharmacy, International Islamic University Malaysia, Kuantan, Pahang, 25200, Malaysia
| | - Hazrina Ab Hadi
- Dermatopharmaceutics Research Group, Faculty of Pharmacy, International Islamic University Malaysia, Kuantan, Pahang, 25200, Malaysia
| | - Ranjit K Harwansh
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, India
| | - Vineet Mittal
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| |
Collapse
|
7
|
Perfluorooctanoic acid induces tight junction injury of Sertoli cells by blocking autophagic flux. Food Chem Toxicol 2023; 173:113649. [PMID: 36736878 DOI: 10.1016/j.fct.2023.113649] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/11/2023] [Accepted: 01/31/2023] [Indexed: 02/04/2023]
Abstract
Perfluorooctanoic acid (PFOA), a man-made chemical widely used in consumers, could cause male reproductive toxicity by disrupting blood-testis barrier (BTB) integrity. Autophagy in Sertoli cells is essential for regulation of spermatogenesis and BTB. However, it remains a mystery that whether PFOA-induced BTB injury is associated with autophagy in Sertoli cells. In this study, we found that PFOA dose-dependently disrupted tight junction (TJ) function in Sertoli cells in vivo and in vitro. Furthermore, the results from transmission electron microscopy, Western blot and immunofluorescence analysis revealed that PFOA induced the accumulation of autophagosome in testicular Sertoli cells as well as TM4 cells. Further study confirmed that autophagosome accumulation resulted from the blockage of autophagic degradation because of disruption of autophagosome and lysosome fusion via downregulation of the expression of α-SNAP. In parallel, the overexpressed MMP9 was also observed in vivo and in vitro. Conversely, overexpression of α-SNAP inhibited the expression of MMP9 in TM4 cells. In conclusion, PFOA blocks autophagic flux through downregulating the expression levels of α-SNAP in Sertoli cells, and then induces the accumulation of MMP9 leading to disruption of TJ function. This finding will provide clues for effective prevention and treatment of PFOA-induced male reproductive toxicity.
Collapse
|
8
|
Chen J, Lin Y, Sun Z. Inhibition of miR-101-3p prevents human aortic valve interstitial cell calcification through regulation of CDH11/SOX9 expression. Mol Med 2023; 29:24. [PMID: 36809926 PMCID: PMC9945614 DOI: 10.1186/s10020-023-00619-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 02/10/2023] [Indexed: 02/24/2023] Open
Abstract
BACKGROUND Calcific aortic valve disease (CAVD) is the second leading cause of adult heart diseases. The purpose of this study is to investigate whether miR-101-3p plays a role in the human aortic valve interstitial cells (HAVICs) calcification and the underlying mechanisms. METHODS Small RNA deep sequencing and qPCR analysis were used to determine changes in microRNA expression in calcified human aortic valves. RESULTS The data showed that miR-101-3p levels were increased in the calcified human aortic valves. Using cultured primary HAVICs, we demonstrated that the miR-101-3p mimic promoted calcification and upregulated the osteogenesis pathway, while anti-miR-101-3p inhibited osteogenic differentiation and prevented calcification in HAVICs treated with the osteogenic conditioned medium. Mechanistically, miR-101-3p directly targeted cadherin-11 (CDH11) and Sry-related high-mobility-group box 9 (SOX9), key factors in the regulation of chondrogenesis and osteogenesis. Both CDH11 and SOX9 expressions were downregulated in the calcified human HAVICs. Inhibition of miR-101-3p restored expression of CDH11, SOX9 and ASPN and prevented osteogenesis in HAVICs under the calcific condition. CONCLUSION miR-101-3p plays an important role in HAVIC calcification through regulation of CDH11/SOX9 expression. The finding is important as it reveals that miR-1013p may be a potential therapeutic target for calcific aortic valve disease.
Collapse
Affiliation(s)
- Jianglei Chen
- Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Yi Lin
- Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Zhongjie Sun
- Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA. .,Department of Physiology, College of Medicine, UT Cardiovascular Institute, University of Tennessee Health Science Center, 956 Court Avenue, Memphis, TN, 38163, USA.
| |
Collapse
|
9
|
Yu H, Yu Q, Mi Y, Wang P, Jin S, Xiao L, Guo Q, Wu Y. Hydrogen Sulfide Inhibited Sympathetic Activation in D-Galactose-Induced Aging Rats by Upregulating Klotho and Inhibiting Inflammation in the Paraventricular Nucleus. Biomedicines 2023; 11:biomedicines11020566. [PMID: 36831102 PMCID: PMC9953477 DOI: 10.3390/biomedicines11020566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/19/2023] [Accepted: 02/08/2023] [Indexed: 02/17/2023] Open
Abstract
The present study aimed to explore the central relationship between cardiovascular conditions and aging. D-galactose (D-gal) was utilized to induce an accelerated aging model and to evaluate the effects of hydrogen sulfide (H2S) on aging-related cardiovascular risk factors and mechanisms. Eight-week-old Sprague Dawley rats were given an intraperitoneal injection of 250 mg/kg D-gal every day with or without H2S (56 μmol/kg) for 12 weeks. We found that D-gal treatment induced a noticeably aging-related increase in p16, p53 and p21 protein levels and senescence-associated beta-galactosidase staining. In addition, the level of noradrenalin was increased, accompanied by enhanced blood pressure and renal sympathetic nerve activity in aged rats. The greater sympathetic responses were related with the increased level of inflammation. The decreased level of klotho in the paraventricular nucleus neuron also contributed to sympathetic activation in D-gal-induced aged rats. However, the exogenous administration of H2S attenuated the sympathetic activity in aged rats, as evidenced by the decreased blood pressure, renal sympathetic nerve activity and noradrenalin level. The ameliorated cellular senescence, inflammation and heightened klotho in the paraventricular nucleus were attributed to the protective effects of H2S. The present study provides further evidence for the drug development of H2S for the prevention or treatment of the aging-associated cardiovascular diseases.
Collapse
Affiliation(s)
- Hao Yu
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
| | - Qiyao Yu
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
- Department of Research, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Yuan Mi
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
- Department of Emergency, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Ping Wang
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
| | - Sheng Jin
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
| | - Lin Xiao
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
| | - Qi Guo
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
- Experimental Center for Teaching, Hebei Medical University, Shijiazhuang 050017, China
- Correspondence: (Q.G.); (Y.W.); Tel./Fax: +86-311-8626-1288 (Y.W.)
| | - Yuming Wu
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
- Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang 050017, China
- Key Laboratory of Vascular Medicine of Hebei Province, Shijiazhuang 050017, China
- Correspondence: (Q.G.); (Y.W.); Tel./Fax: +86-311-8626-1288 (Y.W.)
| |
Collapse
|
10
|
Yan Y, Chen J. Association between serum Klotho concentration and all-cause and cardiovascular mortality among American individuals with hypertension. Front Cardiovasc Med 2022; 9:1013747. [PMID: 36457804 PMCID: PMC9705974 DOI: 10.3389/fcvm.2022.1013747] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 10/31/2022] [Indexed: 08/03/2023] Open
Abstract
BACKGROUND AND AIMS Evidence indicates that serum Klotho concentration is associated with mortality in patients with chronic kidney disease (CKD). However, evidence on this association among people with hypertension is scarce. Therefore, we aimed to examine the association between serum Klotho concentration and all-cause and cardiovascular mortality in American patients with hypertension. METHODS AND RESULTS We included 6,778 participants with hypertension from the National Health and Nutrition Examination Survey (NHANES) 2007-2014. A Cox proportional hazard model was used to compute the hazard ratios (HRs) and 95% confidence intervals (CIs). The correlation between serum Klotho concentration and mortality was determined using restricted cubic spline and piecewise linear regression analyses. During 36,714 person-years of follow-up, 575 deaths were documented. Lower serum Klotho concentration was associated with increased all-cause mortality, but not cardiovascular mortality after multivariate adjustment. According to spline analysis, the correlation between serum Klotho concentration and all-cause mortality was non-linear (P < 0.001), and the threshold value was 574 pg/mL. The HR below the threshold point was 0.79 (95% CI: 0.67-0.93); no significant difference was found above the threshold point. CONCLUSION Higher serum Klotho concentration was associated with lower all-cause mortality, but not cardiovascular mortality in patients with hypertension with or without chronic renal impairment.
Collapse
Affiliation(s)
- Yuqin Yan
- *Correspondence: Yuqin Yan, ; orcid.org/0000-0001-6973-8909
| | | |
Collapse
|
11
|
Klotho Ameliorates Vascular Calcification via Promoting Autophagy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7192507. [PMID: 36338347 PMCID: PMC9629936 DOI: 10.1155/2022/7192507] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 09/01/2022] [Accepted: 09/07/2022] [Indexed: 02/05/2023]
Abstract
Vascular calcification (VC) is regarded as a common feature of vascular aging. Klotho deficiency reportedly contributes to VC, which can be ameliorated by restoration of Klotho expression. However, the specific mechanisms involved remain unclear. Here, we investigated the role of autophagy in the process of Klotho-inhibiting VC. The clinical study results indicated that, based on Agatston score, serum Klotho level was negatively associated with aortic calcification. Then, Klotho-deficient mice exhibited aortic VC, which could be alleviated with the supplementation of Klotho protein. Moreover, autophagy increased in the aorta of Klotho-deficient mice and protected against VC. Finally, we found that Klotho ameliorated calcification by promoting autophagy both in the aorta of Klotho-deficient mice and in mouse vascular smooth muscle cells (MOVAS) under calcifying conditions. These findings indicate that Klotho deficiency induces increased autophagy to protect against VC and that Klotho expression further enhances autophagy to ameliorate calcification. This study is beneficial to exploring the underlying mechanisms of Klotho regulating VC, which has important guiding significance for future clinical studies in the treatment of VC.
Collapse
|
12
|
Yu LX, Li SS, Sha MY, Kong JW, Ye JM, Liu QF. The controversy of klotho as a potential biomarker in chronic kidney disease. Front Pharmacol 2022; 13:931746. [PMID: 36210812 PMCID: PMC9532967 DOI: 10.3389/fphar.2022.931746] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022] Open
Abstract
Klotho is an identified longevity gene with beneficial pleiotropic effects on the kidney. Evidence shows that a decline in serum Klotho level occurs in early chronic kidney disease (CKD) and continues as CKD progresses. Klotho deficiency is associated with poor clinical outcomes and CKD mineral bone disorders (CKD-MBD). Klotho has been postulated as a candidate biomarker in the evaluation of CKD. However, the evidence for the clinical significance of the relationship between Klotho and kidney function, CKD stage, adverse kidney and/or non-kidney outcomes, and CKD-MBD remains inconsistent and in some areas, contradictory. Therefore, there is uncertainty as to whether Klotho is a potential biomarker in CKD; a general consensus regarding the clinical significance of Klotho in CKD has not been reached, and there is limited evidence synthesis in this area. To address this, we have systematically assessed the areas of controversy, focusing on the inconsistencies in the evidence base. We used a PICOM strategy to search for relevant studies and the Newcastle–Ottawa Scale scoring to evaluate included publications. We reviewed the inconsistent clinical findings based on the relationship of Klotho with CKD stage, kidney and/or non-kidney adverse outcomes, and CKD-MBD in human studies. Subsequently, we assessed the underlying sources of the controversies and highlighted future directions to resolve these inconsistencies and clarify whether Klotho has a role as a biomarker in clinical practice in CKD.
Collapse
Affiliation(s)
- Li-Xia Yu
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Sha-Sha Li
- Clinical Research and Lab Center, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Min-Yue Sha
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Jia-Wei Kong
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Jian-Ming Ye
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
- *Correspondence: Jian-Ming Ye, ; Qi-Feng Liu,
| | - Qi-Feng Liu
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
- *Correspondence: Jian-Ming Ye, ; Qi-Feng Liu,
| |
Collapse
|
13
|
Fan J, Wang S, Chen K, Sun Z. Aging impairs arterial compliance via Klotho-mediated downregulation of B-cell population and IgG levels. Cell Mol Life Sci 2022; 79:494. [PMID: 36001158 PMCID: PMC10082671 DOI: 10.1007/s00018-022-04512-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 07/31/2022] [Accepted: 08/01/2022] [Indexed: 11/27/2022]
Abstract
OBJECTIVE Aging is associated with compromised immune function and arterial remodeling and stiffness. The purpose of this study is to investigate whether in vivo AAV-based delivery of secreted Klotho (SKL) gene (AAV-SKL) improves aging- and senescence-associated immune dysfunction and arterial stiffness. METHODS AND RESULTS Senescence-accelerated mice prone strain 1 (SAMP1, 10 months) and old mice (20 months) were used. Serum SKL levels, B-cell population and serum IgG levels were markedly decreased in SAMP1 and old mice. Rescue of downregulation of serum SKL levels by in vivo AAV2-based delivery of SKL gene (AAV-SKL) increased B-cell population and serum IgG levels and attenuated arterial stiffness in SAMP1 and old mice. Thus, Klotho deficiency may play a role in senescence- and aging-associated humoral immune dysfunction and arterial stiffness. Vascular infiltration of inflammatory cells and expression of TGFβ1, collagen 1, scleraxis, MMP-2 and MMP-9 were increased while the elastin level was decreased in aortas of SAMP1 and old mice which can be rescued by AAV-SKL. Interestingly, treatment with IgG effectively rescued arterial inflammation and remodeling and attenuated arterial stiffness and hypertension in aging mice. In cultured B-lymphoblast cells, we further showed that SKL regulates B-cell proliferation and maturation partly via the NFkB pathway. CONCLUSION Aging-associated arterial stiffening may be largely attributed to downregulation of B-cell population and serum IgG levels. AAV-SKL attenuates arterial stiffness in aging mice partly via restoring B-cell population and serum IgG levels which attenuates aging-associated vascular inflammation and arterial remodeling.
Collapse
Affiliation(s)
- Jun Fan
- Department of Physiology, College of Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - Shirley Wang
- Department of Physiology, College of Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - Kai Chen
- Department of Physiology, College of Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, A302 Coleman Building, 956 Court Avenue, Memphis, TN, 38163, USA
| | - Zhongjie Sun
- Department of Physiology, College of Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK, USA.
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, A302 Coleman Building, 956 Court Avenue, Memphis, TN, 38163, USA.
| |
Collapse
|
14
|
Cardioprotection of Klotho against myocardial infarction-induced heart failure through inducing autophagy. Mech Ageing Dev 2022; 207:111714. [PMID: 35931242 DOI: 10.1016/j.mad.2022.111714] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 07/26/2022] [Accepted: 07/29/2022] [Indexed: 11/23/2022]
Abstract
Myocardial infarction (MI) is the most common cause of heart failure (HF) worldwide. The aim of this study was to investigate the role of Klotho in cardiac function and remodeling as well as its underlying mechanism in mice with MI-induced HF. For in vivo analyses, MI or sham MI were established in C57BL/6 mice. For in vitro analyses, the H9C2 cells were used to establish a model of oxygen glucose deprivation (OGD). The In vivo and in vitro models were treated with or without Klotho. 3-methyladenine (3-MA) was used to inhibit autophagy in MI mice and H9C2 cells. Cardiac function, cardiac fibrosis, cardiomyocyte autophagy, inflammatory cytokines and myocardial apoptosis were measured. Our results revealed that Klotho significantly improved cardiac function and remodeling, reduced cardiac fibrosis, and suppressed the levels of myocardial inflammatory factors and apoptosis in MI-induced HF model. Klotho enhanced autophagy in cardiomyocytes and inhibited PI3K/AKT/mTOR signaling pathway in the mouse model of MI. Similar observations were made in the OGD model after treatment with Klotho. However, the cardioprotective effects of Klotho was significantly suppressed by 3-MA. Our data indicate that Klotho exerts its cardioprotective effects against MI-induced HF by inducing autophagy through the inhibition of PI3k/AKT/mTOR signaling pathway.
Collapse
|
15
|
Han X, Sun Z. Adult Mouse Kidney Stem Cells Orchestrate the De Novo Assembly of a Nephron via Sirt2-Modulated Canonical Wnt/β-Catenin Signaling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104034. [PMID: 35315252 PMCID: PMC9130916 DOI: 10.1002/advs.202104034] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 02/24/2022] [Indexed: 06/14/2023]
Abstract
Generation of kidney organoids using autologous kidney stem cells represents an attractive strategy for treating and potentially replacing the failing kidneys. However, whether adult mammalian kidney stem cells have regenerative capacity remains unknown. Here, previously unidentified adult kidney Sca1+ Oct4+ stem/progenitor cells are isolated. Interestingly, culturing these cells leads to generation of kidney-like structures. First, the assembly of self-organizing 3D kidney-like structures is observed. These kidney organoids contain podocytes, proximal tubules, and endothelial cells that form networks of capillary loop-like structures. Second, the differentiation of kidney stem cells into functionally mature tubules and self-organizing kidney-shaped structures in monolayer culture that selectively endocytoses dextran, is shown. Finally, the de novo generation of an entire self-organizing nephron from monolayer cultures is observed. Mechanistically, it is demonstrated that Sirt2-mediated canonical Wnt/β-catenin signaling is critical for the development of kidney organoids. Thus, the first evidence is provided that the adult mouse kidney stem cells are capable of de novo generating kidney organoids.
Collapse
Affiliation(s)
- Xiaobin Han
- Department of PhysiologyUniversity of Tennessee Health Science CenterMemphisTN38163USA
| | - Zhongjie Sun
- Department of PhysiologyUniversity of Tennessee Health Science CenterMemphisTN38163USA
| |
Collapse
|
16
|
Fung TY, Iyaswamy A, Sreenivasmurthy SG, Krishnamoorthi S, Guan XJ, Zhu Z, Su CF, Liu J, Kan Y, Zhang Y, Wong HLX, Li M. Klotho an Autophagy Stimulator as a Potential Therapeutic Target for Alzheimer’s Disease: A Review. Biomedicines 2022; 10:biomedicines10030705. [PMID: 35327507 PMCID: PMC8945569 DOI: 10.3390/biomedicines10030705] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/17/2022] [Accepted: 03/17/2022] [Indexed: 02/04/2023] Open
Abstract
Alzheimer’s disease (AD) is an age-associated neurodegenerative disease; it is the most common cause of senile dementia. Klotho, a single-pass transmembrane protein primarily generated in the brain and kidney, is active in a variety of metabolic pathways involved in controlling neurodegeneration and ageing. Recently, many studies have found that the upregulation of Klotho can improve pathological cognitive deficits in an AD mice model and have demonstrated that Klotho plays a role in the induction of autophagy, a major contributing factor for AD. Despite the close association between Klotho and neurodegenerative diseases, such as AD, the underlying mechanism by which Klotho contributes to AD remains poorly understood. In this paper, we will introduce the expression, location and structure of Klotho and its biological functions. Specifically, this review is devoted to the correlation of Klotho protein and the AD phenotype, such as the effect of Klotho in upregulating the amyloid-beta clearance and in inducing autophagy for the clearance of toxic proteins, by regulating the autophagy lysosomal pathway (ALP). In summary, the results of multiple studies point out that targeting Klotho would be a potential therapeutic strategy in AD treatment.
Collapse
Affiliation(s)
- Tsz Yan Fung
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (T.Y.F.); (S.G.S.); (S.K.); (X.-J.G.); (Z.Z.); (C.-F.S.); (J.L.); (Y.K.)
| | - Ashok Iyaswamy
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (T.Y.F.); (S.G.S.); (S.K.); (X.-J.G.); (Z.Z.); (C.-F.S.); (J.L.); (Y.K.)
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen 518057, China
- Correspondence: or (A.I.); (H.L.X.W.); (M.L.); Tel.: +852-3411-2919 (M.L.); Fax: +852-3411-2461 (M.L.)
| | - Sravan G. Sreenivasmurthy
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (T.Y.F.); (S.G.S.); (S.K.); (X.-J.G.); (Z.Z.); (C.-F.S.); (J.L.); (Y.K.)
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen 518057, China
| | - Senthilkumar Krishnamoorthi
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (T.Y.F.); (S.G.S.); (S.K.); (X.-J.G.); (Z.Z.); (C.-F.S.); (J.L.); (Y.K.)
- Centre for Trans-Disciplinary Research, Department of Pharmacology, Saveetha Dental College and Hospitals, Chennai 600077, Tamil Nadu, India
| | - Xin-Jie Guan
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (T.Y.F.); (S.G.S.); (S.K.); (X.-J.G.); (Z.Z.); (C.-F.S.); (J.L.); (Y.K.)
| | - Zhou Zhu
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (T.Y.F.); (S.G.S.); (S.K.); (X.-J.G.); (Z.Z.); (C.-F.S.); (J.L.); (Y.K.)
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen 518057, China
| | - Cheng-Fu Su
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (T.Y.F.); (S.G.S.); (S.K.); (X.-J.G.); (Z.Z.); (C.-F.S.); (J.L.); (Y.K.)
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen 518057, China
| | - Jia Liu
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (T.Y.F.); (S.G.S.); (S.K.); (X.-J.G.); (Z.Z.); (C.-F.S.); (J.L.); (Y.K.)
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen 518057, China
| | - Yuxuan Kan
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (T.Y.F.); (S.G.S.); (S.K.); (X.-J.G.); (Z.Z.); (C.-F.S.); (J.L.); (Y.K.)
| | - Yuan Zhang
- Shenzhen Key Laboratory of Neurosurgery, Department of Neurosurgery, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518025, China;
| | - Hoi Leong Xavier Wong
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
- Correspondence: or (A.I.); (H.L.X.W.); (M.L.); Tel.: +852-3411-2919 (M.L.); Fax: +852-3411-2461 (M.L.)
| | - Min Li
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China; (T.Y.F.); (S.G.S.); (S.K.); (X.-J.G.); (Z.Z.); (C.-F.S.); (J.L.); (Y.K.)
- Institute for Research and Continuing Education, Hong Kong Baptist University, Shenzhen 518057, China
- Correspondence: or (A.I.); (H.L.X.W.); (M.L.); Tel.: +852-3411-2919 (M.L.); Fax: +852-3411-2461 (M.L.)
| |
Collapse
|
17
|
Serum klotho and pulse pressure; insight from NHANES. Int J Cardiol 2022; 355:54-58. [DOI: 10.1016/j.ijcard.2022.02.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/04/2022] [Accepted: 02/16/2022] [Indexed: 11/20/2022]
|
18
|
Zeng Y, Du X, Yao X, Qiu Y, Jiang W, Shen J, Li L, Liu X. Mechanism of cell death of endothelial cells regulated by mechanical forces. J Biomech 2021; 131:110917. [PMID: 34952348 DOI: 10.1016/j.jbiomech.2021.110917] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 12/26/2022]
Abstract
Cell death of endothelial cells (ECs) is a common devastating consequence of various vascular-related diseases. Atherosclerosis, hypertension, sepsis, diabetes, cerebral ischemia and cardiac ischemia/reperfusion injury, and chronic kidney disease remain major causes of morbidity and mortality worldwide, in which ECs are constantly subjected to a great amount of dynamic changed mechanical forces including shear stress, extracellular matrix stiffness, mechanical stretch and microgravity. A thorough understanding of the regulatory mechanisms by which the mechanical forces controlled the cell deaths including apoptosis, autophagy, and pyroptosis is crucial for the development of new therapeutic strategies. In the present review, experimental and clinical data highlight that nutrient depletion, oxidative stress, tumor necrosis factor-α, high glucose, lipopolysaccharide, and homocysteine possess cytotoxic effects in many tissues and induce apoptosis of ECs, and that sphingosine-1-phosphate protects ECs. Nevertheless, EC apoptosis in the context of those artificial microenvironments could be enhanced, reduced or even reversed along with the alteration of patterns of shear stress. An appropriate level of autophagy diminishes EC apoptosis to some extent, in addition to supporting cell survival upon microenvironment challenges. The intervention of pyroptosis showed a profound effect on atherosclerosis. Further cell and animal studies are required to ascertain whether the alterations in the levels of cell deaths and their associated regulatory mechanisms happen at local lesion sites with considerable mechanical force changes, for preventing senescence and cell deaths in the vascular-related diseases.
Collapse
Affiliation(s)
- Ye Zeng
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Xiaoqiang Du
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xinghong Yao
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yan Qiu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Wenli Jiang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Junyi Shen
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Liang Li
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
19
|
Li-Zhen L, Chen ZC, Wang SS, Liu WB, Zhuang XD. Klotho deficiency causes cardiac ageing by impairing autophagic and activating apoptotic activity. Eur J Pharmacol 2021; 911:174559. [PMID: 34637700 DOI: 10.1016/j.ejphar.2021.174559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 10/03/2021] [Accepted: 10/06/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVE In this study, it was hypothesized that klotho deficiency plays an essential role in cardiac ageing in vivo and demonstrated that supplementation with exogenous klotho protects against cardiomyocyte ageing in vitro. METHODS We measured the lifespan of wild-type (WT) and klotho-hypomorphic mutant (KL-/-) mice and recorded the cardiac function of the mice through echocardiography. We used immunofluorescence staining to detect the LC3B (microtubule-associated protein light chain 3 B), Beclin 1, Bax and Bcl 2 proteins. In vitro, H9c2 cells were incubated with different levels of D-galactose (D-gal) with or without klotho. SA-β-galactosidase staining and western blotting were performed to detect ageing-associated proteins (P53, P21 and P16), autophagy-associated proteins (LC3 II/LC3 I and Beclin 1) and apoptosis-associated proteins (Bax and Bcl 2). Moreover, one-step TUNEL apoptosis, CCK-8, cell morphology, Hoechst 33258 staining, lactate dehydrogenase (LDH) release, and caspase-3 activity assays were performed, and intracellular reactive oxygen species (ROS) levels were measured. RESULTS Genetic klotho deficiency decreased lifespan and cardiac function in mice, impaired autophagic activity and increased apoptotic activity. Exogenous klotho attenuated cardiomyocyte ageing and reversed changes in autophagic and apoptotic activity caused by D-gal. Moreover, klotho supplementation prevented D-gal-induced oxidative stress and cytotoxicity. CONCLUSIONS Klotho might have a protective effect on cardiac ageing via autophagy activation and apoptosis inhibition.
Collapse
Affiliation(s)
- Liao Li-Zhen
- Guangdong Engineering Research Center for Light and Health, Guangzhou Higher Education Mega Center, Guangzhou, Guangdong, PR China; Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, Guangdong, PR China
| | - Zhi-Chong Chen
- Cardiovascular Department, The Sixth Affiliated Hospital of Sun Yat-sen University, No. 26, Erheng Road, Yuan Village, Tianhe District, Guangzhou, Guangdong Province, PR China
| | - Sui-Sui Wang
- Guangdong Engineering Research Center for Light and Health, Guangzhou Higher Education Mega Center, Guangzhou, Guangdong, PR China; Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, Guangdong, PR China
| | - Wen-Bin Liu
- Guangdong Engineering Research Center for Light and Health, Guangzhou Higher Education Mega Center, Guangzhou, Guangdong, PR China; Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, Guangdong, PR China
| | - Xiao-Dong Zhuang
- Cardiology Department, The First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou, Guangdong, PR China.
| |
Collapse
|
20
|
Zhou H, Pu S, Zhou H, Guo Y. Klotho as Potential Autophagy Regulator and Therapeutic Target. Front Pharmacol 2021; 12:755366. [PMID: 34737707 PMCID: PMC8560683 DOI: 10.3389/fphar.2021.755366] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 10/07/2021] [Indexed: 12/22/2022] Open
Abstract
The protein Klotho can significantly delay aging, so it has attracted widespread attention. Abnormal downregulation of Klotho has been detected in several aging-related diseases, such as Alzheimer’s disease, kidney injury, cancer, chronic obstructive pulmonary disease (COPD), vascular disease, muscular dystrophy and diabetes. Conversely, many exogenous and endogenous factors, several drugs, lifestyle changes and genetic manipulations were reported to exert therapeutic effects through increasing Klotho expression. In recent years, Klotho has been identified as a potential autophagy regulator. How Klotho may contribute to reversing the effects of aging and disease became clearer when it was linked to autophagy, the process in which eukaryotic cells clear away dysfunctional proteins and damaged organelles: the abovementioned diseases involve abnormal autophagy. Interestingly, growing evidence indicates that Klotho plays a dual role as inducer or inhibitor of autophagy in different physiological or pathological conditions through its influence on IGF-1/PI3K/Akt/mTOR signaling pathway, Beclin 1 expression and activity, as well as aldosterone level, which can help restore autophagy to beneficial levels. The present review examines the role of Klotho in regulating autophagy in Alzheimer’s disease, kidney injury, cancer, COPD, vascular disease, muscular dystrophy and diabetes. Targeting Klotho may provide a new perspective for preventing and treating aging-related diseases.
Collapse
Affiliation(s)
- Hongjing Zhou
- Department of Pharmacy, Chengdu Fifth People's Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shiyun Pu
- Department of Pharmacy, Chengdu Fifth People's Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Houfeng Zhou
- Department of Pharmacy, Chengdu Fifth People's Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuanxin Guo
- Department of Pharmacy, Chengdu Fifth People's Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
21
|
Li C, Lin L, Zhang L, Xu R, Chen X, Ji J, Li Y. Long noncoding RNA p21 enhances autophagy to alleviate endothelial progenitor cells damage and promote endothelial repair in hypertension through SESN2/AMPK/TSC2 pathway. Pharmacol Res 2021; 173:105920. [PMID: 34601081 DOI: 10.1016/j.phrs.2021.105920] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/22/2021] [Accepted: 09/28/2021] [Indexed: 12/11/2022]
Abstract
Vascular damage of hypertension has been the focus of hypertension treatment, and endothelial progenitor cells (EPCs) play an important role in the repair of vascular endothelial damage. Functional damage and decreased number of EPCs are observed in the peripheral circulation of hypertensive patients, but its mechanism is not yet elucidated. Here, we show that the number of EPCs in hypertensive patients is significantly lower than that of normal population, and the cell function decreases with a higher proportion of EPCs at later stages. A decrease in autophagy is responsible for the senescence and damage of EPCs induced by AngII. Moreover, lncRNA-p21 plays a critical regulator role in EPCs' senescence and dysfunction. Furthermore, lncRNA-p21 activates SESN2/AMPK/TSC2 pathway by promoting the transcriptional activity of p53 and enhances autophagy to protect against AngII-induced EPC damage. The data provide evidence that a reversal of decreased autophagy serves as the protective mechanism of EPC injury in hypertensive patients, and lncRNA-p21 is a new therapeutic target for vascular endothelial repair in hypertension.
Collapse
Affiliation(s)
- Chao Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Lin Lin
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Lei Zhang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Ran Xu
- Tianqiao District People's Hospital, Jinan 250031, China
| | - Xiaoqing Chen
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Jingkang Ji
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yunlun Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250000, China.
| |
Collapse
|
22
|
Chen K, Sun Z. Estrogen inhibits renal Na-Pi Co-transporters and improves klotho deficiency-induced acute heart failure. Redox Biol 2021; 47:102173. [PMID: 34678656 PMCID: PMC8577443 DOI: 10.1016/j.redox.2021.102173] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/17/2021] [Accepted: 10/17/2021] [Indexed: 01/06/2023] Open
Abstract
Objective and hypothesis Klotho is an aging-suppressor gene. Mutation of Klotho gene causes hyperphosphatemia and acute heart failure. However, the relationship of hyperphosphatemia and acute heart failure is unclear. We hypothesize that hyperphosphatemia mediates Klotho deficiency-induced acute heart failure and further that therapeutic reduction of hyperphosphatemia prevents acute heart failure in Klotho mutant (KL(−/−)) mice. Methods and results A significant elevation of serum phosphorus levels and a large reduction of heart function were found in KL(−/−) mice by six weeks of age. Normalization of serum phosphorus levels by low phosphate diet (LPD) rescued Klotho deficiency-induced heart failure and extended lifespan in male mice. Klotho deficiency impaired cardiac mitochondrial respiratory enzyme function and increased superoxide production, oxidative stress, and cardiac cell apoptosis in male KL(−/−) mice which can be eliminated by LPD. LPD, however, did not rescue hyperphosphatemia or heart failure in female KL(−/−) mice. LPD did not affect estrogen depletion in female KL(−/−) mice. Normalization of serum estrogen levels by treatment with 17β-estradiol prevented hyperphosphatemia and heart failure in female KL(−/−) mice. Mechanistically, treatment with 17β-estradiol rescued hyperphosphatemia via inhibiting renal Na-Pi co-transporter expression. Normalization of serum phosphorus levels by treatment with 17β-estradiol also abolished cardiac mitochondrial respiratory enzyme dysfunction, ROS overproduction, oxidative stress and cardiac cell apoptosis in female KL(−/−) mice. Conclusion Klotho deficiency causes acute heart failure via hyperphosphatemia in male mice which can be prevented by LPD. 17β-estradiol prevents Klotho deficiency-induced hyperphosphatemia and heart failure by eliminating upregulation of renal Na-Pi co-transporter expression in female mice.
Collapse
Affiliation(s)
- Kai Chen
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Zhongjie Sun
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
| |
Collapse
|
23
|
Xue M, Yang F, Le Y, Yang Y, Wang B, Jia Y, Zheng Z, Xue Y. Klotho protects against diabetic kidney disease via AMPK- and ERK-mediated autophagy. Acta Diabetol 2021; 58:1413-1423. [PMID: 34046744 DOI: 10.1007/s00592-021-01736-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 05/06/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND Diabetic kidney disease (DKD) is a serious complication of diabetes mellitus and results in serious public health problems. Although a great number of studies have been performed to elucidate the mechanisms of this disease, these mechanisms remain largely unknown. METHODS Cell and animal models were first constructed using human renal proximal tubule cells stimulated by high glucose (HG) and mice induced by streptozotocin (STZ). After Klotho overexpression, Klotho expression was assessed by RT-PCR and western blot, immunofluorescence; autophagy and AMPK/ERK proteins were confirmed using western blot or immunohistochemical assay; the autophagosomes were observed by transmission electron microscope; the pathological structure, fibrosis, polysaccharides and glycogen of kidney were evaluated by H&E staining, Masson staining and PAS staining. RESULTS We first confirmed that Klotho expression and autophagic activity were reduced in DM mice and HG-induced human renal proximal tubule cells. Besides, overexpression of Klotho could significantly enhance autophagy and AMPK and ERK1/2 activities in vivo and in vitro, which also could be abolished by selective AMPK inhibitor and ERK activator. Moreover, we proved that Klotho could inhibit hyperglycemia-induced renal tubular damage. CONCLUSION In summary, our results proved that Klotho improved renal tubular cell autophagy via the AMPK and ERK pathways and played a role in renal protection. These findings provide new insight into the mechanism of Klotho and autophagy in DKD.
Collapse
Affiliation(s)
- Meng Xue
- Department of Endocrinology and Metabolism, The Second Clinical Medical College, Shenzhen People's HospitalJinan UniversityThe First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China
| | - Feng Yang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ying Le
- Department of Endocrinology and Metabolism, The Second Clinical Medical College, Shenzhen People's HospitalJinan UniversityThe First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China
| | - Yanlin Yang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Bingsen Wang
- Department of Endocrinology and Metabolism, The Second Clinical Medical College, Shenzhen People's HospitalJinan UniversityThe First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China
| | - Yijie Jia
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zongji Zheng
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yaoming Xue
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
24
|
Tyurenkov IN, Perfilova VN, Nesterova AA, Glinka Y. Klotho Protein and Cardio-Vascular System. BIOCHEMISTRY (MOSCOW) 2021; 86:132-145. [PMID: 33832412 DOI: 10.1134/s0006297921020024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Klotho protein affects a number of metabolic pathways essential for pathogenesis of cardio-vascular diseases and their prevention. It inhibits lipid peroxidation and inflammation, as well as prevents endothelial injury and calcification of blood vessels. Klotho decreases rigidity of blood vessels and suppresses development of the heart fibrosis. Low level of its expression is associated with a number of diseases. Cardioprotective effect of klotho is based on its ability to interact with multiple receptors and ion channels. Being a pleiotropic protein, klotho could be a useful target for therapeutic intervention in the treatment of cardio-vascular diseases. In this review we present data on pharmaceuticals that stimulate klotho expression and suggest some promising research directions.
Collapse
Affiliation(s)
- Ivan N Tyurenkov
- Volgograd State Medical University, Ministry of Health of the Russian Federation, Volgograd, 400066, Russia
| | - Valentina N Perfilova
- Volgograd State Medical University, Ministry of Health of the Russian Federation, Volgograd, 400066, Russia.
| | - Alla A Nesterova
- Pyatigorsk Medical and Pharmaceutical Institute, Branch of the Volgograd State Medical University, Ministry of Health of the Russian Federation, Pyatigorsk, 357500, Russia
| | - Yelena Glinka
- Keenan Research Centre, St. Michael's Hospital, Toronto, ON M5B 1W8, Canada
| |
Collapse
|
25
|
Wang Q, Wang S, Sun Z. Kidney-Specific Klotho Gene Deletion Causes Aortic Aneurysm via Hyperphosphatemia. Hypertension 2021; 78:308-319. [PMID: 34176284 DOI: 10.1161/hypertensionaha.121.17299] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Qiongxin Wang
- Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center (Q.W., S.W., Z.S.)
| | - Shirley Wang
- Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center (Q.W., S.W., Z.S.).,Department of Physiology, College of Medicine, The University of Tennessee Health Science Center, Memphis (S.W., Z.S.)
| | - Zhongjie Sun
- Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center (Q.W., S.W., Z.S.).,Department of Physiology, College of Medicine, The University of Tennessee Health Science Center, Memphis (S.W., Z.S.)
| |
Collapse
|
26
|
Chen K, Zhang B, Sun Z. MicroRNA 379 Regulates Klotho Deficiency-Induced Cardiomyocyte Apoptosis Via Repression of Smurf1. Hypertension 2021; 78:342-352. [PMID: 34120450 DOI: 10.1161/hypertensionaha.120.16888] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Kai Chen
- From the Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis (K.C., B.Z., Z.S.).,Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center (K.C., Z.S.)
| | - Bo Zhang
- From the Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis (K.C., B.Z., Z.S.)
| | - Zhongjie Sun
- From the Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis (K.C., B.Z., Z.S.).,Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center (K.C., Z.S.)
| |
Collapse
|
27
|
Tang J, Zhang JF, Yang RQ, Chen YL, Ni B. A conserved klo-1-mpk-1 pathway regulates autophagy and modulates longevity in Caenorhabditis elegans. Biochem Biophys Res Commun 2021; 562:36-42. [PMID: 34034091 DOI: 10.1016/j.bbrc.2021.05.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 10/21/2022]
Abstract
There are six different longevity models in Caenorhabditis elegans. Previous studies have identified several convergence points, such as hlh-30, daf-16, and klf-3, required for lifespan extension in these longevity models. However, it is not clear whether there other such convergence points. In this study, based on analysis of transcriptome data, we found that the expression of klo-1/klotho was elevated in several longevity models. klo-1 was required for lifespan extension in the glp-1(e2141) and isp-1(qm150) mutants. klo-1 extended the lifespan of glp-1(e2141) and isp-1(qm150) worms by activating extracellular-signal-regulated kinase (ERK). In addition, klo-1 and mpk-1 (the homologous gene encoding ERK) regulated autophagy in glp-1(e2141) mutants, suggesting that klo-1 regulates lifespan by activating autophagy.
Collapse
Affiliation(s)
- Jie Tang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, Yunnan, 650091, China
| | - Jian-Fan Zhang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, Yunnan, 650091, China
| | - Rui-Qiu Yang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming, Yunnan, 650091, China
| | - Yuan-Li Chen
- Faculty of Basic Medicine, Kunming Medical University, Kunming, 650500, China
| | - Baosen Ni
- Institute of Biology and Environmental Engineering, School of Chemistry, Biology & Environment, Yuxi Normal University, Yuxi, 653100, China.
| |
Collapse
|
28
|
Hendrickx JO, Martinet W, Van Dam D, De Meyer GRY. Inflammation, Nitro-Oxidative Stress, Impaired Autophagy, and Insulin Resistance as a Mechanistic Convergence Between Arterial Stiffness and Alzheimer's Disease. Front Mol Biosci 2021; 8:651215. [PMID: 33855048 PMCID: PMC8039307 DOI: 10.3389/fmolb.2021.651215] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/03/2021] [Indexed: 12/12/2022] Open
Abstract
The average age of the world's elderly population is steadily increasing. This unprecedented rise in the aged world population will increase the prevalence of age-related disorders such as cardiovascular disease (CVD) and neurodegeneration. In recent years, there has been an increased interest in the potential interplay between CVDs and neurodegenerative syndromes, as several vascular risk factors have been associated with Alzheimer's disease (AD). Along these lines, arterial stiffness is an independent risk factor for both CVD and AD. In this review, we discuss several inflammaging-related disease mechanisms including acute tissue-specific inflammation, nitro-oxidative stress, impaired autophagy, and insulin resistance which may contribute to the proposed synergism between arterial stiffness and AD.
Collapse
Affiliation(s)
- Jhana O. Hendrickx
- Laboratory of Physiopharmacology, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Wim Martinet
- Laboratory of Physiopharmacology, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Debby Van Dam
- Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen, Groningen, Netherlands
| | - Guido R. Y. De Meyer
- Laboratory of Physiopharmacology, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
29
|
Kanbay M, Demiray A, Afsar B, Covic A, Tapoi L, Ureche C, Ortiz A. Role of Klotho in the Development of Essential Hypertension. Hypertension 2021; 77:740-750. [PMID: 33423524 DOI: 10.1161/hypertensionaha.120.16635] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Klotho has antiaging properties, and serum levels decrease with physiological aging and aging-related diseases, such as hypertension, cardiovascular, and chronic kidney disease. Klotho deficiency in mice results in accelerated aging and cardiovascular injury, whereas Klotho supplementation slows down the progression of aging-related diseases. The pleiotropic functions of Klotho include, but are not limited to, inhibition of insulin/IGF-1 (insulin-like growth factor 1) and WNT (wingless-related integration site) signaling pathways, suppression of oxidative stress and aldosterone secretion, regulation of calcium-phosphate homeostasis, and modulation of autophagy with inhibition of apoptosis, fibrosis, and cell senescence. Accumulating evidence shows an interconnection between Klotho deficiency and hypertension, and Klotho gene polymorphisms are associated with hypertension in humans. In this review, we critically review the current understanding of the role of Klotho in the development of essential hypertension and the most important underlying pathways involved, such as the FGF23 (fibroblast growth factor 23)/Klotho axis, aldosterone, Wnt5a/RhoA, and SIRT1 (Sirtuin1). Based on this critical review, we suggest avenues for further research.
Collapse
Affiliation(s)
- Mehmet Kanbay
- From the Division of Nephrology, Department of Medicine (M.K.), Koc University School of Medicine, Istanbul, Turkey
| | - Atalay Demiray
- Department of Medicine (A.D.), Koc University School of Medicine, Istanbul, Turkey
| | - Baris Afsar
- Division of Nephrology, Department of Internal Medicine, Suleyman Demirel University School of Medicine, Isparta Turkey (B.A.)
| | - Adrian Covic
- Department of Nephrology, Grigore T. Popa University of Medicine, Iasi, Romania (A.C., L.T., C.U.)
| | - Laura Tapoi
- Department of Nephrology, Grigore T. Popa University of Medicine, Iasi, Romania (A.C., L.T., C.U.)
| | - Carina Ureche
- Department of Nephrology, Grigore T. Popa University of Medicine, Iasi, Romania (A.C., L.T., C.U.)
| | - Alberto Ortiz
- Cardiovascular Diseases Institute, Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania (A.O.)
- IIS-Fundacion Jimenez Diaz, Department of Medicine, School of Medicine, Universidad Autonoma de Madrid, Spain (A.O.)
| |
Collapse
|
30
|
Russell DL, Oates JC, Markiewicz M. Association Between the Anti-Aging Gene Klotho and Selected Rheumatologic Autoimmune Diseases. Am J Med Sci 2021; 361:169-175. [PMID: 33349438 PMCID: PMC9741923 DOI: 10.1016/j.amjms.2020.10.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 08/25/2020] [Accepted: 10/22/2020] [Indexed: 12/14/2022]
Abstract
Klotho long recognized for its role in anti-aging, is potentially implicated in the pathogenesis of rheumatoid arthritis, systemic lupus erythematosus, and systemic sclerosis. Aging of the immune system coincides with the inability of the body to recognize self-antigens, which often leads to autoimmune responses. The role of Klotho in these autoimmune diseases should be of high interest; however, few articles have been published exploring the role of Klotho in the pathogenesis, organ involvement, or clinical manifestation of rheumatoid arthritis, systemic lupus erythematosus, and systemic sclerosis. Herein, we discuss information gathered from peer-reviewed publications to describe the emerging role of Kl in these select rheumatologic autoimmune diseases.
Collapse
Affiliation(s)
| | - Jim C Oates
- Ralph H. Johnson VA Medical Center, Charleston, South Carolina;,Division of Rheumatology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Margaret Markiewicz
- Division of Rheumatology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
31
|
Liang WY, Wang LH, Wei JH, Li QL, Li QY, Liang Q, Hu NQ, Li LH. No significant association of serum klotho concentration with blood pressure and pulse wave velocity in a Chinese population. Sci Rep 2021; 11:2374. [PMID: 33504927 PMCID: PMC7840754 DOI: 10.1038/s41598-021-82258-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 01/19/2021] [Indexed: 01/14/2023] Open
Abstract
Klotho, an important anti-aging protein, may be related to elevated blood pressure (BP) and arterial stiffness. We aimed to investigate associations between the serum klotho concentration and peripheral/central BP and arterial stiffness based on the carotid–femoral pulse wave velocity (cfPWV) in a Chinese population. We invited all inhabitants aged ≥ 18 years in two Dali communities for participation. The SphygmoCor system was used to record radial arterial waveforms. Aortic waveforms were derived using a generalized transfer function. The central BP was assessed by calibrating the brachial BP, which was measured using an oscillometric device. The serum klotho concentration was measured using an enzyme-linked immunosorbent assay and logarithmically transformed. Of the 716 participants (mean age: 51.9 ± 12.6 years), 467 (65.2%) were women. The median serum klotho concentration was 381.8 pg/mL. The serum klotho concentration did not significantly differ between patients with and without hypertension (P > 0.05) and between those with and without arterial stiffness (cfPWV ≥ 10 m/s) (P > 0.05). After adjusting for confounders, the serum klotho concentration was not significantly associated with the peripheral or central BP (P > 0.05) and cfPWV (P > 0.05). Our data indicated that the serum klotho concentration was not associated with BP or cfPWV in the general Chinese population.
Collapse
Affiliation(s)
- Wan-Ying Liang
- Department of Gerontology, The First Affiliated Hospital of Dali University, Jiashibo Road 32, Dali, 671000, Yunnan Province, China
| | - Li-Hong Wang
- Department of Gerontology, The First Affiliated Hospital of Dali University, Jiashibo Road 32, Dali, 671000, Yunnan Province, China
| | - Jian-Hang Wei
- Department of Gerontology, The First Affiliated Hospital of Dali University, Jiashibo Road 32, Dali, 671000, Yunnan Province, China
| | - Qing-Lu Li
- Department of Gerontology, The First Affiliated Hospital of Dali University, Jiashibo Road 32, Dali, 671000, Yunnan Province, China
| | - Qi-Yan Li
- Department of Gerontology, The First Affiliated Hospital of Dali University, Jiashibo Road 32, Dali, 671000, Yunnan Province, China
| | - Quan Liang
- Department of Gerontology, The First Affiliated Hospital of Dali University, Jiashibo Road 32, Dali, 671000, Yunnan Province, China
| | - Nai-Qing Hu
- Department of Gerontology, The First Affiliated Hospital of Dali University, Jiashibo Road 32, Dali, 671000, Yunnan Province, China
| | - Li-Hua Li
- Department of Gerontology, The First Affiliated Hospital of Dali University, Jiashibo Road 32, Dali, 671000, Yunnan Province, China.
| |
Collapse
|
32
|
Hu J, Chu C, Shi T, Yan Y, Mu J. Effects of salt intervention on serum levels of Klotho influenced by salt sensitivity. J Clin Hypertens (Greenwich) 2020; 22:2051-2058. [PMID: 33164306 DOI: 10.1111/jch.14044] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/20/2020] [Accepted: 08/22/2020] [Indexed: 12/21/2022]
Affiliation(s)
- Jia‐Wen Hu
- Department of Cardiovascular Surgery First Affiliated Hospital of Medical School Xi’an Jiaotong University Xi’an China
| | - Chao Chu
- Department of Cardiology First Affiliated Hospital of Medical School Xi’an Jiaotong University Xi’an China
- Key Laboratory of Molecular Cardiology of Shaanxi Province Xi’an China
| | - Tao Shi
- Department of Cardiovascular Surgery First Affiliated Hospital of Medical School Xi’an Jiaotong University Xi’an China
| | - Yang Yan
- Department of Cardiovascular Surgery First Affiliated Hospital of Medical School Xi’an Jiaotong University Xi’an China
| | - Jian‐Jun Mu
- Department of Cardiology First Affiliated Hospital of Medical School Xi’an Jiaotong University Xi’an China
- Key Laboratory of Molecular Cardiology of Shaanxi Province Xi’an China
| |
Collapse
|
33
|
Gao D, Wang S, Lin Y, Sun Z. In vivo AAV delivery of glutathione reductase gene attenuates anti-aging gene klotho deficiency-induced kidney damage. Redox Biol 2020; 37:101692. [PMID: 32863229 PMCID: PMC7476318 DOI: 10.1016/j.redox.2020.101692] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/27/2020] [Accepted: 08/17/2020] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Klotho is an aging-suppressor gene which leads to accelerated aging when disrupted. This study was designed to investigate whether glutathione reductase (GR), a critical intracellular antioxidant enzyme, is involved in the pathogenesis of kidney damages associated with accelerated aging in Klotho-haplodeficient (KL+/-) mice. METHODS AND RESULTS Klotho-haplodeficient (KL+/-) mice and WT mice were used. We found that Klotho haplodeficiency impaired kidney function as evidenced by significant increases in plasma urea and creatinine and a decrease in urinary creatinine in KL+/- mice. The expression and activity of GR was decreased significantly in renal tubular epithelial cells of KL+/- mice, suggesting that Klotho deficiency downregulated GR. We constructed adeno-associated virus 2 (AAV2) carrying GR full-length cDNA (AAV-GR). Interestingly, in vivo AAV-GR delivery significantly improved Klotho deficiency-induced renal functional impairment and structural remodeling. Furthermore, in vivo expression of GR rescued the downregulation of the reduced glutathione/oxidized glutathione (GSH/GSSG) ratio, which subsequently diminished oxidative damages in kidneys, as evidenced by significant decreases in renal 4-HNE expression and urinary 8-isoprostane levels in KL mice. CONCLUSION This study provides the first evidence that Klotho deficiency-induced kidney damage may be partly attributed to downregulation of GR expression. In vivo delivery of AAV-GR may be a promising therapeutic approach for aging-related kidney damage.
Collapse
Affiliation(s)
- Diansa Gao
- Department of Cardiology, The First Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Shirley Wang
- Department of Physiology, College of Medicine, University of Tennessee Health Sciences Center, Memphis, TN, 38163, USA
| | - Yi Lin
- Department of Physiology, College of Medicine, University of Tennessee Health Sciences Center, Memphis, TN, 38163, USA
| | - Zhongjie Sun
- Department of Physiology, College of Medicine, University of Tennessee Health Sciences Center, Memphis, TN, 38163, USA.
| |
Collapse
|
34
|
Feng R, Ullah M, Chen K, Ali Q, Lin Y, Sun Z. Stem cell-derived extracellular vesicles mitigate ageing-associated arterial stiffness and hypertension. J Extracell Vesicles 2020; 9:1783869. [PMID: 32939234 PMCID: PMC7480600 DOI: 10.1080/20013078.2020.1783869] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 02/01/2020] [Accepted: 04/24/2020] [Indexed: 12/19/2022] Open
Abstract
The prevalence of arterial stiffness and hypertension increases with age. This study investigates the effect of induced pluripotent mesenchymal stem cell-derived extracellular vesicles (EVs) on ageing-associated arterial stiffness and hypertension. EVs were collected and purified from induced pluripotent stem cell-derived mesenchymal stem cells (iPS-MSCs). Young and old male C57BL/6 mice were used. Mice in the EVs group were injected via tail vein once a week for four weeks (18 x 106 EVs/mouse/injection). Blood pressure (BP) was measured using the tail-cuff method and validated by direct cannulation. Pulse wave velocity (PWV) was measured using a Doppler workstation. PWV and BP were increased significantly in the old mice, indicating arterial stiffness and hypertension. Intravenous administration of EVs significantly attenuated ageing-related arterial stiffness and hypertension, while enhancing endothelium-dependent vascular relaxation and arterial compliance in the old EVs mice. Elastin degradation and collagen I deposition (fibrosis) were increased in aortas of the old mice, but EVs substantially improved ageing-associated structural remodelling. Mechanistically, EVs abolished downregulation of sirtuin type 1 (SIRT1), and endothelial nitric oxide synthase (eNOS) protein expression in aortas of the older mice. In cultured human aortic endothelial cells, EVs promoted the expression of SIRT1, AMP-activated protein kinase alpha (AMPKα), and eNOS. In conclusion, iPS-MSC-derived EVs attenuated ageing-associated vascular endothelial dysfunction, arterial stiffness, and hypertension, likely via activation of the SIRT1-AMPKα-eNOS pathway and inhibition of MMPs and elastase. Thus, EVs mitigate arterial ageing. This finding also sheds light into the therapeutic potential of EVs for ageing-related vascular diseases. ABBREVIATIONS EV: Extracellular vesicles; iPS: induced pluripotent stem cell; MSC: mesenchymal stem cell; AMPKα: AMP activated protein kinase α; eNOS: endothelial nitric oxide synthase; Sirt1: sirtuin 1; JNC7: Seventh Report of the Joint National Committee; CVD: cardiovascular disease; PWV: pulse wave velocity; BP: blood pressure; SNP: sodium nitroprusside.
Collapse
Affiliation(s)
- Rui Feng
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mujib Ullah
- Department of Physiology, College of Medicine, University of Tennessee Health Sciences Center, Memphis, TN, USA
| | - Kai Chen
- Department of Physiology, College of Medicine, University of Tennessee Health Sciences Center, Memphis, TN, USA
| | - Quaisar Ali
- Department of Physiology, College of Medicine, University of Tennessee Health Sciences Center, Memphis, TN, USA
| | - Yi Lin
- Department of Physiology, College of Medicine, University of Tennessee Health Sciences Center, Memphis, TN, USA
| | - Zhongjie Sun
- Department of Physiology, College of Medicine, University of Tennessee Health Sciences Center, Memphis, TN, USA
| |
Collapse
|